Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Am J Hum Genet ; 110(1): 71-91, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36493769

ABSTRACT

Cleft lip with or without cleft palate (CL/P) is a common birth defect with a complex, heterogeneous etiology. It is well established that common and rare sequence variants contribute to the formation of CL/P, but the contribution of copy-number variants (CNVs) to cleft formation remains relatively understudied. To fill this knowledge gap, we conducted a large-scale comparative analysis of genome-wide CNV profiles of 869 individuals from the Philippines and 233 individuals of European ancestry with CL/P with three primary goals: first, to evaluate whether differences in CNV number, amount of genomic content, or amount of coding genomic content existed within clefting subtypes; second, to assess whether CNVs in our cohort overlapped with known Mendelian clefting loci; and third, to identify unestablished Mendelian clefting genes. Significant differences in CNVs across cleft types or in individuals with non-syndromic versus syndromic clefts were not observed; however, several CNVs in our cohort overlapped with known syndromic and non-syndromic Mendelian clefting loci. Moreover, employing a filtering strategy relying on population genetics data that rare variants are on the whole more deleterious than common variants, we identify several CNV-associated gene losses likely driving non-syndromic clefting phenotypes. By prioritizing genes deleted at a rare frequency across multiple individuals with clefts yet enriched in our cohort of individuals with clefts compared to control subjects, we identify COBLL1, RIC1, and ARHGEF38 as clefting genes. CRISPR-Cas9 mutagenesis of these genes in Xenopus laevis and Danio rerio yielded craniofacial dysmorphologies, including clefts analogous to those seen in human clefting disorders.


Subject(s)
Cleft Lip , Cleft Palate , DNA Copy Number Variations , Humans , Cleft Lip/genetics , Cleft Palate/genetics , Genome-Wide Association Study , Guanine Nucleotide Exchange Factors/genetics , Phenotype , Transcription Factors/genetics
2.
Hum Mol Genet ; 29(5): 845-858, 2020 03 27.
Article in English | MEDLINE | ID: mdl-31943082

ABSTRACT

SPECC1L mutations have been identified in patients with rare atypical orofacial clefts and with syndromic cleft lip and/or palate (CL/P). These mutations cluster in the second coiled-coil and calponin homology domains of SPECC1L and severely affect the ability of SPECC1L to associate with microtubules. We previously showed that gene-trap knockout of Specc1l in mouse results in early embryonic lethality. We now present a truncation mutant mouse allele, Specc1lΔC510, that results in perinatal lethality. Specc1lΔC510/ΔC510 homozygotes showed abnormal palate rugae but did not show cleft palate. However, when crossed with a gene-trap allele, Specc1lcGT/ΔC510 compound heterozygotes showed a palate elevation delay with incompletely penetrant cleft palate. Specc1lcGT/ΔC510 embryos exhibit transient oral epithelial adhesions at E13.5, which may delay shelf elevation. Consistent with oral adhesions, we show periderm layer abnormalities, including ectopic apical expression of adherens junction markers, similar to Irf6 hypomorphic mutants and Arhgap29 heterozygotes. Indeed, SPECC1L expression is drastically reduced in Irf6 mutant palatal shelves. Finally, we wanted to determine if SPECC1L deficiency also contributed to non-syndromic (ns) CL/P. We sequenced 62 Caucasian, 89 Filipino, 90 Ethiopian, 90 Nigerian and 95 Japanese patients with nsCL/P and identified three rare coding variants (p.Ala86Thr, p.Met91Iso and p.Arg546Gln) in six individuals. These variants reside outside of SPECC1L coiled-coil domains and result in milder functional defects than variants associated with syndromic clefting. Together, our data indicate that palate elevation is sensitive to deficiency of SPECC1L dosage and function and that SPECC1L cytoskeletal protein functions downstream of IRF6 in palatogenesis.


Subject(s)
Cleft Palate/pathology , Interferon Regulatory Factors/metabolism , Mutation , Phosphoproteins/physiology , Animals , Cleft Palate/genetics , Cleft Palate/metabolism , Female , Humans , Interferon Regulatory Factors/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoproteins/genetics , Phosphoproteins/metabolism
3.
Am J Hum Genet ; 102(6): 1143-1157, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29805042

ABSTRACT

Non-syndromic cleft lip with or without cleft palate (NS-CL/P) is one of the most common human birth defects and is generally considered a complex trait. Despite numerous loci identified by genome-wide association studies, the effect sizes of common variants are relatively small, with much of the presumed genetic contribution remaining elusive. We report exome-sequencing results in 209 people from 72 multi-affected families with pedigree structures consistent with autosomal-dominant inheritance and variable penetrance. Herein, pathogenic variants are described in four genes encoding components of the p120-catenin complex (CTNND1, PLEKHA7, PLEKHA5) and an epithelial splicing regulator (ESRP2), in addition to the known CL/P-associated gene, CDH1, which encodes E-cadherin. The findings were also validated in a second cohort of 497 people with NS-CL/P, comprising small families and singletons with pathogenic variants in these genes identified in 14% of multi-affected families and 2% of the replication cohort of smaller families. Enriched expression of each gene/protein in human and mouse embryonic oro-palatal epithelia, demonstration of functional impact of CTNND1 and ESRP2 variants, and recapitulation of the CL/P spectrum in Ctnnd1 knockout mice support a causative role in CL/P pathogenesis. These data show that primary defects in regulators of epithelial cell adhesion are the most significant contributors to NS-CL/P identified to date and that inherited and de novo single gene variants explain a substantial proportion of NS-CL/P.


Subject(s)
Cadherins/genetics , Catenins/genetics , Cleft Lip/genetics , Cleft Palate/genetics , Genetic Predisposition to Disease , Mutation/genetics , Alleles , Amino Acid Sequence , Animals , Biotinylation , Epithelium/metabolism , Epithelium/pathology , Female , Gene Deletion , Humans , Infant , Infant, Newborn , Male , Mice , Palate/pathology , Pedigree , Syndrome , Exome Sequencing , Delta Catenin
4.
Hum Mutat ; 40(10): 1813-1825, 2019 10.
Article in English | MEDLINE | ID: mdl-31215115

ABSTRACT

Cleft lip with or without cleft palate (CL/P) is generally viewed as a complex trait with multiple genetic and environmental contributions. In 70% of cases, CL/P presents as an isolated feature and/or deemed nonsyndromic. In the remaining 30%, CL/P is associated with multisystem phenotypes or clinically recognizable syndromes, many with a monogenic basis. Here we report the identification, via exome sequencing, of likely pathogenic variants in two genes that encode interacting proteins previously only linked to orofacial clefting in mouse models. A variant in GDF11 (encoding growth differentiation factor 11), predicting a p.(Arg298Gln) substitution at the Furin protease cleavage site, was identified in one family that segregated with CL/P and both rib and vertebral hypersegmentation, mirroring that seen in Gdf11 knockout mice. In the second family in which CL/P was the only phenotype, a mutation in FST (encoding the GDF11 antagonist, Follistatin) was identified that is predicted to result in a p.(Cys56Tyr) substitution in the region that binds GDF11. Functional assays demonstrated a significant impact of the specific mutated amino acids on FST and GDF11 function and, together with embryonic expression data, provide strong evidence for the importance of GDF11 and Follistatin in the regulation of human orofacial development.


Subject(s)
Bone Morphogenetic Proteins/genetics , Cleft Lip/diagnosis , Cleft Lip/genetics , Follistatin/metabolism , Genetic Association Studies , Genetic Predisposition to Disease , Growth Differentiation Factors/genetics , Mutation , Alleles , Amino Acid Substitution , Bone Morphogenetic Proteins/antagonists & inhibitors , Cell Line , Computational Biology/methods , Follistatin/chemistry , Genetic Association Studies/methods , Genomics/methods , Growth Differentiation Factors/antagonists & inhibitors , Humans , Models, Molecular , Pedigree , Protein Conformation , Exome Sequencing
5.
Am J Hum Genet ; 98(4): 744-54, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27018472

ABSTRACT

Cleft palate (CP) is a common birth defect occurring in 1 in 2,500 live births. Approximately half of infants with CP have a syndromic form, exhibiting other physical and cognitive disabilities. The other half have nonsyndromic CP, and to date, few genes associated with risk for nonsyndromic CP have been characterized. To identify such risk factors, we performed a genome-wide association study of this disorder. We discovered a genome-wide significant association with a missense variant in GRHL3 (p.Thr454Met [c.1361C>T]; rs41268753; p = 4.08 × 10(-9)) and replicated the result in an independent sample of case and control subjects. In both the discovery and replication samples, rs41268753 conferred increased risk for CP (OR = 8.3, 95% CI 4.1-16.8; OR = 2.16, 95% CI 1.43-3.27, respectively). In luciferase transactivation assays, p.Thr454Met had about one-third of the activity of wild-type GRHL3, and in zebrafish embryos, perturbed periderm development. We conclude that this mutation is an etiologic variant for nonsyndromic CP and is one of few functional variants identified to date for nonsyndromic orofacial clefting. This finding advances our understanding of the genetic basis of craniofacial development and might ultimately lead to improvements in recurrence risk prediction, treatment, and prognosis.


Subject(s)
Cleft Palate/genetics , DNA-Binding Proteins/genetics , Polymorphism, Single Nucleotide , Transcription Factors/genetics , Animals , Case-Control Studies , Cleft Palate/diagnosis , Disease Models, Animal , Ethnicity/genetics , Genetic Loci , Genome-Wide Association Study , Genotyping Techniques , Humans , Mutation, Missense , Risk Factors , Zebrafish/embryology , Zebrafish/genetics
6.
Genet Epidemiol ; 41(8): 887-897, 2017 12.
Article in English | MEDLINE | ID: mdl-29124805

ABSTRACT

Orofacial clefts (OFCs) are common, complex birth defects with extremely heterogeneous phenotypic presentations. Two common subtypes-cleft lip alone (CL) and CL plus cleft palate (CLP)-are typically grouped into a single phenotype for genetic analysis (i.e., CL with or without cleft palate, CL/P). However, mounting evidence suggests there may be unique underlying pathophysiology and/or genetic modifiers influencing expression of these two phenotypes. To this end, we performed a genome-wide scan for genetic modifiers by directly comparing 450 CL cases with 1,692 CLP cases from 18 recruitment sites across 13 countries from North America, Central or South America, Asia, Europe, and Africa. We identified a region on 16q21 that is strongly associated with different cleft type (P = 5.611 × 10-8 ). We also identified significant evidence of gene-gene interactions between this modifier locus and two recognized CL/P risk loci: 8q21 and 9q22 (FOXE1) (P = 0.012 and 0.023, respectively). Single nucleotide polymorphism (SNPs) in the 16q21 modifier locus demonstrated significant association with CL over CLP. The marker alleles on 16q21 that increased risk for CL were found at highest frequencies among individuals with a family history of CL (P = 0.003). Our results demonstrate the existence of modifiers for which type of OFC develops and suggest plausible elements responsible for phenotypic heterogeneity, further elucidating the complex genetic architecture of OFCs.


Subject(s)
Brain/abnormalities , Chromosomes, Human, Pair 16 , Cleft Lip/genetics , Cleft Palate/genetics , Alleles , Brain/pathology , Cleft Lip/pathology , Cleft Palate/pathology , Female , Forkhead Transcription Factors/genetics , Genetic Loci , Genome-Wide Association Study , Humans , Male , Phenotype , Polymorphism, Single Nucleotide , Racial Groups/genetics , Risk Factors
7.
Am J Hum Genet ; 96(3): 397-411, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25704602

ABSTRACT

Although genome-wide association studies (GWASs) for nonsyndromic orofacial clefts have identified multiple strongly associated regions, the causal variants are unknown. To address this, we selected 13 regions from GWASs and other studies, performed targeted sequencing in 1,409 Asian and European trios, and carried out a series of statistical and functional analyses. Within a cluster of strongly associated common variants near NOG, we found that one, rs227727, disrupts enhancer activity. We furthermore identified significant clusters of non-coding rare variants near NTN1 and NOG and found several rare coding variants likely to affect protein function, including four nonsense variants in ARHGAP29. We confirmed 48 de novo mutations and, based on best biological evidence available, chose two of these for functional assays. One mutation in PAX7 disrupted the DNA binding of the encoded transcription factor in an in vitro assay. The second, a non-coding mutation, disrupted the activity of a neural crest enhancer downstream of FGFR2 both in vitro and in vivo. This targeted sequencing study provides strong functional evidence implicating several specific variants as primary contributory risk alleles for nonsyndromic clefting in humans.


Subject(s)
Brain/abnormalities , Carrier Proteins/genetics , Cleft Lip/genetics , Cleft Palate/genetics , PAX7 Transcription Factor/genetics , Polymorphism, Single Nucleotide , Receptor, Fibroblast Growth Factor, Type 2/genetics , Alleles , Amino Acid Sequence , Animals , Asian People/genetics , Carrier Proteins/metabolism , Cell Line , Epithelial Cells/metabolism , Gene Expression Regulation , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Mutation, Missense , PAX7 Transcription Factor/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Sequence Analysis, DNA , Transcription Factors/genetics , Transcription Factors/metabolism , White People/genetics , Zebrafish/genetics
8.
Cleft Palate Craniofac J ; 54(3): 281-286, 2017 05.
Article in English | MEDLINE | ID: mdl-27115562

ABSTRACT

OBJECTIVE: Interferon Regulatory Factor 6 (IRF6) is critical for craniofacial development, epidermal differentiation, and tissue repair. IRF6 mutations cause Van der Woude Syndrome (VWS) and Popliteal Pterygium Syndrome. Individuals with VWS exhibit craniofacial anomalies, including cleft lip and palate and lip pits. Furthermore, they have an increased risk for wound-healing complications following surgical repair when compared with patients with nonsyndromic cleft lip and palate (NSCLP). However, nothing is known about the skin of these patients. The objective was to characterize the skin of patients with VWS. We hypothesize that IRF6 is required for proper skin homeostasis in humans. DESIGN: Discarded tissue from a hip was collected during surgical alveolar bone graft. Samples from children with VWS harboring IRF6 mutations (n = 2) were compared with samples from children with NSCLP (n = 7). Histology was assessed following hematoxylin and eosin staining. The expressions of Proliferating Cell Nuclear Antigen, IRF6, P63, and Keratin 10 were determined by immunofluorescence. Keratinocytes were isolated and their proliferation potential was assessed by colony-forming efficiency assay. RESULTS: Hip skin from children with VWS showed a thicker epidermis when compared with that from children with NSCLP. Proliferating Cell Nuclear Antigen staining revealed an increase in proliferation in syndromic tissues when compared with controls. However, P63 and Keratin 10 expression were similar between groups. Finally, keratinocytes from VWS showed increased long-term proliferation when compared with NSCLP. CONCLUSIONS: These results support, in vivo and in vitro, a previously described role for IRF6 in epidermal proliferation in humans. They further demonstrate a critical function for IRF6 in cutaneous homeostasis.


Subject(s)
Abnormalities, Multiple/genetics , Cleft Lip/genetics , Cleft Palate/genetics , Cysts/genetics , Interferon Regulatory Factors/genetics , Keratinocytes/cytology , Lip/abnormalities , Skin Abnormalities/genetics , Cell Proliferation , Child , Female , Fluorescent Antibody Technique , Humans , Male , Mutation , Phenotype
9.
Am J Med Genet A ; 167A(5): 1054-1060, 2015 May.
Article in English | MEDLINE | ID: mdl-25786657

ABSTRACT

Genome wide association (GWA) studies have successfully identified at least a dozen loci associated with orofacial clefts. However, these signals may be unique to specific populations and require replication to validate and extend findings as a prelude to etiologic SNP discovery. We attempted to replicate the findings of a recent meta-analysis of orofacial cleft GWA studies using four different ancestral populations. We studied 946 pedigrees (3,436 persons) of European (US white and Danish) and Asian (Japanese and Mongolian) origin. We genotyped six SNPs that represented the most significant P-value associations identified in published studies: rs742071 (1p36), rs7590268 (2p21), rs7632427 (3p11.1), rs12543318 (8q21.3), rs8001641 (13q31.1), and rs7179658 (15q22.2). We directly sequenced three non-coding conserved regions 200 kb downstream of SPRY2 in 713 cases, 438 controls, and 485 trios from the US, Mongolia, and the Philippines. We found rs8001641 to be significantly associated with nonsyndromic cleft lip with cleft palate (NSCLP) in Europeans (P-value = 4 × 10(-5), ORtransmission = 1.86 with 95% confidence interval: 1.38-2.52). We also found several novel sequence variants in the conserved regions in Asian and European samples, which may help to localize common variants contributing directly to the risk for NSCLP. This study confirms the prior association between rs8001641 and NSCLP in European populations.


Subject(s)
Chromosomes, Human, Pair 13/genetics , Cleft Lip/genetics , Cleft Palate/genetics , Genetic Predisposition to Disease , Asian People/genetics , Cleft Lip/physiopathology , Cleft Palate/physiopathology , Female , Genome-Wide Association Study , Humans , Male , Pedigree , Polymorphism, Single Nucleotide , Risk , White People/genetics
10.
Am J Med Genet A ; 167A(3): 545-52, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25691407

ABSTRACT

The popliteal pterygia syndromes are a distinct subset of the hundreds of Mendelian orofacial clefting syndromes. Popliteal pterygia syndromes have considerable variability in severity and in the associated phenotypic features but are all characterized by cutaneous webbing across one or more major joints, cleft lip and/or palate, syndactyly, and genital malformations. Heterozygous mutations in IRF6 cause popliteal pterygium syndrome (PPS) while homozygous mutations in RIPK4 or CHUK (IKKA) cause the more severe Bartsocas-Papas syndrome (BPS) and Cocoon syndrome, respectively. In this study, we report mutations in six pedigrees with children affected with PPS or BPS. Using a combination of Sanger and exome sequencing, we report the first case of an autosomal recessive popliteal pterygium syndrome caused by homozygous mutation of IRF6 and the first case of uniparental disomy of chromosome 21 leading to a recessive disorder. We also demonstrate that mutations in RIPK4 can cause features with a range of severity along the PPS-BPS spectrum and that mutations in IKKA can cause a range of features along the BPS-Cocoon spectrum. Our findings have clinical implications for genetic counseling of families with pterygia syndromes and further implicate IRF6, RIPK4, and CHUK (IKKA) in potentially interconnected pathways governing epidermal and craniofacial development.


Subject(s)
Cleft Lip/diagnosis , Cleft Lip/genetics , Cleft Palate/diagnosis , Cleft Palate/genetics , Eye Abnormalities/diagnosis , Eye Abnormalities/genetics , Fingers/abnormalities , Genetic Association Studies , Knee Joint/abnormalities , Lower Extremity Deformities, Congenital/diagnosis , Lower Extremity Deformities, Congenital/genetics , Phenotype , Syndactyly/diagnosis , Syndactyly/genetics , Urogenital Abnormalities/diagnosis , Urogenital Abnormalities/genetics , Comparative Genomic Hybridization , DNA Mutational Analysis , Exome , Female , Genes, Recessive , Genome-Wide Association Study , High-Throughput Nucleotide Sequencing , Humans , I-kappa B Kinase/genetics , Infant , Infant, Newborn , Interferon Regulatory Factors/genetics , Knee/abnormalities , Male , Mutation , Pedigree , Protein Serine-Threonine Kinases/genetics
11.
Genet Med ; 15(5): 338-44, 2013 May.
Article in English | MEDLINE | ID: mdl-23154523

ABSTRACT

PURPOSE: Mutations in the transcription factor IRF6 cause allelic autosomal dominant clefting syndromes, Van der Woude syndrome, and popliteal pterygium syndrome. We compared the distribution of IRF6 coding and splice-site mutations from 549 families with Van der Woude syndrome or popliteal pterygium syndrome with that of variants from the 1000 Genomes and National Heart, Lung, and Blood Institute Exome Sequencing Projects. METHODS: We compiled all published pathogenic IRF6 mutations and performed direct sequencing of IRF6 in families with Van der Woude syndrome or popliteal pterygium syndrome. RESULTS: Although mutations causing Van der Woude syndrome or popliteal pterygium syndrome were nonrandomly distributed with significantly increased frequencies in the DNA-binding domain (P = 0.0001), variants found in controls were rare and evenly distributed in IRF6. Of 194 different missense or nonsense variants described as potentially pathogenic, we identified only two in more than 6,000 controls. PolyPhen and SIFT (sorting intolerant from tolerant) reported 5.9% of missense mutations in patients as benign, suggesting that use of current in silico prediction models to determine function can have significant false negatives. CONCLUSION: Mutation of IRF6 occurs infrequently in controls, suggesting that for IRF6 there is a high probability that disruption of the coding sequence, particularly the DNA-binding domain, will result in syndromic features. Prior associations of coding sequence variants in IRF6 with clefting syndromes have had few false positives.


Subject(s)
Abnormalities, Multiple/genetics , Cleft Lip/genetics , Cleft Palate/genetics , Computational Biology , Cysts/genetics , Exome , Eye Abnormalities/genetics , Interferon Regulatory Factors/genetics , Lower Extremity Deformities, Congenital/genetics , Mutation , Syndactyly/genetics , Urogenital Abnormalities/genetics , Computational Biology/methods , Databases, Nucleic Acid , Female , Fingers/abnormalities , Humans , Knee Joint/abnormalities , Lip/abnormalities , Mutation Rate , Mutation, Missense , Protein Interaction Domains and Motifs/genetics
12.
Biol Res Nurs ; 21(2): 157-165, 2019 03.
Article in English | MEDLINE | ID: mdl-30700110

ABSTRACT

OBJECTIVE: We examined genomic variation potentially associated with the cortisol stress response in children having a painful medical procedure. DESIGN: Children 4-10 years old having a peripheral intravenous line inserted provided saliva samples for evaluation of the cortisol response as a biological measure of distress: two on the day of the procedure and two at home on a nonstressful day for comparison values. Children and biological parents also provided samples for genotyping of variants with known or suspected association with the cortisol stress response. Analysis included child-only association and family-based transmission disequilibrium tests (TDTs). RESULTS: Genotype and phenotype data on the cortisol stress response were available from 326 children for child-only association analyses and 376 complete family trios for TDTs. Children were 50% female, an average of 7.5 years old, and mostly (83%) White/non-Hispanic. We identified four single-nucleotide polymorphisms (SNPs) potentially associated with the cortisol stress response: rs1176744 ( HTR3B), rs10062367 ( CRHBP), rs634479 ( OPRM1), and rs8030107 ( NTRK3). Family-based analysis identified a two-SNP haplotype in HTR1B suggestive for association with the cortisol response (rs6296, rs11568817). Allelic TDTs identified rs7897947 ( NFKB2) as potentially related to cortisol response. CONCLUSIONS: Findings provide preliminary evidence for genes potentially important in cortisol response to an acute stressor in children in the serotonin, dopamine, and brain-derived neurotrophic factor pathways, the hypothalamic-pituitary-adrenal axis, and the inflammatory response. Combined with analyses of related phenotypes and clinical data, these results could help identify patients at increased risk of adverse responses to painful medical procedures who might benefit from tailored interventions.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/physiology , Genetic Variation , Hydrocortisone/genetics , Hydrocortisone/metabolism , Stress, Psychological/genetics , Stress, Psychological/physiopathology , Adult , Alleles , Child , Child, Preschool , Female , Genotype , Haplotypes , Humans , Hypothalamo-Hypophyseal System/physiology , Male , Parents , Phenotype , Pituitary-Adrenal System/physiology , Polymorphism, Single Nucleotide
13.
Genetics ; 208(1): 283-296, 2018 01.
Article in English | MEDLINE | ID: mdl-29162626

ABSTRACT

Orofacial clefts are one of the most common birth defects, affecting 1-2 per 1000 births, and have a complex etiology. High-resolution array-based comparative genomic hybridization has increased the ability to detect copy number variants (CNVs) that can be causative for complex diseases such as cleft lip and/or palate. Utilizing this technique on 97 nonsyndromic cleft lip and palate cases and 43 cases with cleft palate only, we identified a heterozygous deletion of Isthmin 1 in one affected case, as well as a deletion in a second case that removes putative 3' regulatory information. Isthmin 1 is a strong candidate for clefting, as it is expressed in orofacial structures derived from the first branchial arch and is also in the same "synexpression group" as fibroblast growth factor 8 and sprouty RTK signaling antagonist 1a and 2, all of which have been associated with clefting. CNVs affecting Isthmin 1 are exceedingly rare in control populations, and Isthmin 1 scores as a likely haploinsufficiency locus. Confirming its role in craniofacial development, knockdown or clustered randomly interspaced short palindromic repeats/Cas9-generated mutation of isthmin 1 in Xenopus laevis resulted in mild to severe craniofacial dysmorphologies, with several individuals presenting with median clefts. Moreover, knockdown of isthmin 1 produced decreased expression of LIM homeobox 8, itself a gene associated with clefting, in regions of the face that pattern the maxilla. Our study demonstrates a successful pipeline from CNV identification of a candidate gene to functional validation in a vertebrate model system, and reveals Isthmin 1 as both a new human clefting locus as well as a key craniofacial patterning gene.


Subject(s)
Cleft Lip/genetics , Cleft Palate/genetics , Craniofacial Abnormalities/genetics , Morphogenesis/genetics , Organogenesis/genetics , Thrombospondins/genetics , CRISPR-Cas Systems , Case-Control Studies , Comparative Genomic Hybridization , Craniofacial Abnormalities/embryology , DNA Copy Number Variations , Gene Deletion , Haploinsufficiency , Humans , Quantitative Trait Loci
14.
Biol Res Nurs ; 19(3): 339-349, 2017 05.
Article in English | MEDLINE | ID: mdl-28413930

ABSTRACT

OBJECTIVE: This study used a candidate gene approach to examine genomic variation associated with pain, anxiety, and distress in children undergoing a medical procedure. STUDY DESIGN: Children aged 4-10 years having an IV catheter insertion were recruited from three Midwestern children's hospitals. Self-report measures of pain, anxiety, and distress were obtained as well as an observed measure of distress. Samples were collected from children and biological parents for analysis of genomic variation. Genotyped variants had known or suspected association with phenotypes of interest. Analyses included child-only association and family-based transmission disequilibrium tests. RESULTS: Genotype and phenotype data were available from 828 children and 376 family trios. Children were 50% male, had a mean age of 7.2 years, and were 84% White/non-Hispanic. In family-based analysis, one single-nucleotide polymorphism (SNP; rs1143629, interleukin ( IL1B) 1ß) was associated with observed child distress at Bonferroni-corrected levels of significance ( p = .00013), while two approached significance for association with high state anxiety (rs6330 Nerve Growth Factor, Beta Subunit, [ NGFB]) and high trait anxiety (rs6265 brain-derived neurotrophic factor [ BDNF]). In the child-only analysis, multiple SNPs showed nominal evidence of relationships with phenotypes of interest. rs6265 BDNF and rs2941026 cholecystokinin B receptor had possible relationships with trait anxiety in child-only and family-based analyses. CONCLUSIONS: Exploring genomic variation furthers our understanding of pain, anxiety, and distress and facilitates genomic screening to identify children at high risk of procedural pain, anxiety, and distress. Combined with clinical observations and knowledge, such explorations could help guide tailoring of interventions to limit procedure-related distress and identify genes and pathways of interest for future genotype-phenotype studies.


Subject(s)
Catheterization, Peripheral/adverse effects , Child, Hospitalized/psychology , Genetic Variation , Anxiety/genetics , Brain-Derived Neurotrophic Factor/genetics , Catheterization, Peripheral/psychology , Child , Depression/genetics , Female , Humans , Male , Pain/genetics , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL