Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Dev Dyn ; 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38958410

ABSTRACT

Primary cilia are antenna-like sensory organelles that are evolutionarily conserved in nearly all modern eukaryotes, from the single-celled green alga, Chlamydomonas reinhardtii, to vertebrates and mammals. Cilia are microtubule-based cellular projections that have adapted to perform a broad range of species-specific functions, from cell motility to detection of light and the transduction of extracellular mechanical and chemical signals. These functions render cilia essential for organismal development and survival. The high conservation of cilia has allowed for discoveries in C. reinhardtii to inform our understanding of the basic biology of mammalian primary cilia, and to provide insight into the genetic etiology of ciliopathies. Over the last two decades, a growing number of studies has revealed that multiple aspects of ciliary homeostasis are regulated by the actin cytoskeleton, including centrosome migration and positioning, vesicle transport to the basal body, ectocytosis, and ciliary-mediated signaling. Here, we review actin regulation of ciliary homeostasis, and highlight conserved and divergent mechanisms in C. reinhardtii and mammalian cells. Further, we compare the disease manifestations of patients with ciliopathies to those with mutations in actin and actin-associated genes, and propose that primary cilia defects caused by genetic alteration of the actin cytoskeleton may underlie certain birth defects.

2.
Biochem Soc Trans ; 51(3): 949-958, 2023 06 28.
Article in English | MEDLINE | ID: mdl-37345651

ABSTRACT

Many structural birth defects occur due to failure of tissue movement and fusion events during embryogenesis. Examples of such birth defects include failure of closure of the neural tube, palate, and ventral body wall. Actomyosin forces play a pivotal role in these closure processes, making proteins that regulate actomyosin dynamics a priority when studying the etiology of structural birth defects. SPECC1L (sperm antigen with calponin homology and coiled-coil domains 1 like) cytoskeletal protein associates with microtubules, filamentous actin, non-muscle myosin II (NMII), as well as membrane-associated components of adherens junctions. Patients with SPECC1L mutations show a range of structural birth defects affecting craniofacial development (hypertelorism, cleft palate), ventral body wall (omphalocele), and internal organs (diaphragmatic hernia, bicornuate uterus). Characterization of mouse models indicates that these syndromic mutations utilize a gain-of-function mechanism to affect intra- and supra-cellular actin organization. Interestingly, SPECC1L deficiency appears to affect the efficiency of tissue dynamics, making it an important cytoskeletal regulator to study tissue movement and fusion events during embryonic development. Here we summarize the SPECC1L-related syndrome mutations, phenotypes of Specc1l mouse models, and cellular functions of SPECC1L that highlight how it may regulate embryonic tissue dynamics.


Subject(s)
Actins , Actomyosin , Animals , Female , Mice , Male , Actins/metabolism , Actomyosin/metabolism , Semen , Cytoskeletal Proteins/metabolism , Cytoskeleton/metabolism
3.
Development ; 149(15)2022 08 01.
Article in English | MEDLINE | ID: mdl-35781329

ABSTRACT

Cell fate determination is a necessary and tightly regulated process for producing different cell types and structures during development. Cranial neural crest cells (CNCCs) are unique to vertebrate embryos and emerge from the neural plate borders into multiple cell lineages that differentiate into bone, cartilage, neurons and glial cells. We have previously reported that Irf6 genetically interacts with Twist1 during CNCC-derived tissue formation. Here, we have investigated the mechanistic role of Twist1 and Irf6 at early stages of craniofacial development. Our data indicate that TWIST1 is expressed in endocytic vesicles at the apical surface and interacts with ß/δ-catenins during neural tube closure, and Irf6 is involved in defining neural fold borders by restricting AP2α expression. Twist1 suppresses Irf6 and other epithelial genes in CNCCs during the epithelial-to-mesenchymal transition (EMT) process and cell migration. Conversely, a loss of Twist1 leads to a sustained expression of epithelial and cell adhesion markers in migratory CNCCs. Disruption of TWIST1 phosphorylation in vivo leads to epidermal blebbing, edema, neural tube defects and CNCC-derived structural abnormalities. Altogether, this study describes a previously uncharacterized function of mammalian Twist1 and Irf6 in the neural tube and CNCCs, and provides new target genes for Twist1 that are involved in cytoskeletal remodeling.


Subject(s)
Neural Crest , Neural Tube , Animals , Catenins , Gene Expression Regulation, Developmental , Mammals/genetics , Skull/metabolism , Delta Catenin
4.
NPJ Precis Oncol ; 6(1): 3, 2022 Jan 18.
Article in English | MEDLINE | ID: mdl-35042970

ABSTRACT

Gene fusions are known to drive many human cancers. Therefore, the functional characterization of newly discovered fusions is critical to understanding the oncobiology of these tumors and to enable therapeutic development. NPM1-TYK2 is a novel fusion identified in CD30 + lymphoproliferative disorders, and here we present the functional evaluation of this fusion gene as an oncogene. The chimeric protein consists of the amino-terminus of nucleophosmin 1 (NPM1) and the carboxyl-terminus of tyrosine kinase 2 (TYK2), including the kinase domain. Using in vitro lymphoid cell transformation assays and in vivo tumorigenic xenograft models we present direct evidence that the fusion gene is an oncogene. NPM1 fusion partner provides the critical homodimerization needed for the fusion kinase constitutive activation and downstream signaling that are responsible for cell transformation. As a result, our studies identify NPM1-TYK2 as a novel fusion oncogene and suggest that inhibition of fusion homodimerization could be a precision therapeutic approach in cutaneous T-cell lymphoma patients expressing this chimera.

5.
Hum Mol Genet ; 31(1): 18-31, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34302166

ABSTRACT

Patients with autosomal dominant SPECC1L variants show syndromic malformations, including hypertelorism, cleft palate and omphalocele. These SPECC1L variants largely cluster in the second coiled-coil domain (CCD2), which facilitates association with microtubules. To study SPECC1L function in mice, we first generated a null allele (Specc1lΔEx4) lacking the entire SPECC1L protein. Homozygous mutants for these truncations died perinatally without cleft palate or omphalocele. Given the clustering of human variants in CCD2, we hypothesized that targeted perturbation of CCD2 may be required. Indeed, homozygotes for in-frame deletions involving CCD2 (Specc1lΔCCD2) resulted in exencephaly, cleft palate and ventral body wall closure defects (omphalocele). Interestingly, exencephaly and cleft palate were never observed in the same embryo. Further examination revealed a narrower oral cavity in exencephalic embryos, which allowed palatal shelves to elevate and fuse despite their defect. In the cell, wild-type SPECC1L was evenly distributed throughout the cytoplasm and colocalized with both microtubules and filamentous actin. In contrast, mutant SPECC1L-ΔCCD2 protein showed abnormal perinuclear accumulation with diminished overlap with microtubules, indicating that SPECC1L used microtubule association for trafficking in the cell. The perinuclear accumulation in the mutant also resulted in abnormally increased actin and non-muscle myosin II bundles dislocated to the cell periphery. Disrupted actomyosin cytoskeletal organization in SPECC1L CCD2 mutants would affect cell alignment and coordinated movement during neural tube, palate and ventral body wall closure. Thus, we show that perturbation of CCD2 in the context of full SPECC1L protein affects tissue fusion dynamics, indicating that human SPECC1L CCD2 variants are gain-of-function.


Subject(s)
Cleft Palate , Gain of Function Mutation , Animals , Cleft Palate/genetics , Cleft Palate/metabolism , Mice , Microtubules/genetics , Microtubules/metabolism , Palate , Phenotype , Phosphoproteins/genetics
6.
J Vis Exp ; (168)2021 02 13.
Article in English | MEDLINE | ID: mdl-33645552

ABSTRACT

Development of the palate is a dynamic process, which involves vertical growth of bilateral palatal shelves next to the tongue followed by elevation and fusion above the tongue. Defects in this process lead to cleft palate, a common birth defect. Recent studies have shown that palatal shelf elevation involves a remodeling process that transforms the orientation of the shelf from a vertical to a horizontal one. The role of the palatal shelf mesenchymal cells in this dynamic remodeling has been difficult to study. Time-lapse-imaging-based quantitative analysis has been recently used to show that primary mouse embryonic palatal mesenchymal (MEPM) cells can self-organize into a collective movement. Quantitative analyses could identify differences in mutant MEPM cells from a mouse model with palate elevation defects. This paper describes methods to isolate and culture MEPM cells from E13.5 embryos-specifically for time-lapse imaging-and to determine various cellular attributes of collective movement, including measures for stream formation, shape alignment, and persistence of direction. It posits that MEPM cells can serve as a proxy model for studying the role of palatal shelf mesenchyme during the dynamic process of elevation. These quantitative methods will allow investigators in the craniofacial field to assess and compare collective movement attributes in control and mutant cells, which will augment the understanding of mesenchymal remodeling during palatal shelf elevation. Furthermore, MEPM cells provide a rare mesenchymal cell model for investigation of collective cell movement in general.


Subject(s)
Cell Movement , Cell Separation/methods , Embryo, Mammalian/cytology , Mesoderm/cytology , Palate/cytology , Time-Lapse Imaging , Animals , Cell Tracking , Cells, Cultured , Cryopreservation , Disease Models, Animal , Dissection , Female , Mice , Wound Healing
7.
PLoS One ; 16(2): e0246989, 2021.
Article in English | MEDLINE | ID: mdl-33577554

ABSTRACT

Topiramate is an anti-epileptic drug that is commonly prescribed not just to prevent seizures but also migraine headaches, with over 8 million prescriptions dispensed annually. Topiramate use during pregnancy has been linked to significantly increased risk of babies born with orofacial clefts (OFCs). However, the exact molecular mechanism of topiramate teratogenicity is unknown. In this study, we first used an unbiased antibody array analysis to test the effect of topiramate on human embryonic palatal mesenchyme (HEPM) cells. This analysis identified 40 differentially expressed proteins, showing strong connectivity to known genes associated with orofacial clefts. However, among known OFC genes, only TGFß1 was significantly upregulated in the antibody array analysis. Next, we validated that topiramate could increase expression of TGFß1 and of downstream target phospho-SMAD2 in primary mouse embryonic palatal mesenchyme (MEPM) cells. Furthermore, we showed that topiramate treatment of primary MEPM cells increased expression of SOX9. SOX9 overexpression in chondrocytes is known to cause cleft palate in mouse. We propose that topiramate mediates upregulation of TGFß1 signaling through activation of γ-aminobutyric acid (GABA) receptors in the palate. TGFß1 and SOX9 play critical roles in orofacial morphogenesis, and their abnormal overexpression provides a plausible etiologic molecular mechanism for the teratogenic effects of topiramate.


Subject(s)
Anticonvulsants/pharmacology , Palate/embryology , SOX9 Transcription Factor/genetics , Teratogens/pharmacology , Topiramate/pharmacology , Transforming Growth Factor beta1/genetics , Animals , Cell Line , Cells, Cultured , Cleft Lip/chemically induced , Cleft Lip/genetics , Cleft Palate/chemically induced , Cleft Palate/genetics , Gene Expression Regulation, Developmental/drug effects , Humans , Mice , Palate/cytology , Palate/drug effects , Palate/metabolism , Up-Regulation/drug effects
8.
Sci Rep ; 11(1): 1452, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33446878

ABSTRACT

Cleft lip and/or palate (CL/P) are common anomalies occurring in 1/800 live-births. Pathogenic SPECC1L variants have been identified in patients with CL/P, which signifies a primary role for SPECC1L in craniofacial development. Specc1l mutant mouse embryos exhibit delayed palatal shelf elevation accompanied by epithelial defects. We now posit that the process of palate elevation is itself abnormal in Specc1l mutants, due to defective remodeling of palatal mesenchyme. To characterize the underlying cellular defect, we studied the movement of primary mouse embryonic palatal mesenchyme (MEPM) cells using live-imaging of wound-repair assays. SPECC1L-deficient MEPM cells exhibited delayed wound-repair, however, reduced cell speed only partially accounted for this delay. Interestingly, mutant MEPM cells were also defective in coordinated cell movement. Therefore, we used open-field 2D cultures of wildtype MEPM cells to show that they indeed formed cell streams at high density, which is an important attribute of collective movement. Furthermore, activation of the PI3K-AKT pathway rescued both cell speed and guidance defects in Specc1l mutant MEPM cells. Thus, we show that live-imaging of primary MEPM cells can be used to assess mesenchymal remodeling defects during palatal shelf elevation, and identify a novel role for SPECC1L in collective movement through modulation of PI3K-AKT signaling.


Subject(s)
Cleft Lip/embryology , Cleft Palate/embryology , Embryo, Mammalian/embryology , Gene Expression Regulation, Developmental , Palate/embryology , Phosphoproteins/deficiency , Animals , Cleft Lip/genetics , Cleft Palate/genetics , Mice , Mice, Knockout , Phosphoproteins/metabolism
9.
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
10.
Dev Biol ; 458(2): 246-256, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31765609

ABSTRACT

In this study, we investigated the role of the transcription factor Six2 in palate development. Six2 was selected using the SysFACE tool to predict genes from the 2p21 locus, a region associated with clefting in humans by GWAS, that are likely to be involved in palatogenesis. We functionally validated the predicted role of Six2 in palatogenesis by showing that 22% of Six2 null embryos develop cleft palate. Six2 contributes to palatogenesis by promoting mesenchymal cell proliferation and regulating bone formation. The clefting phenotype in Six2-/- embryos is similar to Pax9 null embryos, so we examined the functional relationship of these two genes. Mechanistically, SIX2 binds to a PAX9 5' upstream regulatory element and activates PAX9 expression. In addition, we identified a human SIX2 coding variant (p.Gly264Glu) in a proband with cleft palate. We show this missense mutation affects the stability of the SIX2 protein and leads to decreased PAX9 expression. The low penetrance of clefting in the Six2 null mouse combined with the mutation in one patient with cleft palate underscores the potential combinatorial interactions of other genes in clefting. Our study demonstrates that Six2 interacts with the developmental gene regulatory network in the developing palate.


Subject(s)
Homeodomain Proteins/metabolism , PAX9 Transcription Factor/genetics , Transcription Factors/metabolism , Animals , Cleft Palate/embryology , Cleft Palate/genetics , Craniofacial Abnormalities/embryology , Female , Gene Expression Regulation, Developmental/genetics , Genes, Homeobox , Homeodomain Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Morphogenesis , Nerve Tissue Proteins/metabolism , Osteogenesis , PAX9 Transcription Factor/metabolism , Paired Box Transcription Factors , Palate/metabolism , Signal Transduction/genetics , Transcription Factors/genetics
11.
Genet Epidemiol ; 43(6): 704-716, 2019 09.
Article in English | MEDLINE | ID: mdl-31172578

ABSTRACT

Phenotypic heterogeneity is a hallmark of complex traits, and genetic studies of such traits may focus on them as a single diagnostic entity or by analyzing specific components. For example, in orofacial clefting (OFC), three subtypes-cleft lip (CL), cleft lip and palate (CLP), and cleft palate (CP) have been studied separately and in combination. To further dissect the genetic architecture of OFCs and how a given associated locus may be contributing to distinct subtypes of a trait we developed a framework for quantifying and interpreting evidence of subtype-specific or shared genetic effects in complex traits. We applied this technique to create a "cleft map" of the association of 30 genetic loci with three OFC subtypes. In addition to new associations, we found loci with subtype-specific effects (e.g., GRHL3 [CP], WNT5A [CLP]), as well as loci associated with two or all three subtypes. We cross-referenced these results with mouse craniofacial gene expression datasets, which identified additional promising candidate genes. However, we found no strong correlation between OFC subtypes and expression patterns. In aggregate, the cleft map revealed that neither subtype-specific nor shared genetic effects operate in isolation in OFC architecture. Our approach can be easily applied to any complex trait with distinct phenotypic subgroups.


Subject(s)
Brain/abnormalities , Cleft Lip/classification , Cleft Lip/genetics , Cleft Palate/classification , Cleft Palate/genetics , Genetic Loci , Genetic Markers , Genetic Testing/methods , Genome-Wide Association Study/methods , Phenotype , Brain/pathology , Cleft Lip/pathology , Cleft Palate/pathology , Humans , Transcriptome
12.
Eur J Med Genet ; 62(12): 103588, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30472488

ABSTRACT

The SPECC1L protein plays a role in adherens junctions involved in cell adhesion, actin cytoskeleton organization, microtubule stabilization, spindle organization and cytokinesis. It modulates PI3K-AKT signaling and controls cranial neural crest cell delamination during facial morphogenesis. SPECC1L causative variants were first identified in individuals with oblique facial clefts. Recently, causative variants in SPECC1L were reported in a pedigree reported in 1988 as atypical Opitz GBBB syndrome. Six families with SPECC1L variants have been reported thus far. We report here eight further pedigrees with SPECC1L variants, including a three-generation family, and a further individual of a previously published family. We discuss the nosology of Teebi and GBBB, and the syndromes related to SPECC1L variants. Although the phenotype of individuals with SPECC1L mutations shows overlap with Opitz syndrome in its craniofacial anomalies, the canonical laryngeal malformations and male genital anomalies are not observed. Instead, individuals with SPECCL1 variants have branchial fistulae, omphalocele, diaphragmatic hernias, and uterus didelphis. We also point to the clinical overlap of SPECC1L syndrome with mild Baraitser-Winter craniofrontofacial syndrome: they share similar dysmorphic features (wide, short nose with a large tip, cleft lip and palate, blepharoptosis, retrognathia, and craniosynostosis), although intellectual disability, neuronal migration defect, and muscular problems remain largely specific to Baraitser-Winter syndrome. In conclusion, we suggest that patients with pathogenic variants in SPECC1L should not be described as "dominant (or type 2) Opitz GBBB syndrome", and instead should be referred to as "SPECC1L syndrome" as both disorders show distinctive, non overlapping developmental anomalies beyond facial communalities.


Subject(s)
Abnormalities, Multiple/genetics , Craniofacial Abnormalities/genetics , Esophagus/abnormalities , Foot Deformities, Congenital/genetics , Growth Disorders/genetics , Hand Deformities, Congenital/genetics , Hydrocephalus/genetics , Hypertelorism/genetics , Hypospadias/genetics , Mental Retardation, X-Linked/genetics , Obesity/genetics , Phenotype , Phosphoproteins/genetics , Abnormalities, Multiple/pathology , Adolescent , Adult , Child , Child, Preschool , Craniofacial Abnormalities/pathology , Esophagus/pathology , Facies , Female , Foot Deformities, Congenital/pathology , Growth Disorders/pathology , Hand Deformities, Congenital/pathology , Humans , Hydrocephalus/pathology , Hypertelorism/pathology , Hypospadias/pathology , Male , Mental Retardation, X-Linked/pathology , Mutation , Obesity/pathology , Pedigree
13.
Hum Genet ; 137(11-12): 941-954, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30417254

ABSTRACT

Isolated or syndromic congenital cataracts are heterogeneous developmental defects, making the identification of the associated genes challenging. In the past, mouse lens expression microarrays have been successfully applied in bioinformatics tools (e.g., iSyTE) to facilitate human cataract-associated gene discovery. To develop a new resource for geneticists, we report high-throughput RNA sequencing (RNA-seq) profiles of mouse lens at key embryonic stages (E)10.5 (lens pit), E12.5 (primary fiber cell differentiation), E14.5 and E16.5 (secondary fiber cell differentiation). These stages capture important events as the lens develops from an invaginating placode into a transparent tissue. Previously, in silico whole-embryo body (WB)-subtraction-based "lens-enriched" expression has been effective in prioritizing cataract-linked genes. To apply an analogous approach, we generated new mouse WB RNA-seq datasets and show that in silico WB subtraction of lens RNA-seq datasets successfully identifies key genes based on lens-enriched expression. At ≥2 counts-per-million expression, ≥1.5 log2 fold-enrichment (p < 0.05) cutoff, E10.5 lens exhibits 1401 enriched genes (17% lens-expressed genes), E12.5 lens exhibits 1937 enriched genes (22% lens-expressed genes), E14.5 lens exhibits 2514 enriched genes (31% lens-expressed genes), and E16.5 lens exhibits 2745 enriched genes (34% lens-expressed genes). Biological pathway analysis identified genes associated with lens development, transcription regulation and signaling pathways, among other functional groups. Furthermore, these new RNA-seq data confirmed high expression of established cataract-linked genes and identified new potential regulators in the lens. Finally, we developed new lens stage-specific UCSC Genome Brower annotation tracks and made these publicly accessible through iSyTE ( https://research.bioinformatics.udel.edu/iSyTE/ ) for user-friendly visualization of lens gene expression/enrichment to prioritize genes from high-throughput data from cataract cases.


Subject(s)
Cataract/genetics , Cell Differentiation/genetics , Embryonic Development/genetics , Gene Expression Regulation/genetics , Animals , Cataract/pathology , Computational Biology , Genetic Association Studies , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Lens, Crystalline/pathology , Mice , Sequence Analysis, RNA
14.
Birth Defects Res ; 109(1): 27-37, 2017 01 20.
Article in English | MEDLINE | ID: mdl-28029220

ABSTRACT

BACKGROUND: Recent advances in genomics methodologies, in particular the availability of next-generation sequencing approaches have made it possible to identify risk loci throughout the genome, in particular the exome. In the current study, we present findings from an exome study conducted in five affected individuals of a multiplex family with cleft palate only. METHODS: The GEnome MINIng (GEMINI) pipeline was used to functionally annotate the single nucleotide polymorphisms, insertions and deletions. Filtering methods were applied to identify variants that are clinically relevant and present in affected individuals at minor allele frequencies (≤1%) in the 1000 Genomes Project single nucleotide polymorphism database, Exome Aggregation Consortium, and Exome Variant Server databases. The bioinformatics tool Systems Tool for Craniofacial Expression-Based Gene Discovery was used to prioritize cleft candidates in our list of variants, and Sanger sequencing was used to validate the presence of identified variants in affected and unaffected relatives. RESULTS: Our analyses approach narrowed the candidates down to the novel missense variant in ARHGAP29 (GenBank: NM_004815.3, NP_004806.3;c.1654T>C [p.Ser552Pro]. A functional assay in zebrafish embryos showed that the encoded protein lacks the activity possessed by its wild-type counterpart, and migration assays revealed that keratinocytes transfected with wild-type ARHGAP29 migrated faster than counterparts transfected with the p.Ser552Pro ARHGAP29 variant or empty vector (control). CONCLUSION: These findings reveal ARHGAP29 to be a regulatory protein essential for proper development of the face, identifies an amino acid that is key for this, and provides a potential new diagnostic tool.Birth Defects Research 109:27-37, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cleft Palate/genetics , GTPase-Activating Proteins/genetics , Alleles , Animals , Cleft Lip/genetics , Computational Biology , Disease Models, Animal , Exome , Female , GTPase-Activating Proteins/metabolism , Gene Frequency/genetics , Genome-Wide Association Study , High-Throughput Nucleotide Sequencing , Humans , Male , Pedigree , Polymorphism, Single Nucleotide/genetics , Risk Factors , Sequence Analysis, DNA/methods , Exome Sequencing , Zebrafish/embryology , Zebrafish/genetics
15.
Sci Rep ; 6: 17735, 2016 Jan 20.
Article in English | MEDLINE | ID: mdl-26787558

ABSTRACT

Cranial neural crest cells (CNCCs) delaminate from embryonic neural folds and migrate to pharyngeal arches, which give rise to most mid-facial structures. CNCC dysfunction plays a prominent role in the etiology of orofacial clefts, a frequent birth malformation. Heterozygous mutations in SPECC1L have been identified in patients with atypical and syndromic clefts. Here, we report that in SPECC1L-knockdown cultured cells, staining of canonical adherens junction (AJ) components, ß-catenin and E-cadherin, was increased, and electron micrographs revealed an apico-basal diffusion of AJs. To understand the role of SPECC1L in craniofacial morphogenesis, we generated a mouse model of Specc1l deficiency. Homozygous mutants were embryonic lethal and showed impaired neural tube closure and CNCC delamination. Staining of AJ proteins was increased in the mutant neural folds. This AJ defect is consistent with impaired CNCC delamination, which requires AJ dissolution. Further, PI3K-AKT signaling was reduced and apoptosis was increased in Specc1l mutants. In vitro, moderate inhibition of PI3K-AKT signaling in wildtype cells was sufficient to cause AJ alterations. Importantly, AJ changes induced by SPECC1L-knockdown were rescued by activating the PI3K-AKT pathway. Together, these data indicate SPECC1L as a novel modulator of PI3K-AKT signaling and AJ biology, required for neural tube closure and CNCC delamination.


Subject(s)
Adherens Junctions/metabolism , Neural Crest/embryology , Neural Crest/metabolism , Phosphoproteins/deficiency , Animals , Apoptosis/genetics , Biomarkers , Cell Adhesion Molecules/metabolism , Cell Lineage/genetics , Gene Expression , Gene Knockout Techniques , Humans , Mice , Models, Biological , Mutation , Neural Tube Defects/genetics , Neural Tube Defects/pathology , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
16.
J Med Genet ; 52(2): 104-10, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25412741

ABSTRACT

BACKGROUND: Opitz G/BBB syndrome is a heterogeneous disorder characterised by variable expression of midline defects including cleft lip and palate, hypertelorism, laryngealtracheoesophageal anomalies, congenital heart defects, and hypospadias. The X-linked form of the condition has been associated with mutations in the MID1 gene on Xp22. The autosomal dominant form has been linked to chromosome 22q11.2, although the causative gene has yet to be elucidated. METHODS AND RESULTS: In this study, we performed whole exome sequencing on DNA samples from a three-generation family with characteristics of Opitz G/BBB syndrome with negative MID1 sequencing. We identified a heterozygous missense mutation c.1189A>C (p.Thr397Pro) in SPECC1L, located at chromosome 22q11.23. Mutation screening of an additional 19 patients with features of autosomal dominant Opitz G/BBB syndrome identified a c.3247G>A (p.Gly1083Ser) mutation segregating with the phenotype in another three-generation family. CONCLUSIONS: Previously, SPECC1L was shown to be required for proper facial morphogenesis with disruptions identified in two patients with oblique facial clefts. Collectively, these data demonstrate that SPECC1L mutations can cause syndromic forms of facial clefting including some cases of autosomal dominant Opitz G/BBB syndrome and support the original linkage to chromosome 22q11.2.


Subject(s)
Calcium-Binding Proteins/chemistry , Esophagus/abnormalities , Genes, Dominant , Genetic Predisposition to Disease , Hypertelorism/genetics , Hypospadias/genetics , Microfilament Proteins/chemistry , Mutation/genetics , Phosphoproteins/chemistry , Phosphoproteins/genetics , Adult , Base Sequence , DNA Mutational Analysis , Exons/genetics , Family , Female , Genetic Testing , Humans , Infant , Male , Microtubule Proteins/genetics , Molecular Sequence Data , Nuclear Proteins/genetics , Pedigree , Phenotype , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Transcription Factors/genetics , Ubiquitin-Protein Ligases , Calponins
17.
Hematol Oncol Stem Cell Ther ; 8(1): 6-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25482588

ABSTRACT

Single or multilineage bone marrow failure can be a serious health problem caused by hereditary and non-hereditary causes such as exposure to drugs or environmental toxins. Normal hematopoiesis requires the integrity of several pathways including the THPO-MPL pathway. Over the last two decades, significant advances in the understanding of normal and abnormal functions of this and related pathways have led to novel diagnostic and therapeutic options.


Subject(s)
Bone Marrow/physiology , Hematopoiesis/physiology , Signal Transduction/physiology , Thrombopoietin/metabolism , Humans
18.
Eur J Med Genet ; 57(2-3): 76-80, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24462885

ABSTRACT

Recently, 3 unrelated children with a potentially novel 3q26.33-3q27.2 microdeletion syndrome were reported. We now report a new 9 ½ years old Caucasian boy with a 2 Mb deletion of the same genomic region in combination with Klinefelter syndrome. He presented with facial dysmorphism, developmental delay, Asperger syndrome, thrombocytopenia, recurrent infections and hypogammaglobulinemia. The deletion in our patient improves upon the minimum region of the novel 3q26.33-3q27.2 microdeletion, and provides additional insights into the underlying genetic basis of the observed phenotypes. Consistent with two of three previously described patients, our patient also presents with thrombocytopenia, which we postulate is caused by haploinsufficiency of THPO. In addition, haploinsufficiency of LAMP3, a lymphoid and dendritic cell expressed protein that is implicated in bacterial and viral infections, pulmonary surfactant protein transport and amelogenin degradation, may be a novel cause for the immune deficiency, lung disease and dental abnormalities respectively as seen in these patients.


Subject(s)
Chromosome Deletion , Chromosome Disorders/diagnosis , Chromosome Disorders/genetics , Chromosomes, Human, Pair 3 , Child , Comparative Genomic Hybridization , Facies , Humans , In Situ Hybridization, Fluorescence , Male , Syndrome
19.
Blood ; 122(20): 3440-9, 2013 Nov 14.
Article in English | MEDLINE | ID: mdl-24085763

ABSTRACT

We recently identified 2 siblings afflicted with idiopathic, autosomal recessive aplastic anemia. Whole-exome sequencing identified a novel homozygous missense mutation in thrombopoietin (THPO, c.112C>T) in both affected siblings. This mutation encodes an arginine to cysteine substitution at residue 38 or residue 17 excluding the 21-amino acid signal peptide of THPO receptor binding domain (RBD). THPO has 4 conserved cysteines in its RBD that form 2 disulfide bonds. Our in silico modeling predicts that introduction of a fifth cysteine may disrupt normal disulfide bonding to cause poor receptor binding. In functional assays, the mutant-THPO-containing media shows two- to threefold reduced ability to sustain UT7-TPO cells, which require THPO for proliferation. Both parents and a sibling with heterozygous R17C change have reduced platelet counts, whereas a sibling with wild-type sequence has normal platelet count. Thus, the R17C partial loss-of-function allele results in aplastic anemia in the homozygous state and mild thrombocytopenia in the heterozygous state in our family. Together with the recent identification of THPO receptor (MPL) mutations and the effects of THPO agonists in aplastic anemia, our results have clinical implications in the diagnosis and treatment of patients with aplastic anemia and highlight a role for the THPO-MPL pathway in hematopoiesis in vivo.


Subject(s)
Anemia, Aplastic/genetics , Exome/genetics , Thrombopoietin/genetics , Adolescent , Adult , Amino Acid Substitution , Anemia, Aplastic/drug therapy , Base Sequence , Cells, Cultured , Child , Cloning, Molecular , Comparative Genomic Hybridization , Cystine/chemistry , Exons/genetics , Female , Genes, Recessive , Genotype , Humans , Male , Micronesia , Middle Aged , Models, Molecular , Molecular Sequence Data , Molecular Targeted Therapy , Mutation, Missense , Pedigree , Protein Binding , Protein Conformation , Receptors, Thrombopoietin/metabolism , Sequence Alignment , Sequence Homology, Nucleic Acid , Structure-Activity Relationship , Thrombopoietin/chemistry , Thrombopoietin/metabolism , Young Adult
20.
Development ; 140(13): 2697-702, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23720046

ABSTRACT

Bmp4 expression is tightly regulated during embryonic tooth development, with early expression in the dental epithelial placode leading to later expression in the dental mesenchyme. Msx1 is among several transcription factors that are induced by epithelial Bmp4 and that, in turn, are necessary for the induction and maintenance of dental mesenchymal Bmp4 expression. Thus, Msx1(-/-) teeth arrest at early bud stage and show loss of Bmp4 expression in the mesenchyme. Ectopic expression of Bmp4 rescues this bud stage arrest. We have identified Tbx2 expression in the dental mesenchyme at bud stage and show that this can be induced by epithelial Bmp4. We also show that endogenous Tbx2 and Msx1 can physically interact in mouse C3H10T1/2 cells. In order to ascertain a functional relationship between Msx1 and Tbx2 in tooth development, we crossed Tbx2 and Msx1 mutant mice. Our data show that the bud stage tooth arrest in Msx1(-/-) mice is partially rescued in Msx1(-/-);Tbx2(+/-) compound mutants. This rescue is accompanied by formation of the enamel knot (EK) and by restoration of mesenchymal Bmp4 expression. Finally, knockdown of Tbx2 in C3H10T1/2 cells results in an increase in Bmp4 expression. Together, these data identify a novel role for Tbx2 in tooth development and suggest that, following their induction by epithelial Bmp4, Msx1 and Tbx2 in turn antagonistically regulate odontogenic activity that leads to EK formation and to mesenchymal Bmp4 expression at the key bud-to-cap stage transition.


Subject(s)
Bone Morphogenetic Protein 4/metabolism , MSX1 Transcription Factor/metabolism , T-Box Domain Proteins/metabolism , Tooth/embryology , Tooth/metabolism , Animals , Bone Morphogenetic Protein 4/genetics , Cell Line , Immunohistochemistry , Immunoprecipitation , In Situ Hybridization , MSX1 Transcription Factor/genetics , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, Mutant Strains , Odontogenesis/genetics , Odontogenesis/physiology , Protein Binding , T-Box Domain Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...