Involvement of the WNT and FGF signaling pathways in non-isolated anorectal malformations: Sequencing analysis of WNT3A, WNT5A, WNT11, DACT1, FGF10, FGFR2 and the T gene

  • Authors:
    • Markus Draaken
    • Wiebke Prins
    • Claudia Zeidler
    • Alina Hilger
    • Sadaf S. Mughal
    • Jeanette Latus
    • Thomas M. Boemers
    • Dominik Schmidt
    • Eberhard Schmiedeke
    • Nicole Spychalski
    • Enrika Bartels
    • Markus M. Nöthen
    • Heiko Reutter
    • Michael Ludwig
  • View Affiliations

  • Published online on: September 7, 2012     https://doi.org/10.3892/ijmm.2012.1124
  • Pages: 1459-1464
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Anorectal malformations (ARMs) comprise a broad spectrum of anomalies, including anal atresia, congenital anal fistula and persistence of the cloaca. Research suggests that genetic factors play an important role in ARM development. However, few genetic variants have been identified. Embryogenesis is orchestrated by crosstalk of the wingless-type MMTV integration site family (WNT) and fibroblast growth factor (FGF) signaling pathways in a process that involves several intracellular cascades. Studies in mice have implicated several genes from these pathways in the etiology of ARMs. We performed sequencing analysis of seven of these previously reported genes in 78 patients with ARMs occurring within the context of at least one additional congenital anomaly. No associations were identified with variants in WNT3A, WNT5A, WNT11, DACT1, FGF10 or the T gene. In the FGFR2 gene, three novel heterozygous nucleotide substitutions were identified. Further investigations, including the study of family members, revealed that these variants were not causally related to the phenotype in the present ARM cohort. Mutations in the seven investigated genes may nonetheless be a cause of ARMs in rare cases. However, further studies should consider genes encoding other proteins in the WNT/FGF signaling pathways as possible candidates.

Introduction

Anorectal malformations (ARMs) comprise a broad spectrum of anomalies including anal atresia, congenital anal fistula and persistence of the cloaca. The estimated incidence of ARMs is approximately 1 in 2,500 to 3,000 live births (15) and a male to female ratio of 1.7 has been reported for isolated (non-syndromic) forms (6). Isolated forms account for 40–50% of all ARM cases, and these are sometimes associated with other developmental anomalies, such as renal and urogenital malformations (5,6). In the remaining cases, ARMs occur within the context of defined genetic syndromes or multiple congenital anomalies.

Research suggests that ARMs have a heterogeneous etiology and include Mendelian and multifactorial forms. The latter probably arise as a result of a complex interplay between genetic risk variants and environmental factors. Possible maternal risk factors include use of multivitamins or medications for severe chronic dyspepsia and asthma during pregnancy (7,8), periconceptional injuries or pyrexia, obesity and diabetes. Research also suggests that smoking and certain occupational exposures in either parent may be associated with a higher risk of ARMs (912).

Although mutations for some ARM-related syndromes have been identified, the majority of the underlying genetic factors for ARMs remain unknown (13,14). Previously reported genetic factors include the homeobox gene MNX1 in Currarino syndrome (15), the SALL1 zinc-finger protein in Townes-Brocks syndrome (16) and the GLI3 gene in Pallister-Hall syndrome (17).

Since many ARM phenotypes are negatively associated with reproductive fitness, it is reasonable to assume that a substantial proportion of ARM patients carry de novo mutations. Thus candidate gene sequencing to identify rare, high-penetrance mutations is a rational approach.

The processes of urogenital and anorectal embryogenesis involves the wingless-type MMTV integration site family (WNT)/fibroblast growth factor (FGF) signaling pathways. Mammalian WNT proteins constitute a family of roughly 20 secreted glycoproteins (18), which act as short-range paracrine signaling effectors. The FGF family comprises 22 extracellular ligands, whose signals are mediated through a family of tyrosine kinase receptors. These are termed the five FGF receptors (FGFR1-5) (19). Alternative splicing generates multiple isoforms for each FGFR. Each isoform is characterized by a differing affinity for the respective ligand (20).

Multiple lines of evidence from mouse models suggest that genes in these signaling pathways (Fig. 1) are implicated in the etiology of ARMs. Mice that are homozygous for a hypomorphic Wnt3a allele display vertebral defects, a short tail due to loss of caudal vertebrae, deficient cloacal development and incomplete uro-rectal septation (21). Moreover, studies involving human pluripotent stem cells have shown that WNT3A is required for hindgut specification (22). Wnt5a is expressed in the embryonic colon and rectum and affects the development of the proximal cloacal plate (23). Wnt5a-knockout mice display ARMs such as imperforate anus and the presence of fistulas between the urinary and intestinal tracts (24). As with Wnt3a and Wnt5a, Wnt11 has been identified in the developing mouse urogenital tract (25). Two studies have reported expression of human WNT11 in the embryonic uro-rectal septum, the urogenital folds, the labioscrotal swellings and the epithelium of the esophagus and colon (26,27). Studies in Chinese hamster ovary cells have shown that Wnt11 signaling leads to downregulation of the key signaling pathways Wnt/β-catenin, JNK/AP-1 and NF-κB (28).

Figure 1

Molecular FGF/WNT signaling factor pathways as candidates for ARM. Selected proteins and interactions are shown. FGF signal transduction (binding of FGF10 to its receptor FGFR2) is shown. Formation of the FGF:FGFR:HSPG (heparan sulfate proteoglycans) signaling complex activates extra-cellular signal-regulated kinases (ERK)/mitogen-activated protein kinases (MAPK) and phosphoinositide-3 kinase (PI3K). PI3K activates protein kinase AKT or protein kinase B (PKB), with subsequent inhibition of glycogen synthase kinase 3β (GSK3β) by phosphorylation. MAPK dependent phosphorylation of transcription factors allows transcription of FGF target genes. In addition, phosphorylation may promote the release of transcriptional repressor Groucho from transcription factor (TCF) 1. This allows interaction between TCF/lymphoid enhancer-binding factor 1 (LEF1) and β-catenin (βCN) and stimulation of the transcription of WNT-dependent genes, e.g. the T-gene (brachyury). In the absence of nuclear βCN, TCF1/LEF1 act as transcriptional repressors by binding to Groucho. βCN can also displace Groucho from TCF1/LEF1. Stabilization of βCN is the major effect of WNT signaling. Absence of this effect leads to phosphorylation of βCN via a destruction complex including axin, the APC gene (mutant in adenomatous polyposis) product and GSK3β. This mechanism primes βCN-Pi for degradation by the ubiquitin pathway. WNT ligands include the Frizzled (FZD) family of receptors and these signal through co-receptors such as low-density lipoprotein receptor related protein 5/6 (LRP5/6) and the orphan tyrosine kinase receptors ROR1 and ROR2. Binding of WNT3A (inhibited by WNT5A) to a receptor from the Frizzled (FZD) family leads to the activation of Dishellved (DSH), a core component of WNT signaling, thereby enhancing the phosphorylation and subsequent inhibition of GSK3β. In addition to FZD receptors, WNT5A can also bind and activate ROR2, resulting in the activation of the actin-binding protein filamin A and the JNK signaling pathway. WNT3A and WNT5A exert reciprocal pathway inhibition. WNT11 binds to several FZD (type 4, 5 and 7) receptors. Inhibition of WNT/βCN signaling may be mediated by competition for FZD receptors. DACT1 can bind βCN and this complex then inhibits GSK3β. This inhibition represses the destruction complex and leads to the release of βCN, thereby increasing its nuclear and cytoplasmic fraction. The figure is adapted from previous studies (31,60–62).

Dapper homolog 1 (Dact1) also functions as a negative regulator of Wnt signaling (29), and its inactivation leads to perinatal lethality in Dact−/− mice. Wen et al (30) reported that the phenotype of Dact null embryos resembled human congenital caudal regression syndrome, including features such as caudal vertebrae agenesis, ARMs, renal agenesis/dysplasia, fused kidneys and absence of the bladder.

The orchestration of embryogenesis via crosstalk of the WNT and FGF signaling pathways is mediated by several intracellular cascades (19,31), and integration of WNT/FGF signaling can also occur at the level of an individual gene promoter (32). The T gene (brachyury; T-box containing transcription factor) is a direct target of WNT3A signaling during paraxial mesoderm specification (33), and a T missense mutation (p.Ala338Val) has been observed in four patients with sacral agenesis/anorectal atresia (34,35). Bagai et al (36) reported that FGF10 plays an important role in regulating the growth, differentiation and repair of the urothelium. Other studies found that complete invalidation of fibroblast growth factor 10 (FGF10) in mice resulted in a genetically reproducible ARM (37) and failure of ventral fusion in the urethral plate (38).

FGF10 signaling is mediated by the FGFR2 protein, which is encoded by the receptor encoding gene FGFR2. Mutations in this gene cause various forms of autosomal dominant craniosynostosis syndrome and have been associated with ARMs in patients with Apert syndrome (acrocephalosyndactyly type I; MIM, 101200), Pfeiffer syndrome types 1 and 2 (acrocephalosyndactyly type V; MIM, 101600), Crouzon syndrome (craniofacial dysostosis; MIM, 123500) and Beare-Stevenson cutis gyrata syndrome (MIM, 123790) (3947).

To explore the possible involvement of the above genes in the etiology of human ARMs, we performed sequencing analysis in a sample of 78 patients with ARMs occurring within the context of multiple congenital anomalies.

Patients and methods

Subjects

Patients were contacted and recruited through the German self-help organization for patients with anorectal malformations (SoMA e.V.) as well as the Departments of (i) Neonatology, Children’s Hospital, University of Bonn, (ii) Paediatric Surgery and Urology, Centre for Child and Adolescent Health, Hospital Bremen-Mitte, (iii) Pediatric Surgery and Pediatric Urology, Children’s Hospital, Cologne, (iv) Pediatric Surgery, Campus Virchow Clinic, Charité University Hospital Berlin and (v) Pediatric Surgery, Cnopf’sche Kinderklinik, Nuremberg. Blood samples were taken from the patients and (if available) their parents (n=55 trios). In 18 cases only one parent agreed to participate and in five cases no parental sample could be obtained.

All 78 ARM patients were of European descent and had a normal karyotype. The study was approved by the Ethics Committee of the University of Bonn and written informed consent was obtained from all subjects prior to inclusion and blood sampling.

Gene analysis

Standard procedures were used for the isolation of genomic DNA, amplification of DNA via polymerase chain reaction (PCR) and performance of the automated sequencing analyses. In brief, primers (sequences available on request) were directed to all exons of the genes WNT3A, WNT5A, WNT11, DACT1, FGF10, FGFR2 and T (GenBank acc. nos.: NM_033131.3, NM_001256105.1, NM_004626.2, NM_016651.5, NM_004465.1, NM_000141.4, NM_001144913.1-001144919.1, NM_022970.3 and NM_003181.2). The resultant PCR products were subjected to direct automated sequencing (3130xl Genetic Analyzer; Applied Biosystems, Foster City, CA, USA). For each patient, both strands of each amplicon were sequenced. All nucleotide variations were confirmed via the performance of independent PCR reactions. Segregation of these variants in family members was investigated by sequencing the respective PCR products. In the course of direct sequencing, information was obtained concerning various single nucleotide polymorphisms (SNPs) in the analyzed genes.

RNA analysis

RNA analyses were performed to determine the effect of the DNA variation observed in index patient (case B10) and his mother. The respective blood samples were collected in PAXgene tubes (PreAnalytiX, Hombrechtikon, Switzerland) in order to stabilize the intracellular RNA. RNA for reverse transcription (RT) PCR was prepared with the RNeasy Plus Micro kit (Qiagen, Hilden, Germany) in accordance with the manufacturer’s instructions. Primers were directed to FGFR2 exons 10/11 (10/11F-cDNA: 5′-GACAGTTCTGCCAGCGCCTG-3′) and 13 (13R-cDNA: 5′-GCCTCCTTGGGCTTGTCTTTG-3′). This allowed analysis of the effect of the c.G2012A (p.Thr454=) variant (numbering according to GenBank acc. no. NM_022970.3 and Swiss-Prot entry P21802) and detection of alternatively spliced mRNAs.

Results

In our candidate gene approach no variant of likely causal relevance was detected in the genes encoding WNT3A, WNT5A, WNT11, DACT1, FGF10 and T protein. Sequencing analysis of the FGFR2 gene revealed the presence of novel heterozygous variants in three patients (Fig. 2). These variants are listed neither in the current SNP database (dbSNP136) nor in the 1,000 Genomes catalog.

A synonymous p.Thr454= variant (c.G2012A transition in exon 12) was identified in patient B10 and his mother. Patient B10 presented with anal atresia with fistula, hypospadias, left renal agenesis, rib and vertebral column malformations, lumbar spina bifida occulta and hexadactylism of the right foot. Interestingly, his mother also had spina bifida occulta. A closer look at the sequence surroundings affected by this silent substitution, revealed the formation of a potential novel 3′ acceptor splice site in exon 12. Calculation of the consensus value (CV) for splice site recognition (48) revealed a CV of 0.913 for the wild-type sequence (5′-cattttgtatccag^G; exonic base in capital letter) and a CV of 0.917 for the novel variant (5′-cctctcttcaacag^C; substitution underlined). The finding of nearly identical CVs suggested an alternative usage of these splice sites with the possible consequence of a deletion of 76 bp (p.Ala455GlnfsX26). Therefore, RNA analysis was performed. However, no novel 425-bp fragment (normal, 501 bp) was detected in the RT-PCR experiments (data not shown). The only detected variation was due to alternative splicing at the exon 11 donor splice site, which has been shown to account for the absence of residues Val428 and Thr429 in several FGFR2 isoforms (see Swiss-Prot entry P21802).

Another heterozygous exon 12 FGFR2 variant (c.C2032T) was detected in patient G10. This variant is predicted to result in a p.Ala461Val amino acid substitution. Patient G10 presented with perineal fistula, hypoplasia of the left thumb, pre-axial polydactyly of the left hand, wedged vertebra (thoracic and cervical), rib malformations, dextroversio cordis and double kidney (left). The mother showed wild-type sequence only, and no DNA was available from the father. To test if this variant had arisen de novo on the maternal allele, a search for neutral heterozygous variants 5 kb upstream and downstream of exon 12 was performed. Detection of such variants would have allowed allele-specific PCR, and a distinction as to whether the variant resided on the maternal or the paternal allele. However, no heterozygous SNP or private variant was detected in the 10-kb flanking exon 12. Interestingly, pathogenicity prediction of this sequence alteration varied depending on the program used. According to Mutation Taster (www.mutationtaster.org), this variant had a 0.981 probability of being disease-causing. In contrast, three other programs predicted that it was benign: [PolyPhen-2 (http://genetics.bwh.harvard.edu/pph2/, benign with a score of 0.005); MutPred (http://mutpred.mutdb.org, probability of being deleterious 0.235); and SNPs&Go (http://snps-and-go.biocomp.unibo.it/cgi-bin/snps-and-go/runpred.cgi, reliability index 1].

A third variant was identified in patient A02, who presented with anal atresia with recto-urethral prostatic fistula, ventricular septal defect, subaortic stenosis and dystopic kidney. This synonymous FGFR2 exon 18 variant (c.C2717T, p.Phe689=) was assumed to have no effect. Hence, no further analyses of this variant were performed.

Direct sequencing also generated information concerning several SNPs (dbSNP136) in the investigated genes. For all genes, similar haplotype data were found in the European Population of the International HapMap Project, the CEPH (Centre d’Etude du Polymorphisme Human) pedigrees and the PDR90 (The NIH Polymorphism Discovery Resource; 90 individual screenings) subset.

Discussion

WNT/FGF signaling pathways (Fig. 1) orchestrate correct patterning, cell specification and tissue differentiation during embryogenesis (19,31). Research has shown that disruption of this coordinated interplay can result in severe malformations in mice and humans (49,50), including urogenital and anorectal anomalies. The present study investigated selected candidate genes from these pathways, chosen on the basis of observations in mice and human cell lines and/or their involvement in diseases associated with ARMs. However, no potential causal variant for ARMs was detected in the genes encoding WNT3A, WNT5A, WNT11, DACT1, FGF10 and T protein in the present cohort.

In contrast, FGFR2 analysis revealed the presence of novel heterozygous variations in three patients (Fig. 2). We initially considered the two exon 12 variants to be of potential causal relevance. Our rationale for this hypothesis was that as well as affecting all FGFR2 isoforms, a mutation in this exon would affect a cytoplasmic part of the protein which has not yet been implicated in the various forms of autosomal dominant craniosynostosis syndrome. Moreover, the program Mutation Taster (51) predicted that the p.Ala461Val amino acid substitution was disease-causing. However, Thusberg et al (52) reported on the suboptimal performance of this program and in line with their findings of the performance of mutation pathogenicity prediction methods, several other - more reliable - programs classified it as benign. Furthermore, the results of our mRNA experiments suggest that the p.Thr454= variant had no effect on correct splicing.

Despite these negative findings, the possibility remains that these nucleotide substitutions contribute to the ARM phenotype through as-yet-unknown mechanisms. As FGF10 is expressed in the mesenchyme that lies adjacent to the urethral plate, a plausible hypothesis is that it is important in the regulation of endoderm and/or mesenchyme growth, and thus in proliferation-driven urogenital and anorectal development (37,38). Interestingly, ARM patient B10 and his mother - both of whom showed spina bifida occulta - carried the silent p.Thr454= variant. Severe spinal dysraphism has been observed in association with an FGFR2 p.Ser351Cys mutation (53) and spina bifida occulta occurs in the mouse mutant Brachyury curtailed (Tc/+) (54). However, whereas Tc/+ mice are tailless, several of the FGFR2 amino acid substitutions observed in patients with Beare-Stevenson (55), Crouzon and Pfeiffer syndromes (56) are associated with sacral appendage. These findings, together with the repeated observation of ARMs in mice (37,38) and patients with FGFR2 defects (3947), imply that this FGF signaling pathway is of crucial importance in normal caudal development, rendering the coincidental co-occurrence of these defects unlikely.

In summary, no significant association between ARMs and mutations in WNT3A, WNT5A, WNT11, DACT1, FGF10, FGFR2 or the T gene was found in the present cohort. However, although our patient cohort was larger than those used in previous candidate gene studies of ARMs (34,5759), the sample size may have been too small to detect rare causal mutational events. Furthermore, we cannot exclude the possibility that mutations in the promoter region, in as yet unknown regulatory sequences or in non-coding regions that are not detectable with the method applied were overlooked. Future studies should consider additional proteins of the WNT/FGF signaling pathways as possible candidates.

Acknowledgements

M.D., A.H., S.S.M., E.S., E.B., M.M.N., H.R. and M.L. are members of the ‘Network for the Systematic Investigation of the Molecular Causes, Clinical Implications and Psychosocial Outcome of Congenital Uro-Rectal Malformations (CURE-Net)’, which is supported by a research grant (01GM08107) from the German Federal Ministry of Education and Research (Bundesministerium für Bildung und Forschung, BMBF). S.S.M. is supported by a research grant from the Richard-Winter-Stiftung. G.D. and E.B. are supported by the BONFOR program of the University of Bonn, grant nos. O-149.0096 and O-149.0099, respectively. We thank all patients and family members for their cooperation, as well as the German Self-Help Organization for People with Anorectal Malformations (SoMA e.V.). We thank Pia Uerdingen for her excellent technical assistance and Dr Christine Schmael for her expert advice regarding the manuscript.

References

1. 

ED SmithIncidence, frequency of types and aetiology of anorectal malformationsBirth Defects Orig Artic Ser2423124619883067766

2. 

S ChoSP MooreT FangmanOne hundred three consecutive patients with anorectal malformations and their associated anomaliesArch Pediatr Adolesc Med155587591200110.1001/archpedi.155.5.58711343503

3. 

A CuschieriEUROCAT Working GroupDescriptive epidemiology of isolated anal anomalies: a survey of 4.6 million births in EuropeAm J Med Genet103207215200110.1002/ajmg.153211745992

4. 

MA LevittA PeñaAnorectal malformationsOrphanet J Rare Dis233200710.1186/1750-1172-2-33

5. 

C StollY AlembikB DottMP RothAssociated malformations in patients with anorectal anomaliesEur J Med Genet50281290200710.1016/j.ejmg.2007.04.00217572165

6. 

A CuschieriEUROCAT Working GroupAnorectal anomalies associated with or as part of other anomaliesAm J Med Genet110122130200210.1002/ajmg.1037112116249

7. 

N AcsF BánhidyEH PuhóAE CzeizelA possible association between maternal dyspepsia and congenital rectal/anal atresia/stenosis in their children: a population-based case-control studyActa Obstet Gynecol Scand8810171023200910.1080/00016340903128447

8. 

S LinJPW MunsieML Herdt-LosavioCM DruschelK CampbellML BrownePA RomittiRS OlneyEM BellNational Birth Defects Prevention StudyMaternal asthma medication use and the risk of selected birth defectsPediatrics129e317e324201210.1542/peds.2010-266022250027

9. 

IA Van RooijCH WijersPN RieuHS HendriksMM BrouwersNV KnoersI de BlaauwN RoeleveldMaternal and paternal risk factors for anorectal malformations: a Dutch case-control studyBirth Defects Res A Clin Mol Teratol88152158201020073076

10. 

A HackshawC RodeckS BonifaceMaternal smoking in pregnancy and birth defects: a systematic review based on 173 687 malformed cases and 11.7 million controlsHum Reprod Update17589604201110.1093/humupd/dmr02221747128

11. 

SC TinkerJ ReefhuisAM DellingerDJ JamiesonMaternal injuries during the periconceptional period and the risk of birth defects, National Birth Defects Prevention Study, 1997–2005Paediatr Perinat Epidemiol25487496201121819430

12. 

N ZwinkE JenetzkyH BrennerParental risk factors and anorectal malformations: systematic review and meta-analysisOrphanet J Rare Dis625201110.1186/1750-1172-6-2521586115

13. 

B SolomonVACTERL/VATER AssociationOrphanet J Rare Dis656201110.1186/1750-1172-6-56

14. 

E MundtMD BatesGenetics of Hirschsprung disease and anorectal malformationsSemin Pediatr Surg19107117201010.1053/j.sempedsurg.2009.11.01520307847

15. 

E BelloniG MartuccielloD VerderioE PontiM SeriV JasonniM TorreM FerrariLC TsuiSW SchererInvolvement of the HLXB9 homeobox gene in Currarino syndromeAm J Hum Genet66312319200010.1086/30272310631160

16. 

J KohlhaseA WischermannH ReichenbachU FrosterW EngelMutations in the SALL1 putative transcription factor gene cause Townes-Brocks syndromeNat Genet188183199810.1038/ng0198-819425907

17. 

S KangJM Graham JrAH OlneyLG BieseckerGLI3 frameshift mutations cause autosomal dominant Pallister-Hall syndromeNat Genet15266268199710.1038/ng0397-266

18. 

H CleversWnt/beta-catenin signaling in development and diseaseCell127469480200610.1016/j.cell.2006.10.01817081971

19. 

K DoreyE AmayaFGF signalling: diverse roles during early vertebrate embryogenesisDevelopment13737313742201010.1242/dev.03768920978071

20. 

X ZhangOA IbrahimiSK OlsenH UmemoriM MohammadiDM OrnitzReceptor specificity of the fibroblast growth factor family. The complete mammalian FGF familyJ Biol Chem2811569415700200610.1074/jbc.M60125220016597617

21. 

C Van de VenM BialeckaR NeijtsT YoungJE RowlandEJ StringerC van RooijenF MeijlinkA NóvoaJN FreundConcerted involvement of Cdx/Hox genes and Wnt signaling in morphogenesis of the caudal neural tube and cloacal derivatives from the posterior growth zoneDevelopment138345134622011

22. 

JR SpenceCN MayhewSA RankinMW KuharJE VallanceK TolleEE HoskinsVV KalinichenkoSI WellsAM ZornNF ShroyerJM WellsDirected differentiation of human pluripotent stem cells into intestinal tissue in vitroNature470105109201110.1038/nature0969121151107

23. 

M NakataY TakadaT HishikiT SaitoK TeruiY SatoH KosekiH YoshidaInduction of Wnt5a-expressing mesencymal cells adjacent to the cloacal plate is an essential process for its proximodistal elongation and subsequent anorectal developmentPediatr Res661491542009

24. 

CC TaiFG SalaHR FordKS WangC LiP MinooTC GrikscheitS BellusciWnt5a knock-out mouse as a new model of anorectal malformationJ Surg Res156278282200910.1016/j.jss.2009.03.08719577771

25. 

V MehtaLL AblerKP KeilCT SchmitzPS JoshiCM VezinaAtlas of Wnt and R-spondin gene expression in the developing male mouse lower urogenital tractDev Dyn24025482560201121936019

26. 

M LakoT StrachanP BullenDI WilsonSC RobsonS LindsayIsolation, characterisation and embryonic expression of WNT11, a gene which maps to 11q13.5 and has possible roles in the development of skeleton, kidney and lungGene21910111019989757009

27. 

H LickertA KispertS KutschR KemlerExpression patterns of Wnt genes in mouse gut developmentMech Dev105181184200110.1016/S0925-4773(01)00390-211429295

28. 

A RailoII NagyP KilpeläinenS VainioWnt-11 signaling leads to down-regulation of the Wnt/β-catenin, JNK/AP-1 and NF- B pathways and promotes viability in the CHO-K1 cellsExp Cell Res31423892399200818572162

29. 

BN CheyetteJS WaxmanJR MillerK TakemaruLC SheldahlN KhlebtsovaEP FoxT EarnestRT MoonDapper, a Dishevelled-associated antagonist of beta-catenin and JNK signaling, is required for notochord formationDev Cell2449461200210.1016/S1534-5807(02)00140-511970895

30. 

J WenYJ ChiangC GaoH XueJ XuY NingRJ HodesX GaoYG ChenLoss of Dact1 disrupts planar cell polarity signaling by altering dishevelled activity and leads to posterior malformation in miceJ Biol Chem2851102311030201010.1074/jbc.M109.08538120145239

31. 

ME PownallHV IsaacsFGF signalling in vertebrate developmentDevelopmental Biology, Book 2DS KesslerMorgan & Claypool Life SciencesCA14162010

32. 

T HaremakiY TanakaI HongoM YugeH OkamotoIntegration of multiple signal transducing pathways on Fgf response elements of the Xenopus caudal homologue Xcad3Development13049074917200310.1242/dev.0071812930781

33. 

TP YamaguchiS TakadaY YoshikawaN WuAP McMahonT (brachyury) is a direct target of Wnt3a during paraxial mesoderm specificationGenes Dev1331853190199910.1101/gad.13.24.318510617567

34. 

C PapapetrouF DrummondW ReardonR WinterL SpitzYH EdwardsA genetic study of the human T gene and its exclusion as a major candidate gene for sacral agenesis with anorectal atresiaJ Med Genet36208213199910204846

35. 

N GhebraniousRD BlankCL RaggioJ StaubliE McPhersonL IvacicK RasmussenFS JacobsenT FaciszewskiJK BurmesterA missense T (Brachyury) mutation contributes to vertebral malformationsJ Bone Miner Res23157615832008

36. 

S BagaiE RubioJF ChengR SweetR ThomasE FuchsR GradyM MitchellJA BassukFibroblast growth factor-10 is a mitogen for urothelial cellsJ Biol Chem2772382823837200210.1074/jbc.M20165820011923311

37. 

TJ FairbanksS De LangheFG SalaD WarburtonKD AndersonS BellusciRC BurnsFibroblast growth factor 10 (Fgf10) invalidation results in anorectal malformation in miceJ Pediatr Surg39360365200410.1016/j.jpedsurg.2003.11.03415017552

38. 

S YucelW LiuD CorderoA DonjacourG CunhaLS BaskinAnatomical studies of the fibroblast growth factor-10 mutant, Sonic Hedge Hog mutant and androgen receptor mutant mouse genital tubercleAdv Exp Med Biol545123148200410.1007/978-1-4419-8995-6_815086024

39. 

H OhashiH NishimotoJ NishimuraM SatoS ImaizumiT AiharaY FukushimaAnorectal anomaly in Pfeiffer syndromeClin Dysmorphol2283319938298735

40. 

WJ ParkGA MeyersX LiC ThedaD DaySJ OrlowMC JonesEW JabsNovel FGFR2 mutations in Crouzon and Jackson-Weiss syndromes show allelic heterogeneity and phenotypic variabilityHum Mol Genet412291233199510.1093/hmg/4.7.12298528214

41. 

WJ ParkC ThedaNE MaestriGA MeyersJS FryburgC DufresneMM Cohen JrEW JabsAnalysis of phenotypic features and FGFR2 mutations in Apert syndromeAm J Hum Genet5732132819957668257

42. 

RA PfeifferS RinnertR PoppG RöckeleinAsymmetrical coronal synostosis, cutaneous syndactyly of the fingers and toes, and jejunal atresia in a male infantAm J Med Genet63175176199610.1002/(SICI)1096-8628(19960503)63:1%3C175::AID-AJMG30%3E3.0.CO;2-J8723105

43. 

KA PrzylepaW PaznekasM ZhangM GolabiW BiasMJ BamshadJC CareyBD HallR StevensonSJ OrlowFibroblast growth factor receptor 2 mutations in Beare-Stevenson cutis gyrata syndromeNat Genet13492494199610.1038/ng0896-4928696350

44. 

BP LeHeupJP MasuttiP DroulléJ TisserandThe Antley-Bixler syndrome: report of two familial cases with severe renal and anal anomaliesEur J Pediatr154130133199510.1007/BF019919167720741

45. 

F SchaeferC AndersonB CanB SayNovel mutation in the FGFR2 gene at the same codon as the Crouzon syndrome mutations in a severe Pfeiffer syndrome type 2 caseAm J Med Genet75252255199810.1002/(SICI)1096-8628(19980123)75:3%3C252::AID-AJMG4%3E3.0.CO;2-S9475591

46. 

A KřepelováA BaxováP CaldaR PlavkaJ KaprasFGFR2 gene mutation (Tyr375Cys) in a new case of Beare-Stevenson syndromeAm J Med Genet7636236419989545103

47. 

T KodakaY KanamoriM SugiyamaK HashizumeA case of acrocephalosyndactyly with low imperforate anusJ Pediatr Surg39E32E34200410.1016/j.jpedsurg.2003.09.03714694405

48. 

MB ShapiroP SenapathyRNA splice junctions of different classes of eukaryotes: sequence statistics and functional implications in gene expressionNucleic Acids Res1571557174198710.1093/nar/15.17.71553658675

49. 

AO WilkieBad bones, absent smell, selfish testes: The pleiotropic consequences of human FGF receptor mutationsCytokine Growth Factor Rev16187203200510.1016/j.cytogfr.2005.03.00115863034

50. 

ET ShifleySE ColeThe vertebrate segmentation clock and its role in skeletal birth defectsBirth Defects Res C Embryo Today81121133200710.1002/bdrc.2009017600784

51. 

JM SchwarzC RödelspergerM SchuelkeD SeelowMutationTaster evaluates disease-causing potential of sequence alterationsNat Methods7575576201010.1038/nmeth0810-57520676075

52. 

J ThusbergA OlatubosunM VihinenPerformance of mutation pathogenicity prediction methods on missense variantsHum Mutat32358368201110.1002/humu.2144521412949

53. 

K ChunJ Siegel-BarteltD ChitayatJ PhillipsPN RayFGFR2 mutation associated with clinical manifestations consistent with Antley-Bixler syndromeAm J Med Genet77219224199810.1002/(SICI)1096-8628(19980518)77:3%3C219::AID-AJMG6%3E3.0.CO;2-K9605588

54. 

CHT ParkJH PruittD BennettA mouse model for neural tube defects: The curtailed (Tc) mutation produces spina bifida occulta in Tc/+ animals and spina bifida with meningomyelocele in Tc/tTeratology3930331219892658196

55. 

J McGaughranS SinnottR SusmanMF BuckleyG ElakisT CoxT RoscioliA case of Beare-Stevenson syndrome with a broad spectrum of features and a review of the FGFR2 Y375C mutation phenotypeClin Dysmorphol158993200610.1097/01.mcd.0000194407.92676.9d16531735

56. 

AL ShanskeD StaffenbergJT GoodrichSacral appendage in a child with an FGFR2 mutation: a report and reviewAm J Med Genet A146A21722175200810.1002/ajmg.a.3243618629881

57. 

M SeriG MartuccielloL PaleariA BolinoM PrioloG SalemiP ForaboscoF CaroliR CusanoT ToccoExclusion of the Sonic Hedgehog gene as responsible for Currarino syndrome and anorectal malformations with sacral hypodevelopmentHum Genet104108110199910.1007/s00439005091910071202

58. 

V KrügerM KhoshvaghtiH ReutterH VogtTM BoemersM LudwigInvestigation of FGF10 as a candidate gene in patients with anorectal malformations and exstrophy of the cloacaPediatr Surg Int248938972008

59. 

NB AgochukwuDE Pineda-AlvarezAA KeatonN Warren-MoraMS RaamA KamatSC ChandrasekharappaBD SolomonAnalysis of FOXF1 and the FOX gene cluster in patients with VACTERL associationEur J Med Genet54323328201110.1016/j.ejmg.2011.01.00721315191

60. 

M CatalaGenetic control of caudal developmentClin Genet618996200210.1034/j.1399-0004.2002.610202.x11940082

61. 

L GrumolatoG LiuP MongR MudbharyR BiswasR ArroyaveS VijayakumarAN EconomidesSA AaronsonCanonical and noncanonical Wnts use a common mechanism to activate completely unrelated coreceptorsGenes Dev2425172530201010.1101/gad.195771021078818

62. 

P Uysal-OnganerRM KyptaWnt11 in 2011 - the regulation and function of a non-canonical WntActa Physiol (Oxf)2045264201210.1111/j.1748-1716.2011.02297.x21447091

Related Articles

Journal Cover

December 2012
Volume 30 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Draaken M, Prins W, Zeidler C, Hilger A, Mughal SS, Latus J, Boemers TM, Schmidt D, Schmiedeke E, Spychalski N, Spychalski N, et al: Involvement of the WNT and FGF signaling pathways in non-isolated anorectal malformations: Sequencing analysis of WNT3A, WNT5A, WNT11, DACT1, FGF10, FGFR2 and the T gene. Int J Mol Med 30: 1459-1464, 2012
APA
Draaken, M., Prins, W., Zeidler, C., Hilger, A., Mughal, S.S., Latus, J. ... Ludwig, M. (2012). Involvement of the WNT and FGF signaling pathways in non-isolated anorectal malformations: Sequencing analysis of WNT3A, WNT5A, WNT11, DACT1, FGF10, FGFR2 and the T gene. International Journal of Molecular Medicine, 30, 1459-1464. https://doi.org/10.3892/ijmm.2012.1124
MLA
Draaken, M., Prins, W., Zeidler, C., Hilger, A., Mughal, S. S., Latus, J., Boemers, T. M., Schmidt, D., Schmiedeke, E., Spychalski, N., Bartels, E., Nöthen, M. M., Reutter, H., Ludwig, M."Involvement of the WNT and FGF signaling pathways in non-isolated anorectal malformations: Sequencing analysis of WNT3A, WNT5A, WNT11, DACT1, FGF10, FGFR2 and the T gene". International Journal of Molecular Medicine 30.6 (2012): 1459-1464.
Chicago
Draaken, M., Prins, W., Zeidler, C., Hilger, A., Mughal, S. S., Latus, J., Boemers, T. M., Schmidt, D., Schmiedeke, E., Spychalski, N., Bartels, E., Nöthen, M. M., Reutter, H., Ludwig, M."Involvement of the WNT and FGF signaling pathways in non-isolated anorectal malformations: Sequencing analysis of WNT3A, WNT5A, WNT11, DACT1, FGF10, FGFR2 and the T gene". International Journal of Molecular Medicine 30, no. 6 (2012): 1459-1464. https://doi.org/10.3892/ijmm.2012.1124