Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Nov 1.
Published in final edited form as: Birth Defects Res. 2020 Jul 15;112(19):1588–1634. doi: 10.1002/bdr2.1754

Genetics and signaling mechanisms of orofacial clefts

Kurt Reynolds 1,2,3,*, Shuwen Zhang 1,2, Bo Sun 1,2, Michael Garland 1,2, Yu Ji 1,2,3, Chengji J Zhou 1,2,3,*
PMCID: PMC7883771  NIHMSID: NIHMS1669913  PMID: 32666711

Abstract

Craniofacial development involves several complex tissue movements including several fusion processes to form the frontonasal and maxillary structures, including the upper lip and palate. Each of these movements are controlled by many different factors that are tightly regulated by several integral morphogenetic signaling pathways. Subject to both genetic and environmental influences, interruption at nearly any stage can disrupt lip, nasal, or palate fusion and result in a cleft. Here, we discuss many of the genetic risk factors that may contribute to the presentation of orofacial clefts in patients, and several of the key signaling pathways and underlying cellular mechanisms that control lip and palate formation, as identified primarily through investigating equivalent processes in animal models are examined.

Keywords: Cleft lip/palate, Syndromic/non-syndromic, Human genetics, Mouse models, Bmp/Tgfb signaling, Fgf signaling, Retinoic acid signaling, Shh signaling, Wnt signaling, Signaling crosstalk

1. Introduction

Orofacial clefts (OFCs) are among the most common birth defects worldwide and contribute a heavy burden on patients and their families as well as healthcare systems. Clefts can be observed in the context of other syndromic characteristics, but frequently occur as an isolated, nonsyndromic presentation. During embryonic development, several midfacial primordia fuse together to form orofacial structures, including the nose, upper lip, and the palate separating the oral and nasal cavities. Complex regulatory mechanisms control these processes and are subject to both genetic and environmental influences. When they fail, defective closure of facial tissues may manifest as a structural cleft at birth. OFCs are typically classified as either cleft lip with or without cleft palate (CL/P) or cleft palate only (CPO). As the processes that govern lip and palate formation differ along with their respective causes and risk factors, it is becoming more common for studies to group CL/P patients with cleft lip only (CLO) separately from those with cleft lip with cleft palate (CLP) to more accurately investigate the causal relationships between potential influences and specific types of resulting OFCs (L. Huang et al., 2019). This review is a part of a series discussing the underlying mechanisms that contribute to OFC formation. The developmental processes and germ layers involved in orofacial formation and their role in the presentation of clefts are reviewed in greater detail within a companion article (Ji et al., 2020 in revision). Here, we will discuss some of the known genetic influences and risk factors in oral clefts, as well as the underlying signaling pathways that govern lip and palate formation and their role in OFCs.

2. Genetics of human orofacial clefts

Prior to the genomic era, early segregation studies recognized that while many OFCs appear sporadically, they often can be attributed to heritable alleles and occur in multiple cases within the same family. Studies commonly include large multiplex or consanguineous families or smaller case-parent trios to identify inherited causes, but they may compare patients with sporadic cases and unrelated controls. Attempts to identify genetic causes of OFCs began with association of markers such as variable serum antigens and restriction site polymorphisms which segregated with OFCs within familial genetic pedigrees. These were followed by genome-wide scans to identify regions of the genome that associated with clefts and targeted genotyping and studies to pinpoint the sequence variants that were linked with OFCs. Since the completion of the human genome project, reference genome and single nucleotide polymorphism (SNP) databases have been compiled and are continuing to be improved. The availability of these tools to investigators has allowed for more efficient identification of specific variants that may be linked with traits, and evidence for the association of many important candidate genes and loci in the etiology of OFCs, both syndromic and nonsyndromic, has been amassed. The history of genetics studies of OFCs has been previously reviewed (Mary L. Marazita, 2012). Modern sequencing technologies have accelerated our ability to identify specific sequences and variants that are linked with clefts, and at least 350 potential candidate genes have been identified through association studies in human OFC patients alone. Sufficient evidence has accumulated for many genes and loci involved in craniofacial development that may contribute to OFCs, such that we can begin to describe a network of key factors that have clear implications in governing lip and/or palate formation and fusion.

2.1. Genetics of nonsyndromic OFCs

OFCs are often observed in conjunction with other abnormal morphological characteristics as part of a syndrome, and syndromic defects frequently are caused by mutations and deletions that result in a loss of function of one or more genes. However, more commonly seen are nonsyndromic OFCs, which are often not attributable to a single easily identifiable mutation, but are likely controlled by many different risk factors and genetic variants that do not result in a loss of function and do not cause an observable phenotype on their own. As such, attempts to determine genetic causes of nonsyndromic clefts have proven in many cases to be difficult and results can be highly variable, even in studies assessing linkage of the same genomic variants with OFCs. This may be due to differences between genetic backgrounds, environmental influences across patient cohorts, or experimental techniques and analysis methods.

Genome-wide association studies (GWAS) provide an unbiased approach to identify genetic risk factors and several have been performed to determine genomic regions that segregate with clefts, most seeking linkage with nonsyndromic CL/P (NSCL/P), the most common type of OFC (Table 1). Many of the earlier genome scans provided suggestive evidence for cleft association with loci but were unable to establish genome-wide significance. However, a meta-analysis of 13 scans including data from several of these earlier studies demonstrated the strongest evidence at the time for NSCL/P association with several regions. Highly significant results were obtained at 9q21, so linkage with key candidate genes at this locus was assessed and positive associations reported with patched (PTCH1), receptor tyrosine kinase (RTK)-like orphan receptor 2 (ROR2), transforming growth factor-beta receptor 1 (TGFBR1), and forkhead box E1 (FOXE1) (Mary L. Marazita et al., 2004). A subsequent GWAS and fine-mapping in 2009 by the same group also showed evidence for NSCL/P association with FOXE1, which has since been strengthened by multiple other studies (Beaty et al., 2013; Elizabeth J. Leslie et al., 2017; M. L. Marazita et al., 2009; Moreno et al., 2009; Nikopensius et al., 2011), as well as several new loci including 1q32 in which interferon regulatory factor 6 (IRF6) lies, in important craniofacial regulator that had previously been implicated in targeted studies but not genome scans (Blanton et al., 2005; Ji Wan Park et al., 2007; Scapoli et al., 2005; Zucchero et al., 2004). Several other GWAS around this time also identified other important loci involved in NSCL/P, including strong association with the genomic region at 8q24.21, which lies in a gene desert (Beaty et al., 2010; Birnbaum et al., 2009; Grant et al., 2009). While the role this locus plays in NSCL/P is not entirely clear, variants may affect expression of transcription factor c-MYC (MYC), which maps to 8q24 and is involved in craniofacial development (Uslu et al., 2014).

Table 1.

Major GWAS to identify genomic regions associated with nonsyndromic OFC risk

Study Details Associated Loci or Candidate Genes if Identified Reference
GWAS for NSCL/P; UK sibling pairs Suggestive evidence for association at 1p36; 2p13; 2q37; 6p23; 6q25; 8q23–24; 11p12-q14; 12q13; 16p13; 16q24; Xcen-q21 (Prescott, Lees, Winter, & Malcolm, 2000)
GWAS for NSCL/P in Chinese families Positive results for potential NSCL/P linkage with markers on chromosomes 1, 2, 3, 4, 5, 6, 7, 9, 11, 12, 16, 20, and 21. (Mary L. Marazita et al., 2002)
NSCL/P GWAS Meta-Analysis using data from 13 prior scans 1p12–13; 1q32; 2q32–35; 3p25; 6p23; 6q23–25; 7p12; 8q21; 8q23; 9q21; 12p11; 14q21–24; 15q15; 17q21; 18q21; 20q13 (Mary L. Marazita et al., 2004)
GWAS, fine-mapping and candidate analysis in multi-ethnic NSCL/P cohort IRF6; 2p13; 3q27–28; FOXE1; 12p11; 14q21–24; 16q24 (M. L. Marazita et al., 2009)
GWAS for NSCL/P; German cohort 8q24.21 (Birnbaum et al., 2009)
GWAS for NSCL/P; US patients of European ancestry 8q24.21 (Grant et al., 2009)
European and Asian Ancestry groups 8q24; IRF6; MAFB; 1p22 (Reported as ABCA4; Variants may affect ARHGAP29) (Beaty et al., 2010)
GWAS for NSCL/P; Central European ancestry, enlarged dataset from (Birnbaum et al., 2009) VAX1; NOG (Mangold et al., 2010)
GWAS for NSCPO and gene-environment interaction; multi-ethnic group MLLT3, SMC2 × Maternal Alcohol; TBK1, ZNF236 × Maternal Smoking; BAALC × Maternal Multivitamin Supplementation (decreased risk) [No Loci Yielded Significant Association Alone] (Beaty et al., 2011)
GWAS meta-analysis; multi-ethnic ancestry ARHGAP29; PAX7; IRF6; THADA; EPHA3; 8q21.3; 8q24; VAX1; SPRY2; TPM1; NOG; MAFB (Kerstin U. Ludwig et al., 2012)
NSCL/P GWAS in Chinese population IRF6; VAX1; 16p13.3 (CREBBP/ADCY9); NTN1; MAFB (Sun et al., 2015)
NSCL/P GWAS meta-analysis GREM1 (Ludwig et al., 2016)
NSCPO GWAS; patients of European ancestry GRHL3 (Elizabeth J. Leslie et al., 2016)
NSCL/P GWAS in multi-ethnic group ARHGAP29, PAX7, IRF6, FAM49A, 8q24, NTN1, 17q23 (TANC2/DCAF7), RHPN2 (E. J. Leslie et al., 2016)
GWAS and meta-analysis, Han Chinese cohort ARHGAP29; IRF6; FAM49A; MYC; MMP16; GADD45G; VAX1; SPRY2; SPRY1; CREBBP; NTN1; NOG; MAFB; TAF1B; MSX1; FGF10; TFAP2A; FGFR1; RAD54B; PTCH1; KRT18; RPS26; TMEM19; LINC00640; GSC/DICER1; WNT9B (Y. Yu et al., 2017)
GWAS meta-analysis for NSCPO and NSCL/P; multi-ethnic groups NSCPO: GRHL3
NSCL/P: ARHGAP29; IRF6; PAX7; 2p24; TP63; 8q21; 8q24; VAX1; 13q31; ARID3B; NTN1; TANC2; MAFB
All clefts combined: IRF6; ARHGAP29; PAX7; 2p24; 8q21; 8q24; FOXE1; VAX1; 13q31; NTN1; MAFB
(Elizabeth J. Leslie et al., 2017)
NSCL/P GWAS and replication analysis in Polish population DLG1 (A. Mostowska et al., 2018)
GWAS and gene ontology analysis; 5 Chinese cohorts with CLO, CPO, and CLP groups NSCPO: IRF6; POMGNT2; NSD2/MSX1; DOCK9; PAX9; DLK1; FOXC2-FOXL1; MAU2.
NSCLO: IRF6; MYCN; VAX1; GRM5; ALX1; DLK1; MAFB
(L. Huang et al., 2019)
NSCL/P GWAS and replication in patients of northeastern European ancestry 8q24.21; 12q23.1 (ANO4) (van Rooij et al., 2019)

One 2010 GWAS found genome-wide association with markers near ventral anterior homeobox 1 (VAX1) and the bone morphogenetic protein (BMP) regulator noggin (NOG), each of which has been identified in subsequent scans (Mangold et al., 2010) (Table 1). The muscle segment homeobox (Msx) genes are regulated by Bmp signaling, and several targeted studies have implicated MSX1 variants in the etiology of NSCL/P prior to being linked in GWA studies (Jezewski et al., 2003; Lidral et al., 1998; J. Suazo, Santos, Carreno, Jara, & Blanco, 2004; Y. Suzuki et al., 2004). VAX1 is an important transcription factor with roles in craniofacial development, and targeted studies have further strengthened the evidence for association of VAX1 SNPs with NSCL/P as well (A. Butali et al., 2013; de Araujo et al., 2016; B.-H. Zhang, Shi, Lin, Shi, & Jia, 2018). Another 2010 study reported association with 4 regions including two novel loci, V-maf musculoaponeurotic fibrosarcoma oncogene homolog B (MAFB) and ATP-binding cassette, sub-family A, member 4 (ABCA4), which were confirmed in replication studies. MAFB has been identified in multiple genome scans since, while ABCA4 variants may contribute to NSCL/P by altering expression of ARHGAP29 at the same locus (Beaty et al., 2010; Beaty et al., 2013; Azeez Butali et al., 2014; Elizabeth J. Leslie et al., 2012). One of the earlier candidate genes identified that has been thoroughly examined is transforming growth factor-alpha (TGFA), but inconsistent results have been unable to provide strong evidence for its role in OFCs (Vieira, 2006).

A 2017 GWAS and meta-analysis in a Chinese population linked many previously known genes as well as several novel loci with NSCL/P. These authors also performed a network analysis to describe interactions between NSCL/P-associated genes, linking several signaling pathways and transcription factors that govern lip and palate fusion (Y. Yu et al., 2017). One novel gene linked in this study, transcription factor AP-2 alpha (TFAP2A), regulates craniofacial IRF6 and had previously been implicated in NSCL/P through targeted studies as a strong candidate based on its role in animal OFC models (de Araujo et al., 2016; Martinelli, Masiero, et al., 2011; M. Shi et al., 2009). Among signaling pathway genes identified are several fibroblast growth factor (FGF) pathway components with extensive network interaction, including FGF10, the receptor FGFR1, and antagonists Sprouty 1 and 2 (SPRY1/2), the first three of which were novel. Components of the Sonic hedgehog (SHH) and wingless-type MMTV integration site family (WNT) pathways were also identified, including PTCH1 and WNT9B, and the latter is a homolog of an important CL/P mouse model gene (Juriloff, Harris, McMahon, Carroll, & Lidral, 2006; Y. Yu et al., 2017). Many genes previously linked with OFCs were also identified/confirmed in the Yu et al. study, as well as another recently performed extensive GWAS (L. Huang et al., 2019).

Tumor protein p63 (TP63) encodes another transcription factor associated with multiple syndromes that can include OFCs (Table 2). TP63 is a canonical WNT signaling target, and has been recently implicated in NSCL/P in both a GWA meta-analysis and exome-sequencing study (Basha et al., 2018; Elizabeth J. Leslie et al., 2017). A recent study that identified the first OFC association with Pre-B cell leukemia homeobox 1 (PBX1) and PBX2 mutations also identified gene-gene interactions between SNPs in PBX1 and WNT9B, and between IRF6 and each of PBX1, PBX2, and TP63 in the etiology of NSCL/P (Maili et al., 2019). These findings are consistent with a prior study that linked Pbx, p63, and Irf6 with Wnt signaling in midfacial development in animal models (Ferretti et al., 2011).

Table 2.

Summary of syndromic OFCs and candidate genes

SYNDROME CLEFT GENE NOTES ON CLEFT IF AVAILABLE REFERENCES
3MC Syndrome COLEC11, MASP1 CL/P reported in 40–68% of patients (Gardner et al., 2017; Rooryck et al., 2011)
Andersen-Tawil Syndrome KCNJ2 CPO in 8% of patients (n=36) (Andelfinger et al., 2002; Tristani-Firouzi et al., 2002)
Apert Syndrome FGFR2 CPO frequency depends on mutation type; 17–58%, 43% overall (n=85) (Slaney et al., 1996; Wilkie et al., 1995)
Auriculocondylar Syndrome PLCB4 Often includes CPO (Rieder et al., 2012)
Bamforth-Lazarus Syndrome FOXE1 Thyroid dysgenesis that includes CP (Carre et al., 2014; Clifton-Bligh et al., 1998)
Baraitser-Winter Syndrome ACTB Very rare condition; ACTB G74S substitution in patient with CPO (Di Donato et al., 2014)
Bartocas Papas/Recessive Popliteal Pterygium Syndrome RIPK4 Severe phenotypes frequently include bilateral CLP (Karen W. Gripp, Ennis, & Napoli, 2013; Mitchell et al., 2012)
Blepharocheilodontic Syndrome CDH1, CTNND1 Rare condition characterized by CL/P, usually bilateral (Ghoumid et al., 2017)
Branchio-oculo-facial Syndrome TFAP2A Usually includes bilateral CL/P (Milunsky et al., 2008)
Burn-McKeown Syndrome/Oculo-oto-facial Dysplasia TXNL4A 8/14 patients with compound heterozygous or homozygous mutations display CL/P or CPO (Goos et al., 2017; Wieczorek et al., 2014)
Campomelic Dysplasia (PRS) SOX9 Skeletal disorder that includes CPO in ~66% of patients (Foster et al., 1994; Mansour, Hall, Pembrey, & Young, 1995; Wagner et al., 1994)
Carey-Fineman-Ziter Syndrome MYMK Myopathy that includes high-arched or cleft palate in most patients (Di Gioia et al., 2017)
Catel-Manzke Syndrome TGDS PRS with hyperphalangy (Ehmke et al., 2014)
CHARGE Syndrome CHD7 25% of patients exhibit CL/P (L. E. Vissers et al., 2004)
Chondroplasia with Joint Dislocations, GPAPP Type IMPAD1 Includes posterior cleft palate (L. E. L. M. Vissers et al., 2011)
Cleft Palate, Cardiac Defects, and Mental Retardation (CPCMR) MEIS2 CLP or CPO among defects caused by MEIS2 haploinsufficiency (Johansson et al., 2014)
Cornelia de Lange Syndrome NIPBL Craniofacial phenotypes can include CP (Krantz et al., 2004)
Craniofrontonasal Dysplasia EFNB1 Characterized by midline defects that may include CL/P (Twigg et al., 2004)
Crouzon Syndrome FGFR2 Craniosynostosis with high-arched palate, occasional CP (Reardon et al., 1994)
De la Chapelle Dysplasia/Atelosteogenesi s Type II SLC26A2 Rare skeletal dysplasia includes CP (Bonafé et al., 2008)
Desmosterolosis DHCR24 33–40% CPO reported (Andersson, Kratz, & Kelley, 2002; Rohanizadegan & Sacharow, 2018)
Diamond-Blackfan Anemia (DBA) RPL5, RPL19, RPL26 Associated with at least 8 ribosomal protein genes, CP observed in patients with RPL5, RPL19, and RPL26 mutations (Hanna T. Gazda et al., 2012; H. T. Gazda et al., 2008; Konno et al., 2010)
DiGeorge and 22q11.2 Deletion Velo-cardio-facial Syndromes 22q11.2, includes TBX1 55–100% of patients exhibit palatal anomalies, 7.3–11% exhibit overt CPO. (Yagi et al., 2003)
Ectrodactyly, Ectodermal Dysplasia, and Orofacial Clefts (EEC) Syndrome TP63 Characterized by the presence of CLO, CPO, or CLP (Barrow et al., 2002; Celli et al., 1999)
Ehlers-Danlos Syndrome FKBP14 May include mild palate defects such as bifid uvula, submucous CP, or cleft soft palate (Baumann et al., 2012)
Emanuel Syndrome Supernumary chromosomal derivative Malsegregation of 11;22 translocation; 54% CP reported (Carter, St Pierre, Zackai, Emanuel, & Boycott, 2009)
Femoral-facial Syndrome Unknown CL/P reported in 63% of patients (Luisin et al., 2017)
Fraser Syndrome FRAS1 CL/P reported in 7–13% of patients (Hoefele et al., 2013; van Haelst, Scambler, Fraser Syndrome Collaboration, & Hennekam, 2007)
Frontonasal Dysplasia ALX1, ALX3, ALX4 Midline facial defects can include nasal cleft, CL, and CP (Kayserili et al., 2009; Twigg et al., 2009; Uz et al., 2010)
Goltz-Gorlin Syndrome/Focal Dermal Hypoplasia PORCN Can include CL/P, ~15% (Bornholdt et al., 2009; X. Wang et al., 2007)
Gordon Syndrome/Distal Arthrogryposis Type 3 PIEZO2 Most cases include CPO (Alisch et al., 2017; McMillin et al., 2014)
Gorlin-Goltz Syndrome/Nevoid Basal Cell Carcinoma PTCH1 CL/P reported in ~8.5% of patients (Hahn et al., 1996; Lambrecht & Kreusch, 1997)
Hay-Wells/Ankyloblepharon-Ectodermal Dysplasia-Clefting Syndrome TP63 Typified by presence of OFCs (McGrath et al., 2001)
Hereditary Neuralgic Amyotrophy SEPT9 May include CP (Laccone et al., 2008)
Holoprosencephaly + CL/P SIX3, TGIF1, SHH, PTCH1, GLI2, ZIC2, DISP1 Prevalence and severity of OFC varies with associated gene and causal mutation (Aguilella et al., 2003; K. W. Gripp et al., 2000; Ribeiro et al., 2006; E. Roessler et al., 1996; Erich Roessler et al., 2003; Erich Roessler et al., 2009; Solomon et al., 2010; Wallis et al., 1999)
Hyperphosphatasia Mental Retardation Syndrome PGAP3, PIGL, PIGV Can include CP, frequency depends on associated gene (Abdel-Hamid et al., 2018; Altassan, Fox, Poulin, & Buhas, 2018; Horn, Krawitz, Mannhardt, Korenke, & Meinecke, 2011)
Kabuki Syndrome KMT2D (MLL2) CL/P is observed in ~40% of patients (Hannibal et al., 2011)
Kallmann Syndrome/Idiopathic Hypogonadotrophic Hypogonadism FGF8, FGFR1 Occasionally includes CPO (Dode et al., 2003; Falardeau et al., 2008; Pitteloud et al., 2006)
Klippel-Feil Syndrome MEOX1, GDF6 May include CP (Mohamed et al., 2013; May Tassabehji et al., 2008)
Larsen Syndrome FLNB Osteochondroplasia with ~15% CPO (Bicknell et al., 2007)
Loeys-Dietz Syndrome TGFBR1, TGFBR2 Commonly includes bifid uvula, occasional CPO (Loeys et al., 2005)
Marshall’s/Stickler Syndrome COL2A1, COL11A1, LOXL3 Variable phenotypic spectrum, can include CP (Alzahrani et al., 2015; L. Guo et al., 2017)
Meckel-Gruber Syndrome RPGRIP1L, TCTN2, TMEM67, TMEM216 Ciliopathy with many causal genes, can include CL/P (Delous et al., 2007; Shaheen et al., 2011; U. M. Smith et al., 2006; Valente et al., 2010)
Moebius Syndrome Likely combination of Genetic and Environmental Influences Affects cranial nerves VI/VII, can include CPO (Rizos, Negrón, & Serman, 1998)
Mohr-Majewski Syndrome/Oral-facial-digital (OFD) Syndrome Type IV TCTN3 Severe ciliopathy which typically includes CL/P (Thomas et al., 2012)
Mowat-Wilson Syndrome ZEB2 (ZFHX1B) Occasional CPO or mild palatal anomalies (Dastot-Le Moal et al., 2007; M. Wilson et al., 2003)
Muenke Syndrome FGFR3 Craniosynostosis that occasionally includes CPO or CLP (Anderson et al., 2013)
Multiple Epiphyseal Dysplasia SLC26A2 Out of 6 known causal genes, CPO only reported in cases with recessive SLC26A2 mutations (Briggs & Chapman, 2002; Mäkitie et al., 2015)
Multiple Pterygium Syndrome MYH3, CHRNG Occasional cleft palate (Chong et al., 2015; Morgan et al., 2006)
Myhre Syndrome SMAD4 Missense SMAD4 SNPs in patients with CL/P and CPO (Caputo et al., 2012)
Nager Syndrome SF3B4 CP is present in most patients (Czeschik et al., 2013)
Native American/Bailey-Bloch Myopathy STAC3 Patients exhibit CPO or high arched palate (Horstick et al., 2013; Stamm et al., 2008)
Oculo-dento-digital Dysplasia GJA1 Uncommon CPO or CLP observed in ~3% of reported cases (Amano et al., 2012; Paznekas et al., 2009)
Oculo-facio-cardio-dental Syndrome BCOR Palate defects from bifid uvula to overt cleft observed in 31% of patients (n=26) (Davoody, Chen, Nanda, Uribe, & Reichenberger, 2012; Hilton et al., 2009)
OFD Syndrome Type IX TBC1D32, SCLT1 Ciliopathy; commonly includes midline CLP (Adly, Alhashem, Ammari, & Alkuraya, 2014)
OFD Syndrome Type XIV C2CD3 Ciliopathy includes CPO (Thauvin-Robinet et al., 2014)
Opitz G/BBB Syndrome MID1, SPECC1L Often includes CL/P (Kruszka et al., 2015; Quaderi et al., 1997)
Osteopathia striata with cranial sclerosis AMER1 (WTX) 52% of patients with AMER1 mutation exhibited OFC (Jenkins et al., 2009)
Otopalatodigital Syndrome FLNA Both types 1 and 2 caused by FLNA muations and commonly include CPO (Robertson et al., 2003)
Papillon-Leage-Psaume/OFD Syndrome Type I OFD1 CPO or CL/P frequency > 80% (Ferrante et al., 2001)
Peters-Plus Syndrome B3GLCT CL/P is reported in nearly half of patients (Lesnik Oberstein et al., 2006)
Pfeiffer Syndrome FGFR1, FGFR2 Most patients have high arched palate, occasional CP (Stoler et al., 2009)
Pierre Robin Sequence (Isolated) SOX9, BMPR1B PRS is typified by cleft palate (Benko et al., 2009; Jakobsen et al., 2007; Y. Yang et al., 2017)
Popliteal Pterygium Syndrome IRF6 Phenotypic overlap with the allelic VWS, usually including CLP or CPO (Kondo et al., 2002)
Rapp-Hodgkin Syndrome TP63 Ectodermal Dysplasia that includes CLP (Kantaputra, Hamada, Kumchai, & McGrath, 2003; van Straten & Butow, 2013)
Renpenning Syndrome PQBP1 10% reported CPO (Stevenson et al., 2005)
Richieri-Costa-Pereira EIF4A3 78.5% reported CPO; Often includes mandibular cleft (F. P. Favaro et al., 2014; Francine Pinheiro Favaro et al., 2011)
Roberts/SC Phocomelia Syndrome ESCO2 CLP in >50% of patients (H. Vega et al., 2010; Hugo Vega et al., 2005)
Robinow Syndrome ROR2, WNT5A, DVL1, DVL3 Characteristic facial features, sometimes include CP (Person et al., 2010; van Bokhoven et al., 2000; J. White et al., 2015; J. J. White et al., 2016)
SATB2-Associated Syndrome/Glass Syndrome SATB2 59% of patients include CPO overall, varies by mutation type. (Zarate & Fish, 2017)
Saethre-Chotzen Syndrome TWIST1 Craniosynostosis can include CP (Howard et al., 1997)
Schilbach-Rott Syndrome 9q22.32–33 dup./PTCH1 PTCH1 overexpression may contribute; commonly includes CL/P (Prontera et al., 2019)
Simpson-Golabi-Behmel Syndrome GPC3 Cleft palate in 13% of patients (Tenorio et al., 2014)
Smith-Lemli-Opitz Syndrome DHCR7 Approximately half of patients have CP (Fitzky et al., 1998; Wassif et al., 1998; Waterham et al., 1998)
TARP Syndrome RBM10 Characterized by PRS (Johnston et al., 2010)
Tetra-Amelia Syndrome WNT3, RSPO2 Often includes bilateral CLP (Niemann et al., 2004; Szenker-Ravi et al., 2018)
Thurston/OFD Syndrome Type V DDX59 Includes CPO/bifid uvula (Shamseldin et al., 2013)
Treacher-Collins Syndrome TCOF1, POLR1C, POLR1D 93% of cases attributed to TCOF1; high-arched or cleft palate in 28% (Dauwerse et al., 2011; Group, 1996; Trainor, Dixon, & Dixon, 2009)
Van der Woude Syndrome IRF6, GRHL3 CL/P or CPO that includes usually lower lip pits and hypodontia (Kondo et al., 2002; Peyrard-Janvid et al., 2014)
Varadi-Papp/OFD Syndrome Type VI CPLANE1 (C5orf42), TMEM216 Very rare form of OFDS which can include CLP or CPO; Can be caused by CPLANE1 mutations, 2 patients with TMEM216 mutations were reported wifh OFD6 (Valente et al., 2010; Váradi, Szabó, & Papp, 1980)
Vici Syndrome EPG5 CP observed in 6% (n=50) of patients (Byrne et al., 2016)
Waardenburg Syndrome PAX3 High degree of genetic heterogeneity, Type I associated with PAX3, occasionally includes CL/P (Da-Silva, 1991; M. Tassabehji et al., 1992)
Walker-Warburg Syndrome/Muscular Dystrophy-Dystroglycanopathy POMT1 Type of muscular dystrophy with occasional CLP; Many subtypes and causal genes, POMT1, POMT2 mutations identified in CLP patients (Vajsar et al., 2008; van Reeuwijk et al., 2005)
Wolf-Hirschhorn Syndrome NSD2, WHSC2,
LETM1 (deletion)
CL/P commonly observed among associated craniofacial malformations (Rutherford & Lowery, 2016)
Wolff-Parkinson-White/20p12.3 Microdeletion Syndrome BMP2 PRS/CPO common characteristic; BMP2 likely key gene contributing to craniofacial phenotype (Lalani et al., 2009; Sahoo et al., 2011; Williams, Uhas, Bunke, Garber, & Martin, 2012)
X-Linked Mental Retardation + CL/P PQBP1, PHF8 Several dozen genes are implicated in XLMR; PQBP1, PHF8 mutations identified in cases that included CL/P (Kalscheuer et al., 2003; Laumonnier et al., 2005)

The extracellular matrix (ECM) plays many important roles in the tissue movements that occur during craniofacial morphogenesis, and as such its function is intimately linked with OFCs. The mechanisms by which ECM activity contributes to OFCs is discussed in greater detail within a companion review article (Ji et al., 2020 in revision), though several genes involved in ECM regulation have been linked to NSCL/P, including the laminin-related netrin 1 (NTN1), implicated by both genome-wide and targeted studies (Beaty et al., 2010; Beaty et al., 2013; Q. Guo et al., 2017; S. Jiang et al., 2019; E. J. Leslie et al., 2016; Y. Yu et al., 2017). Other ECM regulatory molecule genes for which possible association with nonsyndromic OFCs have been found include those encoding the ECM remodeling proteins matrix metalloproteinase 3 and 16 (MMP3/16) (Kumari, Singh, & Raman, 2018; Y. Yu et al., 2017), tissue inhibitor of metalloproteinase 2 (TIMP2) (Nikopensius et al., 2011), and ADAM Metallopeptidase with thrombospondin type 1 motif 20 (ADAMTS20) (Wolf et al., 2015). The collagen genes COL4A3 and COL4A4 showed linkage disequilibrium with NSCL/P in case-parent trios in 3 populations (Beaty et al., 2006), while COL2A1 and COL11A2 were linked with NSCPO in a case-control genotyping study (Nikopensius et al., 2010). Fgf signaling interacts with the ECM and plays key roles in lip and palate fusion, will be discussed in greater detail below. Notch signaling does so as well, and several studies have linked polymorphisms in the Notch ligand gene jagged 2 (JAG2) with nonsyndromic clefts (de Araujo et al., 2016; Jagomägi et al., 2010; Vieira et al., 2005).

Environmental exposures are strongly linked with OFCs, which will be discussed extensively in another companion review (Garland et al., 2020 in revision). As a result, mutations in many of the genes that code for enzymes that metabolize or detoxify chemicals associated with such exposure can also contribute to the presentation of clefts. Retinoic acid/vitamin A metabolism and signaling are especially integral to craniofacial development and deviations in cellular metabolite levels can result in birth defects including OFCs. As such, several genes associated with retinoid activity have been linked with nonsyndromic clefts, including retinoic acid receptor alpha (RARA) (Fan, Li, & Wu, 2007; Peanchitlertkajorn, Cooper, Liu, Field, & Marazita, 2003; Xavier et al., 2013) and possibly ABCA4, which acts as a flippase-type transporter for retinoids (Beaty et al., 2010; Beaty et al., 2013; Fontoura, Silva, Granjeiro, & Letra, 2012; Q. Yuan, Blanton, & Hecht, 2011).

Folates are another key vitamin required for production of the amino acid methionine and many other important cellular compounds, and insufficient dietary intake can result in OFCs. One of the most well-studied genes in NSCL/P encodes the folate metabolism enzyme methylenetetrahydrofolate reductase (MTHFR), and in particular it is maternal rather than fetal missense mutations that have been shown to contribute to OFC risk (Martinelli et al., 2001; Pan et al., 2015; Pezzetti et al., 2004). Genes encoding several other enzymes involved in the folate/homocysteine metabolic pathway have been associated with NSCL/P with a similar importance of maternal genotype, further demonstrating the importance of maternal folate metabolism in modulating fetal lip and palate fusion. These include methionine synthase (MTR), methionine synthase reductase (MTRR), methylenetetrahydrofolate dehydrogenase (MTHFD1), reduced folate carrier 1 (SLC19A1), betaine-homocysteine methyltransferase (BHMT), and a gene-gene interaction between BHMT and dimethylglycine dehydrogenase (DMGDH), which uses folate as a cofactor, was determined to significantly contribute to risk of NSCL/P in an international cohort (Bufalino et al., 2010; Mills et al., 2008; A. Mostowska, Hozyasz, & Jagodzinski, 2006; Soghani et al., 2017; P. Wang et al., 2018; W. Wang, Jiao, Wang, Sun, & Dong, 2016).

Lipid metabolism plays important roles during craniofacial morphogenesis, and is regulated by Tgf-β signaling during secondary palatogenesis in animal models (J. Iwata et al., 2014). Several genes involved in lipid metabolism have been implicated in nonsyndromic cleft studies, including the fatty acid hydrolase androgen-dependent TFPI-regulating protein (ADTRP) (J. W. Park et al., 2006), apolipoprotein C2 (APOC2) (Mary L. Marazita et al., 2002), and stearoyl-CoA desaturase 5 (SCD5) (Beiraghi et al., 2003), and a gene-gene interaction between variants in the paraoxonases 1 and 2 (PON1/2), which protect against damage due to low-density lipoprotein oxidation, was determined to contribute to NSCL/P risk (Machado et al., 2019). Variants in genes that encode enzymes involved in metabolizing drug and environmental toxins may contribute to formation of OFCs, including aryl hydrocarbon receptor nuclear transporter (ARNT) (Kayano et al., 2004), alcohol dehydrogenase 1C (ADH1C) (Jugessur et al., 2009) and sulfotransferase 2A1 (SULT2A1) (A. Butali et al., 2019). Further, studies investigating the role toxin metabolism plays in OFC development have demonstrated gene-environment interactions contribute to NSCL/P risk, including between maternal smoking and fetal variants in glutathione S-transferase T1 (GSTT1), arylamine N-acetyltransferase 1 (NAT1), and NAT2 (Lammer, Shaw, Iovannisci, Van Waes, & Finnell, 2004; M. Shi et al., 2007), and between maternal occupational chemical exposures and fetal NAT2 and GSTM1 variants (Shaw, Nelson, Iovannisci, Finnell, & Lammer, 2003).

2.2. Genetics of syndromic OFCs

While syndromic OFCs are more often attributable to one congenital cause or disrupted gene than NSOFCs, other challenges arise in determining the underlying mechanisms. Many of the syndromic diseases that can include OFCs are heterogenous in both genotype and phenotype, and often the presentation of a cleft in conjunction with other defining characteristics can be variable in penetrance. For many syndromic conditions in which clefts are mild or uncommon, the underlying etiologies associated with an OFC phenotype may be more difficult to determine. Often multiple disease-causing genes and factors can exist, and the role of specific genes in cleft-associated cases may not be described as thoroughly, especially if clefts are a minor feature and not the primary focus of studies into a particular condition. Additionally, many syndromes are caused by deletions that disrupt several genes, further complicating the connection between specific loci and lip or palate fusion. However, many instances in which a case study of a smaller group or individual patient is available with the mutation data, it is more often possible to attribute to the OFC to the role of a specific disrupted gene or locus.

Van der Woude syndrome (VWS) accounts for the most common form of syndromic clefts, which are often indistinguishable from NSCL/P but often includes lip pits. The majority of cases are caused by mutations in IRF6, which are also linked with dominant Popliteal pterygium syndrome (PPS), a condition that affects the skin and genitals and also frequently includes CL/P (Kondo et al., 2002). Mutations in Grainyhead-like 3 (GRHL3), an ectodermal IRF6 target (Kousa et al., 2019), were identified as the cause of many of the VWS cases in which no causative variant in IRF6 could be identified (Peyrard-Janvid et al., 2014). Cytosolic IRF6 interacts with regulators of cytoskeletal remodeling and cell adhesion, nonmetastatic expressed 1 (NME1) and NME2. IRF6 VWS mutations interrupt this interaction, and recently a missense mutation in NME1 was found in a patient with VWS, while a missense NME2 variant was identified in a patient with NSCL/P (Parada-Sanchez et al., 2017).

The Pierre-Robin Sequence (PRS) refers to a set of characteristic craniofacial phenotypes that are commonly observed together: glossoptosis, cleft palate, micrognathia, and upper airway obstruction. Multiple models for an underlying mechanism exist, and palate defects appear be secondary effects, resulting from altered tongue and mandible positioning, rather than intrinsic defects within the palatal shelves (PS) themselves. The dominant model proposes that mandibular hypoplasia causes a high, retrotransposed tongue to block PS elevation and the upper airway. Alternatively, intrauterine mandible compression may restrict its growth and alter tongue development, similarly blocking the palate and airway. A third model suggests that delayed neuromuscular development reduces the tongue’s ability to stimulate mandible and palate growth, causing the observed phenotypes (Giudice et al., 2018). This sequence is often observed as part of a broader syndrome, but not always. Isolated PRS has been associated with mutations in or near the SRY-related HMG box 9 (SOX9) gene (Benko et al., 2009; Jakobsen et al., 2007). SOX9 mutations also cause Campomelic Dysplasia, which affects skeletal and genital development and can include PRS (Foster et al., 1994; Wagner et al., 1994). More recently, mutations in BMPR1B were also reported to be the cause of PRS in two unrelated families (Y. Yang et al., 2017). Several genes that code for ECM components and ECM-interacting proteins have also been associated with syndromes that include clefts. Stickler syndrome and the similar Marshall syndrome frequently include PRS and can be caused by mutations in several collagen genes, including COL2A1, COL11A1, COL11A2, COL9A1, COL9A2, and the collagen crosslinking enzyme gene lysyl oxidase-like 3 (LOXL3). COL2A1,COL11A1, and LOXL3 mutations have so far been identified in patients with cleft-associated forms (Alzahrani, Al Hazzaa, Tayeb, & Alkuraya, 2015; L. Guo et al., 2017). Filamins are involved in cell-ECM adhesion, and FLNA mutations cause Oto-palato-digital Syndrome, which is typified by cleft palate, while the glypican gene GPC3 is associated with Simpson-Golabi-Behmel Syndrome with CL/P (Robertson et al., 2003; Tenorio et al., 2014).

Fgf signaling is integral to lip and palate development and will be discussed in further detail later, and several syndromic conditions associated with OFCs can result from mutations in Fgf pathway components. Craniosynostosis is caused by the premature fusion of cranial bones and syndromic craniosynostoses are commonly accompanied by oral clefts. These syndromes are often associated with altered Fgf signaling, especially with FGFR2 variants. Both of Apert and Crouzon syndromes are typified by craniosynostosis and can include cleft palate, and both syndromes are linked with mutations in FGFR2 (Reardon et al., 1994; Slaney et al., 1996; Wilkie et al., 1995). Apert syndrome is a condition marked by craniosynostosis and syndactyly that often includes CP. Clefting is most prevalent (~59%) in Apert patients with a S252W substitution near the linker region between extracellular immunoglobulin domains II and III, which eliminates ligand specificity and causes ectopic activation (Slaney et al., 1996; K. Yu, Herr, Waksman, & Ornitz, 2000). Another type of FGFR2 gain-of-function mutation causes Crouzon syndrome, which also presents with frequent CP, and mice carrying the Crouzon mutation similarly exhibit CP (Snyder-Warwick et al., 2010). FGFR3 mutations also can cause a form Crouzon syndrome, but patients with this form do not present with oral clefts (Meyers, Orlow, Munro, Przylepa, & Jabs, 1995). However, a missense FGFR3 mutation was identified in a Mexican family with an atypical craniosynostosis with hypochondroplasia including CPO (Gonzalez-Del Angel et al., 2018). CLP or CPO has also been occasionally reported in patients with Muencke syndrome, another craniosynostosis caused by mutant FGFR3 (Anderson, Snell, & Moore, 2013). Several causative mutations affecting Fgf signaling have been identified in patients with idiopathic hypogonadotropic hypogonadism (IHH) and autosomal dominant Kallmann syndrome (IHH with anosmia), which can include OFCs. FGFR1 mutations are associated with forms of both syndromes that include CL/P, and more recently, mutations in FGF8 have also been linked with Kallmann syndrome (Pitteloud et al., 2006; Villanueva & de Roux, 2010).

WNT signaling is also implicated in syndromic clefts. Mutations in both of WNT3 and the WNT modulator gene R-spondin 2 (RSPO2) have been identified in tetra-amelia patients with CLP (Niemann et al., 2004; Szenker-Ravi et al., 2018). Robinow syndrome is a type of skeletal dysplasia that affects limb and genital development, and characteristic craniofacial features frequently include CL/P or CPO. It is caused by mutations in several genes associated with noncanonical Wnt signaling, which is discussed further below. The more severe recessive form is associated with mutations in ROR2 (van Bokhoven et al., 2000), while dominant forms are caused by mutations in the ligand WNT5A and signal transducers disheveled 1 (DVL1) and DVL3 (Bunn et al., 2015; Person et al., 2010; J. White et al., 2015; J. J. White et al., 2016).

22q11.2 deletion syndrome, or velo-cardio-facial syndrome, includes a spectrum of disorders associated with a genomic deletion on chromosome 22. It affects neural crest development, resulting in a characteristic craniofacial phenotype including CP. DiGeorge Syndrome patients share phenotypic characteristics and causal mutations map to the same genomic region but patients do not have the deleted chromosome band (McDonald-McGinn et al., 2015). The affected region of these syndromes includes T-box transcription factor 1 (TBX1), which may be the gene responsible for associated craniofacial defects, which frequently includes palatal phenotypes ranging from arched palate or bifid uvula to overt CPO (Herman et al., 2012; Yagi et al., 2003). Opitz G/BBB syndrome, which also can include CL/P or CPO, can be classified as X-linked or autosomal dominant. However, the latter form has been determined to be similarly due to a 22q11.2 deletion, while X-Linked Opitz G/BBB is caused by mutations in MID1 and SPECCL1 (McDonald-McGinn et al., 1995).

Holoprosencephaly is characterized by the failure of the forebrain to develop into two hemispheres and often includes defects in the medial craniofacial structures. Alteration of genes encoding the transcriptional factor Sine oculis homeobox 3 (SIX3) and the Nodal/TGF-B modulator transforming growth-interacting factor (TGIF1) have been demonstrated to cause holoprosencephaly with CL/P (Aguilella et al., 2003; K. W. Gripp et al., 2000; Lacbawan et al., 2009; Wallis et al., 1999). Additionally, hedgehog signaling is key to craniofacial patterning and mutations affecting several genes in the Hedgehog pathway are associated with holoprosencephaly with CL/P, including SHH, PTCH1, and glioma-associated oncogene 2 (GLI2) (Ribeiro, Murray, & Richieri-Costa, 2006; E. Roessler et al., 1996; Erich Roessler et al., 2003). PTCH1 mutations also cause nevoid basal cell carcinoma syndrome (also known as Gorlin-Goltz Syndrome), which frequently includes OFCs (Hahn et al., 1996). Several other syndromes in which a causative gene has been identified in cases that include OFCs are listed below (Table 2).

3. Bmp/Tgf-β signaling in orofacial clefts

3.1. Bmp/Tgf-β superfamily

Tgf-β signaling is an embryonic patterning mechanism conserved across metazoa, and includes several signaling pathways that regulate a number of cellular processes to control early vertebrate development, including proliferation, apoptosis, and tissue specification (Adamska et al., 2007; Zinski, Tajer, & Mullins, 2018). The Tgf-β superfamily encompasses several classes of signaling ligand, including two which play indispensable roles in lip and palate formation and are implicated in the presentation of OFCs, the Bmp and the Tgf-β subfamilies (J. Iwata, Parada, & Chai, 2011; C. Parada & Chai, 2012) (Figure 1). The Tgf-β pathway is activated when secreted ligand dimers are recognized by multimeric serine/threonine kinase Tgf-β receptor complex consisting of two type I and two type II receptors (Budi, Duan, & Derynck, 2017; Massagué, 2012; Y. Shi & Massagué, 2003). The ligand-induced receptor complex assembly allows the phosphorylation of the type I receptors by the type II receptors. The activated type I receptor then phosphorylates and activates intracellular signaling Smad proteins (Figure 1). Two of these receptor-activated Smads (R-Smads) interact with a co-Smad, Smad4, to translocate to the nucleus and regulate transcription. Smad2/3 act as R-Smads for Tgf-β signaling, while Smad1/5/8 are R-Smads for Bmp signaling. Smad6/7 are inhibitory Smads that antagonize signaling by competitively binding to the receptor complex (Weiss & Attisano, 2013). Inhibition of Tgf-β signaling upregulates canonical Bmp activity in the posterior PS, likely because Tgf-β-specific Smad2/3 and Bmp-specific Smad1/5/8 must compete for a binding partner within the same pool of Smad4, reflecting a mechanism of mutual antagonism that occurs between the two pathways (G. Yuan, Zhan, Gou, Chen, & Yang, 2018).

Figure 1. Bmp/Tgf-β signaling and OFCs.

Figure 1.

The canonical Tgf-β and Bmp pathways with OFC phenotypes of the signaling molecules demonstrated in mutant mouse models in red. clp, cleft lip/palate; cp, cleft palate.

3.2. Bmp signaling genes associated with OFCs

Several components of the Bmp/Tgf-β superfamily contribute to the development of oral tissues, and their dysregulation can result in clefts in patients and in animal models. The human genome contains 7 type I (TGFBR1, BMPR1A, BMPR1B, ACVRL1, ACVR1, ACVR1B, and ACVR1C) and 5 type II (ACVR2A, ACVR2B, TGFBR2, BMPR2, and AMHR2) receptors, which transduce signal from at least 30 ligands, including 11 BMPs and 3 TGF-βs (Weiss & Attisano, 2013). BMP2 and BMP4 are closely related genes and comprise a subclass of BMPs (McCauley & Bronner-Fraser, 2004). Both genes play a role in mediating lip and palate formation, and mutations in both have been implicated in clefts. BMP4 SNPs have been extensively linked with NSCL/P (Antunes et al., 2013; Q. Chen et al., 2014; Y.-Y. Hu, Qin, Deng, Niu, & Long, 2015; Jianyan et al., 2010; Kempa et al., 2014; J.-Y. Lin et al., 2008; José Suazo, Santos, Jara, & Blanco, 2009), and conditional deletion of Bmp4 by Nestin-Cre in mice causes delayed fusion between the medial and lateral nasal processes (MNP and LNP, respectively), which may result in CL/P (W. Liu et al., 2005). Bmp2 deletion in Wnt1-expressing neural crest cells causes CP with failed PS elevation. Bmp2 activity in the palate likely does not directly contribute to elevation, as mandibular osteogenesis and chondrogenesis are disrupted, resulting in altered positioning of craniofacial structures that blocks PS movement. Bmp/Smad activity is only reduced in the posterior nasal region of the PS of these embryos despite a much larger Bmp2 expression domain, likely reflecting a functional redundancy between different Bmp ligands in other regions of the palate (Y. Chen, Wang, Chen, & Zhang, 2019).

Msx1 is important for neural crest specification, and Bmp signaling regulates Msx1 upstream of Snai1/2 in the neural crest ectoderm (Tríbulo, Aybar, Nguyen, Mullins, & Mayor, 2003). Msx1 knockout mice exhibit cleft palate phenotype as well as dysregulated palatal Bmp2 and Shh expression, which can be rescued by Bmp4 supplementation, evidencing a possible regulatory loop or reinforcing role for Msx1 upstream of Bmp signaling during palatogenesis (Z. Zhang et al., 2002). Bmp2 is a direct target of Wnt/β-catenin in osteoblasts (R. Zhang et al., 2013), but Bmp4 expression is not affected by Wnt signaling during lip and primary palate fusion (L. Song et al., 2009). Bmp7 also plays a role in palatogenesis. A BMP7 SNP has been linked with NSCL/P in a Han Chinese cohort (Q. Yu et al., 2015), while a frameshift mutation was identified in a European patient with syndromic defects that included secondary CPO (Wyatt, Osborne, Stewart, & Ragge, 2010). Bmp7 knockout also causes cleft palate in mice, and it is expressed in both epithelium and mesenchyme, but neither epithelialnor neural crest-specific ablation alone results in CP (Kouskoura et al., 2013). Bmp7 acts downstream of Wnt, and inhibition of Wnt signaling reduces its expression domain in the converging palatal shelves (C. Lin et al., 2011).

Bmpr1a conditional knockout (cKO) by Nestin-Cre also results in cleft lip and secondary palate in mice (W. Liu et al., 2005). These embryos display expanded expression of Pax9 and Barx1, but the conserved Bmp target Msx1 is not affected (Furuta, Piston, & Hogan, 1997; J. S. Hu et al., 2008; A. Suzuki, Ueno, & Hemmati-Brivanlou, 1997; Tríbulo et al., 2003). This may reflect incomplete redundancy with other Bmp receptors such as Bmpr1b, overexpression of which can rescue tooth (but not palate) defects in Bmpr1a mutants (L. Li et al., 2011). The CP phenotype in these embryos is primarily due to defective PS outgrowth, which are still able to fuse in vitro. However, fusion between the maxillary process (MxP) and LNP are affected, and the fusing lip displays reduced expression of p63, and both Fgf8 and its target pituitary homeobox transcription factor 1 (Pitx1) at the edge ectoderm, along with increased apoptosis (W. Liu et al., 2005). While Nestin-Cre activates prior to neural crest migration, deletion of Bmpr1a later in neural crest-derived mesenchyme by Osr2-Cre results in delayed secondary palatogenesis, causing an anterior cleft between the primary and secondary palates, as is also observed in Bmp4 cKO mutants (Baek et al., 2011). Deletion of another type I Bmp receptor, activin A receptor 1 (Acvr1), in Wnt1-expressing neural crest causes severe craniofacial defects including cleft secondary palate with no PS elevation (Dudas, Sridurongrit, Nagy, Okazaki, & Kaartinen, 2004). Overexpression, on the other hand, results in palatal fusion failure with a persistent medial epithelial seam (MES), leaving a submucous cleft of both hard and soft palates (Noda, Mishina, & Komatsu, 2016).

Nog, homolog of the human gene associated with NSCL/P (Table 1), encodes a pathway antagonist that preferentially binds and inhibits the three OFC-linked Bmps, Bmp2, Bmp4 and Bmp7, and is largely expressed in the palatal epithelium. Conventional knockout of Nog in mice causes embryos to exhibit increased epithelial Bmp/Smad activity and proliferation at E13.5, resulting in complete CP with aberrant maxillary-mandibular fusion at the posterior PS (He et al., 2010). Hypomorphic expression of the gene encoding heart-and neural crest derivatives-expressed protein 2 (Hand2) causes full CP with a similar posterior PS-mandibular fusion phenotype (Xiong et al., 2009). It was previously demonstrated that Bmp4 regulates epithelial Shh, which in turn regulates Bmp2 to control PS growth (Z. Zhang et al., 2002). Hand2 is expressed in both palatal epithelium and mesenchyme, and both sources of expression are regulated by mesenchymal Nog, likely via Bmp4. Hand2 itself activates Shh in the medial edge epithelium (MEE) of the PS, linking these two critical pathways. While mesenchymal expression is dispensable for palatogenesis, epithelium-specific Hand2 ablation phenocopies the hypomorphic allele, reflecting its importance in activating epithelial Shh (Xiong et al., 2009).

3.3. Tgf-β signaling and OFCs

Mutations in the common co-SMAD between Bmp and Tgf-β signaling, SMAD4, cause Myhre syndrome, and missense SNPs that cause an amino acid change at Ile500 were identified both in patients with CLO and with CPO (Caputo et al., 2012). Conventional Smad4 knockout causes early embryonic lethality in mice (Sirard et al., 1998; X. Yang, Li, Xu, & Deng, 1998), as does conditional knockout by Wnt1-Cre in neural crest cells. However, these cKO embryos survive until around E12.0 and exhibit failed midline fusion of the MNPs and dysgenesis of the first branchial arch (BA1). Epithelium-specific Smad4 deletion alone does not inhibit palatogenesis, illustrating the importance of Tgf signaling in craniofacial mesenchyme growth (Ko et al., 2007). P38 mitogen-activated protein kinase (MAPK) can transduce Tgf-β signals independent of Smads (L. Yu, Hebert, & Zhang, 2002). SiRNA targeting p38 does not affect palate fusion in vitro, but when p38 is knocked down by siRNA in palatal shelves of epithelial Smad4-knockout embryos in culture, fusion fails, potentially implying a partial redundancy between epithelial Smad4 and p38 (X. Xu et al., 2008). Smad4 deletion in Osr2+ mesenchyme does cause complete secondary palatal cleft, but altered expression of known Bmp targets is not detected in these embryos. However, connective tissue growth factor (Ctgf), is downregulated, which is also observed in Tgfbr2;Wnt1-Cre cKO mutants. Exogenous application of Ctgf can rescue defective mesenchymal proliferation observed in the Tgfbr2 models, further highlighting the importance of Smad4 in transducing Tgf-β subfamily signals upstream of Ctgf during palate formation (Carolina Parada, Li, Iwata, Suzuki, & Chai, 2013).

Tgf-β signaling also contributes to fusion of the MEE of converging PS. Ablation of murine Tgfbr2 by K14-Cre results in excessive epithelial proliferation and a submucous cleft of the hard palate due to a persistent MES resulting in a cyst at the anterior secondary palate. Fusion between the primary and secondary palate and between the palatal shelves and nasal septum fail, and the posterior soft palate is fully cleft with misdirected muscle attachments. These experiments revealed Tgf-β regulation of key epithelial transcription factor Irf6 (X. Xu et al., 2006). Exogenous Irf6 rescues the submucous cleft of the hard palate, demonstrating its role in epithelial apoptosis and fusion downstream of Tgf-β signaling in the MES. However, it did not rescue the soft palate cleft phenotype, suggesting that Tgfbr2 may mediate soft palate formation via a different mechanism (J.-i. Iwata et al., 2013). Tgfbr1 also appears to play different roles in each of the two major tissue lineages, as epithelial Tgfbr1 inhibition results in fully penetrant posterior CP. However, ablation in neural crest-derived mesenchyme results in a more severe phenotype including full secondary cleft palate and a midline frontonasal cleft (Dudas et al., 2006). The Wnt antagonists Dickkopf 1 (Dkk1) and Dkk4 are upregulated in these embryos, which is in turn accompanied by Wnt repression. Dkk inhibition is able to rescue proliferation and muscle formation defects in vitro, highlighting the importance of crosstalk between the Wnt and Tgf-β pathways in the developing posterior palatal mesenchyme (J.-i. Iwata et al., 2014).

FOXE1 has been strongly linked with human OFCs (Table 1), and a key facet of Foxe1’s role in murine craniofacial development is transcriptional activation of Tgfb3 and Msx1, which regulates Bmp signaling (Venza et al., 2010). Tgfb3-null mice also display cleft palate with similar defective MES fusion and proliferation in the palatal mesenchyme as observed in Tgfbr2 mutants (Kaartinen et al., 1995; Proetzel et al., 1995). In addition to signaling through Smad4, epithelial Tgf-β3 can effect a Smad-independent pathway via Tgf-β-activated kinase 1 (Tak1) (Lane et al., 2015). Recently, it was demonstrated that Irf6 directly activates transcription of Homeodomain-interacting protein kinase 2 (Hipk2), and that Hipk2 protein is activated via phosphorylation by Tak1, subsequently promoting MEE apoptosis and palate fusion (Ke, Mei, Wong, & Lo, 2019). These results imply that epithelial Tgf-β3 signaling through Tgfbr2 contributes to palatal fusion via Irf6-Tak1 activation of Hipk2.

Tgfbr3 mediates both Tgf-β and Bmp signals in secondary palate, and Tgfbr3-null embryos have fully cleft secondary palate in which PS fail to elongate and elevate, as well as showing altered vascularization and osteogenesis (Hill, Jacobs, Brown, Barnett, & Goudy, 2015). It is also required for MEE fusion, and cultured shelves treated with anti-Tgfbr3 siRNA do not fuse in vitro (Nakajima et al., 2007). Tgfbr3 is not thought to transduce signal but rather modulate signaling through other receptors. However, in embryos lacking mesenchymal Tgfbr2, Tgf-β2 is upregulated and activates a Smad-independent cascade of TNF receptor associated factor 6 (Traf6)/Tak1/p38 within the palatal mesenchyme through a receptor complex of Tgfbr1/Tgfbr3 and Tgfb2 inhibition results in partial cleft palate rescue (J.-i. Iwata, Hacia, et al., 2012). While excess activation of this pathway may contribute to the CP observed in Tgfbr2 mutants, Tak1 knockout in mesenchyme by Osr2-Cre (but not in epithelium by K14-Cre) also causes cleft palate, suggesting mesenchymal Tgf-β may control palatogenesis through this MAPK pathway, and that appropriate signaling through both the Smad and p38/Mapk branches of the pathway are required for PS closure (Z. Song et al., 2013). Phosphorylation by Tak1 appears to also activate R-Smads and transduce Smad-dependent Tgf-β signaling, so Tak1 activity may be an important mechanism for Tgf-β target cells to mediate the balance between Smad and p38/Mapk signaling (Yumoto et al., 2013).

4. Fgf signaling in orofacial clefts

4.1. Fgf signaling pathway

Fgf signaling is another morphogenetic pathway that is intimately linked with OFCs (Figure 2). Fgf ligands are recognized by their cognate receptors (Fgfrs), class V RTKs that function as hetero- or homodimers. Binding of a ligand facilitates receptor dimerization and allows their cross-phosphorylation and activation. Phosphorylated receptors are bound by intracellular transducers of several key pathways that regulate fundamental cellular processes, such as proliferation, survival, migration, differentiation, and metabolism, including signal transducer and activator of transcription (STAT), Fgfr substrate 2α (FS2α), which activates RAS/MAPK and PI3K/AKT pathways, and phospholipase C (PLC) pathways (Ornitz & Itoh, 2015). There are 22 mammalian Fgf genes, including 4 that code for intracellular proteins and 3 endocrine Fgfs, in addition to the canonical secreted paracrine Fgfs that facilitate communication between proximal cells in developing tissue. Canonical Fgfs associate with heparin/heparin sulfate, which acts as a cofactor for the Fgf ligand, so Fgf signaling is intricately linked with extracellular matrix assembly and function (Lonai, 2005).

Figure 2. Fgf signaling and OFCs.

Figure 2.

The Fgf signaling and regulatory molecules associated with OFCs in mutant mouse models are listed in red.

4.2. FGF signals involved in midfacial patterning

Many Fgf and Fgfr genes are expressed in the orofacial primordia, and mutations in several are linked with OFCs in human patients or animal models. During midfacial morphogenesis, Fgf3, Fgf8, Fgf9, Fgf10, and Fgf17 are broadly expressed within the facial ectoderm of E9.5 mouse embryos (Bachler & Neubuser, 2001). By E10.5, the MNP and LNP have formed, and their expression domains have become restricted. Fgf9, Fgf10, and the particularly strong Fgf8 are expressed around the ectodermal ridge of the nasal pits, while Fgf3, Fgf17, and a new expression domain of Fgf15 are restricted to the medial side of the nasal pit. At E10.5, Fgf8, Fgf9, and Fgf17 are also expressed at the oral side of the MNP, along with Fgf18, which is at the oral MNP but not the nasal pit. Fgf8, Fgf9, and Fgf17 are also expressed in the oral ectoderm of the MxP at E10.5 as it fuses with the LNP, and by E11.5, there are continuous expression domains of Fgf8, Fgf9, and Fgf17 across the dorsal side of the oral ectoderm derived from both progenitor structures (Bachler & Neubuser, 2001). These Fgf signals are likely transduced through Fgfr1 and Fgfr2, which are expressed in both ectoderm and mesenchyme of the facial prominences, while Fgfr3 and Fgfr4 are not detected (Bachler & Neubuser, 2001). The roles of many of these genes are likely conserved, as possible associations have been found between NSCL/P and SNPs in the human homologs of FGF7, FGF10, FGF18, FGFR1, and FGFR2 (M. Hu et al., 2013; E. J. Leslie et al., 2015; Riley et al., 2007; Y. Yu et al., 2017).

4.3. Fgf signaling in secondary palatogenesis

Fgf7 and Fgf10 are involved in patterning along the oronasal axis of the PS, with Fgf7 primarily expressed at the nasal side and Fgf10 at the oral side of the palatal mesenchyme (which correspond with and are sometimes described as the medial and lateral MxP, respectively, prior to elevation) (Alappat et al., 2005; J. Han et al., 2009). Fgfr1 and Fgfr2 are also expressed in the palatal epithelium and mesenchyme from E13 through midline fusion around E15. Fgfr1 is expressed more broadly throughout the mesenchyme, while Fgfr2 expression is more focused at the lateral/maxillary region. Both are more highly expressed at the nasal side, and epithelial expression is especially strong at the MES during its dissolution (S. Lee et al., 2001; Kai Yu, Karuppaiah, & Ornitz, 2015). Fgf signaling mediates epithelial-mesenchymal interactions (EMI) during palate development, and differential receptor-ligand specificity is integral to maintaining differential signals. Fgfr2 encodes two main isoforms that differ in their immunoglobulin-like domain III, Fgfr2IIIb and Fgfr2IIIc (Fgfr2b/c) (De Moerlooze et al., 2000). Epithelial proliferation is normally activated by mesenchyme-derived Fgf10, which preferentially signals through Fgfr2IIIb, while Fgf8 preferentially activates Fgfr2IIIc, which is distributed throughout the mesenchyme (Rice et al., 2004; X. Zhang et al., 2006).

Fgfr1 is thought to have similar spliceoforms and ligand preferences as Fgfr2. Conditional neural crest (by Wnt1-Cre) deletion of Fgfr1 in mice causes midline clefts including nasal and lip, as well as cleft primary and secondary palate with delayed maxillary growth and no PS elevation (C. Wang et al., 2013). When Fgfr1 is targeted using a Cre driver that activates later (Twist2), penetrance of CP is only 16%, and palatal shelf defects are described only after E13.5, while the Wnt1Cre/+;Fgfr1flox/flox palate exhibits reduced proliferation at least a full day earlier. While mesenchymal Fgfr2 cKO in Twist2+ cells does not cause cleft palate on its own, double cKO of Fgfr1 and Fgfr2 results in completely penetrant CP, reflecting the potential for compensation between the two receptors (K. Yu et al., 2015). Unlike mesenchymal Fgfr2 cKO, a substitution mutation that causes ectopic Fgfr2IIIc activation does result in cleft palate with increased posterior proliferation as a likely mechanism (Snyder-Warwick et al., 2010). Both Fgf10-null and Fgfr2b-deficient mice display cleft palate, highlighting the importance of this ligand-receptor interaction in mediating communication between the palatal epithelium and mesenchyme (Rice et al., 2004).

Epithelial Fgf8 is important for mesenchymal patterning and growth within BA1-derived structures, including the MxPs as they grow to form the PS (Trumpp, Depew, Rubenstein, Bishop, & Martin, 1999). However, when Fgf8 is constitutively expressed in Osr2+ mesenchyme, Fgf7 and Fgf10 expression is diminished, and palatal shelves fail to elevate, demonstrating the ligand specificity of mesenchyme-expressed receptors, as well as a feedback mechanism modulating Fgf expression. Proliferation is increased in the mesenchyme but not the epithelium in response to ectopic Fgf8, consistent with the model that it targets mesenchymal cells from the epithelium to help coordinate EMI during palatogenesis (Wu et al., 2015). Additionally, activation of Fgf effector pathways are altered in the palatal shelves of these mutants, including increased phosphorylated extracellular signal-regulated kinase (p-Erk) within the nasal/medial mesenchyme, and Janus kinase 1 (p-Jak1) throughout the mesenchyme, while p-Jak2 is increased in the epithelium but decreased in the mesenchyme (Wu et al., 2015).

4.4. Fgf regulation and target pathway interactions during lip and palate development

Fgf signaling is a key mediator of several other signaling pathways and factors involved in craniofacial development, and a number of important cleft-associated genes are regulated by craniofacial Fgf signaling (Figure 2). Fgf signaling is activated downstream of canonical Wnt pathway in the MNP and LNP. Fgf8 is a direct transcriptional target of Wnt/β-catenin signaling during lip and primary palate development, and is dysregulated in the facial ectoderm and neural ridge of Ctnnb1 (β-catenin)-mutant mouse embryos, accompanied by increased apoptosis (Y. Wang, Song, & Zhou, 2011). Not surprisingly, Fgf8 expression is also reduced in the BA1 ectoderm of embryos lacking the Wnt signaling amplifier Rspo2 (Jin, Turcotte, Crocker, Han, & Yoon, 2011). Fgf8, Fgf10, and Fgf17 are all downregulated in MNP and LNP ectoderm of Wnt9b-null mouse embryos, while all three are in turn upregulated in embryos with a Ctnnb1 allele that is constitutively activated in Foxg1-expressing ectoderm (Jin, Han, Taketo, & Yoon, 2012). This epithelial Wnt9b-Fgf cascade is further demonstrated to regulate mesenchymal proliferation via Fgfr-dependent Erk1/2 phosphorylation (Jin et al., 2012).

The zinc finger transcription factor Sp8 is integral to craniofacial morphogenesis and establishes signaling centers in the olfactory pit and anterior neural ridge of the developing mouse embryos (Kasberg, Brunskill, & Steven Potter, 2013). Loss of Sp8 function results in CLP, due to its role in activating Fgf8 and Fgf17 to regulate Shh, but not Wnt signaling in the developing maxillary and nasal processes, consistent with Wnt acting upstream of Fgf (Kasberg et al., 2013). Fgf signaling may also be regulated by Bmp, though their relationship in midfacial development is less clear. Liu et al. demonstrated that ectodermal Fgf8 expression is decreased where the LNP and MNP fuse during lip formation in Nestin-Cre;Bmpr1a cKO mouse mutants (W. Liu et al., 2005). However, Fgf8 expression is expanded in chick embryos in which mesenchymal Bmp is inhibited via ectopic Noggin application (Ashique, Fu, & Richman, 2002). Additionally, midfacial Fgf8 is a target of the micro RNA miR-17–92, which is activated by the transcription factor AP-2α (Tfap2a). Tfap2a is critical for craniofacial development and represses Fgf signaling in the MNP and LNP. Loss of function results in bilateral CLP, which can be partially rescued by reduction of Fgf8 (Green et al., 2015; J. Wang et al., 2013). The miR-17-92 promoter also contains a SMAD-binding element, and is directly activated by canonical Bmp signaling (J. Wang et al., 2010).

When Bmpr1a is ablated in Osr2-expressing mesenchyme, Fgf10 is reduced in the palatal shelves of E13.5 and E14.5 mouse embryos (Baek et al., 2011). In Fgf10 mutants, the Notch ligand Jag2 is lost in the palatal epithelium, while Tgfb3 is expanded into the oral PS epithelium where Fgf10 normally signals through Fgfr2IIIb, reflecting their antagonistic relationship (Alappat et al., 2005). Jag2 may be an important negative regulator of oral fusion processes, and Jag2-null mice also have CP associated with aberrant epithelial adhesion between the palatal shelves and tongue (R. Jiang et al., 1998). P63 is a cleft-associated transcription factor activated in the oral ectoderm that may be regulated by both Wnt and Tgf-β signaling (Ferretti et al., 2011; R. Richardson et al., 2017). P63 directly activates expression of Fgfr2 and Fgfr3 in the palatal epithelium and positively regulates Fgf8, as well as both Shh and Bmp signaling, further linking these several key pathways in the developing palatal shelves (Ferone et al., 2012; R. Richardson et al., 2017; H. A. Thomason, M. J. Dixon, & J. Dixon, 2008). Mesenchymal Fgf10 and epithelial Shh reciprocally activate each other at the oral side of the PS to mediate communication between the two tissue layers (Lan & Jiang, 2009; Rice et al., 2004). An important orofacial patterning transcription factor that plays important roles in osteoblast differentiation, distal-less homeobox 5 (Dlx5), activates mesenchymal Fgf7 at the nasal side while restricting Shh expression, establishing the Fgf expression domains across the PS epithelial surface. This axis is interrupted by ectopic activation of Fgf8 in neural crest due to its antagonistic relationship with Fgf7, and both Fgf7 and Shh are diminished (J. Han et al., 2009; Wu et al., 2015).

Runt-related transcription factor 2 (Runx2) and its target Osterix (Sp7) are also key transcription factors that are also involved in promoting osteogenesis. They are activated downstream of Fgf signaling in certain contexts, and are thought to play a role in craniosynostosis associated with excess Fgf signaling (Komori, 2011). When ectopic Fgf8 is activated in cells expressing the anterior palatal mesenchyme transcription factor Short stature homeobox 2 (Shox2), however, Runx2 and Sp7 are inhibited in that region. This may be due to an antagonistic relationship between Fgf8 and a different Fgf that promotes osteoblast differentiation, such as Fgf18 (J. Xu et al., 2018). While these embryos do not display secondary CP, osteogenesis is interrupted and the palatine process of the maxilla is absent, with excess cartilaginous tissue formed in its place (J. Xu et al., 2018).

Fgf18 is expressed in the palatal mesenchyme just under the MEE where it activates epithelial Runx1, which is required for completing palate fusion (Charoenchaikorn et al., 2009). Fgf18 also helps regulate craniofacial chondrogenesis and osteogenesis, and Fgf18-null mice exhibit reduced cranial ossification and cleft secondary palate with PS elevation failure (Z. Liu, Xu, Colvin, & Ornitz, 2002). Foxf1 and Foxf2 activity in the palatal mesenchyme inhibits Fgf18 to maintain expression of its repressive target Shh. Foxf1/2 are partially redundant, and conditional Foxf2 deletion in neural crest lineage results in palate elevation failure with expanded Fgf18 expression domains (J. Xu et al., 2016). CP also presents in Fgf9−/− mice with 40% penetrance, but few details on the cause are available (J.-i. Iwata, Tung, et al., 2012). Fgf9 is expressed in the maxillary epithelium at E13.5 and both mesenchyme and epithelium at E14.5 and more recently, Tgf-β signaling has been demonstrated to regulate PS proliferation through a Fgf9-Pitx2 axis (Colvin, White, Pratt, & Ornitz, 2001; J.-i. Iwata, Tung, et al., 2012). Sox11 also regulates proliferation in the palatal shelves through Fgf9, which can be activated to rescue growth defects in Sox11−/− palate in vitro (H. Huang et al., 2016).

4.5. Fgf signaling inhibitors associated with OFCs

Spry family members are antagonists of Fgf signaling and specifically inhibit Ras-Erk pathway activation by Fgf receptors, and Fgf signaling itself regulates Spry gene expression, which can then act as negative feedback mechanism to modulate signaling activity (Mason, Morrison, Basson, & Licht, 2006). Spry genes are involved in facial morphogenesis and SPRY1, SPRY2, and SPRY4 variants have been implicated in NSCL/P in human patients (K. U. Ludwig et al., 2012; Vieira et al., 2005; Y. Yu et al., 2017; R. Zhou et al., 2019). Ectopic activation of Spry1 in mouse neural crest by Wnt1-Cre causes severe craniofacial defects including CLP, with decreased proliferation and increased apoptosis (X. Yang et al., 2010). Spry2 and Spry4 are integral to craniofacial morphogenesis and exhibit partial redundancy, evidenced by an increased severity of defects in compound mutant mouse embryos, though embryos lacking only Spry4 do not present with clefts (Taniguchi et al., 2007). A >1 Mb genomic deletion including Spry2 among multiple genes in mouse causes cleft palate with occasional cleft lip, with altered expression of Etv5, Msx1, Barx1, and Shh signaling (Welsh, Hagge-Greenberg, & O’Brien, 2007). Transgenic Spry2 expression is able to rescue the cleft palate phenotype from 83% to 8% penetrance, demonstrating Spry2 is the gene likely responsible for the oral clefts observed in this strain (Welsh et al., 2007). Targeted Spry2 deletion further reveals its role, as ossification or MES fusion potential are unaffected, while palatal mesenchyme exhibits excess proliferation due to enhanced Fgf signaling. leading to PS elevation failure (Matsumura et al., 2011). Similarly, Spry2 overexpression results in defective outgrowth of facial prominences but does not affect the chondrogenesis or osteogenesis of mesenchymal cells (Goodnough, Brugmann, Hu, & Helms, 2007).

5. Retinoic acid signaling in orofacial clefts

5.1. RA signaling pathways

Retinoic acid (RA) is a metabolic derivative produced through the retinol (Vitamin A) metabolic pathway (Figure 3), and it is an important signaling molecule in morphogenesis and differentiation during embryonic development. Retinoids cannot be synthesized by animal cells and dietary intake of retinoids or their carotenoid precursors is necessary for RA production. Circulating dietary retinol is bound by a retinol binding protein (RBP) family carrier, and this complex is recognized by the RBP receptor STRA6 and taken up by target cells (Kawaguchi et al., 2007; Quadro et al., 1999). Cellular retinol binding protein (CRBP) recognizes free cytosolic retinol and facilitates its interaction with alcohol dehydrogenase (ADH) or retinol dehydrogenase family (RDH) enzymes that catalyze its oxidization into retinaldehyde (Ang, Deltour, Hayamizu, Žgombic-Knight, & Duester, 1996; P. N. MacDonald & Ong, 1987; Sandell et al., 2007; M. Zhang, Chen, Smith, & Napoli, 2001). Retinaldehyde is subsequently oxidized to RA by one of three tissue-specific retinaldehyde dehydrogenase enzymes, RALDH1, RALDH2, and RALDH3 (encoded by ALDH1A1–3) (Niederreither & Dollé, 2008) (Figure 3). RA is in turn metabolized into 4-hydroxy-RA by one of three members of the Cytochrome P450 26 subfamily (CYP26) (J. A. White et al., 1997). Cytosolic RA signals through the nuclear receptor family retinoic acid receptors (RARs) and retinoid X receptors (RXRs), which are imported to the nucleus upon binding RA. Activated RAR and RXR proteins function as heterodimers to recognize Retinoic Acid Response Element (RARE) genomic sequences and recruit co-regulator proteins to activate transcription of target genes (Pogenberg et al., 2005). Several isoforms of RA exist, including notably all-trans, 9-cis, and 13-cis forms. The RAR and RXR receptor families each contain an α, β, and γ form, of which each has three subtypes (Okano, Udagawa, & Shiota, 2014). While RXR is specifically activated by all-trans retinoic acid (atRA), RAR receptors are activated by both cis- and trans- RA (Leid, Kastner, & Chambon, 1992). Cellular retinoic acid-binding proteins I and II (CRABP-I and CRABP-II) facilitate RA interaction with CYP26 enzymes or RAR/RXR receptors, with CRABP-II associated with receptor activation and CRABP-I favoring interaction with CYP26 (Dong, Ruuska, Levinthal, & Noy, 1999; Rhinn & Dollé, 2012). The metabolic and signaling pathways of RA have previously been reviewed in greater detail (Das et al., 2014; Duester, 2008; Ghyselinck & Duester, 2019; Rhinn & Dollé, 2012). Retinoid metabolism is strongly implicated in OFCs, which is discussed at greater length in a companion article (Garland et al., 2020 in revision), while the following section will discuss some of the underlying genetics involved in RA signaling and its interaction with other signaling pathways during craniofacial development.

Figure 3. RA signaling and OFCs.

Figure 3.

The retinoid signaling and metabolic pathway components associated with OFCs demonstrated in mouse models are listed in red. cp, cleft palate.

5.2. Abnormal RA signaling in the cause of orofacial clefts

RA signaling regulates developmental processes of many embryonic tissues/organs, including the neural tube, somite, foregut, limb, brain, and eye. RA is also required for appropriate craniofacial morphogenesis. Deficiencies of vitamin A and its metabolites have long been known to cause congenital craniofacial defects (J. G. Wilson, Roth, & Warkany, 1953), and excess retinoids similarly induce syndromic defects in rodents, including cleft palate (Harold Kalter, 1960; H. Kalter & Warkany, 1961; Walker & Crain Jr, 1960) (Figure 3). The balance between RA production and degradation by RALDH and CYP26 enzymes mediates cellular RA levels and signaling (Cunningham & Duester, 2015), while RARE-inducible transcriptional activation of Cyp26a1 acts as a negative feedback mechanism to regulate excess RA activity (Loudig, Maclean, Dore, Luu, & Petkovich, 2005).

Excess endogenous RA also can result in cleft palate and palatal shelves fail to elevate in Cyp26b1-null mice, as a reduced ability to metabolize RA results in its accumulation and overactivation of its receptors (Okano et al., 2012). Retinoid signaling deficiency is also detrimental and knockout is lethal, either embryonically or within 12 hours postnatally, for 8 out of the 10 RAR isoforms encoded by Rara, Rarb, and Rarg. Two types of compound RAR mutants (α1−/−;α−/+;γ−/−, and α−/−; γ−/−) display midfacial and secondary palatal clefts, as RARβ is unable to significantly compensate for loss of activity by RARα and RARγ (Lohnes et al., 1994). Aldh1a3-deficient neonatal mice also exhibit reduced RA activity due to deficient retinaldehyde metabolism, and exhibit lethal choanal atresia, associated with over-fusion of the nasal primordia (Dupé et al., 2003). Compound Aldh1a2/Aldh1a3 mutants display a midline facial cleft and bilateral cleft lip, and the midline cleft can be rescued by maternal RA supplementation from E7.5 to E10.5. However, the bilateral lip defect is persistent and more precise modulation of cellular RA levels may be required for the frontonasal structures to fully form (Halilagic et al., 2007).

5.3. RA signaling interactions in orofacial clefts

A reciprocal regulation between the Cyp26 RA degradation enzymes and Tbx1 has been revealed in palatogenesis (Figure 3). Tbx1 regulates oral epithelial differentiation and adhesion, and Tbx1-deficient mice exhibit cleft palate (Funato, Nakamura, Richardson, Srivastava, & Yanagisawa, 2012). All three Cyp26 genes (Cyp26a1, Cyp26b1 and Cyp26c1) are expressed in the pharyngeal regions which give rise to maxillary and palatal tissues and are dysregulated in Tbx1 mutants (Roberts, Ivins, Cook, Baldini, & Scambler, 2006). During palatal development, Cyp26b1 is expressed primarily in mesenchymal cells, while Cyp26a1 is localized to the epithelium (Okano et al., 2012). Tbx1 is downregulated in the maxillary tissue in Cyp26b1-null mice (which also have CP) which accumulate RA in mesenchymal cells, also resulting in reduced expression of key palatal regulators Bmp2 and Fgf10 (Okano et al., 2012), and a reduction in Tbx1 was also reported in the embryonic tongue in response to exogenous RA treatment (Okano, Sakai, & Shiota, 2008).

RA has a mutually inhibitory relationship with Tgf-β signaling in palatogenesis, and atRA treatment of cultured palatal shelves prevents fusion, upregulating expression of the inhibitory Smad7, and reducing Smad2 phosphorylation (Yuming Wang et al., 2011). In mouse embryonic palatal mesenchyme cells (MEPM), atRA similarly antagonizes Tgf-β signaling, reducing Smad2 activation and increasing expression of both Rarb and Smad7, resulting in reduced proliferation. Tgf-β3 treatment in turn antagonizes RA signaling, reducing Rarb and Smad7 expression. TGIF1 interacts with RAR-α in addition to Smad2/3, and is a co-repressor of both signaling pathways. Treatment with either atRA or Tgf-β3 causes a significant increase in Tgif1 expression, which was shown to be a key mechanism of each pathway’s antagonism of the other (X. Liu et al., 2014).

Mutual repression between RA and Wnt signaling may also play a critical role in orofacial development (Figure 3). In Wnt-deficient Lrp6-knockout mice, which present with fully penetrant CLP, the expression domain of Aldh1a3 in the nasal pits is expanded, resulting in increased RA activity (L. Song et al., 2009). In contrast, RA treatment has been shown to repress canonical Wnt/β-catenin signaling, along with alterations in proliferation and apoptosis in mouse embryonic palatal shelves through dysregulation of several cell cycle regulators (including cyclinD1, p21, and p27), and of p38 MAPK signaling, respectively (X. Hu, Gao, Liao, Tang, & Lu, 2013).

RA signaling has been shown to maintain both Shh and Fgf8 expression in the frontonasal process of chick embryos, and chemical disruption of RA signaling results in reduced proliferation and increased apoptosis (Schneider, Hu, Rubenstein, Maden, & Helms, 2001). Similar results are observed in embryos treated with a RALDH antagonist, and exogenous Fgf8 is able to partially rescue many of the craniofacial defects from insufficient RA signaling (Y. Song, Hui, Fu, & Richman, 2004). However, more recently, addition of exogenous RA in mouse embryos also resulted in increased apoptosis in maxillary tissue and downregulation of hedgehog pathway components Shh, Ptch1, and Gli1, while addition of a hedgehog agonist partially rescues the penetrance of RA-induced OFC (Q. Wang et al., 2019). This is consistent with previous studies demonstrating RA treatment repressed Shh signaling, further evidencing the importance of Shh inhibition as a function RA signaling (Helms et al., 1997). Shh in turn antagonizes RA signaling by activating Cyp26a1 and Cyp26b1 to reduce cellular RA levels. Shh also regulates the RA receptors, and cKO induced at E10.5 (Shhflox/Cre-ERT) results in significantly increased RARb and RARg expression (El Shahawy et al., 2019).

6. Shh signaling in orofacial clefts

6.1. Shh signaling

Hedgehog signaling is fundamental to many aspects of animal development, and is implicated in the pathology of numerous congenital disorders and various cancers, which has previously been reviewed by Briscoe and Thérond (Briscoe & Thérond, 2013), and in craniofacial development, specifically reviewed by Abramayan (Abramyan, 2019). Of three mammalian hedgehog ligands (Indian hedgehog, Desert hedgehog, and Sonic hedgehog), Shh is the best studied and the hedgehog gene most commonly associated with congenital defects and disease. Upon synthesis, Shh is post-translationally modified via cleavage and lipidation, by which a cholesterol and palmitoyl group are added to the N-terminal fragment. Lipid modification is required for association with the membrane and secretion, which occurs through activity of dispatched (Disp) and signal peptide CUB EGF-like domain-containing protein (Scube2), allowing secreted ligand to then signal both short- and long-distance to target cells. In the absence of signal, the 12-pass transmembrane receptor patched (Ptch) represses signaling by inhibiting the GPCR family member smoothened (Smo), while the downstream effector glioma-associated oncogene (Gli) is sequestered and inhibited by Suppressor of Fused (Sufu). Upon binding of the Hh ligand to the Ptch receptor, Smo inhibition is relieved, which allows it to accumulate in the distal tip of the primary cilium. There it dismantles the Sufu-Gli complex, allowing Gli to freely enter the nucleus and act as a transcription factor to alter target gene expression (Abramyan, 2019). Several components of the Hedgehog pathway have been associated with OFCs in both human patients and animal models (Figure 4). Hedgehog signaling is intimately linked with primary cilia activity, and many genes that encode ciliary proteins are similarly associated with clefts, which is extensively discussed in an associated review article (Ji et al., 2020 in revision).

Figure 4. Shh signaling and OFCs.

Figure 4.

The OFC phenotypes of the Shh signaling molecules (in red font) which have been demonstrated in mutant mouse models. Cyclopamine and Vismodegib are teratogens that inhibit Shh signaling and can induce OFCs in mouse embryos. clp, cleft lip/palate; cp, cleft palate.

6.2. Shh signaling in craniofacial morphogenesis and OFCs

Shh is expressed in the craniofacial ectoderm and is key for regulating development of the cranial neural crest, and dysregulated signaling causes severe defects of BA1-derived structures and midline patterning (Ahlgren & Bronner-Fraser, 1999; Chiang et al., 1996). Holoprosencephaly occurs when the forebrain fails to develop into two hemispheres, and often involves narrowing of craniofacial midline structures. It is often associated with CL/P, and mutations in several components of the Hh pathway, including SHH, PTCH1, and GLI2, can result in holoprosencephaly in human patients (Table 2). Cyclopamine was identified as a teratogen in sheep who gave birth to holoprosencephalic offspring after exposure, which was later determined to confer this effect via inhibition of hedgehog signaling (Chiang et al., 1996; Keeler, 1978). Both cyclopamine and vismodegib, another hedgehog antagonist, cause cleft lip and palate in mouse embryos when introduced between embryonic days 7 and 10 (Heyne et al., 2015; Lipinski et al., 2010). Hedgehog acyltransferase (Hhat) palmitoylates Shh, a modification that is essential to its long-range signaling capability (Buglino & Resh, 2008; M.-H. Chen, Li, Kawakami, Xu, & Chuang, 2004). Mouse embryos lacking Hhat exhibit severe craniofacial defects including holoprosencephaly, and vertical PS growth from the MxP is disrupted, highlighting the key role hedgehog signaling plays in secondary palatogenesis and cleft palate (Dennis et al., 2012).

Shh contributes to midfacial expansion and excess signaling can result in hypertelorism, which can range from wider spacing between the eyes to complete facial duplication and may also contribute to OFC formation. Conditional targeting of hedgehog repressor Ptch1 in mouse neural crest, which effectively results in constitutive hedgehog activation, results in failed fusion of the MNP and LNP. These embryos exhibit reduced Fgf signaling within the nasal pit and prominences with lower mesenchymal cell density and increased epithelial cell death, which would result in bilateral CLP if embryos survived to term (Metzis et al., 2013). While this model is embryonic lethal prior to secondary palatogenesis, transgenic mice with ectopic Shh expression in Krt14+ basal epithelium survive longer and similarly present with severe craniofacial defects, including fully penetrant cleft secondary palate (Cobourne et al., 2009). Constitutive Smo expression in K14+ cells causes submucous CP due to a persistent MES that is resistant to apoptosis and maintains a proliferative state beyond that which is observed during normal palatogenesis (J. Li et al., 2018). However, constitutive Smo activation in Osr2+ mesenchyme causes reduced vertical growth of palatal shelves with a rounded appearance that fail to elevate, resulting in a wide cleft with maxillary skeletal defects (Hammond, Brookes, & Dixon, 2018). Among upregulated target genes in the palatal tissue of these embryos is Sclerostin domain-containing 1 (Sostdc1), which directly antagonizes both Bmp and Wnt signaling (Ahn, Sanderson, Klein, & Krumlauf, 2010; Hammond et al., 2018; Laurikkala, Kassai, Pakkasjärvi, Thesleff, & Itoh, 2003; Lintern, Guidato, Rowe, Saldanha, & Itasaki, 2009).

Shh is expressed in periodically spaced domains along the anteroposterior axis of palatal epithelium and contributes to the formation of palatal rugae, a series of palatal ridges involved in sensing and manipulating food that are conserved throughout mammals (Economou et al., 2012). New expression domains of the rugae patterning genes are sequentially established as the palatal shelves grow. Each new rugal domain is formed immediately anterior to ruga 8, which is formed first and corresponds with the boundary between anterior Shox2 and posterior Meox2 expression (Pantalacci et al., 2008). This expression pattern is regulated by a reaction-diffusion mechanism involving antagonistic activities between Lrp4 and Sostdc1, which are complementarily expressed in the prospective rugal and inter-rugal domains, respectively (Kawasaki et al., 2018). Lrp4 and Sostdc1 may modulate Wnt signaling and Wnt is required to establish Shh signaling centers during rugae induction. (C. Lin et al., 2011). When either of Lrp4 or Sostdc1 is knocked out, Shh expression domains become disordered, resulting in erratic ridge patterning (Kawasaki et al., 2018).

6.3. Shh signaling interactions during lip and palate formation

Shh acts downstream of Fgf signaling in the oral epithelium during secondary palate formation, activated by signaling through Fgfr2IIIb by mesenchyme-derived Fgf10. This axis is disrupted in either Fgf10 or Fgfr2b mouse mutants, which present with dysregulated Shh and CP. This is likely due to proliferation and growth defects observed in the palatal shelves, though the potential for medial fusion remains in vitro (Rice et al., 2004). Shh is also repressed by retinoic acid signaling in the craniofacial primordium, and the role excess RA plays in causing clefts is due in part to its modulation of Shh polarizing activity (Helms et al., 1997).

Shh mediates EMI in the developing palate, and epithelial Shh activates several downstream palatal regulators within the mesenchyme, including Foxf2, Osr2, Bmp2, and Fgf10. It also controls mesenchymal proliferation by maintaining expression of Ccnd1 and Ccnd2 (Lan & Jiang, 2009). Foxf2 is a direct target and key effector of Shh signaling in lip and primary palate formation. Transient cyclopamine-induced hedgehog inhibition at E9.0 causes a reduction of Foxf2 expression, and Foxf2 was shown to promote Shh-dependent mesenchymal proliferation (Everson et al., 2017). In addition to Foxf2, expression of several other Fox genes in neural crest-derived mesenchyme of facial primordia are dependent on Shh signaling, including Foxc2, Foxd1, Foxd2, and Foxf1 (Jeong, Mao, Tenzen, Kottmann, & McMahon, 2004). Foxf1 and Foxf2 are an important facet of a regulatory feedback loop in the secondary palate between Shh and Fgf, in which Shh-dependent Foxf1/2 activates Fgf18, which feeds back to regulate Shh expression (J. Xu et al., 2016).

In another important EMI loop, Msx1 activates Bmp4 in the palatal mesenchyme to signal to the epithelium and activate Shh. Shh in turn signals back to the mesenchyme to activate Bmp2 and control proliferation (Z. Zhang et al., 2002), as well as positively regulate both Msx1 and Bmp4 (Lan & Jiang, 2009). Ablation of epithelial Shh, but not Smo, causes cleft palate, demonstrating that specifically epithelium-to-mesenchyme hedgehog signaling is required for PS growth (Lan & Jiang, 2009). Tp63-null embryos display bilateral cleft lip and palate, and reduced proliferation in the MxP mesenchyme prevents formation of the region that would otherwise develop into palatal shelves. While Bmp4 positively regulates Shh in the developing craniofacial processes, both of Shh and Fgf8 are diminished in the nasal and lip primordia in these embryos, with Bmp4 upregulated in the posterior LNP and anterior MxP between E10.5 and E11.5. These differential responses may be independent of each other and Bmp and Shh likely act in parallel downstream of Tp63 (Helen A. Thomason, Michael J. Dixon, & Jill Dixon, 2008). Studies in chick demonstrated that blocking Bmp signaling in the frontonasal process with ectopic noggin results in diminished Shh expression (Foppiano, Hu, & Marcucio, 2007). Shh activates all three of Bmp2, Bmp4, and Bmp7 in the frontonasal process, and this positive reinforcement loop between Bmp and Shh signaling contributes to growth of the facial prominences during fusion of the primary palate (D. Hu et al., 2015).

7. Wnt signaling in orofacial clefts

7.1. Wnt lipidation and secretion molecules

Wnt signaling is integral to many aspects of development and is often associated with disease, which has been more extensively reviewed elsewhere (Clevers & Nusse, 2012; Komiya & Habas, 2008; B. T. MacDonald, Tamai, & He, 2009; Nusse & Clevers, 2017; van Amerongen, 2012). Wnt can activate a number of downstream pathways, traditionally classified as canonical or non-canonical depending on the primary response by a target cell, but there are many feedback mechanisms and points of interaction between the pathways that complicate this division. In brief, in the absence of Wnt, cytoplasmic β-catenin is continually targeted for proteasomal degradation. A canonical Wnt signal results in inhibition of the β-catenin destruction complex, allowing accumulation of free β-catenin to act as a transcription factor and alter target gene expression. Pathways that Wnt can activate independent of β-catenin include calcium/calmodulin signaling and the planar cell polarity (PCP) signaling cascades. Wnt signaling interacts with many other signaling axes and regulators during craniofacial development, and several components of the Wnt pathways are implicated in the etiology of OFCs (Reynolds et al., 2019).

In a Wnt-secreting cell, nascent Wnt ligands are post-translationally glycosylated, and then subsequently palmitoylated by the O-acyltransferase Porcupine (Porcn) within the endoplasmic reticulum (Galli, Barnes, Secrest, Kadowaki, & Burrus, 2007; Komekado, Yamamoto, Chiba, & Kikuchi, 2007) (Figure 5). Mutations within or near PORCN cause Goltz-Gorlin syndrome/focal dermal hypoplasia, which can include CL/P (Bornholdt et al., 2009; Lombardi et al., 2011; X. Wang et al., 2007). Wntless (Wls, homologous to the human GPR177) promotes secretion of Wnt ligands and is required for the many of the patterning processes established by Wnt signaling during embryonic development (Bänziger et al., 2006). Wls recognition of a nascent Wnt ligand is dependent on Porcn-mediated acylation, and disrupted signaling in its absence reflects a requirement for lipid linkage in secretion and targeting to the membrane of a recipient cell (Herr & Basler, 2012). Wls cKO in Wnt1-expressing neural crest cells results in complete secondary CP. Both canonical β-catenin-dependent and Wnt5a-mediated noncanonical signaling are disrupted, causing altered patterns of proliferation and excessive cell death in palatal mesenchyme and epithelium. (Y. Liu et al., 2015). On the other hand, Wls deletion in the ectoderm using Foxg1-Cre inhibited the cell adhesion and motility of epithelial cells during nasal pit invagination. This was accompanied by increased cell death and reduced proliferation, leading to severe frontonasal defects that were not observed in Wnt1-Cre-mediated Wls mutants (Zhu et al., 2016). Bmp, Fgf, and Shh signaling pathways, as well as JNK and ERK signaling, were all diminished in the facial prominence of these embryos (Zhu et al., 2016), suggesting multiple key roles for Wnt in modulating a complex signaling network to regulate frontonasal development.

Figure 5. Wnt signaling and OFCs.

Figure 5.

Wnt signaling pathways with components in which OFC phenotypes have been demonstrated in mutant mouse models listed in red. clp, cleft lip/palate; cp, cleft palate; mcl, median cleft lip.

7.2. WNT mutations associated with orofacial clefts

There are 19 Wnt ligands in the mammalian genome, and several have been linked with oral clefts in human patients, in both syndromic (Table 2) and nonsyndromic cases. One NSCL/P association study found evidence of linkage with SNPs in WNT5A, WNT7A, WNT8A, and WNT11 across multiple populations (Brett T. Chiquet et al., 2008). Human WNT6 and WNT10A are clustered together at 2q35 (Kirikoshi, Sekihara, & Katoh, 2001), and an analysis of 3 populations found significant association with SNPs at this cluster for both CPO and CL/P (Beaty et al., 2006). Additionally, suppressing the mouse homolog of either of these genes has been demonstrated to perturb growth of MEPM cells in vitro (Feng et al., 2013; Z. Jiang, Pan, Chen, Chen, & Xu, 2017). Several studies have linked WNT3 mutations with NSCL/P, including haplotype association with WNT3A and COL2A1 (B. T. Chiquet et al., 2008; Y. P. Lu et al., 2015; Adrianna Mostowska et al., 2012; Nikopensius et al., 2010; Nikopensius et al., 2011). WNT3 and WNT9B are clustered at 17q21, and an association with WNT9B, but not WNT3, was identified in a Brazilian NSCL/P cohort (Fontoura, Silva, Granjeiro, & Letra, 2015), while another study found haplotype association with two WNT9B SNPs and epistasis between WNT9B and MSX1 (P = 2.564 × 10−4), a conserved craniofacial BMP and WNT target (Medio et al., 2012; Nikopensius et al., 2011).

Wnt3 is expressed in the maxillary and medial nasal ectoderm during midfacial morphogenesis in mice (Lan et al., 2006), but Wnt3-null mice die early in development (P. Liu et al., 1999), prior to emergence of the palatal shelves from the MxP, and currently no models have yet been generated to describe a role Wnt3 plays in lip and palate formation. Wnt9b is one of two Wnt ligands whose role in the formation of OFCs has been clarified in mouse models, however, along with Wnt5a. The A/WySn mouse strain exhibits spontaneous CL/P due to a mutation that maps to a region previously called the Clf1 locus associated with CL/P. It was subsequently determined that Wnt9b is the affected gene in A/WySn mice and that Clf1 is a mutant allele of Wnt9b (Juriloff et al., 2006). Targeted Wnt9b mutation also causes partially penetrant CL/P in which palatal shelves fail or delay to elevate and converge, but do not show altered proliferation or apoptosis (Carroll, Park, Hayashi, Majumdar, & McMahon, 2005; Jin et al., 2012). While Wnt9b is particularly important for lip and primary palate formation, Wnt5a is integral to secondary palate development. Wnt5a-null mice present with fully penetrant CPO due to directional cell migration defects in the palatal mesenchyme which result in failed PS elevation (He et al., 2008).

7.3. Canonical Wnt signaling pathway and orofacial clefts

Canonical Wnt signals are recognized at a target cell membrane by a Frz (Frizzled) GPCR family receptor and a single pass Lrp (lipoprotein receptor-related protein) 5 or 6 co-receptor. Recognition of a Wnt ligand facilitates binding to the Wnt-Fzd-Lrp complex by disheveled (Dvl), which contains a central PDZ domain that physically interacts with the C terminal tail of Fzd (Wong et al., 2003). Several studies have linked various Wnt receptors to OFCs (Figure 5). A FZD2 nonsense mutation was linked to omodysplasia with CLP (Saal et al., 2015), and a rare intronic SNP in FZD6 was identified in an African American family with NSCLP (Cvjetkovic et al., 2015). In mouse models, Fzd2 knockout causes 50% penetrant CPO in mice, and Fzd1/Fzd2 compound mutants exhibit fully penetrant CPO (Huimin Yu et al., 2010). Mutations in LRP6 have been identified in both sporadic and familial NSCL/P cases (Basha et al., 2018; Ockeloen et al., 2016), while Lrp6-knockout mice have fully penetrant CLP (L. Song et al., 2009).

The β-catenin destruction complex continually targets cytoplasmic β-catenin for proteasomal degradation, reviewed in more detail elsewhere (Stamos & Weis, 2013). The destruction complex contains two scaffold proteins, adenomatous polyposis coli (Apc) and axis inhibition (Axin) (E. Lee, Salic, Krüger, Heinrich, & Kirschner, 2003). Association with Fzd-Wnt allows Dvl to recruit Axin, promoting assembly of a “signalosome” complex at the cell membrane. This assembly is dependent on phosphorylation of the Lrp coreceptor by another destruction complex protein, casein kinase 1 (Ck1) (Cliffe, Hamada, & Bienz, 2003; Kishida et al., 1999). Mammals possess two Axin genes. Mouse models with loss of Axin1 function are early embryonic lethal, due to excess β-catenin activation. Reducing Ctnnb1 gene dosage can partially rescue the severity of defects, allowing further progression of craniofacial development, and these mutants exhibit median cleft lip (Chia, Kim, Itoh, Sokol, & Costantini, 2009). While Axin2 mutant mice also exhibit craniofacial defects, no oral clefts have been reported (H.-M. I. Yu et al., 2005). However, AXIN2 variants may play a role in the etiology of NSCL/P or NSCPO in human patients (Y. Han et al., 2014; A. Letra et al., 2012; Ariadne Letra, Menezes, Granjeiro, & Vieira, 2009). Axin also interacts with glycogen synthase kinase 3 (Gsk-3) and β-transducin repeat-containing protein (β-TrCP), which phosphorylate and ubiquitinate β-catenin, respectively. A significant association between a GSK3B variant and NSCL/P was identified in a Brazilian population (Vijayan, Ummer, Weber, Silva, & Letra, 2017), while Gsk3b knockout in mice causes complete secondary CP in which the shelves elevate, but do not converge toward the midline (K. J. Liu, Arron, Stankunas, Crabtree, & Longaker, 2007), further illustrating the requirement for appropriate β-catenin levels during palatogenesis.

Recruitment of Axin to the membrane facilitates its dephosphorylation, resulting in reduced binding affinity for β-catenin and inhibition of the destruction complex (Willert, Shibamoto, & Nusse, 1999). This allows accumulation of β-catenin, which is imported into the nucleus and can heterodimerize with a T-cell factor/Lymphoid enhancer factor TCF/LEF family transcription factor. TCF/LEF recognizes Wnt Responsive Element (WRE) genomic sequences, and normally represses transcription of target genes. However, nuclear β-catenin converts TCF/LEF into an activator and recruits co-activators to transcribe target genes (Cadigan & Waterman, 2012). Mice with epithelial Ctnnb1 cKO display CP in which shelves elevate but do not fuse, with suppressed apoptosis and downregulated Tgf-β activity at the MEE (He et al., 2011). Conversely, epithelial Ctnnb1 overexpression also causes CP with aberrant fusion events between the PS and mandibular epithelium (He et al., 2011). β-catenin also participates in cell adhesion, though it hasn’t been demonstrated whether this role uniquely contributes to palatogenesis independent of its Wnt-induced function (Yamada, Pokutta, Drees, Weis, & Nelson, 2005).

Tcf/Lef1-β-catenin-dependent transcription is activated in the facial ectoderm in response to Wnt signaling during midfacial development, and Lef1 may provide a mechanism for modulating the Wnt and Tgf-β pathways and can participate in Smad-dependent transcriptional activation in response to Tgf-β signaling in the palatal epithelium as well (Nawshad, Medici, Liu, & Hay, 2007). LEF1 variants have been linked with CL/P and CPO in humans (Martinelli, Carinci, et al., 2011), though CP is not among craniofacial defects seen in Lef1-null mice, possibly reflecting partial redundancy with other Tcf/Lef1 family members (van Genderen et al., 1994).

7.4. Non-canonical Wnt signaling in orofacial clefts

Non-canonical Wnt signaling includes several distinct cellular responses that are independent of β-catenin, and can be transduced through Frz and the RTK family paralogues Ror1/2 and Ryk, and may involve Dvl. The PCP pathway regulates how cells in a tissue align themselves in a uniform orientation and undergo convergent extension, polarized cellular movements during which cells narrow along one axis and extend along the perpendicular axis to change the structure of a tissue (Butler & Wallingford, 2017; Dabdoub et al., 2003; Heisenberg et al., 2000; Qian et al., 2007; Tada & Smith, 2000; Jianbo Wang et al., 2006). Wnt can also activate several Rho family GTPases to regulate cytoarchitecture and the Jun N-terminal kinase (JNK) signaling cascade. The other major noncanonical Wnt pathway, Wnt/Ca2+ signaling, triggers cleavage of phosphatidyl inositol 4,5-bisphosphate (PIP2) by Phospholipase C (PLC) to generate secondary messengers 1,2-diacylglycerol (DAG) and 1,4,5-inositol triphosphate (IP3), which then activate Protein Kinase C (PKC) and calcium channels, which in turn activates calmodulin and Ca2+/calmodulin-dependent protein kinase II (CamKII), with a number of downstream functions including activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) and nuclear factor of activated T-cells (NFAT) transcription factors (De, 2011)..

Multiple ROR2 SNPs were linked with NSCPO in an Asian population (H. Wang et al., 2012), and Ror2-null mice exhibit craniofacial defects including CPO (Schwabe et al., 2004). Ror2 has been demonstrated to transduce Wnt5a signaling through Dvl (Ho et al., 2012), and WNT5A, ROR2, and DVL1/3 contribute to Robinow syndrome (Table 2). While neither of Ror2 or Wnt5a heterozygous mutations cause CPO alone, palatal shelves in compound heterozygous mouse embryos fail to elevate due to mesenchymal migration defects, similar to Wnt5a mutants, demonstrating an epistatic relationship between the two genes and evidencing Ror2 as the primary receptor for Wnt5a during PS elevation (He et al., 2008). Wnt5a and Ror2 are both dysregulated in mice with RA-induced tongue malformation and CPO, suggesting not only that the teratogenic effects on palate development from RA may be at least in part due to an interaction with Wnt signaling, but also strengthening the evidence of Ror2 as a key receptor for Wnt5a during palatogenesis (Cong et al., 2014). In mice with neural crest-specific Wls deletion, both canonical Wnt signaling and activation of noncanonical effectors c-Jun and JNK are dysregulated in the palatal mesenchyme. Addition of exogenous Wnt5a is able to rescue phosphorylation of c-Jun and JNK, evidencing Wnt5a as a primary signal responsible for their activity (Y. Liu et al., 2015). Ryk is an important receptor involved in axon guidance and may transduce Wnt5a signal independent of Frz/Dvl (Keeble et al., 2006), but can also activate Tcf/Lef in response to Wnt1 or Wnt3a (W. Lu, Yamamoto, Ortega, & Baltimore, 2004). RYK mutations have been identified in Asian patients with NSCL/P (Watanabe et al., 2006), and Ryk knockout in mouse results in complete secondary cleft palate, though a mechanism has not been described (Halford et al., 2000).

In addition to Fzd and Dvl, the “core” vertebrate PCP proteins include Vangl, Prickle, Celsr, Ankrd6/Dvsn, Scrib, and Ptk7 (Butler & Wallingford, 2017; Jones & Chen, 2007; Strutt, 2008). Variants in both coding and noncoding regions of PRICKLE1 were linked with NSCL/P in a Filipino cohort, and Prickle1-null and -hypomorphic mouse mutants present with Robinow-like traits including CPO (C. Liu et al., 2014; T. Yang et al., 2014), supporting its role as a possible downstream effector of Wnt5a-Ror2-Dvl in mediating secondary palate formation (Figure 5). Fzd2 knockout mice present with CPO at around 50% penetrance, as do compound Fzd2+/−;Fzd7−/−;Vangl2+/Lp (The Vangl2 Lp allele is hypomorphic), which not observed in Fzd2+/−;Fzd7−/− or Fzd7−/−;Vangl2+/Lp, suggesting Vangl2-dependent PCP signaling may be activated through Fzd2 and possibly Fzd7 in secondary palatogenesis (H. Yu, Ye, Guo, & Nathans, 2012). Ca2+/calmodulin-associated serine/threonine kinase (CASK) associates with activated calmodulin and Syndecan-4, which itself interacts with Vangl2 to regulate PCP. Cask mutant mice exhibit highly penetrant CPO, though it was not demonstrated whether this was specifically related to its activity downstream of noncanonical Wnt signaling (Atasoy et al., 2007; Escobedo et al., 2013).

7.5. Wnt signaling effectors and pathway interactions in orofacial clefts

Several Wnt targets are also associated with orofacial clefts, and Wnt regulates important pathways that provide insight into the mechanisms by which Wnt/β-catenin signaling regulates lip and palate formation. Ectodermal Wls cKO results in reduced proliferation and increased apoptosis in the frontal MNP and LNP, with cell adhesion and elongation defects. These embryos showed drastically reduced expression of several Fgfs, including Fgf3, Fgf4, Fgf7, Fgf8, Fgf9, Fgf10 (Zhu et al., 2016). Fgf8 is an important patterning gene in the frontonasal ectoderm and is directly activated by Wnt/β-catenin signaling. Ctnnb1 mutant mice exhibit reduced Fgf8 expression with excessive apoptosis in both the facial epithelium and mesenchyme (Yongping Wang et al., 2011). In Wnt9b mutants, Fgf8, Fgf10, and Fgf17 expression are reduced in the MNP/LNP at E10.5, with Fgf8 particularly affected and diminished at the nasal pit and lamboidal junction at E11.0 (Jin et al., 2012). In turn, Wnt11 expression is modulated by Fgf signaling during secondary palatogenesis and this interaction is required for PS fusion in vitro (J.-M. Lee et al., 2008).

In addition to Fgf, the ectodermal Wls mutants showed reduced expression of Bmp4 and its regulators Msx1/2 (Zhu et al., 2016). While Msx1 is regulated by canonical Wnt signaling, it also directly activates Wnt5a via an enhancer specific to the frontonasal region and palatal shelves, linking the canonical and noncanonical pathways (Nishihara et al., 2016). β-catenin can activate the Bmp4 promoter (Shu et al., 2005), and when Wnt signaling is blocked by ectopic Dkk in the chick MxP, Bmp4 transcript levels are significantly reduced (Shimomura et al., 2019). However, in Lrp6-knockout mouse embryos, Msx1/2 are significantly reduced in the MNP and LNP during lip formation, while Bmp4 is only mildly affected (L. Song et al., 2009). This may be due to differences between the MxP and MNP/LNP or across species, or could reflect an incomplete share in the role of β-catenin activation by Lrp6.

Loss of Tbx1 function in mouse causes excess maxillary-mandibular fusion, resulting in CP with variable severity, while epithelium-specific cKO causes an anterior cleft at the region where the two shelves meet the primary palate (Funato et al., 2012). Tbx1 is repressed by canonical Wnt/β-catenin signaling (Freyer & Morrow, 2010; Huh & Ornitz, 2010) and by RA signaling (Okano et al., 2008) and may represent a point of convergence between the two pathways during facial development. While both pathways inhibit Tbx1, Wnt also represses RA signaling, and in Lrp6-deficient embryos, the RA-synthesizing gene Aldh1a3 is upregulated in the developing lip (L. Song et al., 2009). Though it has not been demonstrated in palate, Ctnnb1 expression is increased in Tbx1-deficient anterior heart field, suggesting a negative feedback loop between canonical Wnt signaling and Tbx1 may be possible (Racedo et al., 2017).

Several other transcription factors associated with CL/P may be directly activated by canonical Wnt signaling. Gbx2 is a direct Wnt target involved in neural crest induction, and Gbx2-null mice sometimes have CP (Byrd & Meyers, 2005; B. Li, Kuriyama, Moreno, & Mayor, 2009). Pitx2-deficient embryos also display CP, and Pitx2 expression is diminished in Ctnnb1;Shh-Cre cKO mutants. The Pitx2 promoter contains WREs and Wnt-activated Pitx2 controls proliferation in palatal mesenchyme through Tgf-β signaling (J.-i. Iwata, Tung, et al., 2012; Kioussi et al., 2002; C. Lin et al., 2011; M. F. Lu, Pressman, Dyer, Johnson, & Martin, 1999). Snai1 and Snai2 (Slug) promote EMT in part by repressing E-cadherin (Cdh1), a binding partner for β-catenin. Snai2-null and compound Snai1+/−;Snai2−/− mutant mice have CP with elevated shelves that fail to adhere and form a MES (Murray, Oram, & Gridley, 2007). Snai1 is targeted by Gsk3-β and β-TrCP, and was shown to be stabilized by canonical Wnt signaling in a manner similar to β-catenin (Yook, Li, Ota, Fearon, & Weiss, 2005), while Snai1/2 in turn reinforce Wnt signaling by promoting β-catenin/Lef1 complex assembly to activate target transcription (Medici, Hay, & Olsen, 2008).

Hoxa2 is required for palate development in mice (T. M. Smith, Wang, Zhang, Kulyk, & Nazarali, 2009), and regulates many targets involved in Wnt signaling (Donaldson et al., 2012). Barx1 is a key palatogenetic regulator that is inhibited by Hoxa2 (T. M. Smith et al., 2009) and was shown to act downstream of Wnt in chick MxP, along with Sox9 and Tbx22 (Shimomura et al., 2019). TBX22 mutations have been linked with NSCL/P and Tbx22 deletion in mice results in a submucous CP with reduced ossification of the palatal mesenchyme (Marçano et al., 2004; Pauws et al., 2009). Tcf/β-catenin can directly activate Tbx3, and while the mechanism is less well defined, Tbx3 mutant mice display full secondary CP (López et al., 2018; Renard et al., 2007). Wnt signaling also directly activates the osteogenic factor Runx2, and Runx2 deletion similarly causes CPO with reduced ossification in oral tissues (Åberg et al., 2004; Gaur et al., 2005). SOX9 is associated with syndromic clefts (Table 2), and Sox9 mutant mice have altered chondrocyte differentiation and ectopic osteogenesis in neural crest lineage cells, resulting in full secondary CPO, reflecting its role as a chondrogenic specifier (Mori-Akiyama, Akiyama, Rowitch, & de Crombrugghe, 2003). Wnt signaling induces Hand2, which represses Sox9 and a chondrogenic fate in favor of an osteogenic one (Abe et al., 2010). Epithelial Hand2 regulates Shh at the MEE independent of Fgf10 and Bmp4, and hypomorphic Hand2 expression results in complete CPO (Xiong et al., 2009).

Compound Pbx1/2 mutants present with fully penetrant CL/P, and Pbx-Prep binding sites may facilitate transactivation at the Wnt3-Wnt9b locus and activation of Wnt/β-catenin activity in the developing maxillary and nasal processes (Ferretti et al., 2011). Wnt9b subsequently regulates Fgf signaling to control mesenchymal proliferation during lip and primary palate formation (Jin et al., 2012). Both Tp63 and Irf6 are important effectors in the developing lamboidal junction between the MNP, LNP, and MxP. Pbx-induced Wnt signaling activates Tp63, which in turn activates Irf6 to promote epithelial apoptosis, and disrupting these interactions results in CL/P (Ferretti et al., 2011). While this sequence was initially identified in mice, a recent study reported gene-gene interactions between several of these factors in association with NSCL/P in human patients, likely demonstrating a conserved mechanism of regulating lip fusion (Maili et al., 2019). Tp63 also represses Bmp and promotes Shh and Fgf signaling, and Tp63-null mouse embryos have bilateral CPO (H. A. Thomason et al., 2008). p63 directly activates Irf6 during palate development, and opposing activities of p63 and Twist activate and repress, respectively, craniofacial Irf6 expression (Fakhouri et al., 2017; Thomason et al., 2010). Irf6 is required for keratinocyte differentiation, and failed PS elevation is among the severe defects exhibited by Irf6-null embryos (Ingraham et al., 2006; R. J. Richardson et al., 2006). Grhl3 is a key target of Irf6 in oral peridermal development (de la Garza et al., 2013), and the human orthologs of both genes are associated with OFCs (Tables 1 & 2).

7.6. Wnt signaling modulators in orofacial clefts

Several classes of protein also modulate Wnt signaling, including the antagonistic Sostdc1, Dkk, and Secreted frizzled-related protein (Sfrp) families and the R-spondins that amplify Wnt signals. RSPO2 mutations are identified in multiple families with tetra-amelia including CL/P, as with WNT3, though it has not been demonstrated whether the characteristics observed in these two sets of patients are caused the same mechanism (Szenker-Ravi et al., 2018). Rspo2-null mice display several craniofacial defects including 75% penetrant cleft palate, likely an indirect effect from abnormal facial structure due to altered patterning of the BA1, rather than defective activity within the palatal shelves themselves (Jin et al., 2011). R-spondins interact with receptors Lgr4, Lgr5, and Lgr6 to inhibit Wnt receptor degradation by Znrf3/Rnf43 ubiquitin ligases (Zebisch et al., 2013). Lgr5/Lgr6 and Lgr4/Lgr5/Lgr6 compound knockout mutants exhibit CPO in which the palatal shelves fail to elevate, which is not observed in Lgr4/Lgr6 knockouts, implying a particular requirement for at least Lgr5 and possibly Lgr6 in secondary palatogenesis (Szenker-Ravi et al., 2018).

While epithelial Ctnnb1 cKO revealed Wnt regulates Tgfb3 in palatogenesis, mouse embryos with epithelial Tgfbr2 cKO have upregulated Dkk1 and Dkk4. Dkk represses Wnt signaling by binding and inhibiting the Lrp co-receptor. These embryos have dysregulated Wnt activity and cleft soft palate with muscular defects, evidencing a reciprocal positive reinforcement between Wnt signaling and Tgf-β in palatal epithelium via Dkk (J.-i. Iwata et al., 2014). Pax9 regulates a network of several important craniofacial regulators, including Osr2, Msx1, Shh, Fgf10, and Bmp4 (J. Zhou, Gao, Lan, Jia, & Jiang, 2013). Pax9 also functions upstream of Wnt and maintains signaling activity in the developing palatal shelves by repressing Dkk. Pax9-deficient mice have secondary cleft palate due to upregulated Dkk1 and Dkk2 expression, which can be rescued with a small-molecule Dkk inhibitor (Jia et al., 2017). Sostdc1 is a target of Shh signaling, and ectopic Smo activation causes cleft palate, with upregulation of Sostdc1 and downregulation of Sox9 and Runx2 (Hammond et al., 2018). Loss of Sostdc1 (which also suppresses Bmp) function similarly rescues the secondary CP, further illustrating a key role for Pax9 in secondary palate is to modulate Wnt (C. Li, Lan, Krumlauf, & Jiang, 2017).

Acknowledgments

This work is supported by grants from the NIH (R01DE026737, R01NS102261, and R01DE021696 to C.J.Z.) and the Shriners Hospitals for Children (85105 to C.J.Z.). We are grateful to the rest of Zhou lab members for their general supports during the manuscript preparation. We apologize to colleagues whose important work we were unable to cite due to space constraints or inadvertently overlooking.

Frequently used lip/palate abbreviations

BA1

First Brachial Arch

BCL

Bilateral Cleft Lip

CLP

Cleft Lip and Cleft Palate

CL/P

Cleft Lip with or without Cleft Palate

CLO

Cleft Lip Only

CP

Cleft Palate

CPO

Cleft Palate Only

EMI

Epithelial-Mesenchymal Interaction

LNP

Lateral Nasal Prominence

MCL

Median Cleft Lip

MEE

Medial Edge Epithelium

MEPM

Mouse Embryonic Palatal Mesenchyme

MES

Medial Epithelial Seam

MNP

Medial Nasal Prominence

MxP

Maxillary Prominence

NS

Nonsyndromic [CL/P, CPO]

OFC

Orofacial Cleft

PS

Palatal Shelf

UCL

Unilateral Cleft Lip

Footnotes

CONFLICT OF INTEREST STATEMENT

The authors declare no conflict of interest.

DATA SHARING AND DATA ACCESSIBILITY

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

References

  1. Abdel-Hamid MS, Issa MY, Otaify GA, Abdel-Ghafar SF, Elbendary HM, & Zaki MS (2018). PGAP3-related hyperphosphatasia with mental retardation syndrome: Report of 10 new patients and a homozygous founder mutation. Clin Genet, 93(1), 84–91. doi: 10.1111/cge.13033 [DOI] [PubMed] [Google Scholar]
  2. Abe M, Michikami I, Fukushi T, Abe A, Maeda Y, Ooshima T, & Wakisaka S (2010). Hand2 regulates chondrogenesis in vitro and in vivo. Bone, 46(5), 1359–1368. doi: 10.1016/j.bone.2009.11.022 [DOI] [PubMed] [Google Scholar]
  3. Åberg T, Cavender A, Gaikwad JS, Bronckers ALJJ, Wang X, Waltimo-Sirén J, … D’Souza RN (2004). Phenotypic Changes in Dentition of Runx2 Homozygote-null Mutant Mice. Journal of Histochemistry & Cytochemistry, 52(1), 131–139. doi: 10.1177/002215540405200113 10.1177/002215540405200113 [DOI] [PubMed] [Google Scholar]
  4. Abramyan J (2019). Hedgehog Signaling and Embryonic Craniofacial Disorders. Journal of developmental biology, 7(2), 9. doi: 10.3390/jdb7020009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Adamska M, Degnan SM, Green KM, Adamski M, Craigie A, Larroux C, & Degnan BM (2007). Wnt and TGF-β Expression in the Sponge Amphimedon queenslandica and the Origin of Metazoan Embryonic Patterning. PLOS ONE, 2(10), e1031. doi: 10.1371/journal.pone.0001031 [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Adly N, Alhashem A, Ammari A, & Alkuraya FS (2014). Ciliary Genes TBC1D32/C6orf170 and SCLT1 are Mutated in Patients with OFD Type IX. Human Mutation, 35(1), 36–40. doi: 10.1002/humu.22477 [DOI] [PubMed] [Google Scholar]
  7. Aguilella C, Dubourg C, Attia-Sobol J, Vigneron J, Blayau M, Pasquier L, … David V (2003). Molecular screening of the TGIF gene in holoprosencephaly: identification of two novel mutations. Hum Genet, 112(2), 131–134. doi: 10.1007/s00439-002-0862-8 [DOI] [PubMed] [Google Scholar]
  8. Ahlgren SC, & Bronner-Fraser M (1999). Inhibition of sonic hedgehog signaling in vivo results in craniofacial neural crest cell death. Curr Biol, 9(22), 1304–1314. doi: 10.1016/s0960-9822(00)80052-4 [DOI] [PubMed] [Google Scholar]
  9. Ahn Y, Sanderson BW, Klein OD, & Krumlauf R (2010). Inhibition of Wnt signaling by Wise (Sostdc1) and negative feedback from Shh controls tooth number and patterning. Development (Cambridge, England), 137(19), 3221–3231. doi: 10.1242/dev.054668 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Alappat SR, Zhang Z, Suzuki K, Zhang X, Liu H, Jiang R, … Chen Y (2005). The cellular and molecular etiology of the cleft secondary palate in Fgf10 mutant mice. Developmental biology, 277(1), 102–113. doi: 10.1016/j.ydbio.2004.09.010 [DOI] [PubMed] [Google Scholar]
  11. Alisch F, Weichert A, Kalache K, Paradiso V, Longardt AC, Dame C, … Horn D (2017). Familial Gordon syndrome associated with a PIEZO2 mutation. Am J Med Genet A, 173(1), 254–259. doi: 10.1002/ajmg.a.37997 [DOI] [PubMed] [Google Scholar]
  12. Altassan R, Fox S, Poulin C, & Buhas D (2018). Hyperphosphatasia with mental retardation syndrome, expanded phenotype of PIGL related disorders. Molecular genetics and metabolism reports, 15, 46–49. doi: 10.1016/j.ymgmr.2018.01.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Alzahrani F, Al Hazzaa SA, Tayeb H, & Alkuraya FS (2015). LOXL3, encoding lysyl oxidase-like 3, is mutated in a family with autosomal recessive Stickler syndrome. Hum Genet, 134(4), 451–453. doi: 10.1007/s00439-015-1531-z [DOI] [PubMed] [Google Scholar]
  14. Amano K, Ishiguchi M, Aikawa T, Kimata M, Kishi N, Fujimaki T, … Kogo M (2012). Cleft lip in oculodentodigital dysplasia suggests novel roles for connexin43. J Dent Res, 91(7 Suppl), 38s–44s. doi: 10.1177/0022034512447952 [DOI] [PubMed] [Google Scholar]
  15. Andelfinger G, Tapper AR, Welch RC, Vanoye CG, George AL Jr., & Benson DW (2002). KCNJ2 mutation results in Andersen syndrome with sex-specific cardiac and skeletal muscle phenotypes. American journal of human genetics, 71(3), 663–668. doi: 10.1086/342360 [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Anderson PJ, Snell B, & Moore MH (2013). Muencke syndrome with cleft lip and palate. J Craniofac Surg, 24(4), 1484–1485. doi: 10.1097/SCS.0b013e31829035c3 [DOI] [PubMed] [Google Scholar]
  17. Andersson HC, Kratz L, & Kelley R (2002). Desmosterolosis presenting with multiple congenital anomalies and profound developmental delay. Am J Med Genet, 113(4), 315–319. doi: 10.1002/ajmg.b.10873 [DOI] [PubMed] [Google Scholar]
  18. Ang HL, Deltour L, Hayamizu TF, Žgombic-Knight M, & Duester G (1996). Retinoic Acid Synthesis in Mouse Embryos during Gastrulation and Craniofacial Development Linked to Class IV Alcohol Dehydrogenase Gene Expression. Journal of Biological Chemistry, 271(16), 9526–9534. doi: 10.1074/jbc.271.16.9526 [DOI] [PubMed] [Google Scholar]
  19. Antunes LS, Küchler EC, Tannure PN, Costa MC, Gouvêa CVD, Olej B, & Granjeiro JM (2013). BMP4 Polymorphism is Associated with Nonsyndromic Oral Cleft in a Brazilian Population. The Cleft Palate-Craniofacial Journal, 50(6), 633–638. doi: 10.1597/12-048 10.1597/12–048 [DOI] [PubMed] [Google Scholar]
  20. Ashique AM, Fu K, & Richman JM (2002). Endogenous bone morphogenetic proteins regulate outgrowth and epithelial survival during avian lip fusion. Development, 129(19), 4647 Retrieved from http://dev.biologists.org/content/129/19/4647.abstract [DOI] [PubMed] [Google Scholar]
  21. Atasoy D, Schoch S, Ho A, Nadasy KA, Liu X, Zhang W, … Südhof TC (2007). Deletion of CASK in mice is lethal and impairs synaptic function. Proceedings of the National Academy of Sciences of the United States of America, 104(7), 2525–2530. doi: 10.1073/pnas.0611003104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Bachler M, & Neubuser A (2001). Expression of members of the Fgf family and their receptors during midfacial development. Mech Dev, 100(2), 313–316. doi: 10.1016/s0925-4773(00)00518-9 [DOI] [PubMed] [Google Scholar]
  23. Baek J-A, Lan Y, Liu H, Maltby KM, Mishina Y, & Jiang R (2011). Bmpr1a signaling plays critical roles in palatal shelf growth and palatal bone formation. Developmental Biology, 350(2), 520–531. doi: 10.1016/j.ydbio.2010.12.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Bänziger C, Soldini D, Schütt C, Zipperlen P, Hausmann G, & Basler K (2006). Wntless, a Conserved Membrane Protein Dedicated to the Secretion of Wnt Proteins from Signaling Cells. Cell, 125(3), 509–522. doi: 10.1016/j.cell.2006.02.049 [DOI] [PubMed] [Google Scholar]
  25. Barrow LL, van Bokhoven H, Daack-Hirsch S, Andersen T, van Beersum SEC, Gorlin R, & Murray JC (2002). Analysis of the p63 gene in classical EEC syndrome, related syndromes, and non-syndromic orofacial clefts. Journal of medical genetics, 39(8), 559–566. doi: 10.1136/jmg.39.8.559 [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Basha M, Demeer B, Revencu N, Helaers R, Theys S, Bou Saba S, … Vikkula M (2018). 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. Journal of medical genetics, 55(7), 449–458. doi: 10.1136/jmedgenet-2017-105110 [DOI] [PubMed] [Google Scholar]
  27. Baumann M, Giunta C, Krabichler B, Ruschendorf F, Zoppi N, Colombi M, … Fauth C (2012). Mutations in FKBP14 cause a variant of Ehlers-Danlos syndrome with progressive kyphoscoliosis, myopathy, and hearing loss. American journal of human genetics, 90(2), 201–216. doi: 10.1016/j.ajhg.2011.12.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Beaty TH, Hetmanski JB, Fallin MD, Park JW, Sull JW, McIntosh I, … Scott AF (2006). Analysis of candidate genes on chromosome 2 in oral cleft case-parent trios from three populations. Human Genetics, 120(4), 501–518. doi: 10.1007/s00439-006-0235-9 [DOI] [PubMed] [Google Scholar]
  29. Beaty TH, Murray JC, Marazita ML, Munger RG, Ruczinski I, Hetmanski JB, … Scott AF (2010). A genome-wide association study of cleft lip with and without cleft palate identifies risk variants near MAFB and ABCA4. Nature Genetics, 42(6), 525–529. doi: 10.1038/ng.580 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Beaty TH, Ruczinski I, Murray JC, Marazita ML, Munger RG, Hetmanski JB, … Scott AF (2011). Evidence for gene-environment interaction in a genome wide study of nonsyndromic cleft palate. Genetic epidemiology, 35(6), 469–478. doi: 10.1002/gepi.20595 [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Beaty TH, Taub MA, Scott AF, Murray JC, Marazita ML, Schwender H, … Ruczinski I (2013). Confirming genes influencing risk to cleft lip with/without cleft palate in a case-parent trio study. Hum Genet, 132(7), 771–781. doi: 10.1007/s00439-013-1283-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Beiraghi S, Zhou M, Talmadge CB, Went-Sumegi N, Davis JR, Huang D, … Sumegi J (2003). Identification and characterization of a novel gene disrupted by a pericentric inversion inv(4)(p13.1q21.1) in a family with cleft lip. Gene, 309(1), 11–21. doi: 10.1016/S0378-1119(03)00461-X [DOI] [PubMed] [Google Scholar]
  33. Benko S, Fantes JA, Amiel J, Kleinjan DJ, Thomas S, Ramsay J, … Lyonnet S (2009). Highly conserved non-coding elements on either side of SOX9 associated with Pierre Robin sequence. Nat Genet, 41(3), 359–364. doi: 10.1038/ng.329 [DOI] [PubMed] [Google Scholar]
  34. Bicknell LS, Farrington-Rock C, Shafeghati Y, Rump P, Alanay Y, Alembik Y, … Robertson SP (2007). A molecular and clinical study of Larsen syndrome caused by mutations in FLNB. Journal of medical genetics, 44(2), 89–98. doi: 10.1136/jmg.2006.043687 [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Birnbaum S, Ludwig KU, Reutter H, Herms S, Steffens M, Rubini M, … Mangold E (2009). Key susceptibility locus for nonsyndromic cleft lip with or without cleft palate on chromosome 8q24. Nat Genet, 41(4), 473–477. doi: 10.1038/ng.333 [DOI] [PubMed] [Google Scholar]
  36. Blanton SH, Cortez A, Stal S, Mulliken JB, Finnell RH, & Hecht JT (2005). Variation in IRF6 contributes to nonsyndromic cleft lip and palate. American Journal of Medical Genetics Part A, 137A(3), 259–262. doi: 10.1002/ajmg.a.30887 [DOI] [PubMed] [Google Scholar]
  37. Bonafé L, Hästbacka J, de la Chapelle A, Campos-Xavier AB, Chiesa C, Forlino A, … Rossi A (2008). A novel mutation in the sulfate transporter gene SLC26A2 (DTDST) specific to the Finnish population causes de la Chapelle dysplasia. Journal of medical genetics, 45(12), 827–831. doi: 10.1136/jmg.2007.057158 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Bornholdt D, Oeffner F, Konig A, Happle R, Alanay Y, Ascherman J, … Grzeschik KH (2009). PORCN mutations in focal dermal hypoplasia: coping with lethality. Hum Mutat, 30(5), E618–628. doi: 10.1002/humu.20992 [DOI] [PubMed] [Google Scholar]
  39. Briggs MD, & Chapman KL (2002). Pseudoachondroplasia and multiple epiphyseal dysplasia: Mutation review, molecular interactions, and genotype to phenotype correlations. Human Mutation, 19(5), 465–478. doi: 10.1002/humu.10066 [DOI] [PubMed] [Google Scholar]
  40. Briscoe J, & Thérond PP (2013). The mechanisms of Hedgehog signalling and its roles in development and disease. Nature Reviews Molecular Cell Biology, 14, 416 Retrieved from 10.1038/nrm3598 [DOI] [PubMed] [Google Scholar]
  41. Budi EH, Duan D, & Derynck R (2017). Transforming Growth Factor-β Receptors and Smads: Regulatory Complexity and Functional Versatility. Trends in Cell Biology, 27(9), 658–672. doi: 10.1016/j.tcb.2017.04.005 [DOI] [PubMed] [Google Scholar]
  42. Bufalino A, Ribeiro Paranaiba LM, Nascimento de Aquino S, Martelli-Junior H, Oliveira Swerts MS, & Coletta RD (2010). Maternal polymorphisms in folic acid metabolic genes are associated with nonsyndromic cleft lip and/or palate in the Brazilian population. Birth Defects Res A Clin Mol Teratol, 88(11), 980–986. doi: 10.1002/bdra.20732 [DOI] [PubMed] [Google Scholar]
  43. Buglino JA, & Resh MD (2008). Hhat is a palmitoylacyltransferase with specificity for N-palmitoylation of Sonic Hedgehog. The Journal of biological chemistry, 283(32), 22076–22088. doi: 10.1074/jbc.M803901200 [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Bunn Kieran J., Daniel P, Rösken Heleen S., O’Neill Adam C., Cameron-Christie Sophia R., Morgan T, … Robertson Stephen P. (2015). Mutations in DVL1 Cause an Osteosclerotic Form of Robinow Syndrome. The American Journal of Human Genetics, 96(4), 623–630. doi: 10.1016/j.ajhg.2015.02.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Butali A, Mossey P, Adeyemo W, Eshete M, Gaines L, Braimah R, … Murray J (2014). Rare functional variants in genome–wide association identified candidate genes for nonsyndromic clefts in the African population. American Journal of Medical Genetics Part A, 164(10), 2567–2571. doi: 10.1002/ajmg.a.36691 [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Butali A, Mossey PA, Adeyemo WL, Eshete MA, Gowans LJJ, Busch TD, … Adeyemo AA (2019). Genomic analyses in African populations identify novel risk loci for cleft palate. Hum Mol Genet, 28(6), 1038–1051. doi: 10.1093/hmg/ddy402 [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Butali A, Suzuki S, Cooper ME, Mansilla AM, Cuenco K, Leslie EJ, … Murray JC (2013). 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, 161a(5), 965–972. doi: 10.1002/ajmg.a.35749 [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Butler MT, & Wallingford JB (2017). Planar cell polarity in development and disease. Nature reviews. Molecular cell biology, 18(6), 375–388. doi: 10.1038/nrm.2017.11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Byrd NA, & Meyers EN (2005). Loss of Gbx2 results in neural crest cell patterning and pharyngeal arch artery defects in the mouse embryo. Developmental Biology, 284(1), 233–245. doi: 10.1016/j.ydbio.2005.05.023 [DOI] [PubMed] [Google Scholar]
  50. Byrne S, Jansen L, U-King-Im J-M, Siddiqui A, Lidov HGW, Bodi I, … Jungbluth H (2016). EPG5-related Vici syndrome: a paradigm of neurodevelopmental disorders with defective autophagy. Brain : a journal of neurology, 139(Pt 3), 765–781. doi: 10.1093/brain/awv393 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Cadigan KM, & Waterman ML (2012). TCF/LEFs and Wnt signaling in the nucleus. Cold Spring Harbor Perspectives in Biology, 4(11), a007906. doi: 10.1101/cshperspect.a007906 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Caputo V, Cianetti L, Niceta M, Carta C, Ciolfi A, Bocchinfuso G, … Tartaglia M (2012). A restricted spectrum of mutations in the SMAD4 tumor-suppressor gene underlies Myhre syndrome. American journal of human genetics, 90(1), 161–169. doi: 10.1016/j.ajhg.2011.12.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Carre A, Hamza RT, Kariyawasam D, Guillot L, Teissier R, Tron E, … Polak M (2014). A novel FOXE1 mutation (R73S) in Bamforth-Lazarus syndrome causing increased thyroidal gene expression. Thyroid, 24(4), 649–654. doi: 10.1089/thy.2013.0417 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Carroll TJ, Park J-S, Hayashi S, Majumdar A, & McMahon AP (2005). Wnt9b Plays a Central Role in the Regulation of Mesenchymal to Epithelial Transitions Underlying Organogenesis of the Mammalian Urogenital System. Developmental Cell, 9(2), 283–292. doi: 10.1016/j.devcel.2005.05.016 [DOI] [PubMed] [Google Scholar]
  55. Carter MT, St Pierre SA, Zackai EH, Emanuel BS, & Boycott KM (2009). Phenotypic delineation of Emanuel syndrome (supernumerary derivative 22 syndrome): Clinical features of 63 individuals. American journal of medical genetics. Part A, 149A(8), 1712–1721. doi: 10.1002/ajmg.a.32957 [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Celli J, Duijf P, Hamel BC, Bamshad M, Kramer B, Smits AP, … van Bokhoven H (1999). Heterozygous germline mutations in the p53 homolog p63 are the cause of EEC syndrome. Cell, 99(2), 143–153. doi: 10.1016/s0092-8674(00)81646-3 [DOI] [PubMed] [Google Scholar]
  57. Charoenchaikorn K, Yokomizo T, Rice DP, Honjo T, Matsuzaki K, Shintaku Y, … Yamashiro T (2009). Runx1 is involved in the fusion of the primary and the secondary palatal shelves. Developmental biology, 326(2), 392–402. doi: 10.1016/j.ydbio.2008.10.018 [DOI] [PubMed] [Google Scholar]
  58. Chen M-H, Li Y-J, Kawakami T, Xu S-M, & Chuang P-T (2004). Palmitoylation is required for the production of a soluble multimeric Hedgehog protein complex and long-range signaling in vertebrates. Genes & Development, 18(6), 641–659. doi: 10.1101/gad.1185804 [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Chen Q, Wang H, Schwender H, Zhang T, Hetmanski JB, Chou Y-HW, … Beaty TH (2014). Joint testing of genotypic and gene-environment interaction identified novel association for BMP4 with non-syndromic CL/P in an Asian population using data from an International Cleft Consortium. PLOS ONE, 9(10), e109038–e109038. doi: 10.1371/journal.pone.0109038 [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Chen Y, Wang Z, Chen Y, & Zhang Y (2019). Conditional deletion of Bmp2 in cranial neural crest cells recapitulates Pierre Robin sequence in mice. Cell and tissue research, 376(2), 199–210. doi: 10.1007/s00441-018-2944-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Chia IV, Kim MJ, Itoh K, Sokol SY, & Costantini F (2009). Both the RGS Domain and the Six C-Terminal Amino Acids of Mouse Axin Are Required for Normal Embryogenesis. Genetics, 181(4), 1359. doi: 10.1534/genetics.109.101055 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Chiang C, Litingtung Y, Lee E, Young KE, Corden JL, Westphal H, & Beachy PA (1996). Cyclopia and defective axial patterning in mice lacking Sonic hedgehog gene function. Nature, 383(6599), 407–413. doi: 10.1038/383407a0 [DOI] [PubMed] [Google Scholar]
  63. Chiquet BT, Blanton SH, Burt A, Ma D, Stal S, Mulliken JB, & Hecht JT (2008). Variation in WNT genes is associated with non-syndromic cleft lip with or without cleft palate. Human Molecular Genetics, 17(14), 2212–2218. doi: 10.1093/hmg/ddn121 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Chiquet BT, Blanton SH, Burt A, Ma D, Stal S, Mulliken JB, & Hecht JT (2008). Variation in WNT genes is associated with non-syndromic cleft lip with or without cleft palate. Hum Mol Genet, 17(14), 2212–2218. doi: 10.1093/hmg/ddn121 [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Chong JX, Burrage LC, Beck AE, Marvin CT, McMillin MJ, Shively KM, … Bamshad MJ (2015). Autosomal-Dominant Multiple Pterygium Syndrome Is Caused by Mutations in MYH3. American journal of human genetics, 96(5), 841–849. doi: 10.1016/j.ajhg.2015.04.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Clevers H, & Nusse R (2012). Wnt/β-Catenin Signaling and Disease. Cell, 149(6), 1192–1205. doi: 10.1016/j.cell.2012.05.012 [DOI] [PubMed] [Google Scholar]
  67. Cliffe A, Hamada F, & Bienz M (2003). A Role of Dishevelled in Relocating Axin to the Plasma Membrane during Wingless Signaling. Current Biology, 13(11), 960–966. doi: 10.1016/S0960-9822(03)00370-1 [DOI] [PubMed] [Google Scholar]
  68. Clifton-Bligh RJ, Wentworth JM, Heinz P, Crisp MS, John R, Lazarus JH, … Chatterjee VK (1998). Mutation of the gene encoding human TTF-2 associated with thyroid agenesis, cleft palate and choanal atresia. Nat Genet, 19(4), 399–401. doi: 10.1038/1294 [DOI] [PubMed] [Google Scholar]
  69. Cobourne MT, Xavier GM, Depew M, Hagan L, Sealby J, Webster Z, & Sharpe PT (2009). Sonic hedgehog signalling inhibits palatogenesis and arrests tooth development in a mouse model of the nevoid basal cell carcinoma syndrome. Developmental Biology, 331(1), 38–49. doi: 10.1016/j.ydbio.2009.04.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Colvin JS, White AC, Pratt SJ, & Ornitz DM (2001). Lung hypoplasia and neonatal death in Fgf9-null mice identify this gene as an essential regulator of lung mesenchyme. Development, 128(11), 2095–2106. Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/11493531 [DOI] [PubMed] [Google Scholar]
  71. Cong W, Liu B, Liu S, Sun M, Liu H, Yang Y, … Xiao J (2014). Implications of the Wnt5a/CaMKII Pathway in Retinoic Acid-Induced Myogenic Tongue Abnormalities of Developing Mice. Scientific Reports, 4, 6082 Retrieved from 10.1038/srep06082 [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Cunningham TJ, & Duester G (2015). Mechanisms of retinoic acid signalling and its roles in organ and limb development. Nature reviews. Molecular cell biology, 16(2), 110–123. doi: 10.1038/nrm3932 [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Cvjetkovic N, Maili L, Weymouth KS, Hashmi SS, Mulliken JB, Topczewski J, … Hecht JT (2015). Regulatory variant in FZD6 gene contributes to nonsyndromic cleft lip and palate in an African-American family. Molecular Genetics & Genomic Medicine, 3(5), 440–451. doi: 10.1002/mgg3.155 [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Czeschik JC, Voigt C, Alanay Y, Albrecht B, Avci S, Fitzpatrick D, … Wieczorek D (2013). Clinical and mutation data in 12 patients with the clinical diagnosis of Nager syndrome. Hum Genet, 132(8), 885–898. doi: 10.1007/s00439-013-1295-2 [DOI] [PubMed] [Google Scholar]
  75. Da-Silva EO (1991). Waardenburg I syndrome: A clinical and genetic study of two large Brazilian kindreds, and literature review. American Journal of Medical Genetics, 40(1), 65–74. doi: 10.1002/ajmg.1320400113 [DOI] [PubMed] [Google Scholar]
  76. Dabdoub A, Donohue MJ, Brennan A, Wolf V, Montcouquiol M, Sassoon DA, … Kelley MW (2003). Wnt signaling mediates reorientation of outer hair cell stereociliary bundles in the mammalian cochlea. Development, 130(11), 2375. doi: 10.1242/dev.00448 [DOI] [PubMed] [Google Scholar]
  77. Das BC, Thapa P, Karki R, Das S, Mahapatra S, Liu T-C, … Evans T (2014). Retinoic acid signaling pathways in development and diseases. Bioorganic & Medicinal Chemistry, 22(2), 673–683. doi: 10.1016/j.bmc.2013.11.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Dastot-Le Moal F, Wilson M, Mowat D, Collot N, Niel F, & Goossens M (2007). ZFHX1B mutations in patients with Mowat-Wilson syndrome. Human Mutation, 28(4), 313–321. doi: 10.1002/humu.20452 [DOI] [PubMed] [Google Scholar]
  79. Dauwerse JG, Dixon J, Seland S, Ruivenkamp CA, van Haeringen A, Hoefsloot LH, … Wieczorek D (2011). Mutations in genes encoding subunits of RNA polymerases I and III cause Treacher Collins syndrome. Nat Genet, 43(1), 20–22. doi: 10.1038/ng.724 [DOI] [PubMed] [Google Scholar]
  80. Davoody A, Chen IP, Nanda R, Uribe F, & Reichenberger EJ (2012). Oculofaciocardiodental syndrome: a rare case and review of the literature. The Cleft palate-craniofacial journal : official publication of the American Cleft Palate-Craniofacial Association, 49(5), e55–e60. doi: 10.1597/10-256 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. De A (2011). Wnt/Ca2+ signaling pathway: a brief overview. Acta Biochimica et Biophysica Sinica, 43(10), 745–756. doi: 10.1093/abbs/gmr079 [DOI] [PubMed] [Google Scholar]
  82. de Araujo TK, Secolin R, Felix TM, de Souza LT, Fontes MI, Monlleo IL, … Gil-da-Silva-Lopes VL (2016). A multicentric association study between 39 genes and nonsyndromic cleft lip and palate in a Brazilian population. J Craniomaxillofac Surg, 44(1), 16–20. doi: 10.1016/j.jcms.2015.07.026 [DOI] [PubMed] [Google Scholar]
  83. de la Garza G, Schleiffarth JR, Dunnwald M, Mankad A, Weirather JL, Bonde G, … Cornell RA (2013). Interferon Regulatory Factor 6 Promotes Differentiation of the Periderm by Activating Expression of Grainyhead-Like 3. Journal of Investigative Dermatology, 133(1), 68–77. doi: 10.1038/jid.2012.269 [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. De Moerlooze L, Spencer-Dene B, Revest JM, Hajihosseini M, Rosewell I, & Dickson C (2000). An important role for the IIIb isoform of fibroblast growth factor receptor 2 (FGFR2) in mesenchymal-epithelial signalling during mouse organogenesis. Development, 127(3), 483–492. Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/1063116910631169 [Google Scholar]
  85. Delous M, Baala L, Salomon R, Laclef C, Vierkotten J, Tory K, … Saunier S (2007). The ciliary gene RPGRIP1L is mutated in cerebello-oculo-renal syndrome (Joubert syndrome type B) and Meckel syndrome. Nature Genetics, 39(7), 875–881. doi: 10.1038/ng2039 [DOI] [PubMed] [Google Scholar]
  86. Dennis JF, Kurosaka H, Iulianella A, Pace J, Thomas N, Beckham S, … Trainor PA (2012). Mutations in Hedgehog acyltransferase (Hhat) perturb Hedgehog signaling, resulting in severe acrania-holoprosencephaly-agnathia craniofacial defects. PLOS Genetics, 8(10), e1002927–e1002927. doi: 10.1371/journal.pgen.1002927 [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Di Donato N, Rump A, Koenig R, Der Kaloustian VM, Halal F, Sonntag K, … Verloes A (2014). Severe forms of Baraitser-Winter syndrome are caused by ACTB mutations rather than ACTG1 mutations. Eur J Hum Genet, 22(2), 179–183. doi: 10.1038/ejhg.2013.130 [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Di Gioia SA, Connors S, Matsunami N, Cannavino J, Rose MF, Gilette NM, … Engle EC (2017). A defect in myoblast fusion underlies Carey-Fineman-Ziter syndrome. Nature communications, 8, 16077. doi: 10.1038/ncomms16077 [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Dode C, Levilliers J, Dupont JM, De Paepe A, Le Du N, Soussi-Yanicostas N, … Hardelin JP (2003). Loss-of-function mutations in FGFR1 cause autosomal dominant Kallmann syndrome. Nat Genet, 33(4), 463–465. doi: 10.1038/ng1122 [DOI] [PubMed] [Google Scholar]
  90. Donaldson IJ, Amin S, Hensman JJ, Kutejova E, Rattray M, Lawrence N, … Bobola N (2012). Genome-wide occupancy links Hoxa2 to Wnt-β-catenin signaling in mouse embryonic development. Nucleic acids research, 40(9), 3990–4001. doi: 10.1093/nar/gkr1240 [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Dong D, Ruuska SE, Levinthal DJ, & Noy N (1999). Distinct Roles for Cellular Retinoic Acid-binding Proteins I and II in Regulating Signaling by Retinoic Acid. Journal of Biological Chemistry, 274(34), 23695–23698. doi: 10.1074/jbc.274.34.23695 [DOI] [PubMed] [Google Scholar]
  92. Dudas M, Kim J, Li W-Y, Nagy A, Larsson J, Karlsson S, … Kaartinen V (2006). Epithelial and ectomesenchymal role of the type I TGF-beta receptor ALK5 during facial morphogenesis and palatal fusion. Developmental Biology, 296(2), 298–314. doi: 10.1016/j.ydbio.2006.05.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Dudas M, Sridurongrit S, Nagy A, Okazaki K, & Kaartinen V (2004). Craniofacial defects in mice lacking BMP type I receptor Alk2 in neural crest cells. Mechanisms of Development, 121(2), 173–182. doi: 10.1016/j.mod.2003.12.003 [DOI] [PubMed] [Google Scholar]
  94. Duester G (2008). Retinoic acid synthesis and signaling during early organogenesis. Cell, 134(6), 921–931. doi: 10.1016/j.cell.2008.09.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Dupé V, Matt N, Garnier J-M, Chambon P, Mark M, & Ghyselinck NB (2003). A newborn lethal defect due to inactivation of retinaldehyde dehydrogenase type 3 is prevented by maternal retinoic acid treatment. Proceedings of the National Academy of Sciences, 100(24), 14036. doi: 10.1073/pnas.2336223100 [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Economou AD, Ohazama A, Porntaveetus T, Sharpe PT, Kondo S, Basson MA, … Green JBA (2012). Periodic stripe formation by a Turing mechanism operating at growth zones in the mammalian palate. Nature Genetics, 44(3), 348–351. doi: 10.1038/ng.1090 [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Ehmke N, Caliebe A, Koenig R, Kant SG, Stark Z, Cormier-Daire V, … Mundlos S (2014). Homozygous and compound-heterozygous mutations in TGDS cause Catel-Manzke syndrome. American journal of human genetics, 95(6), 763–770. doi: 10.1016/j.ajhg.2014.11.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. El Shahawy M, Reibring C-G, Hallberg K, Neben CL, Marangoni P, Harfe BD, … Gritli-Linde A (2019). Sonic Hedgehog Signaling Is Required for Cyp26 Expression during Embryonic Development. International journal of molecular sciences, 20(9), 2275. doi: 10.3390/ijms20092275 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Escobedo N, Contreras O, Munoz R, Farias M, Carrasco H, Hill C, … Larrain J (2013). Syndecan 4 interacts genetically with Vangl2 to regulate neural tube closure and planar cell polarity. Development (Cambridge, England), 140(14), 3008–3017. doi: 10.1242/dev.091173 [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Everson JL, Fink DM, Yoon JW, Leslie EJ, Kietzman HW, Ansen-Wilson LJ, … Lipinski RJ (2017). Sonic hedgehog regulation of Foxf2 promotes cranial neural crest mesenchyme proliferation and is disrupted in cleft lip morphogenesis. Development (Cambridge, England), 144(11), 2082. doi: 10.1242/dev.149930 [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Fakhouri WD, Metwalli K, Naji A, Bakhiet S, Quispe-Salcedo A, Nitschke L, … Schutte BC (2017). Intercellular Genetic Interaction Between Irf6 and Twist1 during Craniofacial Development. Scientific Reports, 7(1), 7129. doi: 10.1038/s41598-017-06310-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Falardeau J, Chung WCJ, Beenken A, Raivio T, Plummer L, Sidis Y, … Pitteloud N (2008). Decreased FGF8 signaling causes deficiency of gonadotropin-releasing hormone in humans and mice. The Journal of clinical investigation, 118(8), 2822–2831. doi: 10.1172/JCI34538 [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Fan GZ, Li YL, & Wu PA (2007). [Association between retinoic acid receptor alpha gene polymorphisms and nonsyndromic cleft lip with or without cleft palate susceptibility]. Zhonghua Yi Xue Za Zhi, 87(6), 396–398. [PubMed] [Google Scholar]
  104. Favaro FP, Alvizi L, Zechi-Ceide RM, Bertola D, Felix TM, de Souza J, … Passos-Bueno MR (2014). A noncoding expansion in EIF4A3 causes Richieri-Costa-Pereira syndrome, a craniofacial disorder associated with limb defects. American journal of human genetics, 94(1), 120–128. doi: 10.1016/j.ajhg.2013.11.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Favaro FP, Zechi-Ceide RM, Alvarez CW, Maximino LP, Antunes LFBB, Richieri-Costa A, & Guion-Almeida ML (2011). Richieri-Costa–Pereira syndrome: A unique acrofacial dysostosis type. An overview of the Brazilian cases. American Journal of Medical Genetics Part A, 155(2), 322–331. doi: 10.1002/ajmg.a.33806 [DOI] [PubMed] [Google Scholar]
  106. Feng C, Xu Z, Li Z, Zhang D, Liu Q, & Lu L (2013). Down-regulation of Wnt10a by RNA interference inhibits proliferation and promotes apoptosis in mouse embryonic palatal mesenchymal cells through Wnt/beta-catenin signaling pathway. Journal of physiology and biochemistry, 69(4), 855–863. doi: 10.1007/s13105-013-0262-7 [DOI] [PubMed] [Google Scholar]
  107. Ferone G, Thomason HA, Antonini D, De Rosa L, Hu B, Gemei M, … Missero C (2012). Mutant p63 causes defective expansion of ectodermal progenitor cells and impaired FGF signalling in AEC syndrome. EMBO Mol Med, 4(3), 192–205. doi: 10.1002/emmm.201100199 [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Ferrante MI, Giorgio G, Feather SA, Bulfone A, Wright V, Ghiani M, … Franco B (2001). Identification of the gene for oral-facial-digital type I syndrome. American journal of human genetics, 68(3), 569–576. doi: 10.1086/318802 [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Ferretti E, Li B, Zewdu R, Wells V, Hebert Jean M., Karner C, … Selleri L (2011). A Conserved Pbx-Wnt-p63-Irf6 Regulatory Module Controls Face Morphogenesis by Promoting Epithelial Apoptosis. Developmental Cell, 21(4), 627–641. doi: 10.1016/j.devcel.2011.08.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Fitzky BU, Witsch-Baumgartner M, Erdel M, Lee JN, Paik YK, Glossmann H, … Moebius FF (1998). Mutations in the Delta7-sterol reductase gene in patients with the Smith-Lemli-Opitz syndrome. Proceedings of the National Academy of Sciences of the United States of America, 95(14), 8181–8186. doi: 10.1073/pnas.95.14.8181 [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Fontoura C, Silva RM, Granjeiro JM, & Letra A (2012). Further evidence of association of the ABCA4 gene with cleft lip/palate. European Journal of Oral Sciences, 120(6), 553–557. doi: 10.1111/eos.12001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Fontoura C, Silva RM, Granjeiro JM, & Letra A (2015). Association of WNT9B Gene Polymorphisms with Nonsyndromic Cleft Lip with or Without Cleft Palate in Brazilian Nuclear Families. The Cleft Palate-Craniofacial Journal, 52(1), 44–48. doi: 10.1597/13-146 10.1597/13–146 [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Foppiano S, Hu D, & Marcucio RS (2007). Signaling by bone morphogenetic proteins directs formation of an ectodermal signaling center that regulates craniofacial development. Developmental Biology, 312(1), 103–114. doi: 10.1016/j.ydbio.2007.09.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Foster JW, Dominguez-Steglich MA, Guioli S, Kwok C, Weller PA, Stevanovic M, … et al. (1994). Campomelic dysplasia and autosomal sex reversal caused by mutations in an SRY-related gene. Nature, 372(6506), 525–530. doi: 10.1038/372525a0 [DOI] [PubMed] [Google Scholar]
  115. Freyer L, & Morrow BE (2010). Canonical Wnt signaling modulates Tbx1, Eya1, and Six1 expression, restricting neurogenesis in the otic vesicle. Developmental dynamics : an official publication of the American Association of Anatomists, 239(6), 1708–1722. doi: 10.1002/dvdy.22308 [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Funato N, Nakamura M, Richardson JA, Srivastava D, & Yanagisawa H (2012). Tbx1 regulates oral epithelial adhesion and palatal development. Hum Mol Genet, 21(11), 2524–2537. doi: 10.1093/hmg/dds071 [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Furuta Y, Piston DW, & Hogan BL (1997). Bone morphogenetic proteins (BMPs) as regulators of dorsal forebrain development. Development, 124(11), 2203 Retrieved from http://dev.biologists.org/content/124/11/2203.abstract [DOI] [PubMed] [Google Scholar]
  118. Galli LM, Barnes TL, Secrest SS, Kadowaki T, & Burrus LW (2007). Porcupine-mediated lipid-modification regulates the activity and distribution of Wnt proteins in the chick neural tube. Development, 134(18), 3339. doi: 10.1242/dev.02881 [DOI] [PubMed] [Google Scholar]
  119. Gardner OK, Haynes K, Schweitzer D, Johns A, Magee WP, Urata MM, & Sanchez-Lara PA (2017). Familial Recurrence of 3MC Syndrome in Consanguineous Families: A Clinical and Molecular Diagnostic Approach With Review of the Literature. The Cleft palate-craniofacial journal : official publication of the American Cleft Palate-Craniofacial Association, 54(6), 739–748. doi: 10.1597/15-151 [DOI] [PubMed] [Google Scholar]
  120. Gaur T, Lengner CJ, Hovhannisyan H, Bhat RA, Bodine PVN, Komm BS, … Lian JB (2005). Canonical WNT Signaling Promotes Osteogenesis by Directly Stimulating Runx2 Gene Expression. Journal of Biological Chemistry, 280(39), 33132–33140. doi: 10.1074/jbc.M500608200 [DOI] [PubMed] [Google Scholar]
  121. Gazda HT, Preti M, Sheen MR, O’Donohue M-F, Vlachos A, Davies SM, … Beggs AH (2012). Frameshift mutation in p53 regulator RPL26 is associated with multiple physical abnormalities and a specific pre-ribosomal RNA processing defect in diamond–blackfan anemia. Human Mutation, 33(7), 1037–1044. doi: 10.1002/humu.22081 [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Gazda HT, Sheen MR, Vlachos A, Choesmel V, O’Donohue MF, Schneider H, … Beggs AH (2008). Ribosomal protein L5 and L11 mutations are associated with cleft palate and abnormal thumbs in Diamond-Blackfan anemia patients. American journal of human genetics, 83(6), 769–780. doi: 10.1016/j.ajhg.2008.11.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Ghoumid J, Stichelbout M, Jourdain A-S, Frenois F, Lejeune-Dumoulin S, Alex-Cordier M-P, … Manouvrier-Hanu S (2017). Blepharocheilodontic syndrome is a CDH1 pathway–related disorder due to mutations in CDH1 and CTNND1. Genetics in Medicine, 19(9), 1013–1021. doi: 10.1038/gim.2017.11 [DOI] [PubMed] [Google Scholar]
  124. Ghyselinck NB, & Duester G (2019). Retinoic acid signaling pathways. Development, 146(13), dev167502. doi: 10.1242/dev.167502 [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Giudice A, Barone S, Belhous K, Morice A, Soupre V, Bennardo F, … Picard A (2018). Pierre Robin sequence: A comprehensive narrative review of the literature over time. Journal of Stomatology, Oral and Maxillofacial Surgery, 119(5), 419–428. doi: 10.1016/j.jormas.2018.05.002 [DOI] [PubMed] [Google Scholar]
  126. Gonzalez-Del Angel A, Caro-Contreras A, Alcantara-Ortigoza MA, Ramos S, Cruz-Alcivar R, & Moyers-Perez P (2018). Unique association of hypochondroplasia with craniosynostosis and cleft palate in a Mexican family. Am J Med Genet A, 176(1), 161–166. doi: 10.1002/ajmg.a.38526 [DOI] [PubMed] [Google Scholar]
  127. Goodnough LH, Brugmann SA, Hu D, & Helms JA (2007). Stage-dependent craniofacial defects resulting from Sprouty2 overexpression. Developmental dynamics : an official publication of the American Association of Anatomists, 236(7), 1918–1928. doi: 10.1002/dvdy.21195 [DOI] [PubMed] [Google Scholar]
  128. Goos JAC, Swagemakers SMA, Twigg SRF, van Dooren MF, Hoogeboom AJM, Beetz C, … Hurst JA (2017). Identification of causative variants in TXNL4A in Burn-McKeown syndrome and isolated choanal atresia. European Journal of Human Genetics, 25(10), 1126–1133. doi: 10.1038/ejhg.2017.107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Grant SFA, Wang K, Zhang H, Glaberson W, Annaiah K, Kim CE, … Hakonarson H (2009). A Genome-Wide Association Study Identifies a Locus for Nonsyndromic Cleft Lip with or without Cleft Palate on 8q24. The Journal of Pediatrics, 155(6), 909–913. doi: 10.1016/j.jpeds.2009.06.020 [DOI] [PubMed] [Google Scholar]
  130. Green RM, Feng W, Phang T, Fish JL, Li H, Spritz RA, … Williams T (2015). Tfap2a-dependent changes in mouse facial morphology result in clefting that can be ameliorated by a reduction in Fgf8 gene dosage. Disease Models & Mechanisms, 8(1), 31. doi: 10.1242/dmm.017616 [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Gripp KW, Ennis S, & Napoli J (2013). Exome Analysis in Clinical Practice: Expanding the Phenotype of Bartsocas–Papas Syndrome. American Journal of Medical Genetics Part A, 161(5), 1058–1063. doi: 10.1002/ajmg.a.35913 [DOI] [PubMed] [Google Scholar]
  132. Gripp KW, Wotton D, Edwards MC, Roessler E, Ades L, Meinecke P, … Elledge SJ (2000). Mutations in TGIF cause holoprosencephaly and link NODAL signalling to human neural axis determination. Nat Genet, 25(2), 205–208. doi: 10.1038/76074 [DOI] [PubMed] [Google Scholar]
  133. Group TTCSC (1996). Positional cloning of a gene involved in the pathogenesis of Treacher Collins syndrome. Nat Genet, 12(2), 130–136. doi: 10.1038/ng0296-130 [DOI] [PubMed] [Google Scholar]
  134. Guo L, Elcioglu NH, Wang Z, Demirkol YK, Isguven P, Matsumoto N, … Ikegawa S (2017). Novel and recurrent COL11A1 and COL2A1 mutations in the Marshall–Stickler syndrome spectrum. Human Genome Variation, 4(1), 17040. doi: 10.1038/hgv.2017.40 [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Guo Q, Li D, Meng X, Liu T, Shi J, Hao Y, … Song T (2017). Association between PAX7 and NTN1 gene polymorphisms and nonsyndromic orofacial clefts in a northern Chinese population. Medicine, 96(19), e6724–e6724. doi: 10.1097/MD.0000000000006724 [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Hahn H, Wicking C, Zaphiropoulos PG, Gailani MR, Shanley S, Chidambaram A, … Bale AE (1996). Mutations of the Human Homolog of Drosophila patched in the Nevoid Basal Cell Carcinoma Syndrome. Cell, 85(6), 841–851. doi: 10.1016/S0092-8674(00)81268-4 [DOI] [PubMed] [Google Scholar]
  137. Halford MM, Armes J, Buchert M, Meskenaite V, Grail D, Hibbs ML, … Stacker SA (2000). Ryk-deficient mice exhibit craniofacial defects associated with perturbed Eph receptor crosstalk. Nature Genetics, 25(4), 414–418. doi: 10.1038/78099 [DOI] [PubMed] [Google Scholar]
  138. Halilagic A, Ribes V, Ghyselinck NB, Zile MH, Dollé P, & Studer M (2007). Retinoids control anterior and dorsal properties in the developing forebrain. Developmental Biology, 303(1), 362–375. doi: 10.1016/j.ydbio.2006.11.021 [DOI] [PubMed] [Google Scholar]
  139. Hammond NL, Brookes KJ, & Dixon MJ (2018). Ectopic Hedgehog Signaling Causes Cleft Palate and Defective Osteogenesis. Journal of Dental Research, 97(13), 1485–1493. doi: 10.1177/0022034518785336 [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Han J, Mayo J, Xu X, Li J, Bringas P, Maas RL, … Chai Y (2009). Indirect modulation of Shh signaling by Dlx5 affects the oral-nasal patterning of palate and rescues cleft palate in Msx1-null mice. Development, 136(24), 4225. doi: 10.1242/dev.036723 [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Han Y, Zhou L, Ma L, Li D, Xu M, Yuan H, … Pan Y (2014). The axis inhibition protein 2 polymorphisms and non-syndromic orofacial clefts susceptibility in a Chinese Han population. Journal of Oral Pathology & Medicine, 43(7), 554–560. doi: 10.1111/jop.12162 10.1111/jop.12162 [DOI] [PubMed] [Google Scholar]
  142. Hannibal MC, Buckingham KJ, Ng SB, Ming JE, Beck AE, McMillin MJ, … Bamshad MJ (2011). Spectrum of MLL2 (ALR) mutations in 110 cases of Kabuki syndrome. American journal of medical genetics. Part A, 155A(7), 1511–1516. doi: 10.1002/ajmg.a.34074 [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. He F, Xiong W, Wang Y, Li L, Liu C, Yamagami T, … Chen Y (2011). Epithelial Wnt/β-catenin signaling regulates palatal shelf fusion through regulation of Tgfβ3 expression. Developmental Biology, 350(2), 511–519. doi: 10.1016/j.ydbio.2010.12.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. He F, Xiong W, Wang Y, Matsui M, Yu X, Chai Y, … Chen Y (2010). Modulation of BMP signaling by Noggin is required for the maintenance of palatal epithelial integrity during palatogenesis. Developmental Biology, 347(1), 109–121. doi: 10.1016/j.ydbio.2010.08.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. He F, Xiong W, Yu X, Espinoza-Lewis R, Liu C, Gu S, … Chen Y (2008). Wnt5a regulates directional cell migration and cell proliferation via Ror2-mediated noncanonical pathway in mammalian palate development. Development (Cambridge, England), 135(23), 3871–3879. doi: 10.1242/dev.025767 [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Heisenberg C-P, Tada M, Rauch G-J, Saúde L, Concha ML, Geisler R, … Wilson SW (2000). Silberblick/Wnt11 mediates convergent extension movements during zebrafish gastrulation. Nature, 405(6782), 76–81. doi: 10.1038/35011068 [DOI] [PubMed] [Google Scholar]
  147. Helms JA, Kim CH, Hu D, Minkoff R, Thaller C, & Eichele G (1997). Sonic hedgehogParticipates in Craniofacial Morphogenesis and Is Down-regulated by Teratogenic Doses of Retinoic Acid. Developmental Biology, 187(1), 25–35. doi: 10.1006/dbio.1997.8589 [DOI] [PubMed] [Google Scholar]
  148. Herman SB, Guo T, McGinn DMM, Blonska A, Shanske AL, Bassett AS, … International Chromosome 22q, C. (2012). Overt cleft palate phenotype and TBX1 genotype correlations in velo-cardio-facial/DiGeorge/22q11.2 deletion syndrome patients. American journal of medical genetics. Part A, 158A(11), 2781–2787. doi: 10.1002/ajmg.a.35512 [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Herr P, & Basler K (2012). Porcupine-mediated lipidation is required for Wnt recognition by Wls. Developmental Biology, 361(2), 392–402. doi: 10.1016/j.ydbio.2011.11.003 [DOI] [PubMed] [Google Scholar]
  150. Heyne GW, Melberg CG, Doroodchi P, Parins KF, Kietzman HW, Everson JL, … Lipinski RJ (2015). Definition of critical periods for Hedgehog pathway antagonist-induced holoprosencephaly, cleft lip, and cleft palate. PLOS ONE, 10(3), e0120517–e0120517. doi: 10.1371/journal.pone.0120517 [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Hill CR, Jacobs BH, Brown CB, Barnett JV, & Goudy SL (2015). Type III transforming growth factor beta receptor regulates vascular and osteoblast development during palatogenesis. Developmental dynamics : an official publication of the American Association of Anatomists, 244(2), 122–133. doi: 10.1002/dvdy.24225 [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Hilton E, Johnston J, Whalen S, Okamoto N, Hatsukawa Y, Nishio J, … Black G (2009). BCOR analysis in patients with OFCD and Lenz microphthalmia syndromes, mental retardation with ocular anomalies, and cardiac laterality defects. European journal of human genetics : EJHG, 17(10), 1325–1335. doi: 10.1038/ejhg.2009.52 [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Ho H-YH, Susman MW, Bikoff JB, Ryu YK, Jonas AM, Hu L, … Greenberg ME (2012). Wnt5a-Ror-Dishevelled signaling constitutes a core developmental pathway that controls tissue morphogenesis. Proceedings of the National Academy of Sciences of the United States of America, 109(11), 4044–4051. doi: 10.1073/pnas.1200421109 [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Hoefele J, Wilhelm C, Schiesser M, Mack R, Heinrich U, Weber LT, … Rost I (2013). Expanding the mutation spectrum for Fraser syndrome: identification of a novel heterozygous deletion in FRAS1. Gene, 520(2), 194–197. doi: 10.1016/j.gene.2013.02.031 [DOI] [PubMed] [Google Scholar]
  155. Horn D, Krawitz P, Mannhardt A, Korenke GC, & Meinecke P (2011). Hyperphosphatasia-mental retardation syndrome due to PIGV mutations: expanded clinical spectrum. Am J Med Genet A, 155a(8), 1917–1922. doi: 10.1002/ajmg.a.34102 [DOI] [PubMed] [Google Scholar]
  156. Horstick EJ, Linsley JW, Dowling JJ, Hauser MA, McDonald KK, Ashley-Koch A, … Kuwada JY (2013). Stac3 is a component of the excitation-contraction coupling machinery and mutated in Native American myopathy. Nature communications, 4, 1952. doi: 10.1038/ncomms2952 [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Howard TD, Paznekas WA, Green ED, Chiang LC, Ma N, Luna RIOD, … Jabs EW (1997). Mutations in TWIST, a basic helix–loop–helix transcription factor, in Saethre-Chotzen syndrome. Nature Genetics, 15(1), 36–41. doi: 10.1038/ng0197-36 [DOI] [PubMed] [Google Scholar]
  158. Hu D, Young NM, Li X, Xu Y, Hallgrímsson B, & Marcucio RS (2015). A dynamic Shh expression pattern, regulated by SHH and BMP signaling, coordinates fusion of primordia in the amniote face. Development (Cambridge, England), 142(3), 567–574. doi: 10.1242/dev.114835 [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Hu JS, Doan LT, Currle DS, Paff M, Rheem JY, Schreyer R, … Monuki ES (2008). Border formation in a Bmp gradient reduced to single dissociated cells. Proceedings of the National Academy of Sciences, 105(9), 3398. doi: 10.1073/pnas.0709100105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Hu M, Chen X, Zhang J, Wang D, Fang X, Wang X, … Wang Y (2013). Over-expression of regulator of G protein signaling 5 promotes tumor metastasis by inducing epithelial-mesenchymal transition in hepatocellular carcinoma cells. J Surg Oncol, 108(3), 192–196. doi: 10.1002/jso.23367 [DOI] [PubMed] [Google Scholar]
  161. Hu X, Gao J, Liao Y, Tang S, & Lu F (2013). Retinoic acid alters the proliferation and survival of the epithelium and mesenchyme and suppresses Wnt/β-catenin signaling in developing cleft palate. Cell Death & Disease, 4(10), e898–e898. doi: 10.1038/cddis.2013.424 [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Hu Y-Y, Qin C-Q, Deng M-H, Niu Y-M, & Long X (2015). Association between BMP4 rs17563 polymorphism and NSCL/P risk: a meta-analysis. Disease markers, 2015, 763090–763090. doi: 10.1155/2015/763090 [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Huang H, Yang X, Bao M, Cao H, Miao X, Zhang X, … Zhang Z (2016). Ablation of the Sox11 Gene Results in Clefting of the Secondary Palate Resembling the Pierre Robin Sequence. The Journal of biological chemistry, 291(13), 7107–7118. doi: 10.1074/jbc.M115.690875 [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Huang L, Jia Z, Shi Y, Du Q, Shi J, Wang Z, … Yang Z (2019). Genetic factors define CPO and CLO subtypes of nonsyndromicorofacial cleft. PLoS genetics, 15(10), e1008357–e1008357. doi: 10.1371/journal.pgen.1008357 [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Huh S-H, & Ornitz DM (2010). β-catenin deficiency causes DiGeorge syndrome-like phenotypes through regulation of Tbx1. Development, 137(7), 1137. doi: 10.1242/dev.045534 [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Ingraham CR, Kinoshita A, Kondo S, Yang B, Sajan S, Trout KJ, … Schutte BC (2006). Abnormal skin, limb and craniofacial morphogenesis in mice deficient for interferon regulatory factor 6 (Irf6). Nature Genetics, 38(11), 1335–1340. doi: 10.1038/ng1903 [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Iwata J. i., Hacia JG, Suzuki A, Sanchez-Lara PA, Urata M, & Chai Y (2012). Modulation of noncanonical TGF-β signaling prevents cleft palate in Tgfbr2 mutant mice. The Journal of Clinical Investigation, 122(3), 873–885. doi: 10.1172/JCI61498 [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Iwata J. i., Suzuki A, Pelikan RC, Ho T-V, Sanchez-Lara PA, Urata M, … Chai Y (2013). Smad4-Irf6 genetic interaction and TGFβ-mediated IRF6 signaling cascade are crucial for palatal fusion in mice. Development, 140(6), 1220. doi: 10.1242/dev.089615 [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Iwata J. i., Suzuki A, Yokota T, Ho T-V, Pelikan R, Urata M, … Chai Y (2014). TGFβ regulates epithelial-mesenchymal interactions through WNT signaling activity to control muscle development in the soft palate. Development, 141(4), 909. doi: 10.1242/dev.103093 [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Iwata J. i., Tung L, Urata M, Hacia JG, Pelikan R, Suzuki A, … Chai Y (2012). Fibroblast growth factor 9 (FGF9)-pituitary homeobox 2 (PITX2) pathway mediates transforming growth factor β (TGFβ) signaling to regulate cell proliferation in palatal mesenchyme during mouse palatogenesis. The Journal of biological chemistry, 287(4), 2353–2363. doi: 10.1074/jbc.M111.280974 [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Iwata J, Parada C, & Chai Y (2011). The mechanism of TGF-β signaling during palate development. Oral Diseases, 17(8), 733–744. doi: 10.1111/j.1601-0825.2011.01806.x 10.1111/j.1601–0825.2011.01806.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Iwata J, Suzuki A, Pelikan RC, Ho TV, Sanchez-Lara PA, & Chai Y (2014). Modulation of lipid metabolic defects rescues cleft palate in Tgfbr2 mutant mice. Hum Mol Genet, 23(1), 182–193. doi: 10.1093/hmg/ddt410 [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Jagomägi T, Nikopensius T, Krjutškov K, Tammekivi V, Viltrop T, Saag M, & Metspalu A (2010). MTHFR and MSX1 contribute to the risk of nonsyndromic cleft lip/palate. European Journal of Oral Sciences, 118(3), 213–220. doi: 10.1111/j.1600-0722.2010.00729.x [DOI] [PubMed] [Google Scholar]
  174. Jakobsen LP, Ullmann R, Christensen SB, Jensen KE, Mølsted K, Henriksen KF, … Tümer Z (2007). Pierre Robin sequence may be caused by dysregulation of SOX9 and KCNJ2. Journal of medical genetics, 44(6), 381–386. doi: 10.1136/jmg.2006.046177 [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Jenkins ZA, van Kogelenberg M, Morgan T, Jeffs A, Fukuzawa R, Pearl E, … Robertson SP (2009). Germline mutations in WTX cause a sclerosing skeletal dysplasia but do not predispose to tumorigenesis. Nat Genet, 41(1), 95–100. doi: 10.1038/ng.270 [DOI] [PubMed] [Google Scholar]
  176. Jeong J, Mao J, Tenzen T, Kottmann AH, & McMahon AP (2004). Hedgehog signaling in the neural crest cells regulates the patterning and growth of facial primordia. Genes & development, 18(8), 937–951. doi: 10.1101/gad.1190304 [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Jezewski PA, Vieira AR, Nishimura C, Ludwig B, Johnson M, O’Brien SE, … Murray JC (2003). Complete sequencing shows a role for MSX1 in non-syndromic cleft lip and palate. Journal of medical genetics, 40(6), 399–407. doi: 10.1136/jmg.40.6.399 [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Jia S, Zhou J, Fanelli C, Wee Y, Bonds J, Schneider P, … D’Souza RN (2017). Small-molecule Wnt agonists correct cleft palates in Pax9 mutant mice in utero. Development, 144(20), 3819–3828. doi: 10.1242/dev.157750 [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Jiang R, Lan Y, Chapman HD, Shawber C, Norton CR, Serreze DV, … Gridley T (1998). Defects in limb, craniofacial, and thymic development in Jagged2 mutant mice. Genes & development, 12(7), 1046–1057. doi: 10.1101/gad.12.7.1046 [DOI] [PMC free article] [PubMed] [Google Scholar]
  180. Jiang S, Shi JY, Lin YS, Duan SJ, Chen X, Jiao JJ, … Jia ZL (2019). NTN1 gene was risk to non-syndromic cleft lip only among Han Chinese population. Oral Dis, 25(2), 535–542. doi: 10.1111/odi.13009 [DOI] [PubMed] [Google Scholar]
  181. Jiang Z, Pan L, Chen X, Chen Z, & Xu D (2017). Wnt6 influences the viability of mouse embryonic palatal mesenchymal cells via the β-catenin pathway. Experimental and therapeutic medicine, 14(6), 5339–5344. doi: 10.3892/etm.2017.5240 [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Jianyan L, Zeqiang G, Yongjuan C, Kaihong D, Bing D, & Rongsheng L (2010). Analysis of interactions between genetic variants of BMP4 and environmental factors with nonsyndromic cleft lip with or without cleft palate susceptibility. International Journal of Oral and Maxillofacial Surgery, 39(1), 50–56. doi: 10.1016/j.ijom.2009.10.010 [DOI] [PubMed] [Google Scholar]
  183. Jin Y-R, Han XH, Taketo MM, & Yoon JK (2012). Wnt9b-dependent FGF signaling is crucial for outgrowth of the nasal and maxillary processes during upper jaw and lip development. Development, 139(10), 1821. doi: 10.1242/dev.075796 [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Jin Y-R, Turcotte TJ, Crocker AL, Han XH, & Yoon JK (2011). The canonical Wnt signaling activator, R-spondin2, regulates craniofacial patterning and morphogenesis within the branchial arch through ectodermal-mesenchymal interaction. Developmental Biology, 352(1), 1–13. doi: 10.1016/j.ydbio.2011.01.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Johansson S, Berland S, Gradek GA, Bongers E, de Leeuw N, Pfundt R, … Houge G (2014). Haploinsufficiency of MEIS2 is associated with orofacial clefting and learning disability. Am J Med Genet A, 164a(7), 1622–1626. doi: 10.1002/ajmg.a.36498 [DOI] [PubMed] [Google Scholar]
  186. Johnston JJ, Teer JK, Cherukuri PF, Hansen NF, Loftus SK, Chong K, … Biesecker LG (2010). Massively parallel sequencing of exons on the X chromosome identifies RBM10 as the gene that causes a syndromic form of cleft palate. American journal of human genetics, 86(5), 743–748. doi: 10.1016/j.ajhg.2010.04.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Jones C, & Chen P (2007). Planar cell polarity signaling in vertebrates. BioEssays : news and reviews in molecular, cellular and developmental biology, 29(2), 120–132. doi: 10.1002/bies.20526 [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Jugessur A, Shi M, Gjessing HK, Lie RT, Wilcox AJ, Weinberg CR, … Murray JC (2009). Genetic Determinants of Facial Clefting: Analysis of 357 Candidate Genes Using Two National Cleft Studies from Scandinavia. PLOS ONE, 4(4), e5385. doi: 10.1371/journal.pone.0005385 [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Juriloff DM, Harris MJ, McMahon AP, Carroll TJ, & Lidral AC (2006). Wnt9b is the mutated gene involved in multifactorial nonsyndromic cleft lip with or without cleft palate in A/WySn mice, as confirmed by a genetic complementation test. Birth Defects Research Part A: Clinical and Molecular Teratology, 76(8), 574–579. doi: 10.1002/bdra.20302 10.1002/bdra.20302 [DOI] [PubMed] [Google Scholar]
  190. Kaartinen V, Voncken JW, Shuler C, Warburton D, Bu D, Heisterkamp N, & Groffen J (1995). Abnormal lung development and cleft palate in mice lacking TGF–β3 indicates defects of epithelial–mesenchymal interaction. Nature Genetics, 11(4), 415–421. doi: 10.1038/ng1295-415 [DOI] [PubMed] [Google Scholar]
  191. Kalscheuer VM, Freude K, Musante L, Jensen LR, Yntema HG, Gecz J, … Ropers HH (2003). Mutations in the polyglutamine binding protein 1 gene cause X-linked mental retardation. Nat Genet, 35(4), 313–315. doi: 10.1038/ng1264 [DOI] [PubMed] [Google Scholar]
  192. Kalter H (1960). THE TERATOGENIC EFFECTS OF HYPERVITAMINOSIS A UPON THE FACE AND MOUTH OF INBRED MICE*. Annals of the New York Academy of Sciences, 85(1), 42–55. doi: 10.1111/j.1749-6632.1960.tb49947.x 10.1111/j.1749–6632.1960.tb49947.x [DOI] [PubMed] [Google Scholar]
  193. Kalter H, & Warkany J (1961). Experimental production of congenital malformations in strains of inbred mice by maternal treatment with hypervitaminosis A. The American journal of pathology, 38(1), 1–21. Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/13750997https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1942316/ [PMC free article] [PubMed] [Google Scholar]
  194. Kantaputra PN, Hamada T, Kumchai T, & McGrath JA (2003). Heterozygous Mutation in the SAM Domain of p63 Underlies Rapp-Hodgkin Ectodermal Dysplasia. Journal of Dental Research, 82(6), 433–437. doi: 10.1177/154405910308200606 [DOI] [PubMed] [Google Scholar]
  195. Kasberg AD, Brunskill EW, & Steven Potter S (2013). SP8 regulates signaling centers during craniofacial development. Developmental Biology, 381(2), 312–323. doi: 10.1016/j.ydbio.2013.07.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Kawaguchi R, Yu J, Honda J, Hu J, Whitelegge J, Ping P, … Sun H (2007). A Membrane Receptor for Retinol Binding Protein Mediates Cellular Uptake of Vitamin A. Science, 315(5813), 820. doi: 10.1126/science.1136244 [DOI] [PubMed] [Google Scholar]
  197. Kawasaki M, Kawasaki K, Meguro F, Yamada A, Ishikawa R, Porntaveetus T, … Ohazama A (2018). Lrp4/Wise regulates palatal rugae development through Turing-type reaction-diffusion mechanisms. PLOS ONE, 13(9), e0204126. doi: 10.1371/journal.pone.0204126 [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Kayano S, Suzuki Y, Kanno K, Aoki Y, Kure S, Yamada A, & Matsubara Y (2004). Significant association between nonsyndromic oral clefts and arylhydrocarbon receptor nuclear translocator (ARNT). American Journal of Medical Genetics Part A, 130A(1), 40–44. doi: 10.1002/ajmg.a.30023 [DOI] [PubMed] [Google Scholar]
  199. Kayserili H, Uz E, Niessen C, Vargel I, Alanay Y, Tuncbilek G, … Akarsu NA (2009). ALX4 dysfunction disrupts craniofacial and epidermal development. Human Molecular Genetics, 18(22), 4357–4366. doi: 10.1093/hmg/ddp391 [DOI] [PubMed] [Google Scholar]
  200. Ke C-Y, Mei H-H, Wong F-H, & Lo L-J (2019). IRF6 and TAK1 coordinately promote the activation of HIPK2 to stimulate apoptosis during palate fusion. Science Signaling, 12(593), eaav7666. doi: 10.1126/scisignal.aav7666 [DOI] [PubMed] [Google Scholar]
  201. Keeble TR, Halford MM, Seaman C, Kee N, Macheda M, Anderson RB, … Cooper HM (2006). The Wnt Receptor Ryk Is Required for Wnt5a-Mediated Axon Guidance on the Contralateral Side of the Corpus Callosum. The Journal of Neuroscience, 26(21), 5840. doi: 10.1523/JNEUROSCI.1175-06.2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  202. Keeler RF (1978). Cyclopamine and related steroidal alkaloid teratogens: their occurrence, structural relationship, and biologic effects. Lipids, 13(10), 708–715. doi: 10.1007/bf02533750 [DOI] [PubMed] [Google Scholar]
  203. Kempa I, Ambrozaitytė L, Stavusis J, Akota I, Barkane B, Krumina A, … Lace B (2014). Association of BMP4 polymorphisms with non-syndromic cleft lip with or without cleft palate and isolated cleft palate in Latvian and Lithuanian populations. Stomatolgija, 16(3), 94–101. [PubMed] [Google Scholar]
  204. Kioussi C, Briata P, Baek SH, Rose DW, Hamblet NS, Herman T, … Rosenfeld MG (2002). Identification of a Wnt/Dvl/β-Catenin → Pitx2 Pathway Mediating Cell-Type-Specific Proliferation during Development. Cell, 111(5), 673–685. doi: 10.1016/S0092-8674(02)01084-X 10.1016/S0092–8674(02)01084-X [DOI] [PubMed] [Google Scholar]
  205. Kirikoshi H, Sekihara H, & Katoh M (2001). WNT10A and WNT6, Clustered in Human Chromosome 2q35 Region with Head-to-Tail Manner, Are Strongly Coexpressed in SW480 Cells. Biochemical and Biophysical Research Communications, 283(4), 798–805. doi: 10.1006/bbrc.2001.4855 [DOI] [PubMed] [Google Scholar]
  206. Kishida S, Yamamoto H, Hino S. i., Ikeda S, Kishida M, & Kikuchi A (1999). DIX Domains of Dvl and Axin Are Necessary for Protein Interactions and Their Ability To Regulate β-Catenin Stability. Molecular and Cellular Biology, 19(6), 4414. doi: 10.1128/MCB.19.6.4414 [DOI] [PMC free article] [PubMed] [Google Scholar]
  207. Ko SO, Chung IH, Xu X, Oka S, Zhao H, Cho ES, … Chai Y (2007). Smad4 is required to regulate the fate of cranial neural crest cells. Developmental Biology, 312(1), 435–447. doi: 10.1016/j.ydbio.2007.09.050 [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Komekado H, Yamamoto H, Chiba T, & Kikuchi A (2007). Glycosylation and palmitoylation of Wnt-3a are coupled to produce an active form of Wnt-3a. Genes to Cells, 12(4), 521–534. doi: 10.1111/j.1365-2443.2007.01068.x 10.1111/j.1365–2443.2007.01068.x [DOI] [PubMed] [Google Scholar]
  209. Komiya Y, & Habas R (2008). Wnt signal transduction pathways. Organogenesis, 4(2), 68–75. doi: 10.4161/org.4.2.5851 [DOI] [PMC free article] [PubMed] [Google Scholar]
  210. Komori T (2011). Signaling networks in RUNX2-dependent bone development. J Cell Biochem, 112(3), 750–755. doi: 10.1002/jcb.22994 [DOI] [PubMed] [Google Scholar]
  211. Kondo S, Schutte BC, Richardson RJ, Bjork BC, Knight AS, Watanabe Y, … Murray JC (2002). Mutations in IRF6 cause Van der Woude and popliteal pterygium syndromes. Nature Genetics, 32(2), 285–289. doi: 10.1038/ng985 [DOI] [PMC free article] [PubMed] [Google Scholar]
  212. Konno Y, Toki T, Tandai S, Xu G, Wang R, Terui K, … Ito E (2010). Mutations in the ribosomal protein genes in Japanese patients with Diamond-Blackfan anemia. Haematologica, 95(8), 1293–1299. doi: 10.3324/haematol.2009.020826 [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Kousa YA, Zhu H, Fakhouri WD, Lei Y, Kinoshita A, Roushangar RR, … Schutte BC (2019). The TFAP2A–IRF6–GRHL3 genetic pathway is conserved in neurulation. Human Molecular Genetics, 28(10), 1726–1737. doi: 10.1093/hmg/ddz010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Kouskoura T, Kozlova A, Alexiou M, Blumer S, Zouvelou V, Katsaros C, … Graf D (2013). The Etiology of Cleft Palate Formation in BMP7-Deficient Mice. PLOS ONE, 8(3), e59463. doi: 10.1371/journal.pone.0059463 [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Krantz ID, McCallum J, DeScipio C, Kaur M, Gillis LA, Yaeger D, … Jackson LG (2004). Cornelia de Lange syndrome is caused by mutations in NIPBL, the human homolog of Drosophila melanogaster Nipped-B. Nat Genet, 36(6), 631–635. doi: 10.1038/ng1364 [DOI] [PMC free article] [PubMed] [Google Scholar]
  216. Kruszka P, Li D, Harr MH, Wilson NR, Swarr D, McCormick EM, … Zackai EH (2015). Mutations in SPECC1L, encoding sperm antigen with calponin homology and coiled-coil domains 1-like, are found in some cases of autosomal dominant Opitz G/BBB syndrome. Journal of medical genetics, 52(2), 104–110. doi: 10.1136/jmedgenet-2014-102677 [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Kumari P, Singh SK, & Raman R (2018). TGFβ3, MSX1, and MMP3 as Candidates for NSCL±P in an Indian Population. The Cleft Palate-Craniofacial Journal, 56(3), 363–372. doi: 10.1177/1055665618775727 [DOI] [PubMed] [Google Scholar]
  218. Lacbawan F, Solomon BD, Roessler E, El-Jaick K, Domené S, Vélez JI, … Muenke M (2009). Clinical spectrum of SIX3-associated mutations in holoprosencephaly: correlation between genotype, phenotype and function. Journal of medical genetics, 46(6), 389–398. doi: 10.1136/jmg.2008.063818 [DOI] [PMC free article] [PubMed] [Google Scholar]
  219. Laccone F, Hannibal MC, Neesen J, Grisold W, Chance PF, & Rehder H (2008). Dysmorphic syndrome of hereditary neuralgic amyotrophy associated with a SEPT9 gene mutation--a family study. Clin Genet, 74(3), 279–283. doi: 10.1111/j.1399-0004.2008.01022.x [DOI] [PubMed] [Google Scholar]
  220. Lalani SR, Thakuria JV, Cox GF, Wang X, Bi W, Bray MS, … Potocki L (2009). 20p12.3 microdeletion predisposes to Wolff-Parkinson-White syndrome with variable neurocognitive deficits. Journal of medical genetics, 46(3), 168–175. doi: 10.1136/jmg.2008.061002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  221. Lambrecht JT, & Kreusch T (1997). Examine your orofacial cleft patients for Gorlin-Goltz syndrome. The Cleft palate-craniofacial journal : official publication of the American Cleft Palate-Craniofacial Association, 34(4), 342–350. doi: 10.1597/1545-1569_1997_034_0341_eyocpf_2.3.co_2 [DOI] [PubMed] [Google Scholar]
  222. Lammer EJ, Shaw GM, Iovannisci DM, Van Waes J, & Finnell RH (2004). Maternal smoking and the risk of orofacial clefts: Susceptibility with NAT1 and NAT2 polymorphisms. Epidemiology, 15(2), 150–156. doi: 10.1097/01.ede.0000112214.33432.cc [DOI] [PubMed] [Google Scholar]
  223. Lan Y, & Jiang R (2009). Sonic hedgehog signaling regulates reciprocal epithelial-mesenchymal interactions controlling palatal outgrowth. Development (Cambridge, England), 136(8), 1387–1396. doi: 10.1242/dev.028167 [DOI] [PMC free article] [PubMed] [Google Scholar]
  224. Lan Y, Ryan RC, Zhang Z, Bullard SA, Bush JO, Maltby KM, … Jiang R (2006). Expression of Wnt9b and activation of canonical Wnt signaling during midfacial morphogenesis in mice. Developmental dynamics : an official publication of the American Association of Anatomists, 235(5), 1448–1454. doi: 10.1002/dvdy.20723 [DOI] [PMC free article] [PubMed] [Google Scholar]
  225. Lane J, Yumoto K, Azhar M, Ninomiya-Tsuji J, Inagaki M, Hu Y, … Kaartinen V (2015). Tak1, Smad4 and Trim33 redundantly mediate TGF-β3 signaling during palate development. Developmental Biology, 398(2), 231–241. doi: 10.1016/j.ydbio.2014.12.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  226. Laumonnier F, Holbert S, Ronce N, Faravelli F, Lenzner S, Schwartz CE, … Briault S (2005). Mutations in PHF8 are associated with X linked mental retardation and cleft lip/cleft palate. Journal of medical genetics, 42(10), 780–786. doi: 10.1136/jmg.2004.029439 [DOI] [PMC free article] [PubMed] [Google Scholar]
  227. Laurikkala J, Kassai Y, Pakkasjärvi L, Thesleff I, & Itoh N (2003). Identification of a secreted BMP antagonist, ectodin, integrating BMP, FGF, and SHH signals from the tooth enamel knot. Developmental Biology, 264(1), 91–105. doi: 10.1016/j.ydbio.2003.08.011 [DOI] [PubMed] [Google Scholar]
  228. Lee E, Salic A, Krüger R, Heinrich R, & Kirschner MW (2003). The roles of APC and Axin derived from experimental and theoretical analysis of the Wnt pathway. PLoS biology, 1(1), E10–E10. doi: 10.1371/journal.pbio.0000010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  229. Lee J-M, Kim J-Y, Cho K-W, Lee M-J, Cho S-W, Kwak S, … Jung H-S (2008). Wnt11/Fgfr1b cross-talk modulates the fate of cells in palate development. Developmental Biology, 314(2), 341–350. doi: 10.1016/j.ydbio.2007.11.033 [DOI] [PubMed] [Google Scholar]
  230. Lee S, Crisera CA, Erfani S, Maldonado TS, Lee JJ, Alkasab SL, & Longaker MT (2001). Immunolocalization of Fibroblast Growth Factor Receptors 1 and 2 in Mouse Palate Development. Plastic and Reconstructive Surgery, 107(7). Retrieved from https://journals.lww.com/plasreconsurg/Fulltext/2001/06000/Immunolocalization_of_Fibroblast_Growth_Factor.21.aspx [DOI] [PubMed] [Google Scholar]
  231. Leid M, Kastner P, & Chambon P (1992). Multiplicity generates diversity in the retinoic acid signalling pathways. Trends in Biochemical Sciences, 17(10), 427–433. doi: 10.1016/0968-0004(92)90014-Z [DOI] [PubMed] [Google Scholar]
  232. Leslie EJ, Carlson JC, Shaffer JR, Butali A, Buxó CJ, Castilla EE, … Marazita ML (2017). 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. Human Genetics, 136(3), 275–286. doi: 10.1007/s00439-016-1754-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  233. Leslie EJ, Carlson JC, Shaffer JR, Feingold E, Wehby G, Laurie CA, … Marazita ML (2016). A multi-ethnic genome-wide association study identifies novel loci for non-syndromic cleft lip with or without cleft palate on 2p24.2, 17q23 and 19q13. Hum Mol Genet, 25(13), 2862–2872. doi: 10.1093/hmg/ddw104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  234. Leslie EJ, Liu H, Carlson JC, Shaffer JR, Feingold E, Wehby G, … Marazita ML (2016). A Genome-wide Association Study of Nonsyndromic Cleft Palate Identifies an Etiologic Missense Variant in GRHL3. American journal of human genetics, 98(4), 744–754. doi: 10.1016/j.ajhg.2016.02.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  235. Leslie EJ, Mansilla MA, Biggs LC, Schuette K, Bullard S, Cooper M, … Murray JC (2012). Expression and mutation analyses implicate ARHGAP29 as the etiologic gene for the cleft lip with or without cleft palate locus identified by genome-wide association on chromosome 1p22. Birth defects research. Part A, Clinical and molecular teratology, 94(11), 934–942. doi: 10.1002/bdra.23076 [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Leslie EJ, Taub MA, Liu H, Steinberg KM, Koboldt DC, Zhang Q, … Murray JC (2015). 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. American journal of human genetics, 96(3), 397–411. doi: 10.1016/j.ajhg.2015.01.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  237. Lesnik Oberstein SAJ, Kriek M, White SJ, Kalf ME, Szuhai K, den Dunnen JT, … Hennekam RCM (2006). Peters Plus Syndrome Is Caused by Mutations in B3GALTL, a Putative Glycosyltransferase. The American Journal of Human Genetics, 79(3), 562–566. doi: 10.1086/507567 [DOI] [PMC free article] [PubMed] [Google Scholar]
  238. Letra A, Bjork B, Cooper ME, Szabo-Rogers H, Deleyiannis FWB, Field LL, … Silva RM (2012). Association of AXIN2 with Non-syndromic Oral Clefts in Multiple Populations. Journal of Dental Research, 91(5), 473–478. doi: 10.1177/0022034512440578 10.1177/0022034512440578 [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Letra A, Menezes R, Granjeiro JM, & Vieira AR (2009). AXIN2 and CDH1 polymorphisms, tooth agenesis, and oral clefts. Birth Defects Research Part A: Clinical and Molecular Teratology, 85(2), 169–173. doi: 10.1002/bdra.20489 10.1002/bdra.20489 [DOI] [PMC free article] [PubMed] [Google Scholar]
  240. Li B, Kuriyama S, Moreno M, & Mayor R (2009). The posteriorizing gene Gbx2 is a direct target of Wnt signalling and the earliest factor in neural crest induction. Development (Cambridge, England), 136(19), 3267–3278. doi: 10.1242/dev.036954 [DOI] [PMC free article] [PubMed] [Google Scholar]
  241. Li C, Lan Y, Krumlauf R, & Jiang R (2017). Modulating Wnt Signaling Rescues Palate Morphogenesis in Pax9 Mutant Mice. Journal of Dental Research, 96(11), 1273–1281. doi: 10.1177/0022034517719865 [DOI] [PMC free article] [PubMed] [Google Scholar]
  242. Li J, Yuan Y, He J, Feng J, Han X, Jing J, … Chai Y (2018). Constitutive activation of hedgehog signaling adversely affects epithelial cell fate during palatal fusion. Developmental Biology, 441(1), 191–203. doi: 10.1016/j.ydbio.2018.07.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  243. Li L, Lin M, Wang Y, Cserjesi P, Chen Z, & Chen Y (2011). BmprIa is required in mesenchymal tissue and has limited redundant function with BmprIb in tooth and palate development. Developmental Biology, 349(2), 451–461. doi: 10.1016/j.ydbio.2010.10.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  244. Lidral AC, Romitti PA, Basart AM, Doetschman T, Leysens NJ, Daack-Hirsch S, … Murray JC (1998). Association of MSX1 and TGFB3 with Nonsyndromic Clefting in Humans. The American Journal of Human Genetics, 63(2), 557–568. doi: 10.1086/301956 [DOI] [PMC free article] [PubMed] [Google Scholar]
  245. Lin C, Fisher AV, Yin Y, Maruyama T, Veith GM, Dhandha M, … Ma L (2011). The inductive role of Wnt-β-Catenin signaling in the formation of oral apparatus. Developmental Biology, 356(1), 40–50. doi: 10.1016/j.ydbio.2011.05.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  246. Lin J-Y, Chen Y-J, Huang Y-L, Tang G-P, Zhang L, Deng B, … Luan R-S (2008). Association of Bone Morphogenetic Protein 4 Gene Polymorphisms with Nonsyndromic Cleft Lip with or without Cleft Palate in Chinese Children. DNA and Cell Biology, 27(11), 601–605. doi: 10.1089/dna.2008.0777 10.1089/dna.2008.0777 [DOI] [PubMed] [Google Scholar]
  247. Lintern KB, Guidato S, Rowe A, Saldanha JW, & Itasaki N (2009). Characterization of wise protein and its molecular mechanism to interact with both Wnt and BMP signals. The Journal of biological chemistry, 284(34), 23159–23168. doi: 10.1074/jbc.M109.025478 [DOI] [PMC free article] [PubMed] [Google Scholar]
  248. Lipinski RJ, Song C, Sulik KK, Everson JL, Gipp JJ, Yan D, … Rowland IJ (2010). Cleft lip and palate results from Hedgehog signaling antagonism in the mouse: Phenotypic characterization and clinical implications. Birth defects research. Part A, Clinical and molecular teratology, 88(4), 232–240. doi: 10.1002/bdra.20656 [DOI] [PMC free article] [PubMed] [Google Scholar]
  249. Liu C, Lin C, Gao C, May-Simera H, Swaroop A, & Li T (2014). Null and hypomorph Prickle1 alleles in mice phenocopy human Robinow syndrome and disrupt signaling downstream of Wnt5a. Biology Open, 3(9), 861. doi: 10.1242/bio.20148375 [DOI] [PMC free article] [PubMed] [Google Scholar]
  250. Liu KJ, Arron JR, Stankunas K, Crabtree GR, & Longaker MT (2007). Chemical rescue of cleft palate and midline defects in conditional GSK-3β mice. Nature, 446(7131), 79–82. doi: 10.1038/nature05557 [DOI] [PubMed] [Google Scholar]
  251. Liu P, Wakamiya M, Shea MJ, Albrecht U, Behringer RR, & Bradley A (1999). Requirement for Wnt3 in vertebrate axis formation. Nature Genetics, 22(4), 361–365. doi: 10.1038/11932 [DOI] [PubMed] [Google Scholar]
  252. Liu W, Sun X, Braut A, Mishina Y, Behringer RR, Mina M, & Martin JF (2005). Distinct functions for Bmp signaling in lip and palate fusion in mice. Development, 132(6), 1453. doi: 10.1242/dev.01676 [DOI] [PubMed] [Google Scholar]
  253. Liu X, Zhang H, Gao L, Yin Y, Pan X, Li Z, … Yu Z (2014). Negative Interplay of Retinoic Acid and TGF-β Signaling Mediated by TG-Interacting Factor to Modulate Mouse Embryonic Palate Mesenchymal–Cell Proliferation. Birth Defects Research Part B: Developmental and Reproductive Toxicology, 101(6), 403–409. doi: 10.1002/bdrb.21130 [DOI] [PubMed] [Google Scholar]
  254. Liu Y, Wang M, Zhao W, Yuan X, Yang X, Li Y, … Zhang Z (2015). Gpr177-mediated Wnt Signaling Is Required for Secondary Palate Development. Journal of Dental Research, 94(7), 961–967. doi: 10.1177/0022034515583532 10.1177/0022034515583532 [DOI] [PubMed] [Google Scholar]
  255. Liu Z, Xu J, Colvin JS, & Ornitz DM (2002). Coordination of chondrogenesis and osteogenesis by fibroblast growth factor 18. Genes & development, 16(7), 859–869. doi: 10.1101/gad.965602 [DOI] [PMC free article] [PubMed] [Google Scholar]
  256. Loeys BL, Chen J, Neptune ER, Judge DP, Podowski M, Holm T, … Dietz HC (2005). A syndrome of altered cardiovascular, craniofacial, neurocognitive and skeletal development caused by mutations in TGFBR1 or TGFBR2. Nature Genetics, 37(3), 275–281. doi: 10.1038/ng1511 [DOI] [PubMed] [Google Scholar]
  257. Lohnes D, Mark M, Mendelsohn C, Dolle P, Dierich A, Gorry P, … Chambon P (1994). Function of the retinoic acid receptors (RARs) during development (I). Craniofacial and skeletal abnormalities in RAR double mutants. Development, 120(10), 2723 Retrieved from http://dev.biologists.org/content/120/10/2723.abstract [DOI] [PubMed] [Google Scholar]
  258. Lombardi MP, Bulk S, Celli J, Lampe A, Gabbett MT, Ousager LB, … Hennekam RC (2011). Mutation update for the PORCN gene. Human Mutation, 32(7), 723–728. doi: 10.1002/humu.21505 10.1002/humu.21505 [DOI] [PubMed] [Google Scholar]
  259. Lonai P (2005). Fibroblast growth factor signaling and the function and assembly of basement membranes. Curr Top Dev Biol, 66, 37–64. doi: 10.1016/S0070-2153(05)66002-4 [DOI] [PubMed] [Google Scholar]
  260. López SH, Avetisyan M, Wright CM, Mesbah K, Kelly RG, Moon AM, & Heuckeroth RO (2018). Loss of Tbx3 in murine neural crest reduces enteric glia and causes cleft palate, but does not influence heart development or bowel transit. The Neural Crest: 150 years after His’ discovery, 444, S337–S351. doi: 10.1016/j.ydbio.2018.09.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  261. Loudig O, Maclean GA, Dore NL, Luu L, & Petkovich M (2005). Transcriptional co-operativity between distant retinoic acid response elements in regulation of Cyp26A1 inducibility. The Biochemical journal, 392(Pt 1), 241–248. doi: 10.1042/BJ20050874 [DOI] [PMC free article] [PubMed] [Google Scholar]
  262. Lu MF, Pressman C, Dyer R, Johnson RL, & Martin JF (1999). Function of Rieger syndrome gene in left-right asymmetry and craniofacial development. Nature, 401(6750), 276–278. doi: 10.1038/45797 [DOI] [PubMed] [Google Scholar]
  263. Lu W, Yamamoto V, Ortega B, & Baltimore D (2004). Mammalian Ryk Is a Wnt Coreceptor Required for Stimulation of Neurite Outgrowth. Cell, 119(1), 97–108. doi: 10.1016/j.cell.2004.09.019 [DOI] [PubMed] [Google Scholar]
  264. Lu YP, Han WT, Liu Q, Li JX, Li ZJ, Jiang M, & Xu W (2015). Variations in WNT3 gene are associated with incidence of non-syndromic cleft lip with or without cleft palate in a northeast Chinese population. 14(4), 12646–12653. doi: 10.4238/2015.October.19.8 [DOI] [PubMed] [Google Scholar]
  265. Ludwig KU, Ahmed ST, Böhmer AC, Sangani NB, Varghese S, Klamt J, … Peters H (2016). Meta-analysis Reveals Genome-Wide Significance at 15q13 for Nonsyndromic Clefting of Both the Lip and the Palate, and Functional Analyses Implicate GREM1 As a Plausible Causative Gene. PLoS genetics, 12(3), e1005914–e1005914. doi: 10.1371/journal.pgen.1005914 [DOI] [PMC free article] [PubMed] [Google Scholar]
  266. Ludwig KU, Mangold E, Herms S, Nowak S, Reutter H, Paul A, … Nöthen MM (2012). Genome-wide meta-analyses of nonsyndromic cleft lip with or without cleft palate identify six new risk loci. Nature Genetics, 44(9), 968–971. doi: 10.1038/ng.2360 [DOI] [PMC free article] [PubMed] [Google Scholar]
  267. Ludwig KU, Mangold E, Herms S, Nowak S, Reutter H, Paul A, … Nothen MM (2012). Genome-wide meta-analyses of nonsyndromic cleft lip with or without cleft palate identify six new risk loci. Nature genetics, 44(9), 968–971. doi: 10.1038/ng.2360 [DOI] [PMC free article] [PubMed] [Google Scholar]
  268. Luisin M, Chevreau J, Klein C, Naepels P, Demeer B, Mathieu-Dramard M, … Morin G (2017). Prenatal diagnosis of femoral facial syndrome: Three case reports and literature review. American Journal of Medical Genetics Part A, 173(11), 2923–2946. doi: 10.1002/ajmg.a.38420 [DOI] [PubMed] [Google Scholar]
  269. MacDonald BT, Tamai K, & He X (2009). Wnt/beta-catenin signaling: components, mechanisms, and diseases. Developmental Cell, 17(1), 9–26. doi: 10.1016/j.devcel.2009.06.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  270. MacDonald PN, & Ong DE (1987). Binding specificities of cellular retinol-binding protein and cellular retinol-binding protein, type II. Journal of Biological Chemistry, 262(22), 10550–10556. Retrieved from http://www.jbc.org/content/262/22/10550.abstract [PubMed] [Google Scholar]
  271. Machado RA, de Oliveira Silva C, Persuhn DC, de Melo Cavalcanti Dantas V, de Almeida Reis SR, Wu T, … Coletta RD (2019). Interactions between superoxide dismutase and paraoxonase polymorphic variants in nonsyndromic cleft lip with or without cleft palate in the Brazilian population. Environ Mol Mutagen, 60(2), 185–196. doi: 10.1002/em.22239 [DOI] [PubMed] [Google Scholar]
  272. Maili L, Letra A, Silva R, Buchanan EP, Mulliken JB, Greives MR, … Hecht JT (2019). PBX-WNT-P63-IRF6 pathway in nonsyndromic cleft lip and palate. Birth Defects Research, n/a(n/a). doi: 10.1002/bdr2.1630 [DOI] [PMC free article] [PubMed] [Google Scholar]
  273. Mäkitie O, Geiberger S, Horemuzova E, Hagenäs L, Moström E, Nordenskjöld M, … Nordgren A (2015). SLC26A2 disease spectrum in Sweden – high frequency of recessive multiple epiphyseal dysplasia (rMED). Clinical Genetics, 87(3), 273–278. doi: 10.1111/cge.12371 [DOI] [PubMed] [Google Scholar]
  274. Mangold E, Ludwig KU, Birnbaum S, Baluardo C, Ferrian M, Herms S, … Nöthen MM (2010). Genome-wide association study identifies two susceptibility loci for nonsyndromic cleft lip with or without cleft palate. Nature Genetics, 42(1), 24–26. doi: 10.1038/ng.506 [DOI] [PubMed] [Google Scholar]
  275. Mansour S, Hall CM, Pembrey ME, & Young ID (1995). A clinical and genetic study of campomelic dysplasia. Journal of medical genetics, 32(6), 415–420. doi: 10.1136/jmg.32.6.415 [DOI] [PMC free article] [PubMed] [Google Scholar]
  276. Marazita ML (2012). The evolution of human genetic studies of cleft lip and cleft palate. Annual review of genomics and human genetics, 13, 263–283. doi: 10.1146/annurevgenom-090711-163729 [DOI] [PMC free article] [PubMed] [Google Scholar]
  277. Marazita ML, Field LL, Cooper ME, Tobias R, Maher BS, Peanchitlertkajorn S, & Liu Y. e. (2002). Genome Scan for Loci Involved in Cleft Lip With or Without Cleft Palate, in Chinese Multiplex Families. The American Journal of Human Genetics, 71(2), 349–364. doi: 10.1086/341944 [DOI] [PMC free article] [PubMed] [Google Scholar]
  278. Marazita ML, Lidral AC, Murray JC, Field LL, Maher BS, Goldstein McHenry T, … Arcos-Burgos M (2009). Genome scan, fine-mapping, and candidate gene analysis of non-syndromic cleft lip with or without cleft palate reveals phenotype-specific differences in linkage and association results. Hum Hered, 68(3), 151–170. doi: 10.1159/000224636 [DOI] [PMC free article] [PubMed] [Google Scholar]
  279. Marazita ML, Murray JC, Lidral AC, Arcos-Burgos M, Cooper ME, Goldstein T, … Roddick LG (2004). Meta-analysis of 13 genome scans reveals multiple cleft lip/palate genes with novel loci on 9q21 and 2q32–35. American journal of human genetics, 75(2), 161–173. doi: 10.1086/422475 [DOI] [PMC free article] [PubMed] [Google Scholar]
  280. Marçano ACB, Doudney K, Braybrook C, Squires R, Patton MA, Lees MM, … Stanier P (2004). TBX22 mutations are a frequent cause of cleft palate. Journal of Medical Genetics, 41(1), 68. doi: 10.1136/jmg.2003.010868 [DOI] [PMC free article] [PubMed] [Google Scholar]
  281. Martinelli M, Carinci F, Morselli PG, Caramelli E, Palmieri A, Girardi A, … Scapoli L (2011). Evidence of LEF1 Fetal-Maternal Interaction in Cleft Lip with or without Cleft Palate in a Consistent Italian Sample Study. International Journal of Immunopathology and Pharmacology, 24(2_suppl), 15–19. doi: 10.1177/03946320110240S204 10.1177/03946320110240S204 [DOI] [PubMed] [Google Scholar]
  282. Martinelli M, Masiero E, Carinci F, Morselli PG, Palmieri A, Girardi A, … Scapoli L (2011). Evidence of an Involvement of TFAP2A Gene in Nonsyndromic Cleft LIP with or without Cleft Palate: An Italian Study. International Journal of Immunopathology and Pharmacology, 24(2_suppl), 7–10. doi: 10.1177/03946320110240S202 [DOI] [PubMed] [Google Scholar]
  283. Martinelli M, Scapoli L, Pezzetti F, Carinci F, Carinci P, Stabellini G, … Tognon M (2001). C677T variant form at the MTHFR gene and CL/P: A risk factor for mothers? American Journal of Medical Genetics, 98(4), 357–360. doi: [DOI] [PubMed] [Google Scholar]
  284. Mason JM, Morrison DJ, Basson MA, & Licht JD (2006). Sprouty proteins: multifaceted negative-feedback regulators of receptor tyrosine kinase signaling. Trends Cell Biol, 16(1), 45–54. doi: 10.1016/j.tcb.2005.11.004 [DOI] [PubMed] [Google Scholar]
  285. Massagué J (2012). TGFβ signalling in context. Nature Reviews Molecular Cell Biology, 13(10), 616–630. doi: 10.1038/nrm3434 [DOI] [PMC free article] [PubMed] [Google Scholar]
  286. Matsumura K, Taketomi T, Yoshizaki K, Arai S, Sanui T, Yoshiga D, … Nakamura S (2011). Sprouty2 controls proliferation of palate mesenchymal cells via fibroblast growth factor signaling. Biochem Biophys Res Commun, 404(4), 1076–1082. doi: 10.1016/j.bbrc.2010.12.116 [DOI] [PubMed] [Google Scholar]
  287. McCauley DW, & Bronner-Fraser M (2004). Conservation and divergence of BMP2/4 genes in the lamprey: expression and phylogenetic analysis suggest a single ancestral vertebrate gene. Evolution & Development, 6(6), 411–422. doi: 10.1111/j.1525-142X.2004.04054.x 10.1111/j.1525–142X.2004.04054.x [DOI] [PubMed] [Google Scholar]
  288. McDonald-McGinn DM, Driscoll DA, Bason L, Christensen K, Lynch D, Sullivan K, … Zackai EH (1995). Autosomal dominant “Opitz” GBBB syndrome due to a 22q11. 2 deletion. American Journal of Medical Genetics, 59(1), 103–113. doi: 10.1002/ajmg.1320590122 [DOI] [PubMed] [Google Scholar]
  289. McDonald-McGinn DM, Sullivan KE, Marino B, Philip N, Swillen A, Vorstman JAS, … Bassett AS (2015). 22q11.2 deletion syndrome. Nature reviews. Disease primers, 1, 15071–15071. doi: 10.1038/nrdp.2015.71 [DOI] [PMC free article] [PubMed] [Google Scholar]
  290. McGrath JA, Duijf PH, Doetsch V, Irvine AD, de Waal R, Vanmolkot KR, … van Bokhoven H (2001). Hay-Wells syndrome is caused by heterozygous missense mutations in the SAM domain of p63. Hum Mol Genet, 10(3), 221–229. doi: 10.1093/hmg/10.3.221 [DOI] [PubMed] [Google Scholar]
  291. McMillin MJ, Beck AE, Chong JX, Shively KM, Buckingham KJ, Gildersleeve HI, … Bamshad MJ (2014). Mutations in PIEZO2 cause Gordon syndrome, Marden-Walker syndrome, and distal arthrogryposis type 5. American journal of human genetics, 94(5), 734–744. doi: 10.1016/j.ajhg.2014.03.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  292. Medici D, Hay ED, & Olsen BR (2008). Snail and Slug promote epithelial-mesenchymal transition through beta-catenin-T-cell factor-4-dependent expression of transforming growth factor-beta3. Molecular biology of the cell, 19(11), 4875–4887. doi: 10.1091/mbc.e08-05-0506 [DOI] [PMC free article] [PubMed] [Google Scholar]
  293. Medio M, Yeh E, Popelut A, Babajko S, Berdal A, & Helms JA (2012). Wnt/β-catenin signaling and Msx1 promote outgrowth of the maxillary prominences. Frontiers in physiology, 3, 375–375. doi: 10.3389/fphys.2012.00375 [DOI] [PMC free article] [PubMed] [Google Scholar]
  294. Metzis V, Courtney AD, Kerr MC, Ferguson C, Rondón Galeano MC, Parton RG, … Wicking C (2013). Patched1 is required in neural crest cells for the prevention of orofacial clefts. Human Molecular Genetics, 22(24), 5026–5035. doi: 10.1093/hmg/ddt353 [DOI] [PubMed] [Google Scholar]
  295. Meyers GA, Orlow SJ, Munro IR, Przylepa KA, & Jabs EW (1995). Fibroblast growth factor receptor 3 (FGFR3) transmembrane mutation in Crouzon syndrome with acanthosis nigricans. Nature genetics, 11(4), 462–464. doi: 10.1038/ng1295-462 [DOI] [PubMed] [Google Scholar]
  296. Mills JL, Molloy AM, Parle-McDermott A, Troendle JF, Brody LC, Conley MR, … Kirke PN (2008). Folate-related gene polymorphisms as risk factors for cleft lip and cleft palate. Birth defects research. Part A, Clinical and molecular teratology, 82(9), 636–643. doi: 10.1002/bdra.20491 [DOI] [PMC free article] [PubMed] [Google Scholar]
  297. Milunsky JM, Maher TA, Zhao G, Roberts AE, Stalker HJ, Zori RT, … Lin AE (2008). TFAP2A Mutations Result in Branchio-Oculo-Facial Syndrome. The American Journal of Human Genetics, 82(5), 1171–1177. doi: 10.1016/j.ajhg.2008.03.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  298. Mitchell K, O’Sullivan J, Missero C, Blair E, Richardson R, Anderson B, … Dixon MJ (2012). Exome sequence identifies RIPK4 as the Bartsocas-Papas syndrome locus. American journal of human genetics, 90(1), 69–75. doi: 10.1016/j.ajhg.2011.11.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  299. Mohamed JY, Faqeih E, Alsiddiky A, Alshammari MJ, Ibrahim NA, & Alkuraya FS (2013). Mutations in MEOX1, encoding mesenchyme homeobox 1, cause Klippel-Feil anomaly. American journal of human genetics, 92(1), 157–161. doi: 10.1016/j.ajhg.2012.11.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  300. Moreno LM, Mansilla MA, Bullard SA, Cooper ME, Busch TD, Machida J, … Lidral AC (2009). FOXE1 association with both isolated cleft lip with or without cleft palate, and isolated cleft palate. Hum Mol Genet, 18(24), 4879–4896. doi: 10.1093/hmg/ddp444 [DOI] [PMC free article] [PubMed] [Google Scholar]
  301. Morgan NV, Brueton LA, Cox P, Greally MT, Tolmie J, Pasha S, … Maher ER (2006). Mutations in the embryonal subunit of the acetylcholine receptor (CHRNG) cause lethal and Escobar variants of multiple pterygium syndrome. American journal of human genetics, 79(2), 390–395. doi: 10.1086/506256 [DOI] [PMC free article] [PubMed] [Google Scholar]
  302. Mori-Akiyama Y, Akiyama H, Rowitch DH, & de Crombrugghe B (2003). Sox9 is required for determination of the chondrogenic cell lineage in the cranial neural crest. Proceedings of the National Academy of Sciences, 100(16), 9360. doi: 10.1073/pnas.1631288100 [DOI] [PMC free article] [PubMed] [Google Scholar]
  303. Mostowska A, Gaczkowska A, Żukowski K, Ludwig KU, Hozyasz KK, Wójcicki P, … Jagodziński PP (2018). Common variants in DLG1 locus are associated with non-syndromic cleft lip with or without cleft palate. Clinical Genetics, 93(4), 784–793. doi: 10.1111/cge.13141 [DOI] [PubMed] [Google Scholar]
  304. Mostowska A, Hozyasz KK, Biedziak B, Wojcicki P, Lianeri M, & Jagodzinski PP (2012). Genotype and haplotype analysis of WNT genes in non-syndromic cleft lip with or without cleft palate. European Journal of Oral Sciences, 120(1), 1–8. doi: 10.1111/j.1600-0722.2011.00938.x 10.1111/j.1600–0722.2011.00938.x [DOI] [PubMed] [Google Scholar]
  305. Mostowska A, Hozyasz KK, & Jagodzinski PP (2006). Maternal MTR genotype contributes to the risk of non-syndromic cleft lip and palate in the Polish population. Clin Genet, 69(6), 512–517. doi: 10.1111/j.1399-0004.2006.00618.x [DOI] [PubMed] [Google Scholar]
  306. Murray SA, Oram KF, & Gridley T (2007). Multiple functions of Snail family genes during palate development in mice. Development, 134(9), 1789. doi: 10.1242/dev.02837 [DOI] [PubMed] [Google Scholar]
  307. Nakajima A, Ito Y, Asano M, Maeno M, Iwata K, Mitsui N, … Shuler CF (2007). Functional role of transforming growth factor-β type III receptor during palatal fusion. Developmental Dynamics, 236(3), 791–801. doi: 10.1002/dvdy.21090 [DOI] [PubMed] [Google Scholar]
  308. Nawshad A, Medici D, Liu C-C, & Hay ED (2007). TGFbeta3 inhibits E-cadherin gene expression in palate medial-edge epithelial cells through a Smad2-Smad4-LEF1 transcription complex. Journal of cell science, 120(Pt 9), 1646–1653. doi: 10.1242/jcs.003129 [DOI] [PMC free article] [PubMed] [Google Scholar]
  309. Niederreither K, & Dollé P (2008). Retinoic acid in development: towards an integrated view. Nature Reviews Genetics, 9(7), 541–553. doi: 10.1038/nrg2340 [DOI] [PubMed] [Google Scholar]
  310. Niemann S, Zhao C, Pascu F, Stahl U, Aulepp U, Niswander L, … Müller U (2004). Homozygous WNT3 mutation causes tetra-amelia in a large consanguineous family. American journal of human genetics, 74(3), 558–563. doi: 10.1086/382196 [DOI] [PMC free article] [PubMed] [Google Scholar]
  311. Nikopensius T, Jagomägi T, Krjutškov K, Tammekivi V, Saag M, Prane I, … Metspalu A (2010). Genetic variants in COL2A1, COL11A2, and IRF6 contribute risk to nonsyndromic cleft palate. Birth Defects Research Part A: Clinical and Molecular Teratology, 88(9), 748–756. doi: 10.1002/bdra.20700 [DOI] [PubMed] [Google Scholar]
  312. Nikopensius T, Kempa I, Ambrozaitytė L, Jagomägi T, Saag M, Matulevičienė A, … Metspalu A (2011). Variation in FGF1, FOXE1, and TIMP2genes is associated with nonsyndromic cleft lip with or without cleft palate. Birth Defects Research Part A: Clinical and Molecular Teratology, 91(4), 218–225. doi: 10.1002/bdra.20791 [DOI] [PubMed] [Google Scholar]
  313. Nishihara H, Kobayashi N, Kimura-Yoshida C, Yan K, Bormuth O, Ding Q, … Okada N (2016). Coordinately Co-opted Multiple Transposable Elements Constitute an Enhancer for wnt5a Expression in the Mammalian Secondary Palate. PLoS genetics, 12(10), e1006380–e1006380. doi: 10.1371/journal.pgen.1006380 [DOI] [PMC free article] [PubMed] [Google Scholar]
  314. Noda K, Mishina Y, & Komatsu Y (2016). Constitutively active mutation of ACVR1 in oral epithelium causes submucous cleft palate in mice. Animal models of craniofacial anomalies, 415(2), 306–313. doi: 10.1016/j.ydbio.2015.06.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  315. Nusse R, & Clevers H (2017). Wnt/β-Catenin Signaling, Disease, and Emerging Therapeutic Modalities. Cell, 169(6), 985–999. doi: 10.1016/j.cell.2017.05.016 [DOI] [PubMed] [Google Scholar]
  316. Ockeloen CW, Khandelwal KD, Dreesen K, Ludwig KU, Sullivan R, van Rooij IALM, … Carels CEL (2016). Novel mutations in LRP6 highlight the role of WNT signaling in tooth agenesis. Genetics In Medicine, 18, 1158 Retrieved from 10.1038/gim.2016.10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  317. Okano J, Kimura W, Papaionnou VE, Miura N, Yamada G, Shiota K, & Sakai Y (2012). The regulation of endogenous retinoic acid level through CYP26B1 is required for elevation of palatal shelves. Developmental Dynamics, 241(11), 1744–1756. doi: 10.1002/dvdy.23862 10.1002/dvdy.23862 [DOI] [PubMed] [Google Scholar]
  318. Okano J, Sakai Y, & Shiota K (2008). Retinoic acid down-regulates Tbx1 expression and induces abnormal differentiation of tongue muscles in fetal mice. Developmental Dynamics, 237(10), 3059–3070. doi: 10.1002/dvdy.21715 10.1002/dvdy.21715 [DOI] [PubMed] [Google Scholar]
  319. Okano J, Udagawa J, & Shiota K (2014). Roles of retinoic acid signaling in normal and abnormal development of the palate and tongue. Congenital Anomalies, 54(2), 69–76. doi: 10.1111/cga.12049 [DOI] [PubMed] [Google Scholar]
  320. Ornitz DM, & Itoh N (2015). The Fibroblast Growth Factor signaling pathway. Wiley interdisciplinary reviews. Developmental biology, 4(3), 215–266. doi: 10.1002/wdev.176 [DOI] [PMC free article] [PubMed] [Google Scholar]
  321. Pan X, Wang P, Yin X, Liu X, Li D, Li X, … Yu Z (2015). Association between Maternal MTHFR Polymorphisms and Nonsyndromic Cleft Lip with or without Cleft Palate in Offspring, A Meta-Analysis Based on 15 Case-Control Studies. International journal of fertility & sterility, 8(4), 463–480. doi: 10.22074/ijfs.2015.4186 [DOI] [PMC free article] [PubMed] [Google Scholar]
  322. Pantalacci S, Prochazka J, Martin A, Rothova M, Lambert A, Bernard L, … Laudet V (2008). Patterning of palatal rugae through sequential addition reveals an anterior/posterior boundary in palatal development. BMC Developmental Biology, 8(1), 116. doi: 10.1186/1471-213X-8-116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  323. Parada-Sanchez MT, Chu EY, Cox LL, Undurty SS, Standley JM, Murray JC, & Cox TC (2017). Disrupted IRF6-NME1/2 Complexes as a Cause of Cleft Lip/Palate. J Dent Res, 96(11), 1330–1338. doi: 10.1177/0022034517723615 [DOI] [PMC free article] [PubMed] [Google Scholar]
  324. Parada C, & Chai Y (2012). Roles of BMP Signaling Pathway in Lip and Palate Development. In Frontiers of Oral Biology (Vol. 16, pp. 60–70). [DOI] [PMC free article] [PubMed] [Google Scholar]
  325. Parada C, Li J, Iwata J, Suzuki A, & Chai Y (2013). CTGF mediates Smad-dependent transforming growth factor β signaling to regulate mesenchymal cell proliferation during palate development. Molecular and Cellular Biology, 33(17), 3482–3493. doi: 10.1128/MCB.00615-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  326. Park JW, Cai J, McIntosh I, Jabs EW, Fallin MD, Ingersoll R, … Beaty TH (2006). High throughput SNP and expression analyses of candidate genes for nonsyndromic oral clefts. Journal of medical genetics, 43(7), 598–608. doi: 10.1136/jmg.2005.040162 [DOI] [PMC free article] [PubMed] [Google Scholar]
  327. Park JW, McIntosh I, Hetmanski JB, Jabs EW, Kolk CAV, Wu-Chou Y-H, … Beaty TH (2007). Association between IRF6 and nonsyndromic cleft lip with or without cleft palate in four populations. Genetics in Medicine, 9(4), 219–227. doi: 10.1097/GIM.0b013e3180423cca [DOI] [PMC free article] [PubMed] [Google Scholar]
  328. Pauws E, Hoshino A, Bentley L, Prajapati S, Keller C, Hammond P, … Stanier P (2009). Tbx22null mice have a submucous cleft palate due to reduced palatal bone formation and also display ankyloglossia and choanal atresia phenotypes. Human Molecular Genetics, 18(21), 4171–4179. doi: 10.1093/hmg/ddp368 [DOI] [PMC free article] [PubMed] [Google Scholar]
  329. Paznekas WA, Karczeski B, Vermeer S, Lowry RB, Delatycki M, Laurence F, … Wang Jabs E (2009). GJA1 mutations, variants, and connexin 43 dysfunction as it relates to the oculodentodigital dysplasia phenotype. Human Mutation, 30(5), 724–733. doi: 10.1002/humu.20958 [DOI] [PubMed] [Google Scholar]
  330. Peanchitlertkajorn S, Cooper ME, Liu YE, Field LL, & Marazita ML (2003). Chromosome 17: gene mapping studies of cleft lip with or without cleft palate in Chinese families. The Cleft palate-craniofacial journal : official publication of the American Cleft Palate-Craniofacial Association, 40(1), 71–79. doi: 10.1597/1545-1569_2003_040_0071_cgmsoc_2.0.co_2 [DOI] [PubMed] [Google Scholar]
  331. Person AD, Beiraghi S, Sieben CM, Hermanson S, Neumann AN, Robu ME, … Lohr JL (2010). WNT5A mutations in patients with autosomal dominant Robinow syndrome. Dev Dyn, 239(1), 327–337. doi: 10.1002/dvdy.22156 [DOI] [PMC free article] [PubMed] [Google Scholar]
  332. Peyrard-Janvid M, Leslie EJ, Kousa YA, Smith TL, Dunnwald M, Magnusson M, … Schutte BC (2014). Dominant mutations in GRHL3 cause Van der Woude Syndrome and disrupt oral periderm development. American journal of human genetics, 94(1), 23–32. doi: 10.1016/j.ajhg.2013.11.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  333. Pezzetti F, Martinelli M, Scapoli L, Carinci F, Palmieri A, Marchesini J, … Tognon M (2004). Maternal MTHFR variant forms increase the risk in offspring of isolated nonsyndromic cleft lip with or without cleft palate. Human Mutation, 24(1), 104–105. doi: 10.1002/humu.9257 [DOI] [PubMed] [Google Scholar]
  334. Pitteloud N, Acierno JS Jr., Meysing A, Eliseenkova AV, Ma J, Ibrahimi OA, … Crowley WF Jr. (2006). Mutations in fibroblast growth factor receptor 1 cause both Kallmann syndrome and normosmic idiopathic hypogonadotropic hypogonadism. Proceedings of the National Academy of Sciences of the United States of America, 103(16), 6281–6286. doi: 10.1073/pnas.0600962103 [DOI] [PMC free article] [PubMed] [Google Scholar]
  335. Pogenberg V, Guichou J-F, Vivat-Hannah V, Kammerer S, Pérez E, Germain P, … Bourguet W (2005). Characterization of the Interaction between Retinoic Acid Receptor/Retinoid X Receptor (RAR/RXR) Heterodimers and Transcriptional Coactivators through Structural and Fluorescence Anisotropy Studies. Journal of Biological Chemistry, 280(2), 1625–1633. doi: 10.1074/jbc.M409302200 [DOI] [PubMed] [Google Scholar]
  336. Prescott NJ, Lees MM, Winter RM, & Malcolm S (2000). Identification of susceptibility loci for nonsyndromic cleft lip with or without cleft palate in a two stage genome scan of affected sib-pairs. Hum Genet, 106(3), 345–350. doi: 10.1007/s004390051048 [DOI] [PubMed] [Google Scholar]
  337. Proetzel G, Pawlowski SA, Wiles MV, Yin M, Boivin GP, Howles PN, … Doetschman T (1995). Transforming growth factor-beta 3 is required for secondary palate fusion. Nature Genetics, 11(4), 409–414. doi: 10.1038/ng1295-409 [DOI] [PMC free article] [PubMed] [Google Scholar]
  338. Prontera P, Rogaia D, Sallicandro E, Mencarelli A, Imperatore V, Squeo GM, … Stangoni G (2019). Schilbach-Rott syndrome associated with 9q22.32q22.33 duplication, involving the PTCH1 gene. Eur J Hum Genet, 27(8), 1260–1266. doi: 10.1038/s41431-019-0385-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  339. Qian D, Jones C, Rzadzinska A, Mark S, Zhang X, Steel KP, … Chen P (2007). Wnt5a functions in planar cell polarity regulation in mice. Developmental Biology, 306(1), 121–133. doi: 10.1016/j.ydbio.2007.03.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  340. Quaderi NA, Schweiger S, Gaudenz K, Franco B, Rugarli EI, Berger W, … Ballabio A (1997). Opitz G/BBB syndrome, a defect of midline development, is due to mutations in a new RING finger gene on Xp22. Nature Genetics, 17(3), 285–291. doi: 10.1038/ng1197-285 [DOI] [PubMed] [Google Scholar]
  341. Quadro L, Blaner WS, Salchow DJ, Vogel S, Piantedosi R, Gouras P, … Gottesman ME (1999). Impaired retinal function and vitamin A availability in mice lacking retinol-binding protein. The EMBO journal, 18(17), 4633–4644. doi: 10.1093/emboj/18.17.4633 [DOI] [PMC free article] [PubMed] [Google Scholar]
  342. Racedo SE, Hasten E, Lin M, Devakanmalai GS, Guo T, Ozbudak EM, … Morrow BE (2017). Reduced dosage of β-catenin provides significant rescue of cardiac outflow tract anomalies in a Tbx1 conditional null mouse model of 22q11.2 deletion syndrome. PLOS Genetics, 13(3), e1006687. doi: 10.1371/journal.pgen.1006687 [DOI] [PMC free article] [PubMed] [Google Scholar]
  343. Reardon W, Winter RM, Rutland P, Pulleyn LJ, Jones BM, & Malcolm S (1994). Mutations in the fibroblast growth factor receptor 2 gene cause Crouzon syndrome. Nat Genet, 8(1), 98–103. doi: 10.1038/ng0994-98 [DOI] [PubMed] [Google Scholar]
  344. Renard C-A, Labalette C, Armengol C, Cougot D, Wei Y, Cairo S, … Buendia M-A (2007). Tbx3 Is a Downstream Target of the Wnt/β-Catenin Pathway and a Critical Mediator of β-Catenin Survival Functions in Liver Cancer. Cancer Research, 67(3), 901. doi: 10.1158/0008-5472.CAN-06-2344 [DOI] [PubMed] [Google Scholar]
  345. Reynolds K, Kumari P, Sepulveda Rincon L, Gu R, Ji Y, Kumar S, & Zhou CJ (2019). Wnt signaling in orofacial clefts: crosstalk, pathogenesis and models. Disease Models & Mechanisms, 12(2), dmm037051. doi: 10.1242/dmm.037051 [DOI] [PMC free article] [PubMed] [Google Scholar]
  346. Rhinn M, & Dollé P (2012). Retinoic acid signalling during development. Development, 139(5), 843. doi: 10.1242/dev.065938 [DOI] [PubMed] [Google Scholar]
  347. Ribeiro LA, Murray JC, & Richieri-Costa A (2006). PTCH mutations in four Brazilian patients with holoprosencephaly and in one with holoprosencephaly-like features and normal MRI. Am J Med Genet A, 140(23), 2584–2586. doi: 10.1002/ajmg.a.31369 [DOI] [PubMed] [Google Scholar]
  348. Rice R, Spencer-Dene B, Connor EC, Gritli-Linde A, McMahon AP, Dickson C, … Rice DPC (2004). Disruption of Fgf10/Fgfr2b-coordinated epithelial-mesenchymal interactions causes cleft palate. The Journal of Clinical Investigation, 113(12), 1692–1700. doi: 10.1172/JCI20384 [DOI] [PMC free article] [PubMed] [Google Scholar]
  349. Richardson R, Mitchell K, Hammond NL, Mollo MR, Kouwenhoven EN, Wyatt ND, … Dixon J (2017). p63 exerts spatio-temporal control of palatal epithelial cell fate to prevent cleft palate. PLoS genetics, 13(6), e1006828. doi: 10.1371/journal.pgen.1006828 [DOI] [PMC free article] [PubMed] [Google Scholar]
  350. Richardson RJ, Dixon J, Malhotra S, Hardman MJ, Knowles L, Boot-Handford RP, … Dixon MJ (2006). Irf6 is a key determinant of the keratinocyte proliferation-differentiation switch. Nature Genetics, 38(11), 1329–1334. doi: 10.1038/ng1894 [DOI] [PubMed] [Google Scholar]
  351. Rieder MJ, Green GE, Park SS, Stamper BD, Gordon CT, Johnson JM, … Cunningham ML (2012). A human homeotic transformation resulting from mutations in PLCB4 and GNAI3 causes auriculocondylar syndrome. American journal of human genetics, 90(5), 907–914. doi: 10.1016/j.ajhg.2012.04.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  352. Riley BM, Mansilla MA, Ma J, Daack-Hirsch S, Maher BS, Raffensperger LM, … Murray JC (2007). Impaired FGF signaling contributes to cleft lip and palate. Proceedings of the National Academy of Sciences of the United States of America, 104(11), 4512–4517. doi: 10.1073/pnas.0607956104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  353. Rizos M, Negrón RJ, & Serman N (1998). Möbius Syndrome with Dental Involvement: A Case Report and Literature Review. The Cleft Palate-Craniofacial Journal, 35(3), 262–268. doi: 10.1597/1545-1569_1998_035_0262_mbswdi_2.3.co_2 [DOI] [PubMed] [Google Scholar]
  354. Roberts C, Ivins S, Cook AC, Baldini A, & Scambler PJ (2006). Cyp26 genes a1, b1 and c1 are down-regulated in Tbx1 null mice and inhibition of Cyp26 enzyme function produces a phenocopy of DiGeorge Syndrome in the chick. Human Molecular Genetics, 15(23), 3394–3410. doi: 10.1093/hmg/ddl416 [DOI] [PubMed] [Google Scholar]
  355. Robertson SP, Twigg SRF, Sutherland-Smith AJ, Biancalana V, Gorlin RJ, Horn D, … The, O. P. D. s. D. C. C. G. (2003). Localized mutations in the gene encoding the cytoskeletal protein filamin A cause diverse malformations in humans. Nature Genetics, 33(4), 487–491. doi: 10.1038/ng1119 [DOI] [PubMed] [Google Scholar]
  356. Roessler E, Belloni E, Gaudenz K, Jay P, Berta P, Scherer SW, … Muenke M (1996). Mutations in the human Sonic Hedgehog gene cause holoprosencephaly. Nat Genet, 14(3), 357–360. doi: 10.1038/ng1196-357 [DOI] [PubMed] [Google Scholar]
  357. Roessler E, Du Y-Z, Mullor JL, Casas E, Allen WP, Gillessen-Kaesbach G, … Muenke M (2003). Loss-of-function mutations in the human GLI2 gene are associated with pituitary anomalies and holoprosencephaly-like features. Proceedings of the National Academy of Sciences, 100(23), 13424. doi: 10.1073/pnas.2235734100 [DOI] [PMC free article] [PubMed] [Google Scholar]
  358. Roessler E, Ma Y, Ouspenskaia MV, Lacbawan F, Bendavid C, Dubourg C, … Muenke M (2009). Truncating loss-of-function mutations of DISP1 contribute to holoprosencephaly-like microform features in humans. Human Genetics, 125(4), 393–400. doi: 10.1007/s00439-009-0628-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  359. Rohanizadegan M, & Sacharow S (2018). Desmosterolosis presenting with multiple congenital anomalies. Eur J Med Genet, 61(3), 152–156. doi: 10.1016/j.ejmg.2017.11.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  360. Rooryck C, Diaz-Font A, Osborn DP, Chabchoub E, Hernandez-Hernandez V, Shamseldin H, … Beales PL (2011). Mutations in lectin complement pathway genes COLEC11 and MASP1 cause 3MC syndrome. Nat Genet, 43(3), 197–203. doi: 10.1038/ng.757 [DOI] [PMC free article] [PubMed] [Google Scholar]
  361. Rutherford EL, & Lowery LA (2016). Exploring the developmental mechanisms underlying Wolf-Hirschhorn Syndrome: Evidence for defects in neural crest cell migration. Developmental Biology, 420(1), 1–10. doi: 10.1016/j.ydbio.2016.10.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  362. Saal HM, Prows CA, Guerreiro I, Donlin M, Knudson L, Sund KL, … Stottmann RW (2015). A mutation in FRIZZLED2 impairs Wnt signaling and causes autosomal dominant omodysplasia. Human Molecular Genetics, 24(12), 3399–3409. doi: 10.1093/hmg/ddv088 [DOI] [PMC free article] [PubMed] [Google Scholar]
  363. Sahoo T, Theisen A, Sanchez-Lara PA, Marble M, Schweitzer DN, Torchia BS, … Lacassie Y (2011). Microdeletion 20p12.3 involving BMP2 contributes to syndromic forms of cleft palate. American journal of medical genetics. Part A, 155A(7), 1646–1653. doi: 10.1002/ajmg.a.34063 [DOI] [PMC free article] [PubMed] [Google Scholar]
  364. Sandell LL, Sanderson BW, Moiseyev G, Johnson T, Mushegian A, Young K, … Trainor PA (2007). RDH10 is essential for synthesis of embryonic retinoic acid and is required for limb, craniofacial, and organ development. Genes & Development, 21(9), 1113–1124. doi: 10.1101/gad.1533407 [DOI] [PMC free article] [PubMed] [Google Scholar]
  365. Scapoli L, Palmieri A, Martinelli M, Pezzetti F, Carinci P, Tognon M, & Carinci F (2005). Strong Evidence of Linkage Disequilibrium between Polymorphisms at the IRF6 Locus and Nonsyndromic Cleft Lip With or Without Cleft Palate, in an Italian Population. The American Journal of Human Genetics, 76(1), 180–183. doi: 10.1086/427344 [DOI] [PMC free article] [PubMed] [Google Scholar]
  366. Schneider RA, Hu D, Rubenstein JLR, Maden M, & Helms JA (2001). Local retinoid signaling coordinates forebrain and facial morphogenesis by maintaining FGF8 and SHH. Development, 128(14), 2755 Retrieved from http://dev.biologists.org/content/128/14/2755.abstract [DOI] [PubMed] [Google Scholar]
  367. Schwabe GC, Trepczik B, Süring K, Brieske N, Tucker AS, Sharpe PT, … Mundlos S (2004). Ror2 knockout mouse as a model for the developmental pathology of autosomal recessive Robinow syndrome. Developmental Dynamics, 229(2), 400–410. doi: 10.1002/dvdy.10466 10.1002/dvdy.10466 [DOI] [PubMed] [Google Scholar]
  368. Shaheen R, Faqeih E, Seidahmed MZ, Sunker A, Alali FE, Khadijah A, & Alkuraya FS (2011). A TCTN2 mutation defines a novel Meckel Gruber syndrome locus. Human Mutation, 32(6), 573–578. doi: 10.1002/humu.21507 [DOI] [PubMed] [Google Scholar]
  369. Shamseldin HE, Rajab A, Alhashem A, Shaheen R, Al-Shidi T, Alamro R, … Alkuraya FS (2013). Mutations in DDX59 implicate RNA helicase in the pathogenesis of orofaciodigital syndrome. American journal of human genetics, 93(3), 555–560. doi: 10.1016/j.ajhg.2013.07.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  370. Shaw GM, Nelson V, Iovannisci DM, Finnell RH, & Lammer EJ (2003). Maternal Occupational Chemical Exposures and Biotransformation Genotypes as Risk Factors for Selected Congenital Anomalies. American Journal of Epidemiology, 157(6), 475–484. doi: 10.1093/aje/kwg013 [DOI] [PubMed] [Google Scholar]
  371. Shi M, Christensen K, Weinberg CR, Romitti P, Bathum L, Lozada A, … Murray JC (2007). Orofacial cleft risk is increased with maternal smoking and specific detoxification-gene variants. American journal of human genetics, 80(1), 76–90. doi: 10.1086/510518 [DOI] [PMC free article] [PubMed] [Google Scholar]
  372. Shi M, Mostowska A, Jugessur A, Johnson MK, Mansilla MA, Christensen K, … Murray JC (2009). Identification of microdeletions in candidate genes for cleft lip and/or palate. Birth Defects Res A Clin Mol Teratol, 85(1), 42–51. doi: 10.1002/bdra.20571 [DOI] [PMC free article] [PubMed] [Google Scholar]
  373. Shi Y, & Massagué J (2003). Mechanisms of TGF-β Signaling from Cell Membrane to the Nucleus. Cell, 113(6), 685–700. doi: 10.1016/S0092-8674(03)00432-X [DOI] [PubMed] [Google Scholar]
  374. Shimomura T, Kawakami M, Tatsumi K, Tanaka T, Morita-Takemura S, Kirita T, & Wanaka A (2019). The Role of the Wnt Signaling Pathway in Upper Jaw Development of Chick Embryo. Acta histochemica et cytochemica, 52(1), 19–26. doi: 10.1267/ahc.18038 [DOI] [PMC free article] [PubMed] [Google Scholar]
  375. Shu W, Guttentag S, Wang Z, Andl T, Ballard P, Lu MM, … Morrisey EE (2005). Wnt/β-catenin signaling acts upstream of N-myc, BMP4, and FGF signaling to regulate proximal–distal patterning in the lung. Developmental Biology, 283(1), 226–239. doi: 10.1016/j.ydbio.2005.04.014 [DOI] [PubMed] [Google Scholar]
  376. Sirard C, de la Pompa JL, Elia A, Itie A, Mirtsos C, Cheung A, … Mak TW (1998). The tumor suppressor gene Smad4/Dpc4 is required for gastrulation and later for anterior development of the mouse embryo. Genes & Development, 12(1), 107–119. doi: 10.1101/gad.12.1.107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  377. Slaney SF, Oldridge M, Hurst JA, Moriss-Kay GM, Hall CM, Poole MD, & Wilkie AO (1996). Differential effects of FGFR2 mutations on syndactyly and cleft palate in Apert syndrome. American journal of human genetics, 58(5), 923–932. Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/8651276https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1914616/ [PMC free article] [PubMed] [Google Scholar]
  378. Smith TM, Wang X, Zhang W, Kulyk W, & Nazarali AJ (2009). Hoxa2 plays a direct role in murine palate development. Developmental Dynamics, 238(9), 2364–2373. doi: 10.1002/dvdy.22040 10.1002/dvdy.22040 [DOI] [PubMed] [Google Scholar]
  379. Smith UM, Consugar M, Tee LJ, McKee BM, Maina EN, Whelan S, … Johnson CA (2006). The transmembrane protein meckelin (MKS3) is mutated in Meckel-Gruber syndrome and the wpk rat. Nat Genet, 38(2), 191–196. doi: 10.1038/ng1713 [DOI] [PubMed] [Google Scholar]
  380. Snyder-Warwick AK, Perlyn CA, Pan J, Yu K, Zhang L, & Ornitz DM (2010). Analysis of a gain-of-function FGFR2 Crouzon mutation provides evidence of loss of function activity in the etiology of cleft palate. Proc Natl Acad Sci U S A, 107(6), 2515–2520. doi: 10.1073/pnas.0913985107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  381. Soghani B, Ebadifar A, Khorram Khorshid HR, Kamali K, Hamedi R, & Aghakhani Moghadam F (2017). The study of association between reduced folate carrier 1 (RFC1) polymorphism and non-syndromic cleft lip/palate in Iranian population. Bioimpacts, 7(4), 263–268. doi: 10.15171/bi.2017.31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  382. Solomon BD, Lacbawan F, Mercier S, Clegg NJ, Delgado MR, Rosenbaum K, … Muenke M (2010). Mutations in ZIC2 in human holoprosencephaly: description of a novel ZIC2 specific phenotype and comprehensive analysis of 157 individuals. Journal of medical genetics, 47(8), 513–524. doi: 10.1136/jmg.2009.073049 [DOI] [PMC free article] [PubMed] [Google Scholar]
  383. Song L, Li Y, Wang K, Wang Y-Z, Molotkov A, Gao L, … Zhou CJ (2009). Lrp6-mediated canonical Wnt signaling is required for lip formation and fusion. Development, 136(18), 3161. doi: 10.1242/dev.037440 [DOI] [PubMed] [Google Scholar]
  384. Song Y, Hui JN, Fu KK, & Richman JM (2004). Control of retinoic acid synthesis and FGF expression in the nasal pit is required to pattern the craniofacial skeleton. Developmental Biology, 276(2), 313–329. doi: 10.1016/j.ydbio.2004.08.035 [DOI] [PubMed] [Google Scholar]
  385. Song Z, Liu C, Iwata J, Gu S, Suzuki A, Sun C, … Chen Y (2013). Mice with Tak1 deficiency in neural crest lineage exhibit cleft palate associated with abnormal tongue development. The Journal of biological chemistry, 288(15), 10440–10450. doi: 10.1074/jbc.M112.432286 [DOI] [PMC free article] [PubMed] [Google Scholar]
  386. Stamm DS, Aylsworth AS, Stajich JM, Kahler SG, Thorne LB, Speer MC, & Powell CM (2008). Native American myopathy: Congenital myopathy with cleft palate, skeletal anomalies, and susceptibility to malignant hyperthermia. American Journal of Medical Genetics Part A, 146A(14), 1832–1841. doi: 10.1002/ajmg.a.32370 [DOI] [PubMed] [Google Scholar]
  387. Stamos JL, & Weis WI (2013). The β-catenin destruction complex. Cold Spring Harbor Perspectives in Biology, 5(1), a007898–a007898. doi: 10.1101/cshperspect.a007898 [DOI] [PMC free article] [PubMed] [Google Scholar]
  388. Stevenson RE, Bennett CW, Abidi F, Kleefstra T, Porteous M, Simensen RJ, … Schwartz CE (2005). Renpenning syndrome comes into focus. American Journal of Medical Genetics Part A, 134A(4), 415–421. doi: 10.1002/ajmg.a.30664 [DOI] [PubMed] [Google Scholar]
  389. Stoler JM, Rosen H, Desai U, Mulliken JB, Meara JG, & Rogers GF (2009). Cleft palate in Pfeiffer syndrome. J Craniofac Surg, 20(5), 1375–1377. doi: 10.1097/SCS.0b013e3181ae42e4 [DOI] [PubMed] [Google Scholar]
  390. Strutt D (2008). The planar polarity pathway. Current Biology, 18(19), R898–R902. doi: 10.1016/j.cub.2008.07.055 [DOI] [PubMed] [Google Scholar]
  391. Suazo J, Santos JL, Carreno H, Jara L, & Blanco R (2004). Linkage disequilibrium between MSX1 and non-syndromic cleft lip/palate in the Chilean population. J Dent Res, 83(10), 782–785. doi: 10.1177/154405910408301009 [DOI] [PubMed] [Google Scholar]
  392. Suazo J, Santos JL, Jara L, & Blanco R (2009). Association Between Bone Morphogenetic Protein 4 Gene Polymorphisms with Nonsyndromic Cleft Lip with or without Cleft Palate in a Chilean Population. DNA and Cell Biology, 29(2), 59–64. doi: 10.1089/dna.2009.0944 10.1089/dna.2009.0944 [DOI] [PubMed] [Google Scholar]
  393. Sun Y, Huang Y, Yin A, Pan Y, Wang Y, Wang C, … Yang Y (2015). Genome-wide association study identifies a new susceptibility locus for cleft lip with or without a cleft palate. Nature communications, 6(1), 6414. doi: 10.1038/ncomms7414 [DOI] [PubMed] [Google Scholar]
  394. Suzuki A, Ueno N, & Hemmati-Brivanlou A (1997). Xenopus msx1 mediates epidermal induction and neural inhibition by BMP4. Development, 124(16), 3037 Retrieved from http://dev.biologists.org/content/124/16/3037.abstract [DOI] [PubMed] [Google Scholar]
  395. Suzuki Y, Jezewski PA, Machida J, Watanabe Y, Shi M, Cooper ME, … Murray JC (2004). In a Vietnamese population, MSX1 variants contribute to cleft lip and palate. Genetics in Medicine, 6(3), 117–125. doi: 10.1097/01.GIM.0000127275.52925.05 [DOI] [PubMed] [Google Scholar]
  396. Szenker-Ravi E, Altunoglu U, Leushacke M, Bosso-Lefèvre C, Khatoo M, Thi Tran H, … Reversade B (2018). RSPO2 inhibition of RNF43 and ZNRF3 governs limb development independently of LGR4/5/6. Nature, 557(7706), 564–569. doi: 10.1038/s41586-018-0118-y [DOI] [PubMed] [Google Scholar]
  397. Tada M, & Smith JC (2000). Xwnt11 is a target of Xenopus Brachyury: regulation of gastrulation movements via Dishevelled, but not through the canonical Wnt pathway. Development, 127(10), 2227 Retrieved from http://dev.biologists.org/content/127/10/2227.abstract [DOI] [PubMed] [Google Scholar]
  398. Taniguchi K, Ayada T, Ichiyama K, Kohno R, Yonemitsu Y, Minami Y, … Yoshimura A (2007). Sprouty2 and Sprouty4 are essential for embryonic morphogenesis and regulation of FGF signaling. Biochem Biophys Res Commun, 352(4), 896–902. doi: 10.1016/j.bbrc.2006.11.107 [DOI] [PubMed] [Google Scholar]
  399. Tassabehji M, Fang ZM, Hilton EN, McGaughran J, Zhao Z, de Bock CE, … Clarke RA (2008). Mutations in GDF6 are associated with vertebral segmentation defects in Klippel-Feil syndrome. Human Mutation, 29(8), 1017–1027. doi: 10.1002/humu.20741 [DOI] [PubMed] [Google Scholar]
  400. Tassabehji M, Read AP, Newton VE, Harris R, Balling R, Gruss P, & Strachan T (1992). Waardenburg’s syndrome patients have mutations in the human homologue of the Pax-3 paired box gene. Nature, 355(6361), 635–636. doi: 10.1038/355635a0 [DOI] [PubMed] [Google Scholar]
  401. Tenorio J, Arias P, Martínez-Glez V, Santos F, García-Miñaur S, Nevado J, & Lapunzina P (2014). Simpson-Golabi-Behmel syndrome types I and II. Orphanet Journal of Rare Diseases, 9(1), 138. doi: 10.1186/s13023-014-0138-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  402. Thauvin-Robinet C, Lee JS, Lopez E, Herranz-Pérez V, Shida T, Franco B, … Nachury MV (2014). The oral-facial-digital syndrome gene C2CD3 encodes a positive regulator of centriole elongation. Nature Genetics, 46(8), 905–911. doi: 10.1038/ng.3031 [DOI] [PMC free article] [PubMed] [Google Scholar]
  403. Thomas S, Legendre M, Saunier S, Bessières B, Alby C, Bonnière M, … Attié-Bitach T (2012). TCTN3 mutations cause Mohr-Majewski syndrome. American journal of human genetics, 91(2), 372–378. doi: 10.1016/j.ajhg.2012.06.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  404. Thomason HA, Dixon MJ, & Dixon J (2008). Facial clefting in Tp63 deficient mice results from altered Bmp4, Fgf8 and Shh signaling. Developmental Biology, 321(1), 273–282. doi: 10.1016/j.ydbio.2008.06.030 [DOI] [PubMed] [Google Scholar]
  405. Thomason HA, Dixon MJ, & Dixon J (2008). Facial clefting in Tp63 deficient mice results from altered Bmp4, Fgf8 and Shh signaling. Developmental biology, 321(1), 273–282. doi: 10.1016/j.ydbio.2008.06.030 [DOI] [PubMed] [Google Scholar]
  406. Thomason HA, Zhou H, Kouwenhoven EN, Dotto G-P, Restivo G, Nguyen B-C, … Dixon J (2010). Cooperation between the transcription factors p63 and IRF6 is essential to prevent cleft palate in mice. The Journal of Clinical Investigation, 120(5), 1561–1569. doi: 10.1172/JCI40266 [DOI] [PMC free article] [PubMed] [Google Scholar]
  407. Trainor PA, Dixon J, & Dixon MJ (2009). Treacher Collins syndrome: etiology, pathogenesis and prevention. European Journal of Human Genetics, 17(3), 275–283. doi: 10.1038/ejhg.2008.221 [DOI] [PMC free article] [PubMed] [Google Scholar]
  408. Tríbulo C, Aybar MJ, Nguyen VH, Mullins MC, & Mayor R (2003). Regulation of Msx genes by a Bmp gradient is essential for neural crest specification. Development, 130(26), 6441. doi: 10.1242/dev.00878 [DOI] [PubMed] [Google Scholar]
  409. Tristani-Firouzi M, Jensen JL, Donaldson MR, Sansone V, Meola G, Hahn A, … Tawil R (2002). Functional and clinical characterization of KCNJ2 mutations associated with LQT7 (Andersen syndrome). J Clin Invest, 110(3), 381–388. doi: 10.1172/jci15183 [DOI] [PMC free article] [PubMed] [Google Scholar]
  410. Trumpp A, Depew MJ, Rubenstein JLR, Bishop JM, & Martin GR (1999). Cre-mediated gene inactivation demonstrates that FGF8 is required for cell survival and patterning of the first branchial arch. Genes & Development, 13(23), 3136–3148. Retrieved from http://genesdev.cshlp.org/content/13/23/3136.abstract [DOI] [PMC free article] [PubMed] [Google Scholar]
  411. Twigg SRF, Kan R, Babbs C, Bochukova EG, Robertson SP, Wall SA, … Wilkie AOM (2004). Mutations of ephrin-B1 (EFNB1), a marker of tissue boundary formation, cause craniofrontonasal syndrome. Proceedings of the National Academy of Sciences of the United States of America, 101(23), 8652–8657. doi: 10.1073/pnas.0402819101 [DOI] [PMC free article] [PubMed] [Google Scholar]
  412. Twigg SRF, Versnel SL, Nürnberg G, Lees MM, Bhat M, Hammond P, … Wilkie AOM (2009). Frontorhiny, a distinctive presentation of frontonasal dysplasia caused by recessive mutations in the ALX3 homeobox gene. American journal of human genetics, 84(5), 698–705. doi: 10.1016/j.ajhg.2009.04.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  413. Uslu VV, Petretich M, Ruf S, Langenfeld K, Fonseca NA, Marioni JC, & Spitz F (2014). Long-range enhancers regulating Myc expression are required for normal facial morphogenesis. Nature Genetics, 46(7), 753–758. doi: 10.1038/ng.2971 [DOI] [PubMed] [Google Scholar]
  414. Uz E, Alanay Y, Aktas D, Vargel I, Gucer S, Tuncbilek G, … Akarsu NA (2010). Disruption of ALX1 causes extreme microphthalmia and severe facial clefting: expanding the spectrum of autosomal-recessive ALX-related frontonasal dysplasia. American journal of human genetics, 86(5), 789–796. doi: 10.1016/j.ajhg.2010.04.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  415. Vajsar J, Baskin B, Swoboda K, Biggar DW, Schachter H, & Ray PN (2008). Walker-Warburg Syndrome with POMT1 mutations can be associated with cleft lip and cleft palate. Neuromuscular Disorders, 18(8), 675–677. doi: 10.1016/j.nmd.2008.05.014 [DOI] [PubMed] [Google Scholar]
  416. Valente EM, Logan CV, Mougou-Zerelli S, Lee JH, Silhavy JL, Brancati F, … Gleeson JG (2010). Mutations in TMEM216 perturb ciliogenesis and cause Joubert, Meckel and related syndromes. Nature Genetics, 42(7), 619–625. doi: 10.1038/ng.594 [DOI] [PMC free article] [PubMed] [Google Scholar]
  417. van Amerongen R (2012). Alternative Wnt Pathways and Receptors. Cold Spring Harbor Perspectives in Biology, 4(10). doi: 10.1101/cshperspect.a007914 [DOI] [PMC free article] [PubMed] [Google Scholar]
  418. van Bokhoven H, Celli J, Kayserili H, van Beusekom E, Balci S, Brussel W, … Brunner HG (2000). Mutation of the gene encoding the ROR2 tyrosine kinase causes autosomal recessive Robinow syndrome. Nature Genetics, 25(4), 423–426. doi: 10.1038/78113 [DOI] [PubMed] [Google Scholar]
  419. van Genderen C, Okamura RM, Farinas I, Quo RG, Parslow TG, Bruhn L, & Grosschedl R (1994). Development of several organs that require inductive epithelial-mesenchymal interactions is impaired in LEF-1-deficient mice. Genes & development, 8(22), 2691–2703. doi: 10.1101/gad.8.22.2691 [DOI] [PubMed] [Google Scholar]
  420. van Haelst MM, Scambler PJ, Fraser Syndrome Collaboration G, & Hennekam RCM (2007). Fraser syndrome: A clinical study of 59 cases and evaluation of diagnostic criteria. American Journal of Medical Genetics Part A, 143A(24), 3194–3203. doi: 10.1002/ajmg.a.31951 [DOI] [PubMed] [Google Scholar]
  421. van Reeuwijk J, Janssen M, van den Elzen C, Beltran-Valero de Bernabé D, Sabatelli P, Merlini L, … van Bokhoven H (2005). POMT2 mutations cause alpha-dystroglycan hypoglycosylation and Walker-Warburg syndrome. Journal of medical genetics, 42(12), 907–912. doi: 10.1136/jmg.2005.031963 [DOI] [PMC free article] [PubMed] [Google Scholar]
  422. van Rooij IA, Ludwig KU, Welzenbach J, Ishorst N, Thonissen M, Galesloot TE, … Mangold E (2019). 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, 10(12), 1023. doi: 10.3390/genes10121023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  423. van Straten C, & Butow KW (2013). Gene p63: In ectrodactyly-ectodermal dysplasia clefting, ankyloblepharon-ectodermal dysplasia, Rapp-Hodgkin syndrome. Ann Maxillofac Surg, 3(1), 58–61. doi: 10.4103/2231-0746.110085 [DOI] [PMC free article] [PubMed] [Google Scholar]
  424. Váradi V, Szabó L, & Papp Z (1980). Syndrome of polydactyly, cleft lip/palate or lingual lump, and psychomotor retardation in endogamic gypsies. Journal of medical genetics, 17(2), 119–122. doi: 10.1136/jmg.17.2.119 [DOI] [PMC free article] [PubMed] [Google Scholar]
  425. Vega H, Trainer AH, Gordillo M, Crosier M, Kayserili H, Skovby F, … Jabs EW (2010). Phenotypic variability in 49 cases of ESCO2 mutations, including novel missense and codon deletion in the acetyltransferase domain, correlates with ESCO2 expression and establishes the clinical criteria for Roberts syndrome. Journal of medical genetics, 47(1), 30–37. doi: 10.1136/jmg.2009.068395 [DOI] [PubMed] [Google Scholar]
  426. Vega H, Waisfisz Q, Gordillo M, Sakai N, Yanagihara I, Yamada M, … Joenje H (2005). Roberts syndrome is caused by mutations in ESCO2, a human homolog of yeast ECO1 that is essential for the establishment of sister chromatid cohesion. Nature Genetics, 37(5), 468–470. doi: 10.1038/ng1548 [DOI] [PubMed] [Google Scholar]
  427. Venza I, Visalli M, Parrillo L, De Felice M, Teti D, & Venza M (2010). MSX1 and TGF-β3 are novel target genes functionally regulated by FOXE1. Human Molecular Genetics, 20(5), 1016–1025. doi: 10.1093/hmg/ddq547 [DOI] [PubMed] [Google Scholar]
  428. Vieira AR (2006). Association between the transforming growth factor alpha gene and nonsyndromic oral clefts: a HuGE review. Am J Epidemiol, 163(9), 790–810. doi: 10.1093/aje/kwj103 [DOI] [PubMed] [Google Scholar]
  429. Vieira AR, Avila JR, Daack-Hirsch S, Dragan E, Felix TM, Rahimov F, … Murray JC (2005). Medical sequencing of candidate genes for nonsyndromic cleft lip and palate. PLoS Genet, 1(6), e64. doi: 10.1371/journal.pgen.0010064 [DOI] [PMC free article] [PubMed] [Google Scholar]
  430. Vijayan V, Ummer R, Weber R, Silva R, & Letra A (2017). Association of WNT Pathway Genes With Nonsyndromic Cleft Lip With or Without Cleft Palate. The Cleft Palate-Craniofacial Journal, 55(3), 335–341. doi: 10.1177/1055665617732782 10.1177/1055665617732782 [DOI] [PubMed] [Google Scholar]
  431. Villanueva C, & de Roux N (2010). FGFR1 mutations in Kallmann syndrome. Front Horm Res, 39, 51–61. doi: 10.1159/000312693 [DOI] [PubMed] [Google Scholar]
  432. Vissers LE, van Ravenswaaij CM, Admiraal R, Hurst JA, de Vries BB, Janssen IM, … van Kessel AG (2004). Mutations in a new member of the chromodomain gene family cause CHARGE syndrome. Nat Genet, 36(9), 955–957. doi: 10.1038/ng1407 [DOI] [PubMed] [Google Scholar]
  433. Vissers LELM, Lausch E, Unger S, Campos-Xavier AB, Gilissen C, Rossi A, … Superti-Furga A (2011). Chondrodysplasia and abnormal joint development associated with mutations in IMPAD1, encoding the Golgi-resident nucleotide phosphatase, gPAPP. American journal of human genetics, 88(5), 608–615. doi: 10.1016/j.ajhg.2011.04.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  434. Wagner T, Wirth J, Meyer J, Zabel B, Held M, Zimmer J, … Scherer G (1994). Autosomal sex reversal and campomelic dysplasia are caused by mutations in and around the SRY-related gene SOX9. Cell, 79(6), 1111–1120. doi: 10.1016/0092-8674(94)90041-8 [DOI] [PubMed] [Google Scholar]
  435. Walker BE, & Crain B Jr (1960). Effects of hypervitaminosis a on palate development in two strains of mice. American Journal of Anatomy, 107(1), 49–58. doi: 10.1002/aja.1001070104 10.1002/aja.1001070104 [DOI] [PubMed] [Google Scholar]
  436. Wallis DE, Roessler E, Hehr U, Nanni L, Wiltshire T, Richieri-Costa A, … Muenke M (1999). Mutations in the homeodomain of the human SIX3 gene cause holoprosencephaly. Nat Genet, 22(2), 196–198. doi: 10.1038/9718 [DOI] [PubMed] [Google Scholar]
  437. Wang C, Chang JY, Yang C, Huang Y, Liu J, You P, … Li X (2013). Type 1 fibroblast growth factor receptor in cranial neural crest cell-derived mesenchyme is required for palatogenesis. The Journal of biological chemistry, 288(30), 22174–22183. doi: 10.1074/jbc.M113.463620 [DOI] [PMC free article] [PubMed] [Google Scholar]
  438. Wang H, Hetmanski JB, Ruczinski I, Liang KY, Fallin MD, Redett RJ, … Beaty TH (2012). ROR2 gene is associated with risk of non-syndromic cleft palate in an Asian population. Chinese medical journal, 125(3), 476–480. Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/22490406https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3384720/ [PMC free article] [PubMed] [Google Scholar]
  439. Wang J, Bai Y, Li H, Greene SB, Klysik E, Yu W, … Martin JF (2013). MicroRNA-17–92, a direct Ap-2alpha transcriptional target, modulates T-box factor activity in orofacial clefting. PLoS Genet, 9(9), e1003785. doi: 10.1371/journal.pgen.1003785 [DOI] [PMC free article] [PubMed] [Google Scholar]
  440. Wang J, Greene SB, Bonilla-Claudio M, Tao Y, Zhang J, Bai Y, … Martin JF (2010). Bmp signaling regulates myocardial differentiation from cardiac progenitors through a MicroRNA-mediated mechanism. Developmental cell, 19(6), 903–912. doi: 10.1016/j.devcel.2010.10.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  441. Wang J, Hamblet NS, Mark S, Dickinson ME, Brinkman BC, Segil N, … Wynshaw-Boris A (2006). Dishevelled genes mediate a conserved mammalian PCP pathway to regulate convergent extension during neurulation. Development, 133(9), 1767. doi: 10.1242/dev.02347 [DOI] [PMC free article] [PubMed] [Google Scholar]
  442. Wang P, Wu T, Schwender H, Wang H, Shi B, Wang ZQ, … Beaty TH (2018). Evidence of interaction between genes in the folate/homocysteine metabolic pathway in controlling risk of non-syndromic oral cleft. Oral Dis, 24(5), 820–828. doi: 10.1111/odi.12831 [DOI] [PubMed] [Google Scholar]
  443. Wang Q, Kurosaka H, Kikuchi M, Nakaya A, Trainor PA, & Yamashiro T (2019). Perturbed development of cranial neural crest cells in association with reduced sonic hedgehog signaling underlies the pathogenesis of retinoic-acid-induced cleft palate. Disease Models & Mechanisms, 12(10), dmm040279. doi: 10.1242/dmm.040279 [DOI] [PMC free article] [PubMed] [Google Scholar]
  444. Wang W, Jiao XH, Wang XP, Sun XY, & Dong C (2016). MTR, MTRR, and MTHFR Gene Polymorphisms and Susceptibility to Nonsyndromic Cleft Lip With or Without Cleft Palate. Genet Test Mol Biomarkers, 20(6), 297–303. doi: 10.1089/gtmb.2015.0186 [DOI] [PMC free article] [PubMed] [Google Scholar]
  445. Wang X, Reid Sutton V, Omar Peraza-Llanes J, Yu Z, Rosetta R, Kou YC, … Van den Veyver IB (2007). Mutations in X-linked PORCN, a putative regulator of Wnt signaling, cause focal dermal hypoplasia. Nat Genet, 39(7), 836–838. doi: 10.1038/ng2057 [DOI] [PubMed] [Google Scholar]
  446. Wang Y, Dai Y, Li X, Chen CY, Li W, & Yu Z (2011). Inhibition of Smad Signaling is Implicated in Cleft Palate Induced by All-trans Retinoic Acid. Acta Biologica Hungarica, 62(2), 142–150. doi: 10.1556/ABiol.62.2011.2.4 [DOI] [PubMed] [Google Scholar]
  447. Wang Y, Song L, & Zhou CJ (2011). The canonical Wnt/ß-catenin signaling pathway regulates Fgf signaling for early facial development. Developmental Biology, 349(2), 250–260. doi: 10.1016/j.ydbio.2010.11.004 [DOI] [PubMed] [Google Scholar]
  448. Wassif CA, Maslen C, Kachilele-Linjewile S, Lin D, Linck LM, Connor WE, … Porter FD (1998). Mutations in the human sterol delta7-reductase gene at 11q12–13 cause Smith-Lemli-Opitz syndrome. American journal of human genetics, 63(1), 55–62. doi: 10.1086/301936 [DOI] [PMC free article] [PubMed] [Google Scholar]
  449. Watanabe A, Akita S, Tin NTD, Natsume N, Nakano Y, Niikawa N, … Yoshiura K.-i. (2006). A Mutation in RYK is a Genetic Factor for Nonsyndromic Cleft Lip and Palate. The Cleft Palate-Craniofacial Journal, 43(3), 310–316. doi: 10.1597/04-145.1 10.1597/04–145.1 [DOI] [PubMed] [Google Scholar]
  450. Waterham HR, Wijburg FA, Hennekam RCM, Vreken P, Poll-The BT, Dorland L, … Wanders RJA (1998). Smith-Lemli-Opitz Syndrome Is Caused by Mutations in the 7-Dehydrocholesterol Reductase Gene. The American Journal of Human Genetics, 63(2), 329–338. doi: 10.1086/301982 [DOI] [PMC free article] [PubMed] [Google Scholar]
  451. Weiss A, & Attisano L (2013). The TGFbeta Superfamily Signaling Pathway. Wiley Interdisciplinary Reviews: Developmental Biology, 2(1), 47–63. doi: 10.1002/wdev.86 10.1002/wdev.86 [DOI] [PubMed] [Google Scholar]
  452. Welsh IC, Hagge-Greenberg A, & O’Brien TP (2007). A dosage-dependent role for Spry2 in growth and patterning during palate development. Mech Dev, 124(9–10), 746–761. doi: 10.1016/j.mod.2007.06.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  453. White J, Mazzeu JF, Hoischen A, Jhangiani SN, Gambin T, Alcino MC, … Carvalho CM (2015). DVL1 frameshift mutations clustering in the penultimate exon cause autosomal-dominant Robinow syndrome. American journal of human genetics, 96(4), 612–622. doi: 10.1016/j.ajhg.2015.02.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  454. White JA, Beckett-Jones B, Guo Y-D, Dilworth FJ, Bonasoro J, Jones G, & Petkovich M (1997). cDNA Cloning of Human Retinoic Acid-metabolizing Enzyme (hP450RAI) Identifies a Novel Family of Cytochromes P450 (CYP26). Journal of Biological Chemistry, 272(30), 18538–18541. doi: 10.1074/jbc.272.30.18538 [DOI] [PubMed] [Google Scholar]
  455. White JJ, Mazzeu JF, Hoischen A, Bayram Y, Withers M, Gezdirici A, … Carvalho CMB (2016). DVL3 Alleles Resulting in a −1 Frameshift of the Last Exon Mediate Autosomal-Dominant Robinow Syndrome. American journal of human genetics, 98(3), 553–561. doi: 10.1016/j.ajhg.2016.01.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  456. Wieczorek D, Newman WG, Wieland T, Berulava T, Kaffe M, Falkenstein D, … Strom TM (2014). Compound heterozygosity of low-frequency promoter deletions and rare loss-of-function mutations in TXNL4A causes Burn-McKeown syndrome. American journal of human genetics, 95(6), 698–707. doi: 10.1016/j.ajhg.2014.10.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  457. Wilkie AO, Slaney SF, Oldridge M, Poole MD, Ashworth GJ, Hockley AD, … et al. (1995). Apert syndrome results from localized mutations of FGFR2 and is allelic with Crouzon syndrome. Nat Genet, 9(2), 165–172. doi: 10.1038/ng0295-165 [DOI] [PubMed] [Google Scholar]
  458. Willert K, Shibamoto S, & Nusse R (1999). Wnt-induced dephosphorylation of axin releases beta-catenin from the axin complex. Genes & Development, 13(14), 1768–1773. doi: 10.1101/gad.13.14.1768 [DOI] [PMC free article] [PubMed] [Google Scholar]
  459. Williams ES, Uhas KA, Bunke BP, Garber KB, & Martin CL (2012). Cleft palate in a multigenerational family with a microdeletion of 20p12.3 involving BMP2. American Journal of Medical Genetics Part A, 158A(10), 2616–2620. doi: 10.1002/ajmg.a.35594 10.1002/ajmg.a.35594 [DOI] [PMC free article] [PubMed] [Google Scholar]
  460. Wilson JG, Roth CB, & Warkany J (1953). An analysis of the syndrome of malformations induced by maternal vitamin a deficiency. Effects of restoration of vitamin a at various times during gestation. American Journal of Anatomy, 92(2), 189–217. doi: 10.1002/aja.1000920202 10.1002/aja.1000920202 [DOI] [PubMed] [Google Scholar]
  461. Wilson M, Mowat D, Dastot-Le Moal F, Cacheux V, Kääriäinen H, Cass D, … Goossens M (2003). Further delineation of the phenotype associated with heterozygous mutations in ZFHX1B. American Journal of Medical Genetics Part A, 119A(3), 257–265. doi: 10.1002/ajmg.a.20053 [DOI] [PubMed] [Google Scholar]
  462. Wolf ZT, Brand HA, Shaffer JR, Leslie EJ, Arzi B, Willet CE, … Bannasch DL (2015). Genome-wide association studies in dogs and humans identify ADAMTS20 as a risk variant for cleft lip and palate. PLoS Genet, 11(3), e1005059. doi: 10.1371/journal.pgen.1005059 [DOI] [PMC free article] [PubMed] [Google Scholar]
  463. Wong H-C, Bourdelas A, Krauss A, Lee H-J, Shao Y, Wu D, … Zheng J (2003). Direct binding of the PDZ domain of Dishevelled to a conserved internal sequence in the C-terminal region of Frizzled. Molecular cell, 12(5), 1251–1260. doi: 10.1016/s1097-2765(03)00427-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  464. Wu W, Gu S, Sun C, He W, Xie X, Li X, … Liu C (2015). Altered FGF Signaling Pathways Impair Cell Proliferation and Elevation of Palate Shelves. PLOS ONE, 10(9), e0136951. doi: 10.1371/journal.pone.0136951 [DOI] [PMC free article] [PubMed] [Google Scholar]
  465. Wyatt AW, Osborne RJ, Stewart H, & Ragge NK (2010). Bone morphogenetic protein 7 (BMP7) mutations are associated with variable ocular, brain, ear, palate, and skeletal anomalies. Human Mutation, 31(7), 781–787. doi: 10.1002/humu.21280 10.1002/humu.21280 [DOI] [PubMed] [Google Scholar]
  466. Xavier D, Arif Y, Murali R, Kishore Kumar S, Vipin Kumar S, Tamang R, … Bhaskar L (2013). Analysis of microsatellite polymorphisms in South Indian patients with non syndromic cleft lip and palate. Balkan journal of medical genetics : BJMG, 16(1), 49–54. doi: 10.2478/bjmg-2013-0017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  467. Xiong W, He F, Morikawa Y, Yu X, Zhang Z, Lan Y, … Chen Y (2009). Hand2 is required in the epithelium for palatogenesis in mice. Developmental Biology, 330(1), 131–141. doi: 10.1016/j.ydbio.2009.03.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  468. Xu J, Huang Z, Wang W, Tan X, Li H, Zhang Y, … Chen YP (2018). FGF8 Signaling Alters the Osteogenic Cell Fate in the Hard Palate. Journal of Dental Research, 97(5), 589–596. doi: 10.1177/0022034517750141 [DOI] [PMC free article] [PubMed] [Google Scholar]
  469. Xu J, Liu H, Lan Y, Aronow BJ, Kalinichenko VV, & Jiang R (2016). A Shh-Foxf-Fgf18-Shh Molecular Circuit Regulating Palate Development. PLoS Genet, 12(1), e1005769. doi: 10.1371/journal.pgen.1005769 [DOI] [PMC free article] [PubMed] [Google Scholar]
  470. Xu X, Han J, Ito Y, Bringas P Jr., Deng C, & Chai Y (2008). Ectodermal Smad4 and p38 MAPK are functionally redundant in mediating TGF-beta/BMP signaling during tooth and palate development. Developmental Cell, 15(2), 322–329. doi: 10.1016/j.devcel.2008.06.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  471. Xu X, Han J, Ito Y, Bringas P, Urata MM, & Chai Y (2006). Cell autonomous requirement for Tgfbr2 in the disappearance of medial edge epithelium during palatal fusion. Developmental Biology, 297(1), 238–248. doi: 10.1016/j.ydbio.2006.05.014 [DOI] [PubMed] [Google Scholar]
  472. Yagi H, Furutani Y, Hamada H, Sasaki T, Asakawa S, Minoshima S, … Matsuoka R (2003). Role of TBX1 in human del22q11.2 syndrome. Lancet, 362(9393), 1366–1373. doi: 10.1016/s0140-6736(03)14632-6 [DOI] [PubMed] [Google Scholar]
  473. Yamada S, Pokutta S, Drees F, Weis WI, & Nelson WJ (2005). Deconstructing the Cadherin-Catenin-Actin Complex. Cell, 123(5), 889–901. doi: 10.1016/j.cell.2005.09.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  474. Yang T, Jia Z, Bryant-Pike W, Chandrasekhar A, Murray JC, Fritzsch B, & Bassuk AG (2014). Analysis of PRICKLE1 in human cleft palate and mouse development demonstrates rare and common variants involved in human malformations. Molecular Genetics & Genomic Medicine, 2(2), 138–151. doi: 10.1002/mgg3.53 10.1002/mgg3.53 [DOI] [PMC free article] [PubMed] [Google Scholar]
  475. Yang X, Kilgallen S, Andreeva V, Spicer DB, Pinz I, & Friesel R (2010). Conditional expression of Spry1 in neural crest causes craniofacial and cardiac defects. BMC Dev Biol, 10, 48. doi: 10.1186/1471-213X-10-48 [DOI] [PMC free article] [PubMed] [Google Scholar]
  476. Yang X, Li C, Xu X, & Deng C (1998). The tumor suppressor SMAD4/DPC4 is essential for epiblast proliferation and mesoderm induction in mice. Proceedings of the National Academy of Sciences of the United States of America, 95(7), 3667–3672. doi: 10.1073/pnas.95.7.3667 [DOI] [PMC free article] [PubMed] [Google Scholar]
  477. Yang Y, Yuan J, Yao X, Zhang R, Yang H, Zhao R, … Zhu Y (2017). BMPR1B mutation causes Pierre Robin sequence. Oncotarget, 8(16), 25864–25871. doi: 10.18632/oncotarget.16531 [DOI] [PMC free article] [PubMed] [Google Scholar]
  478. Yook JI, Li X-Y, Ota I, Fearon ER, & Weiss SJ (2005). Wnt-dependent Regulation of the E-cadherin Repressor Snail. Journal of Biological Chemistry, 280(12), 11740–11748. Retrieved from http://www.jbc.org/content/280/12/11740.abstract [DOI] [PubMed] [Google Scholar]
  479. Yu H-MI, Jerchow B, Sheu T-J, Liu B, Costantini F, Puzas JE, … Hsu W (2005). The role of Axin2 in calvarial morphogenesis and craniosynostosis. Development (Cambridge, England), 132(8), 1995–2005. doi: 10.1242/dev.01786 [DOI] [PMC free article] [PubMed] [Google Scholar]
  480. Yu H, Smallwood PM, Wang Y, Vidaltamayo R, Reed R, & Nathans J (2010). Frizzled 1 and frizzled 2 genes function in palate, ventricular septum and neural tube closure: general implications for tissue fusion processes. Development, 137(21), 3707. doi: 10.1242/dev.052001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  481. Yu H, Ye X, Guo N, & Nathans J (2012). Frizzled 2 and frizzled 7 function redundantly in convergent extension and closure of the ventricular septum and palate: evidence for a network of interacting genes. Development (Cambridge, England), 139(23), 4383–4394. doi: 10.1242/dev.083352 [DOI] [PMC free article] [PubMed] [Google Scholar]
  482. Yu K, Herr AB, Waksman G, & Ornitz DM (2000). Loss of fibroblast growth factor receptor 2 ligand-binding specificity in Apert syndrome. Proceedings of the National Academy of Sciences of the United States of America, 97(26), 14536–14541. doi: 10.1073/pnas.97.26.14536 [DOI] [PMC free article] [PubMed] [Google Scholar]
  483. Yu K, Karuppaiah K, & Ornitz DM (2015). Mesenchymal fibroblast growth factor receptor signaling regulates palatal shelf elevation during secondary palate formation. Developmental dynamics : an official publication of the American Association of Anatomists, 244(11), 1427–1438. doi: 10.1002/dvdy.24319 [DOI] [PMC free article] [PubMed] [Google Scholar]
  484. Yu L, Hebert MC, & Zhang YE (2002). TGF-beta receptor-activated p38 MAP kinase mediates Smad-independent TGF-beta responses. Embo j, 21(14), 3749–3759. doi: 10.1093/emboj/cdf366 [DOI] [PMC free article] [PubMed] [Google Scholar]
  485. Yu Q, He S, Zeng N, Ma J, Zhang B, Shi B, & Jia Z (2015). BMP7 Gene involved in nonsyndromic orofacial clefts in Western Han Chinese. Medicina oral, patologia oral y cirugia bucal, 20(3), e298–e304. doi: 10.4317/medoral.20335 [DOI] [PMC free article] [PubMed] [Google Scholar]
  486. Yu Y, Zuo X, He M, Gao J, Fu Y, Qin C, … Bian Z (2017). Genome-wide analyses of non-syndromic cleft lip with palate identify 14 novel loci and genetic heterogeneity. Nature communications, 8, 14364–14364. doi: 10.1038/ncomms14364 [DOI] [PMC free article] [PubMed] [Google Scholar]
  487. Yuan G, Zhan Y, Gou X, Chen Y, & Yang G (2018). TGF-β signaling inhibits canonical BMP signaling pathway during palate development. Cell and tissue research, 371(2), 283–291. doi: 10.1007/s00441-017-2757-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  488. Yuan Q, Blanton SH, & Hecht JT (2011). Association of ABCA4 and MAFB with nonsyndromic cleft lip with or without cleft palate. American journal of medical genetics. Part A, 155A(6), 1469–1471. doi: 10.1002/ajmg.a.33940 [DOI] [PMC free article] [PubMed] [Google Scholar]
  489. Yumoto K, Thomas PS, Lane J, Matsuzaki K, Inagaki M, Ninomiya-Tsuji J, … Kaartinen V (2013). TGF-β-activated kinase 1 (Tak1) mediates agonist-induced Smad activation and linker region phosphorylation in embryonic craniofacial neural crest-derived cells. The Journal of biological chemistry, 288(19), 13467–13480. doi: 10.1074/jbc.M112.431775 [DOI] [PMC free article] [PubMed] [Google Scholar]
  490. Zarate YA, & Fish JL (2017). SATB2-associated syndrome: Mechanisms, phenotype, and practical recommendations. American journal of medical genetics. Part A, 173(2), 327–337. doi: 10.1002/ajmg.a.38022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  491. Zebisch M, Xu Y, Krastev C, MacDonald BT, Chen M, Gilbert RJC, … Jones EY (2013). Structural and molecular basis of ZNRF3/RNF43 transmembrane ubiquitin ligase inhibition by the Wnt agonist R-spondin. Nature communications, 4, 2787–2787. doi: 10.1038/ncomms3787 [DOI] [PMC free article] [PubMed] [Google Scholar]
  492. Zhang B-H, Shi J-Y, Lin Y-S, Shi B, & Jia Z-L (2018). VAX1 gene associated nonsyndromic cleft lip with or without palate in Western Han Chinese. Archives of Oral Biology, 95, 40–43. doi: 10.1016/j.archoralbio.2018.07.014 [DOI] [PubMed] [Google Scholar]
  493. Zhang M, Chen W, Smith SM, & Napoli JL (2001). Molecular Characterization of a Mouse Short Chain Dehydrogenase/Reductase Active with All-trans-retinol in Intact Cells, mRDH1. Journal of Biological Chemistry, 276(47), 44083–44090. doi: 10.1074/jbc.M105748200 [DOI] [PubMed] [Google Scholar]
  494. Zhang R, Oyajobi BO, Harris SE, Chen D, Tsao C, Deng H-W, & Zhao M (2013). Wnt/β-catenin signaling activates bone morphogenetic protein 2 expression in osteoblasts. Bone, 52(1), 145–156. doi: 10.1016/j.bone.2012.09.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  495. Zhang X, Ibrahimi OA, Olsen SK, Umemori H, Mohammadi M, & Ornitz DM (2006). Receptor specificity of the fibroblast growth factor family. The complete mammalian FGF family. The Journal of biological chemistry, 281(23), 15694–15700. doi: 10.1074/jbc.M601252200 [DOI] [PMC free article] [PubMed] [Google Scholar]
  496. Zhang Z, Song Y, Zhao X, Zhang X, Fermin C, & Chen Y (2002). Rescue of cleft palate in Msx1-deficient mice by transgenic Bmp4 reveals a network of BMP and Shh signaling in the regulation of mammalian palatogenesis. Development, 129(17), 4135 Retrieved from http://dev.biologists.org/content/129/17/4135.abstract [DOI] [PubMed] [Google Scholar]
  497. Zhou J, Gao Y, Lan Y, Jia S, & Jiang R (2013). Pax9 regulates a molecular network involving Bmp4, Fgf10, Shh signaling and the Osr2 transcription factor to control palate morphogenesis. Development (Cambridge, England), 140(23), 4709–4718. doi: 10.1242/dev.099028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  498. Zhou R, Wang M, Li W, Wang S, Zheng H, Zhou Z, … Beaty TH (2019). Haplotype and Haplotype-Environment Interaction Analysis Revealed Roles of SPRY2 for NSCL/P among Chinese Populations. Int J Environ Res Public Health, 16(4). doi: 10.3390/ijerph16040557 [DOI] [PMC free article] [PubMed] [Google Scholar]
  499. Zhu X-J, Liu Y, Yuan X, Wang M, Zhao W, Yang X, … Zhang Z (2016). Ectodermal Wnt controls nasal pit morphogenesis through modulation of the BMP/FGF/JNK signaling axis. Developmental Dynamics, 245(3), 414–426. doi: 10.1002/dvdy.24376 10.1002/dvdy.24376 [DOI] [PMC free article] [PubMed] [Google Scholar]
  500. Zinski J, Tajer B, & Mullins MC (2018). TGF-β Family Signaling in Early Vertebrate Development. Cold Spring Harbor Perspectives in Biology, 10(6). doi: 10.1101/cshperspect.a033274 [DOI] [PMC free article] [PubMed] [Google Scholar]
  501. Zucchero TM, Cooper ME, Maher BS, Daack-Hirsch S, Nepomuceno B, Ribeiro L, … Murray JC (2004). Interferon Regulatory Factor 6 (IRF6) Gene Variants and the Risk of Isolated Cleft Lip or Palate. New England Journal of Medicine, 351(8), 769–780. doi: 10.1056/NEJMoa032909 [DOI] [PubMed] [Google Scholar]

RESOURCES