Disruption of an AP-2alpha binding site in an IRF6 enhancer is associated with cleft lip

Fedik Rahimov, Mary L Marazita, Axel Visel, Margaret E Cooper, Michael J Hitchler, Michele Rubini, Frederick E Domann, Manika Govil, Kaare Christensen, Camille Bille, Mads Melbye, Astanand Jugessur, Rolv T Lie, Allen J Wilcox, David R Fitzpatrick, Eric D Green, Peter A Mossey, Julian Little, Regine P Steegers-Theunissen, Len A Pennacchio, Brian C Schutte, Jeffrey C Murray, Fedik Rahimov, Mary L Marazita, Axel Visel, Margaret E Cooper, Michael J Hitchler, Michele Rubini, Frederick E Domann, Manika Govil, Kaare Christensen, Camille Bille, Mads Melbye, Astanand Jugessur, Rolv T Lie, Allen J Wilcox, David R Fitzpatrick, Eric D Green, Peter A Mossey, Julian Little, Regine P Steegers-Theunissen, Len A Pennacchio, Brian C Schutte, Jeffrey C Murray

Abstract

Previously we have shown that nonsyndromic cleft lip with or without cleft palate (NSCL/P) is strongly associated with SNPs in IRF6 (interferon regulatory factor 6). Here, we use multispecies sequence comparisons to identify a common SNP (rs642961, G>A) in a newly identified IRF6 enhancer. The A allele is significantly overtransmitted (P = 1 x 10(-11)) in families with NSCL/P, in particular those with cleft lip but not cleft palate. Further, there is a dosage effect of the A allele, with a relative risk for cleft lip of 1.68 for the AG genotype and 2.40 for the AA genotype. EMSA and ChIP assays demonstrate that the risk allele disrupts the binding site of transcription factor AP-2alpha and expression analysis in the mouse localizes the enhancer activity to craniofacial and limb structures. Our findings place IRF6 and AP-2alpha in the same developmental pathway and identify a high-frequency variant in a regulatory element contributing substantially to a common, complex disorder.

Figures

Figure 1
Figure 1
Transcription factor AP-2α binds to MCS-9.7 and rs642961 disrupts its binding site. (a) Multispecies sequence alignment of the MCS-9.7 segment corresponding to nucleotides from –14470 to –14535, with respect to IRF6 translation initiation codon, that contains variants -14474A>G, rs642961 and -14523G>A and the predicted AP-2α binding site (highlighted in yellow). (b) The double-stranded IRDye-700 labeled oligonucleotide probes used in EMSA. The probes correspond to MCS-9.7 nucleotides –14495 to –14522 and differ only at the rs642961 SNP. (c) EMSA using probe alone (lane 1) and incubated with the human recombinant AP-2α protein, allele G (lane 2) and allele A (lane 3). Competition with increasing amounts of unlabeled probes with allele G (lanes 4–6) and allele A (lanes 7–9). Supershift is formed when AP-2α is pre-incubated with anti-AP-2α antibody (AP-2α-Ab) (lane 10). (d) Quantification of ChIP by real-time PCR. AP-2α-Ab and IgG immunoprecipitated chromatin fragments were amplified using primers flanking the MCS-9.7 segment with four putative AP-2α binding sites shown in Supplementary Figure 1 and a control region of a similar length devoid of predicted AP-2α binding sites. The curves represent the accumulation of PCR products given as log values (logΔR) of SYBR-Green fluorescence intensity plotted against the number of cycles. MCS-9.7 amplification plot of the immunoprecipitated chromatin from uninfected (Plot I) and ad-AP-2α-infected (Plot III) HaCaT cells. Amplification curves of the control region are shown on Plot II for the uninfected cells and on Plot IV for the infected cells.
Figure 2
Figure 2
MCS-9.7 shows IRF6 enhancer activity in transgenic mouse assay. (a-a”) Lateral views of three independent transgenic embryos at embryonic day 11.5 (E11.5) expressing LacZ directed by MCS-9.7. (b) Frontal view and (c) expanded view of the orofacial region of the embryo shown in panel a”. White arrow points to LacZ expression at the fusion sites between the lateral nasal (ln), medial nasal (mn) and maxillary (mx) prominences towards the end of upper lip formation at E11.5. (d) Sagittal virtual section through the orofacial region of the embryo shown in panel a” generated with optical projection tomography. Shown in red (white arrow) is LacZ expression in ectoderm covering fusing facial prominences (ln, mn and mx). See 3D views of embryos a-a″ in Supplementary videos online

References

    1. Jugessur A, Murray JC. Orofacial clefting: recent insights into a complex trait. Curr Opin Genet Dev. 2005;15:270–278.
    1. Zucchero TM, et al. Interferon regulatory factor 6 (IRF6) gene variants and the risk of isolated cleft lip or palate. N Engl J Med. 2004;351:769–780.
    1. Kondo S, et al. Mutations in IRF6 cause Van der Woude and popliteal pterygium syndromes. Nat Genet. 2002;32:285–289.
    1. Scapoli L, et al. Strong evidence of linkage disequilibrium between polymorphisms at the IRF6 locus and nonsyndromic cleft lip with or without cleft palate, in an Italian population. Am J Hum Genet. 2005;76:180–183.
    1. Blanton SH, et al. Variation in IRF6 contributes to nonsyndromic cleft lip and palate. Am J Med Genet A. 2005;137:259–262.
    1. Ghassibe M, et al. Interferon regulatory factor-6: a gene predisposing to isolated cleft lip with or without cleft palate in the Belgian population. Eur J Hum Genet. 2005;13:1239–1242.
    1. Park JW, et al. Association between IRF6 and nonsyndromic cleft lip with or without cleft palate in four populations. Genet Med. 2007;9:219–227.
    1. Schorle H, Meier P, Buchert M, Jaenisch R, Mitchell PJ. Transcription factor AP-2 essential for cranial closure and craniofacial development. Nature. 1996;381:235–238.
    1. Milunsky JM, et al. TFAP2A mutations result in branchio-oculo-facial syndrome. Am J Hum Genet. 2008;82:1171–1177.
    1. Horvath S, Xu X, Laird NM. The family based association test method: strategies for studying general genotype--phenotype associations. Eur J Hum Genet. 2001;9:301–306.
    1. Graham RR, et al. Three functional variants of IFN regulatory factor 5 (IRF5) define risk and protective haplotypes for human lupus. Proc Natl Acad Sci U S A. 2007;104:6758–6763.
    1. Bille C, et al. Oral clefts and life style factors--a case-cohort study based on prospective Danish data. Eur J Epidemiol. 2007;22:173–181.
    1. Nguyen RH, Wilcox AJ, Moen BE, McConnaughey DR, Lie RT. Parent's occupation and isolated orofacial clefts in Norway: a population-based case-control study. Ann Epidemiol. 2007;17:763–771.
    1. Mitchell LE, et al. Guidelines for the design and analysis of studies on nonsyndromic cleft lip and cleft palate in humans: summary report from a Workshop of the International Consortium for Oral Clefts Genetics. Cleft Palate Craniofac J. 2002;39:93–100.
    1. Harville EW, Wilcox AJ, Lie RT, Vindenes H, Abyholm F. Cleft lip and palate versus cleft lip only: are they distinct defects? Am J Epidemiol. 2005;162:448–453.
    1. Marazita ML, et al. Meta-analysis of 13 genome scans reveals multiple cleft lip/palate genes with novel loci on 9q21 and 2q32–35. Am J Hum Genet. 2004;75:161–173.
    1. Mossey P. Epidemiology underpinning research in the aetiology of orofacial clefts. Orthod Craniofac Res. 2007;10:114–120.
    1. Poulin F, et al. In vivo characterization of a vertebrate ultraconserved enhancer. Genomics. 2005;85:774–781.
    1. Knight AS, Schutte BC, Jiang R, Dixon MJ. Developmental expression analysis of the mouse and chick orthologues of IRF6: the gene mutated in Van der Woude syndrome. Dev Dyn. 2006;235:1441–1447.
    1. Sharpe J, et al. Optical projection tomography as a tool for 3D microscopy and gene expression studies. Science. 2002;296:541–545.
    1. Cirulli ET, Goldstein DB. In vitro assays fail to predict in vivo effects of regulatory polymorphisms. Hum Mol Genet. 2007;16:1931–1939.
    1. Vieira AR, et al. Medical sequencing of candidate genes for nonsyndromic cleft lip and palate. PLoS Genet. 2005;1:e64.
    1. Riley BM, et al. Impaired FGF signaling contributes to cleft lip and palate. Proc Natl Acad Sci U S A. 2007;104:4512–4517.
    1. Thomas JW, et al. Comparative analyses of multi-species sequences from targeted genomic regions. Nature. 2003;424:788–793.
    1. Schwartz S, et al. MultiPipMaker and supporting tools: Alignments and analysis of multiple genomic DNA sequences. Nucleic Acids Res. 2003;31:3518–3524.
    1. Margulies EH, Blanchette M, Haussler D, Green ED. Identification and characterization of multi-species conserved sequences. Genome Res. 2003;13:2507–2518.
    1. Nickerson DA, Tobe VO, Taylor SL. PolyPhred: automating the detection and genotyping of single nucleotide substitutions using fluorescence-based resequencing. Nucleic Acids Res. 1997;25:2745–2751.
    1. Weinberg CR. Methods for detection of parent-of-origin effects in genetic studies of case-parents triads. Am J Hum Genet. 1999;65:229–235.
    1. Dudbridge F. Likelihood-based association analysis for nuclear families and unrelated subjects with missing genotype data. Hum Hered. 2008;66:87–98.
    1. Provenzano MJ, et al. AP-2 participates in the transcriptional control of the amyloid precursor protein (APP) gene in oral squamous cell carcinoma. Exp Mol Pathol. 2007;83:277–282.

Source: PubMed

3
Subskrybuj