Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Gene Expr Patterns ; 50: 119344, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37844855

ABSTRACT

Tissue fusion is a critical process that is repeated in multiple contexts during embryonic development and shares common attributes to processes such as wound healing and metastasis. Ocular coloboma is a developmental eye disorder that presents as a physical gap in the ventral eye, and is a major cause of childhood blindness. Coloboma results from fusion failure between opposing ventral retinal epithelia, but there are major knowledge gaps in our understanding of this process at the molecular and cell behavioural levels. Here we catalogue the expression of cell adhesion proteins: N-cadherin, E-cadherin, R-cadherin, ZO-1, and the EMT transcriptional activator and cadherin regulator SNAI2, in the developing chicken embryonic eye. We find that fusion pioneer cells at the edges of the fusing optic fissure have unique and dynamic expression profiles for N-cad, E-cad and ZO-1, and that these are temporally preceded by expression of SNAI2. This highlights the unique properties of these cells and indicates that regulation of cell adhesion factors may be a critical process in optic fissure closure.


Subject(s)
Coloboma , Retina , Animals , Chick Embryo , Female , Pregnancy , Humans , Cell Adhesion , Retina/metabolism , Coloboma/metabolism , Coloboma/pathology , Transcription Factors/metabolism , Chickens/genetics , Chickens/metabolism , Eye/metabolism
2.
Biomolecules ; 13(2)2023 02 04.
Article in English | MEDLINE | ID: mdl-36830662

ABSTRACT

Ocular coloboma (OC) is a failure of complete optic fissure closure during embryonic development and presents as a tissue defect along the proximal-distal axis of the ventral eye. It is classed as part of the clinical spectrum of structural eye malformations with microphthalmia and anophthalmia, collectively abbreviated to MAC. Despite deliberate attempts to identify causative variants in MAC, many patients remain without a genetic diagnosis. To reveal potential candidate genes, we utilised transcriptomes experimentally generated from embryonic eye tissues derived from humans, mice, zebrafish, and chicken at stages coincident with optic fissure closure. Our in-silico analyses found 10 genes with optic fissure-specific enriched expression: ALDH1A3, BMPR1B, EMX2, EPHB3, NID1, NTN1, PAX2, SMOC1, TENM3, and VAX1. In situ hybridization revealed that all 10 genes were broadly expressed ventrally in the developing eye but that only PAX2 and NTN1 were expressed in cells at the edges of the optic fissure margin. Of these conserved optic fissure genes, EMX2, NID1, and EPHB3 have not previously been associated with human MAC cases. Targeted genetic manipulation in zebrafish embryos using CRISPR/Cas9 caused the developmental MAC phenotype for emx2 and ephb3. We analysed available whole genome sequencing datasets from MAC patients and identified a range of variants with plausible causality. In combination, our data suggest that expression of genes involved in ventral eye development is conserved across a range of vertebrate species and that EMX2, NID1, and EPHB3 are candidate loci that warrant further functional analysis in the context of MAC and should be considered for sequencing in cohorts of patients with structural eye malformations.


Subject(s)
Coloboma , Eye Abnormalities , Neuropeptides , Female , Pregnancy , Humans , Animals , Mice , Coloboma/genetics , Coloboma/metabolism , Eye/metabolism , Zebrafish/genetics , Gene Expression Profiling , Membrane Proteins/metabolism , Nerve Tissue Proteins/genetics , Neuropeptides/metabolism , Homeodomain Proteins/metabolism
3.
PLoS One ; 17(11): e0268149, 2022.
Article in English | MEDLINE | ID: mdl-36413568

ABSTRACT

Classical aniridia is a congenital and progressive panocular disorder almost exclusively caused by heterozygous loss-of-function variants at the PAX6 locus. We report nine individuals from five families with severe aniridia and/or microphthalmia (with no detectable PAX6 mutation) with ultrarare monoallelic missense variants altering the Arg51 codon of MAB21L1. These mutations occurred de novo in 3/5 families, with the remaining families being compatible with autosomal dominant inheritance. Mice engineered to carry the p.Arg51Leu change showed a highly-penetrant optic disc anomaly in heterozygous animals with severe microphthalmia in homozygotes. Substitutions of the same codon (Arg51) in MAB21L2, a close homolog of MAB21L1, cause severe ocular and skeletal malformations in humans and mice. The predicted nucleotidyltransferase function of MAB21L1 could not be demonstrated using purified protein with a variety of nucleotide substrates and oligonucleotide activators. Induced expression of GFP-tagged wildtype and mutant MAB21L1 in human cells caused only modest transcriptional changes. Mass spectrometry of immunoprecipitated protein revealed that both mutant and wildtype MAB21L1 associate with transcription factors that are known regulators of PAX6 (MEIS1, MEIS2 and PBX1) and with poly(A) RNA binding proteins. Arg51 substitutions reduce the association of wild-type MAB21L1 with TBL1XR1, a component of the NCoR complex. We found limited evidence for mutation-specific interactions with MSI2/Musashi-2, an RNA-binding proteins with effects on many different developmental pathways. Given that biallelic loss-of-function variants in MAB21L1 result in a milder eye phenotype we suggest that Arg51-altering monoallelic variants most plausibly perturb eye development via a gain-of-function mechanism.


Subject(s)
Aniridia , Microphthalmos , Humans , Animals , Mice , Microphthalmos/genetics , PAX6 Transcription Factor/genetics , Aniridia/genetics , Mutation, Missense , Heterozygote , Transcription Factors/genetics , Homeodomain Proteins/genetics , RNA-Binding Proteins/genetics , Eye Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics
4.
Genes (Basel) ; 13(10)2022 10 05.
Article in English | MEDLINE | ID: mdl-36292683

ABSTRACT

Anophthalmia (missing eye) describes a failure of early embryonic ocular development. Mutations in a relatively small set of genes account for 75% of bilateral anophthalmia cases, yet 25% of families currently are left without a molecular diagnosis. Here, we report our experimental work that aimed to uncover the developmental and genetic basis of the anophthalmia characterising the X-linked Ie (eye-ear reduction) X-ray-induced allele in mouse that was first identified in 1947. Histological analysis of the embryonic phenotype showed failure of normal eye development after the optic vesicle stage with particularly severe malformation of the ventral retina. Linkage analysis mapped this mutation to a ~6 Mb region on the X chromosome. Short- and long-read whole-genome sequencing (WGS) of affected and unaffected male littermates confirmed the Ie linkage but identified no plausible causative variants or structural rearrangements. These analyses did reduce the critical candidate interval and revealed evidence of multiple variants within the ancestral DNA, although none were found that altered coding sequences or that were unique to Ie. To investigate early embryonic events at a genetic level, we then generated mouse ES cells derived from male Ie embryos and wild type littermates. RNA-seq and accessible chromatin sequencing (ATAC-seq) data generated from cultured optic vesicle organoids did not reveal any large differences in gene expression or accessibility of putative cis-regulatory elements between Ie and wild type. However, an unbiased TF-footprinting analysis of accessible chromatin regions did provide evidence of a genome-wide reduction in binding of transcription factors associated with ventral eye development in Ie, and evidence of an increase in binding of the Zic-family of transcription factors, including Zic3, which is located within the Ie-refined critical interval. We conclude that the refined Ie critical region at chrX: 56,145,000-58,385,000 contains multiple genetic variants that may be linked to altered cis regulation but does not contain a convincing causative mutation. Changes in the binding of key transcription factors to chromatin causing altered gene expression during development, possibly through a subtle mis-regulation of Zic3, presents a plausible cause for the anophthalmia phenotype observed in Ie, but further work is required to determine the precise causative allele and its genetic mechanism.


Subject(s)
Anophthalmos , Mice , Male , Animals , Anophthalmos/genetics , Regulatory Sequences, Nucleic Acid , Transcription Factors/genetics , Transcription Factors/metabolism , Chromatin , DNA , Homeodomain Proteins/genetics
5.
Sci Rep ; 10(1): 10814, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32616903

ABSTRACT

Cilia are complex microtubule-based organelles essential to a range of processes associated with embryogenesis and tissue homeostasis. Mutations in components of these organelles or those involved in their assembly may result in a diverse set of diseases collectively known as ciliopathies. Accordingly, many cilia-associated proteins have been described, while those distinguishing cilia subtypes are poorly defined. Here we set out to define genes associated with motile cilia in humans based on their transcriptional signature. To define the signature, we performed network deconvolution of transcriptomics data derived from tissues possessing motile ciliated cell populations. For each tissue, genes coexpressed with the motile cilia-associated transcriptional factor, FOXJ1, were identified. The consensus across tissues provided a transcriptional signature of 248 genes. To validate these, we examined the literature, databases (CilDB, CentrosomeDB, CiliaCarta and SysCilia), single cell RNA-Seq data, and the localisation of mRNA and proteins in motile ciliated cells. In the case of six poorly characterised signature genes, we performed new localisation experiments on ARMC3, EFCAB6, FAM183A, MYCBPAP, RIBC2 and VWA3A. In summary, we report a set of motile cilia-associated genes that helps shape our understanding of these complex cellular organelles.


Subject(s)
Cilia/genetics , Forkhead Transcription Factors/genetics , Transcription, Genetic/genetics , Armadillo Domain Proteins , Calcium-Binding Proteins , Carrier Proteins , Cilia/physiology , Gene Expression , Humans , Membrane Proteins , Repressor Proteins
6.
Front Cell Dev Biol ; 8: 620774, 2020.
Article in English | MEDLINE | ID: mdl-33505973

ABSTRACT

A key embryonic process that occurs early in ocular development is optic fissure closure (OFC). This fusion process closes the ventral optic fissure and completes the circumferential continuity of the 3-dimensional eye. It is defined by the coming together and fusion of opposing neuroepithelia along the entire proximal-distal axis of the ventral optic cup, involving future neural retina, retinal pigment epithelium (RPE), optic nerve, ciliary body, and iris. Once these have occurred, cells within the fused seam differentiate into components of the functioning visual system. Correct development and progression of OFC, and the continued integrity of the fused margin along this axis, are important for the overall structure of the eye. Failure of OFC results in ocular coloboma-a significant cause of childhood visual impairment that can be associated with several complex ocular phenotypes including microphthalmia and anterior segment dysgenesis. Despite a large number of genes identified, the exact pathways that definitively mediate fusion have not yet been found, reflecting both the biological complexity and genetic heterogeneity of the process. This review will highlight how recent developmental studies have become focused specifically on the epithelial fusion aspects of OFC, applying a range of model organisms (spanning fish, avian, and mammalian species) and utilizing emerging high-resolution live-imaging technologies, transgenic fluorescent models, and unbiased transcriptomic analyses of segmentally-dissected fissure tissue. Key aspects of the fusion process are discussed, including basement membrane dynamics, unique cell behaviors, and the identities and fates of the cells that mediate fusion. These will be set in the context of what is now known, and how these point the way to new avenues of research.

7.
Elife ; 82019 06 04.
Article in English | MEDLINE | ID: mdl-31162046

ABSTRACT

Epithelial fusion underlies many vital organogenic processes during embryogenesis. Disruptions to these cause a significant number of human birth defects, including ocular coloboma. We provide robust spatial-temporal staging and unique anatomical detail of optic fissure closure (OFC) in the embryonic chick, including evidence for roles of apoptosis and epithelial remodelling. We performed complementary transcriptomic profiling and show that Netrin-1 (NTN1) is precisely expressed in the chick fissure margin during fusion but is immediately downregulated after fusion. We further provide a combination of protein localisation and phenotypic evidence in chick, humans, mice and zebrafish that Netrin-1 has an evolutionarily conserved and essential requirement for OFC, and is likely to have an important role in palate fusion. Our data suggest that NTN1 is a strong candidate locus for human coloboma and other multi-system developmental fusion defects, and show that chick OFC is a powerful model for epithelial fusion research.


Subject(s)
Coloboma/genetics , Evolution, Molecular , Eye/growth & development , Netrin-1/genetics , Animals , Apoptosis/genetics , Chick Embryo , Chickens , Coloboma/pathology , Conserved Sequence/genetics , Epithelial Cells/metabolism , Eye/pathology , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , Humans , Mice , Palate/growth & development , Palate/pathology , Zebrafish/genetics , Zebrafish/growth & development
8.
G3 (Bethesda) ; 9(3): 943-954, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30696701

ABSTRACT

Goniodysgenesis is a developmental abnormality of the anterior chamber of the eye. It is generally considered to be congenital in dogs (Canis lupus familiaris), and has been associated with glaucoma and blindness. Goniodysgenesis and early-onset glaucoma initially emerged in Border Collies in Australia in the late 1990s and have subsequently been found in this breed in Europe and the USA. The objective of the present study was to determine the genetic basis of goniodysgenesis in Border Collies. Clinical diagnosis was based on results of examinations by veterinary ophthalmologists of affected and unaffected dogs from eleven different countries. Genotyping using the Illumina high density canine single nucleotide variant genotyping chip was used to identify a candidate genetic region. There was a highly significant peak of association over chromosome 17, with a p-value of 2 × 10-13 Expression profiles and evolutionary conservation of candidate genes were assessed using public databases. Whole genome sequences of three dogs with glaucoma, three severely affected by goniodysgenesis and three unaffected dogs identified a missense variant in the olfactomedin like 3 (OLFML3) gene in all six affected animals. This was homozygous for the risk allele in all nine cases with glaucoma and 12 of 14 other severely affected animals. Of 67 reportedly unaffected animals, only one was homozygous for this variant (offspring of parents both with goniodysgenesis who were also homozygous for the variant). Analysis of pedigree information was consistent with an autosomal recessive mode of inheritance for severe goniodysgenesis (potentially leading to glaucoma) in this breed. The identification of a candidate genetic region and putative causative variant will aid breeders to reduce the frequency of goniodysgenesis and the risk of glaucoma in the Border Collie population.


Subject(s)
Anterior Chamber/abnormalities , Extracellular Matrix Proteins/genetics , Glaucoma/genetics , Mutation, Missense , Amino Acid Sequence , Animals , Anterior Chamber/metabolism , Chick Embryo , Dog Diseases/genetics , Dog Diseases/metabolism , Dogs/abnormalities , Eye Proteins/genetics , Female , Gene Expression Regulation , Genome-Wide Association Study , Glaucoma/metabolism , Glaucoma/veterinary , Glycoproteins/genetics , Humans , Male , Mice , Polymorphism, Single Nucleotide , Sequence Alignment , Sequence Analysis, DNA
10.
Int J Dev Biol ; 62(1-2-3): 257-264, 2018.
Article in English | MEDLINE | ID: mdl-29616734

ABSTRACT

After decades of research investment, techniques for the robust and efficient modification of the chicken genome are now with us. The biology of the chicken has provided many challenges, as have the methods by which transgenes can be readily, stably and functionally integrated into the genome. Now that these obstacles have been surmounted and the chicken has been 'updated' to a cutting-edge modern model organism, a future as a central and versatile model in developmental biology beckons. In this review, we describe recent advances in genetic modification of the chicken and some of the many transgenic models developed for the elucidation of the mechanisms of embryogenesis.


Subject(s)
Chick Embryo , Chickens/physiology , Genetic Engineering , Animals , Animals, Genetically Modified , Embryo Culture Techniques , Embryonic Development , Gene Editing , Genome , Germ Cells/cytology , Green Fluorescent Proteins/metabolism , Lentivirus , Models, Genetic , Transgenes
11.
Mech Dev ; 150: 42-49, 2018 04.
Article in English | MEDLINE | ID: mdl-29526791

ABSTRACT

Precise anterior segment (AS) development in the vertebrate eye is essential for maintaining ocular health throughout life. Disruptions to genetic programs can lead to severe structural AS disorders at birth, while more subtle AS defects may disrupt the drainage of ocular fluids and cause dysregulation of intraocular pressure homeostasis, leading to progressive vision loss. To date, the mouse has served as the major model to study AS development and pathogenesis. Here we present an accurate histological atlas of chick AS formation throughout eye development, with a focus on the formation of drainage structures. We performed expression analyses for a panel of known AS disorder genes, and showed that chick PAX6 was localized to cells of neural retina and surface ectoderm derived structures, displaying remarkable similarity to the mouse. We provide a comparison to mouse and humans for chick AS developmental sequences and structures and confirm that AS development shares common features in all three species, although the main AS structures in the chick are developed prior to hatching. These features enable the unique experimental advantages inherent to chick embryos, and we therefore propose the chick as an appropriate additional model for AS development and disease.


Subject(s)
Chickens/growth & development , Eye/growth & development , Homeodomain Proteins/genetics , PAX6 Transcription Factor/genetics , Animals , Chick Embryo , Chickens/genetics , Ectoderm/anatomy & histology , Ectoderm/growth & development , Ectoderm/metabolism , Eye/anatomy & histology , Eye Abnormalities/genetics , Eye Abnormalities/pathology , Gene Expression Regulation, Developmental , Humans , Mice
12.
Hum Mutat ; 38(8): 942-946, 2017 08.
Article in English | MEDLINE | ID: mdl-28493397

ABSTRACT

Ocular coloboma (OC) is a defect in optic fissure closure and is a common cause of severe congenital visual impairment. Bilateral OC is primarily genetically determined and shows marked locus heterogeneity. Whole-exome sequencing (WES) was used to analyze 12 trios (child affected with OC and both unaffected parents). This identified de novo mutations in 10 different genes in eight probands. Three of these genes encoded proteins associated with actin cytoskeleton dynamics: ACTG1, TWF1, and LCP1. Proband-only WES identified a second unrelated individual with isolated OC carrying the same ACTG1 allele, encoding p.(Pro70Leu). Both individuals have normal neurodevelopment with no extra-ocular signs of Baraitser-Winter syndrome. We found this mutant protein to be incapable of incorporation into F-actin. The LCP1 and TWF1 variants each resulted in only minor disturbance of actin interactions, and no further plausibly causative variants were identified in these genes on resequencing 380 unrelated individuals with OC.


Subject(s)
Actins/genetics , Coloboma/etiology , Coloboma/genetics , Animals , Female , Humans , Male , Mice , Microfilament Proteins/genetics , Mutation/genetics , Protein-Tyrosine Kinases/genetics
14.
Nat Genet ; 49(2): 238-248, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28067909

ABSTRACT

Arhinia, or absence of the nose, is a rare malformation of unknown etiology that is often accompanied by ocular and reproductive defects. Sequencing of 40 people with arhinia revealed that 84% of probands harbor a missense mutation localized to a constrained region of SMCHD1 encompassing the ATPase domain. SMCHD1 mutations cause facioscapulohumeral muscular dystrophy type 2 (FSHD2) via a trans-acting loss-of-function epigenetic mechanism. We discovered shared mutations and comparable DNA hypomethylation patterning between these distinct disorders. CRISPR/Cas9-mediated alteration of smchd1 in zebrafish yielded arhinia-relevant phenotypes. Transcriptome and protein analyses in arhinia probands and controls showed no differences in SMCHD1 mRNA or protein abundance but revealed regulatory changes in genes and pathways associated with craniofacial patterning. Mutations in SMCHD1 thus contribute to distinct phenotypic spectra, from craniofacial malformation and reproductive disorders to muscular dystrophy, which we speculate to be consistent with oligogenic mechanisms resulting in pleiotropic outcomes.


Subject(s)
Choanal Atresia/genetics , Chromosomal Proteins, Non-Histone/genetics , Genetic Predisposition to Disease/genetics , Microphthalmos/genetics , Muscular Dystrophies/genetics , Mutation/genetics , Nose/abnormalities , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Male , Phenotype
15.
Am J Hum Genet ; 98(5): 981-992, 2016 05 05.
Article in English | MEDLINE | ID: mdl-27108798

ABSTRACT

Gillespie syndrome (GS) is characterized by bilateral iris hypoplasia, congenital hypotonia, non-progressive ataxia, and progressive cerebellar atrophy. Trio-based exome sequencing identified de novo mutations in ITPR1 in three unrelated individuals with GS recruited to the Deciphering Developmental Disorders study. Whole-exome or targeted sequence analysis identified plausible disease-causing ITPR1 mutations in 10/10 additional GS-affected individuals. These ultra-rare protein-altering variants affected only three residues in ITPR1: Glu2094 missense (one de novo, one co-segregating), Gly2539 missense (five de novo, one inheritance uncertain), and Lys2596 in-frame deletion (four de novo). No clinical or radiological differences were evident between individuals with different mutations. ITPR1 encodes an inositol 1,4,5-triphosphate-responsive calcium channel. The homo-tetrameric structure has been solved by cryoelectron microscopy. Using estimations of the degree of structural change induced by known recessive- and dominant-negative mutations in other disease-associated multimeric channels, we developed a generalizable computational approach to indicate the likely mutational mechanism. This analysis supports a dominant-negative mechanism for GS variants in ITPR1. In GS-derived lymphoblastoid cell lines (LCLs), the proportion of ITPR1-positive cells using immunofluorescence was significantly higher in mutant than control LCLs, consistent with an abnormality of nuclear calcium signaling feedback control. Super-resolution imaging supports the existence of an ITPR1-lined nucleoplasmic reticulum. Mice with Itpr1 heterozygous null mutations showed no major iris defects. Purkinje cells of the cerebellum appear to be the most sensitive to impaired ITPR1 function in humans. Iris hypoplasia is likely to result from either complete loss of ITPR1 activity or structure-specific disruption of multimeric interactions.


Subject(s)
Aniridia/etiology , Aniridia/pathology , Cerebellar Ataxia/etiology , Cerebellar Ataxia/pathology , Genes, Dominant/genetics , Inositol 1,4,5-Trisphosphate Receptors/genetics , Intellectual Disability/etiology , Intellectual Disability/pathology , Mutation/genetics , Adolescent , Adult , Animals , Cells, Cultured , Child , Female , Humans , Inositol 1,4,5-Trisphosphate Receptors/chemistry , Lymphocytes/metabolism , Lymphocytes/pathology , Male , Mice , Microscopy, Confocal , Middle Aged , Pedigree , Protein Conformation
16.
Hum Mol Genet ; 25(7): 1382-91, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26908622

ABSTRACT

Ocular coloboma is a common eye malformation resulting from incomplete fusion of the optic fissure during development. Coloboma is often associated with microphthalmia and/or contralateral anophthalmia. Coloboma shows extensive locus heterogeneity associated with causative mutations identified in genes encoding developmental transcription factors or components of signaling pathways. We report an ultra-rare, heterozygous frameshift mutation in FZD5 (p.Ala219Glufs*49) that was identified independently in two branches of a large family with autosomal dominant non-syndromic coloboma. FZD5 has a single-coding exon and consequently a transcript with this frameshift variant is not a canonical substrate for nonsense-mediated decay. FZD5 encodes a transmembrane receptor with a conserved extracellular cysteine rich domain for ligand binding. The frameshift mutation results in the production of a truncated protein, which retains the Wingless-type MMTV integration site family member-ligand-binding domain, but lacks the transmembrane domain. The truncated protein was secreted from cells, and behaved as a dominant-negative FZD5 receptor, antagonizing both canonical and non-canonical WNT signaling. Expression of the resultant mutant protein caused coloboma and microphthalmia in zebrafish, and disruption of the apical junction of the retinal neural epithelium in mouse, mimicking the phenotype of Fz5/Fz8 compound conditional knockout mutants. Our studies have revealed a conserved role of Wnt-Frizzled (FZD) signaling in ocular development and directly implicate WNT-FZD signaling both in normal closure of the human optic fissure and pathogenesis of coloboma.


Subject(s)
Frameshift Mutation , Frizzled Receptors/genetics , Wnt Signaling Pathway , Animals , DNA Mutational Analysis , Female , Humans , Male , Mice , Microphthalmos/genetics , Microphthalmos/metabolism , Pedigree , Zebrafish/genetics , Zebrafish/metabolism
17.
Am J Hum Genet ; 94(6): 915-23, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24906020

ABSTRACT

We identified four different missense mutations in the single-exon gene MAB21L2 in eight individuals with bilateral eye malformations from five unrelated families via three independent exome sequencing projects. Three mutational events altered the same amino acid (Arg51), and two were identical de novo mutations (c.151C>T [p.Arg51Cys]) in unrelated children with bilateral anophthalmia, intellectual disability, and rhizomelic skeletal dysplasia. c.152G>A (p.Arg51His) segregated with autosomal-dominant bilateral colobomatous microphthalmia in a large multiplex family. The fourth heterozygous mutation (c.145G>A [p.Glu49Lys]) affected an amino acid within two residues of Arg51 in an adult male with bilateral colobomata. In a fifth family, a homozygous mutation (c.740G>A [p.Arg247Gln]) altering a different region of the protein was identified in two male siblings with bilateral retinal colobomata. In mouse embryos, Mab21l2 showed strong expression in the developing eye, pharyngeal arches, and limb bud. As predicted by structural homology, wild-type MAB21L2 bound single-stranded RNA, whereas this activity was lost in all altered forms of the protein. MAB21L2 had no detectable nucleotidyltransferase activity in vitro, and its function remains unknown. Induced expression of wild-type MAB21L2 in human embryonic kidney 293 cells increased phospho-ERK (pERK1/2) signaling. Compared to the wild-type and p.Arg247Gln proteins, the proteins with the Glu49 and Arg51 variants had increased stability. Abnormal persistence of pERK1/2 signaling in MAB21L2-expressing cells during development is a plausible pathogenic mechanism for the heterozygous mutations. The phenotype associated with the homozygous mutation might be a consequence of complete loss of MAB21L2 RNA binding, although the cellular function of this interaction remains unknown.


Subject(s)
Anophthalmos/genetics , Eye Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mutation, Missense , Adult , Alleles , Animals , Brain Diseases, Metabolic, Inborn/genetics , Coloboma/genetics , Corneal Opacity/genetics , Exome , Eye Proteins/metabolism , Female , Gene Expression , HEK293 Cells , Heterozygote , Homozygote , Humans , Intellectual Disability/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Microcephaly/genetics , Microphthalmos/genetics , Pedigree , Phenotype , Protein Conformation , Signal Transduction
18.
PLoS Genet ; 10(5): e1004359, 2014 May.
Article in English | MEDLINE | ID: mdl-24809698

ABSTRACT

Mutations in the LIM-homeodomain transcription factor LMX1B cause nail-patella syndrome, an autosomal dominant pleiotrophic human disorder in which nail, patella and elbow dysplasia is associated with other skeletal abnormalities and variably nephropathy and glaucoma. It is thought to be a haploinsufficient disorder. Studies in the mouse have shown that during development Lmx1b controls limb dorsal-ventral patterning and is also required for kidney and eye development, midbrain-hindbrain boundary establishment and the specification of specific neuronal subtypes. Mice completely deficient for Lmx1b die at birth. In contrast to the situation in humans, heterozygous null mice do not have a mutant phenotype. Here we report a novel mouse mutant Icst, an N-ethyl-N-nitrosourea-induced missense substitution, V265D, in the homeodomain of LMX1B that abolishes DNA binding and thereby the ability to transactivate other genes. Although the homozygous phenotypic consequences of Icst and the null allele of Lmx1b are the same, heterozygous Icst elicits a phenotype whilst the null allele does not. Heterozygous Icst causes glaucomatous eye defects and is semi-lethal, probably due to kidney failure. We show that the null phenotype is rescued more effectively by an Lmx1b transgene than is Icst. Co-immunoprecipitation experiments show that both wild-type and Icst LMX1B are found in complexes with LIM domain binding protein 1 (LDB1), resulting in lower levels of functional LMX1B in Icst heterozygotes than null heterozygotes. We conclude that Icst is a dominant-negative allele of Lmx1b. These findings indicate a reassessment of whether nail-patella syndrome is always haploinsufficient. Furthermore, Icst is a rare example of a model of human glaucoma caused by mutation of the same gene in humans and mice.


Subject(s)
Genes, Dominant , Genes, Lethal , Glaucoma/genetics , LIM-Homeodomain Proteins/genetics , Transcription Factors/genetics , Alleles , Animals , Body Patterning , Dimerization , Heterozygote , Mice , Mice, Transgenic , Mutation, Missense
19.
Am J Hum Genet ; 94(2): 295-302, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24462371

ABSTRACT

Exome sequence analysis of affected individuals from two families with autosomal-dominant inheritance of coloboma identified two different cosegregating heterozygous nonsense mutations (c.370C>T [p.Arg124*] and c. 1066G>T [p.Glu356*]) in YAP1. The phenotypes of the affected families differed in that one included no extraocular features and the other manifested with highly variable multisystem involvement, including hearing loss, intellectual disability, hematuria, and orofacial clefting. A combined LOD score of 4.2 was obtained for the association between YAP1 loss-of-function mutations and the phenotype in these families. YAP1 encodes an effector of the HIPPO-pathway-induced growth response, and whole-mount in situ hybridization in mouse embryos has shown that Yap1 is strongly expressed in the eye, brain, and fusing facial processes. RT-PCR showed that an alternative transcription start site (TSS) in intron 1 of YAP1 and Yap1 is widely used in human and mouse development, respectively. Transcripts from the alternative TSS are predicted to initiate at codon Met179 relative to the canonical transcript (RefSeq NM_001130145). In these alternative transcripts, the c.370C>T mutation in family 1305 is within the 5' UTR and cannot result in nonsense-mediated decay (NMD). The c. 1066G>T mutation in family 132 should result in NMD in transcripts from either TSS. Amelioration of the phenotype by the alternative transcripts provides a plausible explanation for the phenotypic differences between the families.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Codon, Nonsense , Eye Abnormalities/genetics , Heterozygote , Phosphoproteins/genetics , Adolescent , Adult , Aged , Alleles , Animals , Cell Cycle Proteins , Child , Child, Preschool , Exome , Eye Abnormalities/pathology , Female , Humans , Introns , Male , Mice , Middle Aged , Nonsense Mediated mRNA Decay/genetics , Pedigree , Phenotype , Transcription Factors , Transcription Initiation Site , YAP-Signaling Proteins , Young Adult
20.
Hum Mol Genet ; 23(10): 2569-79, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24363063

ABSTRACT

Heterozygous loss-of-function (LOF) mutations in the gene encoding the DNA-binding protein, SATB2, result in micrognathia and cleft palate in both humans and mice. In three unrelated individuals, we show that translocation breakpoints (BPs) up to 896 kb 3' of SATB2 polyadenylation site cause a phenotype which is indistinguishable from that caused by SATB2 LOF mutations. This syndrome comprises long nose, small mouth, micrognathia, cleft palate, arachnodactyly and intellectual disability. These BPs map to a gene desert between PLCL1 and SATB2. We identified three putative cis-regulatory elements (CRE1-3) using a comparative genomic approach each of which would be placed in trans relative to SATB2 by all three BPs. CRE1-3 each bind p300 and mono-methylated H3K4 consistent with enhancer function. In silico analysis suggested that CRE1-3 contain one or more conserved SOX9-binding sites, and this binding was confirmed using chromatin immunoprecipitation on cells derived from mouse embryonic pharyngeal arch. Interphase bacterial artificial chromosome fluorescence in situ hybridization measurements in embryonic craniofacial tissues showed that the orthologous region in mice exhibits Satb2 expression-dependent chromatin decondensation consistent with Satb2 being a target gene of CRE1-3. To assess their in vivo function, we made multiple stable reporter transgenic lines for each enhancer in zebrafish. CRE2 was shown to drive SATB2-like expression in the embryonic craniofacial region. This expression could be eliminated by mutating the SOX9-binding site of CRE2. These observations suggest that SATB2 and SOX9 may be acting together via complex cis-regulation to coordinate the growth of the developing jaw.


Subject(s)
Matrix Attachment Region Binding Proteins/genetics , Pierre Robin Syndrome/diagnosis , SOX9 Transcription Factor/genetics , Transcription Factors/genetics , Adult , Animals , Binding Sites , Cells, Cultured , Child , Child, Preschool , Epistasis, Genetic , Female , Humans , Infant , Male , Mice , Mutation , Pierre Robin Syndrome/genetics , Regulatory Elements, Transcriptional , Young Adult , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...