Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 129
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Development ; 149(22)2022 11 15.
Article in English | MEDLINE | ID: mdl-36408946

ABSTRACT

Craniofacial development requires precise spatiotemporal regulation of multiple signaling pathways that crosstalk to coordinate the growth and patterning of the skull with surrounding tissues. Recent insights into these signaling pathways and previously uncharacterized progenitor cell populations have refined our understanding of skull patterning, bone mineralization and tissue homeostasis. Here, we touch upon classical studies and recent advances with an emphasis on developmental and signaling mechanisms that regulate the osteoblast lineage for the calvaria, which forms the roof of the skull. We highlight studies that illustrate the roles of osteoprogenitor cells and cranial suture-derived stem cells for proper calvarial growth and homeostasis. We also discuss genes and signaling pathways that control suture patency and highlight how perturbing the molecular regulation of these pathways leads to craniosynostosis. Finally, we discuss the recently discovered tissue and signaling interactions that integrate skull and cerebrovascular development, and the potential implications for both cerebrospinal fluid hydrodynamics and brain waste clearance in craniosynostosis.


Subject(s)
Craniosynostoses , Skull , Humans , Skull/metabolism , Cranial Sutures/metabolism , Craniosynostoses/genetics , Craniosynostoses/metabolism , Homeostasis , Signal Transduction
2.
Development ; 149(1)2022 01 01.
Article in English | MEDLINE | ID: mdl-34878091

ABSTRACT

A major feature of Saethre-Chotzen syndrome is coronal craniosynostosis, the fusion of the frontal and parietal bones at the coronal suture. It is caused by heterozygous loss-of-function mutations in either of the bHLH transcription factors TWIST1 and TCF12. Although compound heterozygous Tcf12; Twist1 mice display severe coronal synostosis, the individual role of Tcf12 had remained unexplored. Here, we show that Tcf12 controls several key processes in calvarial development, including the rate of frontal and parietal bone growth, and the boundary between sutural and osteogenic cells. Genetic analysis supports an embryonic requirement for Tcf12 in suture formation, as combined deletion of Tcf12 in embryonic neural crest and mesoderm, but not in postnatal suture mesenchyme, disrupts the coronal suture. We also detected asymmetric distribution of mesenchymal cells on opposing sides of the wild-type frontal and parietal bones, which prefigures later bone overlap at the sutures. In Tcf12 mutants, reduced asymmetry is associated with bones meeting end-on-end, possibly contributing to synostosis. Our results support embryonic requirements of Tcf12 in proper formation of the overlapping coronal suture.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Craniosynostoses/metabolism , Osteogenesis , Skull/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Craniosynostoses/embryology , Craniosynostoses/genetics , Mesenchymal Stem Cells/metabolism , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Neural Crest/metabolism , Skull/metabolism
3.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Article in English | MEDLINE | ID: mdl-34548398

ABSTRACT

Skeletal ciliopathies (e.g., Jeune syndrome, short rib polydactyly syndrome, and Sensenbrenner syndrome) are frequently associated with nephronophthisis-like cystic kidney disease and other organ manifestations. Despite recent progress in genetic mapping of causative loci, a common molecular mechanism of cartilage defects and cystic kidneys has remained elusive. Targeting two ciliary chondrodysplasia loci (ift80 and ift172) by CRISPR/Cas9 mutagenesis, we established models for skeletal ciliopathies in Xenopus tropicalis Froglets exhibited severe limb deformities, polydactyly, and cystic kidneys, closely matching the phenotype of affected patients. A data mining-based in silico screen found ttc30a to be related to known skeletal ciliopathy genes. CRISPR/Cas9 targeting replicated limb malformations and renal cysts identical to the models of established disease genes. Loss of Ttc30a impaired embryonic renal excretion and ciliogenesis because of altered posttranslational tubulin acetylation, glycylation, and defective axoneme compartmentalization. Ttc30a/b transcripts are enriched in chondrocytes and osteocytes of single-cell RNA-sequenced embryonic mouse limbs. We identify TTC30A/B as an essential node in the network of ciliary chondrodysplasia and nephronophthisis-like disease proteins and suggest that tubulin modifications and cilia segmentation contribute to skeletal and renal ciliopathy manifestations of ciliopathies in a cell type-specific manner. These findings have implications for potential therapeutic strategies.


Subject(s)
Bone and Bones/abnormalities , Ciliopathies/pathology , Craniosynostoses/pathology , Cytoskeletal Proteins/metabolism , Ectodermal Dysplasia/pathology , Embryo, Nonmammalian/pathology , Musculoskeletal Abnormalities/pathology , Polycystic Kidney Diseases/pathology , Tubulin/chemistry , Animals , Bone and Bones/metabolism , Bone and Bones/pathology , Ciliopathies/genetics , Ciliopathies/metabolism , Craniosynostoses/genetics , Craniosynostoses/metabolism , Cytoskeletal Proteins/genetics , Disease Models, Animal , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/metabolism , Embryo, Nonmammalian/metabolism , Musculoskeletal Abnormalities/genetics , Musculoskeletal Abnormalities/metabolism , Phenotype , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/metabolism , Tubulin/metabolism , Xenopus laevis
4.
Hum Mol Genet ; 30(3-4): 213-225, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33517396

ABSTRACT

Primary cilia contain specific proteins to achieve their functions as cellular antennae. Ciliary protein trafficking is mediated by the intraflagellar transport (IFT) machinery containing the IFT-A and IFT-B complexes. Mutations in genes encoding the IFT-A subunits (IFT43, IFT121/WDR35, IFT122, IFT139/TTC21B, IFT140 and IFT144/WDR19) often result in skeletal ciliopathies, including cranioectodermal dysplasia (CED). We here characterized the molecular and cellular defects of CED caused by compound heterozygous mutations in IFT144 [the missense variant IFT144(L710S) and the nonsense variant IFT144(R1103*)]. These two variants were distinct with regard to their interactions with other IFT-A subunits and with the IFT-B complex. When exogenously expressed in IFT144-knockout (KO) cells, IFT144(L710S) as well as IFT144(WT) rescued both moderately compromised ciliogenesis and the abnormal localization of ciliary proteins. As the homozygous IFT144(L710S) mutation was found to cause autosomal recessive retinitis pigmentosa, IFT144(L710S) is likely to be hypomorphic at the cellular level. In striking contrast, the exogenous expression of IFT144(R1103*) in IFT144-KO cells exacerbated the ciliogenesis defects. The expression of IFT144(R1103*) together with IFT144(WT) restored the abnormal phenotypes of IFT144-KO cells. However, the coexpression of IFT144(R1103*) with the hypomorphic IFT144(L710S) variant in IFT144-KO cells, which mimics the genotype of compound heterozygous CED patients, resulted in severe ciliogenesis defects. Taken together, these observations demonstrate that compound heterozygous mutations in IFT144 cause severe ciliary defects via a complicated mechanism, where one allele can cause severe ciliary defects when combined with a hypomorphic allele.


Subject(s)
Bone and Bones/abnormalities , Cilia/metabolism , Craniosynostoses/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Ectodermal Dysplasia/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mutation , Bone and Bones/metabolism , Bone and Bones/physiopathology , Cilia/pathology , Ciliopathies/genetics , Ciliopathies/metabolism , Ciliopathies/physiopathology , Codon, Nonsense , Craniosynostoses/genetics , Craniosynostoses/physiopathology , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/physiopathology , HEK293 Cells , Humans , Mutation, Missense
5.
Hum Genet ; 142(1): 21-32, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35997807

ABSTRACT

Lambdoid craniosynostosis (CS) is a congenital anomaly resulting from premature fusion of the cranial suture between the parietal and occipital bones. Predominantly sporadic, it is the rarest form of CS and its genetic etiology is largely unexplored. Exome sequencing of 25 kindreds, including 18 parent-offspring trios with sporadic lambdoid CS, revealed a marked excess of damaging (predominantly missense) de novo mutations that account for ~ 40% of sporadic cases. These mutations clustered in the BMP signaling cascade (P = 1.6 × 10-7), including mutations in genes encoding BMP receptors (ACVRL1 and ACVR2A), transcription factors (SOX11, FOXO1) and a transcriptional co-repressor (IFRD1), none of which have been implicated in other forms of CS. These missense mutations are at residues critical for substrate or target sequence recognition and many are inferred to cause genetic gain-of-function. Additionally, mutations in transcription factor NFIX were implicated in syndromic craniosynostosis affecting diverse sutures. Single cell RNA sequencing analysis of the mouse lambdoid suture identified enrichment of mutations in osteoblast precursors (P = 1.6 × 10-6), implicating perturbations in the balance between proliferation and differentiation of osteoprogenitor cells in lambdoid CS. The results contribute to the growing knowledge of the genetics of CS, have implications for genetic counseling, and further elucidate the molecular etiology of premature suture fusion.


Subject(s)
Craniosynostoses , Mice , Animals , Craniosynostoses/genetics , Craniosynostoses/metabolism , Mutation , Signal Transduction/genetics , Transcription Factors/genetics , Cell Differentiation , NFI Transcription Factors/genetics , NFI Transcription Factors/metabolism
6.
J Cell Mol Med ; 26(8): 2428-2437, 2022 04.
Article in English | MEDLINE | ID: mdl-35285136

ABSTRACT

Craniosynostosis is the premature fusion of skull sutures and has a severe pathological impact on childrens' life. Mechanical forces are capable of triggering biological responses in bone cells and regulate osteoblastogenesis in cranial sutures, leading to premature closure. The mechanosensitive proteins polycystin-1 (PC1) and polycystin-2 (PC2) have been documented to play an important role in craniofacial proliferation and development. Herein, we investigated the contribution of PC1 to the pathogenesis of non-syndromic craniosynostosis and the associated molecular mechanisms. Protein expression of PC1 and PC2 was detected in bone fragments derived from craniosynostosis patients via immunohistochemistry. To explore the modulatory role of PC1 in primary cranial suture cells, we further abrogated the function of PC1 extracellular mechanosensing domain using a specific anti-PC1 IgPKD1 antibody. Effect of IgPKD1 treatment was evaluated with cell proliferation and migration assays. Activation of PI3K/AKT/mTOR pathway components was further detected via Western blot in primary cranial suture cells following IgPKD1 treatment. PC1 and PC2 are expressed in human tissues of craniosynostosis. PC1 functional inhibition resulted in elevated proliferation and migration of primary cranial suture cells. PC1 inhibition also induced activation of AKT, exhibiting elevated phospho (p)-AKT (Ser473) levels, but not 4EBP1 or p70S6K activation. Our findings indicate that PC1 may act as a mechanosensing molecule in cranial sutures by modulating osteoblastic cell proliferation and migration through the PC1/AKT/mTORC2 cascade with a potential impact on the development of non-syndromic craniosynostosis.


Subject(s)
Craniosynostoses , Proto-Oncogene Proteins c-akt , Cell Proliferation , Child , Craniosynostoses/genetics , Craniosynostoses/metabolism , Humans , Mechanistic Target of Rapamycin Complex 2/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism
7.
J Cell Mol Med ; 25(7): 3216-3225, 2021 04.
Article in English | MEDLINE | ID: mdl-33656806

ABSTRACT

Craniosynostosis refers to the premature fusion of one or more cranial sutures leading to skull shape deformities and brain growth restriction. Among the many factors that contribute to abnormal suture fusion, mechanical forces seem to play a major role. Nevertheless, the underlying mechanobiology-related mechanisms of craniosynostosis still remain unknown. Understanding how aberrant mechanosensation and mechanotransduction drive premature suture fusion will offer important insights into the pathophysiology of craniosynostosis and result in the development of new therapies, which can be used to intervene at an early stage and prevent premature suture fusion. Herein, we provide evidence for the first time on the role of polycystin-1 (PC1), a key protein in cellular mechanosensitivity, in craniosynostosis, using primary cranial suture cells isolated from patients with trigonocephaly and dolichocephaly, two common types of craniosynostosis. Initially, we showed that PC1 is expressed at the mRNA and protein level in both trigonocephaly and dolichocephaly cranial suture cells. Followingly, by utilizing an antibody against the mechanosensing extracellular N-terminal domain of PC1, we demonstrated that PC1 regulates runt-related transcription factor 2 (RUNX2) activation and osteocalcin gene expression via extracellular signal-regulated kinase (ERK) signalling in our human craniosynostosis cell model. Altogether, our study reveals a novel mechanotransduction signalling axis, PC1-ERK-RUNX2, which affects osteoblastic differentiation in cranial suture cells from trigonocephaly and dolichocephaly patients.


Subject(s)
Craniosynostoses/metabolism , TRPP Cation Channels/metabolism , Cells, Cultured , Child , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Fibroblasts/metabolism , Humans , MAP Kinase Signaling System , Male , Mechanotransduction, Cellular , Osteoblasts/metabolism , Osteocalcin/genetics , Osteocalcin/metabolism , TRPP Cation Channels/genetics
8.
Hum Mol Genet ; 28(15): 2501-2513, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31067316

ABSTRACT

Craniosynostosis, the premature ossification of cranial sutures, is a developmental disorder of the skull vault, occurring in approximately 1 in 2250 births. The causes are heterogeneous, with a monogenic basis identified in ~25% of patients. Using whole-genome sequencing, we identified a novel, de novo variant in BCL11B, c.7C>A, encoding an R3S substitution (p.R3S), in a male patient with coronal suture synostosis. BCL11B is a transcription factor that interacts directly with the nucleosome remodelling and deacetylation complex (NuRD) and polycomb-related complex 2 (PRC2) through the invariant proteins RBBP4 and RBBP7. The p.R3S substitution occurs within a conserved amino-terminal motif (RRKQxxP) of BCL11B and reduces interaction with both transcriptional complexes. Equilibrium binding studies and molecular dynamics simulations show that the p.R3S substitution disrupts ionic coordination between BCL11B and the RBBP4-MTA1 complex, a subassembly of the NuRD complex, and increases the conformational flexibility of Arg-4, Lys-5 and Gln-6 of BCL11B. These alterations collectively reduce the affinity of BCL11B p.R3S for the RBBP4-MTA1 complex by nearly an order of magnitude. We generated a mouse model of the BCL11B p.R3S substitution using a CRISPR-Cas9-based approach, and we report herein that these mice exhibit craniosynostosis of the coronal suture, as well as other cranial sutures. This finding provides strong evidence that the BCL11B p.R3S substitution is causally associated with craniosynostosis and confirms an important role for BCL11B in the maintenance of cranial suture patency.


Subject(s)
Chromatin Assembly and Disassembly , Cranial Sutures/growth & development , Craniosynostoses/metabolism , Mutation, Missense , Nucleosomes/metabolism , Osteogenesis , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Animals , Cranial Sutures/metabolism , Craniosynostoses/genetics , Craniosynostoses/physiopathology , DNA Mutational Analysis , Disease Models, Animal , Humans , Infant , Male , Mice , Protein Binding , Protein Conformation , Repressor Proteins/metabolism , Repressor Proteins/physiology , Retinoblastoma-Binding Protein 4/metabolism , Trans-Activators/metabolism , Tumor Suppressor Proteins/metabolism , Tumor Suppressor Proteins/physiology , White People , Whole Genome Sequencing
9.
Hum Genet ; 139(4): 461-472, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31980905

ABSTRACT

SKI pathogenic variations are associated with Shprintzen-Goldberg Syndrome (SGS), a rare systemic connective tissue disorder characterized by craniofacial, skeletal and cardiovascular features. So far, the clinical description, including intellectual disability, has been relatively homogeneous, and the known pathogenic variations were located in two different hotspots of the SKI gene. In the course of diagnosing Marfan syndrome and related disorders, we identified nine sporadic probands (aged 2-47 years) carrying three different likely pathogenic or pathogenic variants in the SKI gene affecting the same amino acid (Thr180). Seven of these molecular events were confirmed de novo. All probands displayed a milder morphological phenotype with a marfanoid habitus that did not initially lead to a clinical diagnosis of SGS. Only three of them had learning disorders, and none had intellectual disability. Six out of nine presented thoracic aortic aneurysm, which led to preventive surgery in the oldest case. This report extends the phenotypic spectrum of variants identified in the SKI gene. We describe a new mutational hotspot associated with a marfanoid syndrome with no intellectual disability. Cardiovascular involvement was confirmed in a significant number of cases, highlighting the importance of accurately diagnosing SGS and ensuring appropriate medical treatment and follow-up.


Subject(s)
Arachnodactyly , Craniosynostoses , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Marfan Syndrome , Mutation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Adolescent , Adult , Arachnodactyly/diagnosis , Arachnodactyly/genetics , Arachnodactyly/metabolism , Child , Child, Preschool , Craniosynostoses/diagnosis , Craniosynostoses/genetics , Craniosynostoses/metabolism , Female , Humans , Male , Marfan Syndrome/diagnosis , Marfan Syndrome/genetics , Marfan Syndrome/metabolism , Middle Aged , Pathology, Molecular
10.
Development ; 144(21): 4026-4036, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28947535

ABSTRACT

Craniosynostosis is a prevalent human birth defect characterized by premature fusion of calvarial bones. In this study, we show that tight regulation of endogenous PDGFRα activity is required for normal calvarium development in the mouse and that dysregulated PDGFRα activity causes craniosynostosis. Constitutive activation of PDGFRα leads to expansion of cartilage underlying the coronal sutures, which contribute to suture closure through endochondral ossification, in a process regulated in part by PI3K/AKT signaling. Our results thus identify a novel mechanism underlying calvarial development in craniosynostosis.


Subject(s)
Cartilage/embryology , Cranial Sutures/embryology , Cranial Sutures/metabolism , Craniosynostoses/metabolism , Morphogenesis , Osteogenesis , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Alleles , Animals , Cartilage/abnormalities , Cartilage/metabolism , Cell Lineage , Chondrogenesis , Cranial Sutures/pathology , Gene Expression Regulation, Developmental , Ligands , Mesoderm/metabolism , Mice, Inbred C57BL , Neural Crest/metabolism , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , Signal Transduction , Skull/abnormalities , Skull/pathology
11.
PLoS Genet ; 13(3): e1006679, 2017 03.
Article in English | MEDLINE | ID: mdl-28301481

ABSTRACT

3MC syndrome is an autosomal recessive heterogeneous disorder with features linked to developmental abnormalities. The main features include facial dysmorphism, craniosynostosis and cleft lip/palate; skeletal structures derived from cranial neural crest cells (cNCC). We previously reported that lectin complement pathway genes COLEC11 and MASP1/3 are mutated in 3MC syndrome patients. Here we define a new gene, COLEC10, also mutated in 3MC families and present novel mutations in COLEC11 and MASP1/3 genes in a further five families. The protein products of COLEC11 and COLEC10, CL-K1 and CL-L1 respectively, form heteromeric complexes. We show COLEC10 is expressed in the base membrane of the palate during murine embryo development. We demonstrate how mutations in COLEC10 (c.25C>T; p.Arg9Ter, c.226delA; p.Gly77Glufs*66 and c.528C>G p.Cys176Trp) impair the expression and/or secretion of CL-L1 highlighting their pathogenicity. Together, these findings provide further evidence linking the lectin complement pathway and complement factors COLEC11 and COLEC10 to morphogenesis of craniofacial structures and 3MC etiology.


Subject(s)
Abnormalities, Multiple/genetics , Cleft Palate/genetics , Collectins/genetics , Craniofacial Abnormalities/genetics , Craniosynostoses/genetics , Mutation , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology , Animals , Base Sequence , Blotting, Western , Cell Line , Cleft Palate/metabolism , Collectins/metabolism , Craniofacial Abnormalities/metabolism , Craniosynostoses/metabolism , Exome/genetics , Family Health , Female , Genetic Predisposition to Disease/genetics , HEK293 Cells , HeLa Cells , Humans , Male , Mice , Sequence Analysis, DNA/methods , Syndrome
12.
Proc Natl Acad Sci U S A ; 114(35): E7341-E7347, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28808027

ABSTRACT

Non-syndromic craniosynostosis (NSC) is a frequent congenital malformation in which one or more cranial sutures fuse prematurely. Mutations causing rare syndromic craniosynostoses in humans and engineered mouse models commonly increase signaling of the Wnt, bone morphogenetic protein (BMP), or Ras/ERK pathways, converging on shared nuclear targets that promote bone formation. In contrast, the genetics of NSC is largely unexplored. More than 95% of NSC is sporadic, suggesting a role for de novo mutations. Exome sequencing of 291 parent-offspring trios with midline NSC revealed 15 probands with heterozygous damaging de novo mutations in 12 negative regulators of Wnt, BMP, and Ras/ERK signaling (10.9-fold enrichment, P = 2.4 × 10-11). SMAD6 had 4 de novo and 14 transmitted mutations; no other gene had more than 1. Four familial NSC kindreds had mutations in genes previously implicated in syndromic disease. Collectively, these mutations contribute to 10% of probands. Mutations are predominantly loss-of-function, implicating haploinsufficiency as a frequent mechanism. A common risk variant near BMP2 increased the penetrance of SMAD6 mutations and was overtransmitted to patients with de novo mutations in other genes in these pathways, supporting a frequent two-locus pathogenesis. These findings implicate new genes in NSC and demonstrate related pathophysiology of common non-syndromic and rare syndromic craniosynostoses. These findings have implications for diagnosis, risk of recurrence, and risk of adverse neurodevelopmental outcomes. Finally, the use of pathways identified in rare syndromic disease to find genes accounting for non-syndromic cases may prove broadly relevant to understanding other congenital disorders featuring high locus heterogeneity.


Subject(s)
Craniosynostoses/genetics , Craniosynostoses/physiopathology , Adult , Animals , Bone Morphogenetic Proteins/antagonists & inhibitors , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Child , Child, Preschool , Cranial Sutures , Craniosynostoses/metabolism , Exome/genetics , Female , Humans , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Male , Mutation/genetics , Osteogenesis/genetics , Penetrance , Phenotype , Sequence Analysis, DNA/methods , Signal Transduction , Smad6 Protein/genetics , Smad6 Protein/physiology , Exome Sequencing/methods , ras Proteins/antagonists & inhibitors , ras Proteins/genetics , ras Proteins/metabolism
13.
Int J Mol Sci ; 21(12)2020 Jun 19.
Article in English | MEDLINE | ID: mdl-32575385

ABSTRACT

All skeletal bones house osteogenic stem cell niches, in which mesenchymal stromal cells (MSC) provide progenitors for tissue growth and regeneration. They have been widely studied in long bones formed through endochondral ossification. Limited information is available on the composition of the osteogenic niche in flat bones (i.e., skull vault bones) that develop through direct membranous ossification. Craniosynostosis (CS) is a congenital craniofacial defect due to the excessive and premature ossification of skull vault sutures. This study aimed at analysing the expression of GLI1, AXIN2 and THY1 in the context of the human skull vault, using nonsyndromic forms of CS (NCS) as a model to test their functional implication in the aberrant osteogenic process. The expression of selected markers was studied in NCS patients' calvarial bone specimens, to assess the in vivo location of cells, and in MSC isolated thereof. The marker expression profile was analysed during in vitro osteogenic differentiation to validate the functional implication. Our results show that GLI1 and AXIN2 are expressed in periosteal and endosteal locations within the osteogenic niche of human calvarial bones. Their expression is higher in MSC isolated from calvarial bones than in those isolated from long bones and tends to decrease upon osteogenic commitment and differentiation. In particular, AXIN2 expression was lower in cells isolated from prematurely fused sutures than in those derived from patent sutures of NCS patients. This suggests that AXIN2 could reasonably represent a marker for the stem cell population that undergoes depletion during the premature ossification process occurring in CS.


Subject(s)
Axin Protein/metabolism , Biomarkers/metabolism , Craniosynostoses/metabolism , Skull/cytology , Zinc Finger Protein GLI1/metabolism , Axin Protein/genetics , Cell Differentiation , Cells, Cultured , Craniosynostoses/genetics , Down-Regulation , Female , Humans , Infant , Infant, Newborn , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteogenesis , Primary Cell Culture , Skull/metabolism , Stem Cell Niche , Zinc Finger Protein GLI1/genetics
14.
Nanomedicine ; 14(7): 2365-2374, 2018 10.
Article in English | MEDLINE | ID: mdl-28648641

ABSTRACT

Advances in molecular biology and nanomedicine based therapies hold promise to obviate the need of multiple surgical interventions (associated with current management) in craniosynostosis by preventing bone re-ossification. One such adjunctive therapy involves application of glypicans 1 and 3 (GPC1 and GPC3) that are BMP inhibitors implicated in downregulating the BMP2 activity in prematurely fusing sutures. Electrochemically anodized Titania nanotube (TNT) arrays have been recognized as a promising localized, long-term drug delivery platform for bone-related therapies. This study presents the application of nanoengineered TNT/Ti implants loaded with recombinant glypicans for craniosynostosis therapy. By using Dual luciferase Reporter assay, we tested the biofunctionality of eluted glypicans from the TNT/Ti implants for BMP2 bioactivity regulation in C2C12 murine myoblast cell line. BMP2 activity was inhibited significantly for up to 15days by the glypicans released from polymer-coated TNT/Ti implants, indicating their potential application in adjunctive craniosynostosis treatment.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Craniosynostoses/drug therapy , Drug Liberation , Glypicans/administration & dosage , Myoblasts/drug effects , Prostheses and Implants , Titanium/chemistry , Animals , Cells, Cultured , Craniosynostoses/metabolism , Craniosynostoses/pathology , Drug Delivery Systems , Gene Expression Regulation/drug effects , Glypicans/chemistry , Mice , Myoblasts/cytology , Myoblasts/metabolism , Polymers/chemistry
15.
Adv Exp Med Biol ; 1046: 249-268, 2018.
Article in English | MEDLINE | ID: mdl-29442326

ABSTRACT

Zic genes are strongly expressed in the cerebellum. This feature leads to their initial identification and their name "zic," as the abbreviation of "zinc finger protein of the cerebellum." Zic gene function in cerebellar development has been investigated mainly in mice. However, association of heterozygous loss of ZIC1 and ZIC4 with Dandy-Walker malformation, a structural birth defect of the human cerebellum, highlights the clinical relevance of these studies. Two proposed mechanisms for Zic-mediated cerebellar developmental control have been documented: regulation of neuronal progenitor proliferation-differentiation and the patterning of the cerebellar primordium. Clinical studies have also revealed that ZIC1 gain of function mutations contribute to coronal craniosynostosis, a rare skull malformation. The molecular pathways contributing to these phenotypes are not fully explored; however, embryonic interactions with sonic hedgehog signaling, retinoic acid signaling, and TGFß signaling have been described during mouse cerebellar development. Further, Zic1/2 target a multitude of genes associated with cerebellar granule cell maturation during postnatal mouse cerebellar development.


Subject(s)
Cerebellum , Craniosynostoses , Dandy-Walker Syndrome , Neural Stem Cells , Signal Transduction/genetics , Transcription Factors , Animals , Cerebellum/growth & development , Cerebellum/physiology , Craniosynostoses/genetics , Craniosynostoses/metabolism , Craniosynostoses/pathology , Dandy-Walker Syndrome/genetics , Dandy-Walker Syndrome/metabolism , Dandy-Walker Syndrome/pathology , Humans , Mice , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Transcription Factors/genetics , Transcription Factors/metabolism
16.
J Cell Mol Med ; 21(11): 2782-2795, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28470873

ABSTRACT

The cranial suture complex is a heterogeneous tissue consisting of osteogenic progenitor cells and mesenchymal stem cells (MSCs) from bone marrow and suture mesenchyme. The fusion of cranial sutures is a highly coordinated and tightly regulated process during development. Craniosynostosis is a congenital malformation caused by premature fusion of cranial sutures. While the progenitor cells derived from the cranial suture complex should prove valuable for studying the molecular mechanisms underlying suture development and pathogenic premature suture fusion, primary human cranial suture progenitors (SuPs) have limited life span and gradually lose osteoblastic ability over passages. To overcome technical challenges in maintaining sufficient and long-term culture of SuPs for suture biology studies, we establish and characterize the reversibly immortalized human cranial suture progenitors (iSuPs). Using a reversible immortalization system expressing SV40 T flanked with FRT sites, we demonstrate that primary human suture progenitor cells derived from the patent sutures of craniosynostosis patients can be efficiently immortalized. The iSuPs maintain long-term proliferative activity, express most of the consensus MSC markers and can differentiate into osteogenic and adipogenic lineages upon BMP9 stimulation in vitro and in vivo. The removal of SV40 T antigen by FLP recombinase results in a decrease in cell proliferation and an increase in the endogenous osteogenic and adipogenic capability in the iSuPs. Therefore, the iSuPs should be a valuable resource to study suture development, intramembranous ossification and the pathogenesis of craniosynostosis, as well as to explore cranial bone tissue engineering.


Subject(s)
Cranial Sutures/metabolism , Craniosynostoses/genetics , Founder Effect , Growth Differentiation Factors/genetics , Mesenchymal Stem Cells/metabolism , Osteogenesis/genetics , Adipocytes/cytology , Adipocytes/metabolism , Cell Differentiation , Cell Line, Transformed , Cell Proliferation , Cranial Sutures/pathology , Craniosynostoses/metabolism , Craniosynostoses/pathology , Gene Expression , Growth Differentiation Factor 2 , Growth Differentiation Factors/metabolism , Humans , Infant , Male , Mesenchymal Stem Cells/cytology , Osteoblasts/cytology , Osteoblasts/metabolism , Simian virus 40/genetics , Simian virus 40/metabolism , Transformation, Genetic
17.
Hum Mol Genet ; 24(23): 6699-710, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26362256

ABSTRACT

Patients with 46,XY gonadal dysgenesis (GD) exhibit genital anomalies, which range from hypospadias to complete male-to-female sex reversal. However, a molecular diagnosis is made in only 30% of cases. Heterozygous mutations in the human FGFR2 gene cause various craniosynostosis syndromes including Crouzon and Pfeiffer, but testicular defects were not reported. Here, we describe a patient whose features we would suggest represent a new FGFR2-related syndrome, craniosynostosis with XY male-to-female sex reversal or CSR. The craniosynostosis patient was chromosomally XY, but presented as a phenotypic female due to complete GD. DNA sequencing identified the FGFR2c heterozygous missense mutation, c.1025G>C (p.Cys342Ser). Substitution of Cys342 by Ser or other amino acids (Arg/Phe/Try/Tyr) has been previously reported in Crouzon and Pfeiffer syndrome. We show that the 'knock-in' Crouzon mouse model Fgfr2c(C342Y/C342Y) carrying a Cys342Tyr substitution displays XY gonadal sex reversal with variable expressivity. We also show that despite FGFR2c-Cys342Tyr being widely considered a gain-of-function mutation, Cys342Tyr substitution in the gonad leads to loss of function, as demonstrated by sex reversal in Fgfr2c(C342Y/-) mice carrying the knock-in allele on a null background. The rarity of our patient suggests the influence of modifier genes which exacerbated the testicular phenotype. Indeed, patient whole exome analysis revealed several potential modifiers expressed in Sertoli cells at the time of testis determination in mice. In summary, this study identifies the first FGFR2 mutation in a 46,XY GD patient. We conclude that, in certain rare genetic contexts, maintaining normal levels of FGFR2 signaling is important for human testis determination.


Subject(s)
Craniosynostoses/genetics , Gonadal Dysgenesis, 46,XY/genetics , Mutation, Missense , Receptor, Fibroblast Growth Factor, Type 2/genetics , Adolescent , Animals , Craniosynostoses/metabolism , DNA Mutational Analysis , Disease Models, Animal , Female , Gene Knock-In Techniques , Humans , Male , Mice , Mice, Mutant Strains , Syndrome
18.
Dev Dyn ; 245(2): 175-82, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26605996

ABSTRACT

BACKGROUND: Inactivating mutations in tissue-nonspecific alkaline phosphatase (TNAP) cause hypophosphatasia (HPP), which is commonly characterized by decreased bone mineralization. Infants and mice with HPP can also develop craniosynostosis and craniofacial shape abnormalities, although the mechanism by which TNAP deficiency causes these craniofacial defects is not yet known. Manifestations of HPP are heterogeneous in severity, and evidence from the literature suggests that much of this variability is mutation dependent. Here, we performed a comprehensive analysis of craniosynostosis and craniofacial shape variation in the Alpl(-/-) mouse model of murine HPP as an initial step toward better understanding penetrance of the HPP craniofacial phenotype. RESULTS: Despite similar deficiencies in alkaline phosphatase, Alpl(-/-) mice develop craniosynostosis and a brachycephalic/acrocephalic craniofacial shape of variable penetrance. Only those Alpl(-/-) mice with a severe bone hypomineralization defect develop craniosynostosis and an abnormal craniofacial shape. CONCLUSIONS: These results indicate that variability of the HPP phenotype is not entirely dependent upon the type of genetic mutation and level of residual alkaline phosphatase activity. Additionally, despite a severity continuum of the bone hypomineralization phenotype, craniofacial skeletal shape abnormalities and craniosynostosis occur only in the context of severely diminished bone mineralization in the Alpl(-/-) mouse model of HPP.


Subject(s)
Alkaline Phosphatase/genetics , Craniofacial Abnormalities/genetics , Craniosynostoses/genetics , Hypophosphatasia/genetics , Alkaline Phosphatase/metabolism , Animals , Craniofacial Abnormalities/metabolism , Craniosynostoses/metabolism , Disease Models, Animal , Hypophosphatasia/metabolism , Mice , Mice, Knockout , Phenotype
19.
BMC Dev Biol ; 16(1): 37, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27756203

ABSTRACT

BACKGROUND: Increased apposition of the frontal and parietal bones of the skull during embryogenesis may be a risk factor for the subsequent development of premature skull fusion, or craniosynostosis. Human craniosynostosis is a prevalent, and often serious embryological and neonatal pathology. Other than known mutations in a small number of contributing genes, the aetiology of craniosynostosis is largely unknown. Therefore, the identification of novel genes which contribute to normal skull patterning, morphology and premature suture apposition is imperative, in order to fully understand the genetic regulation of cranial development. RESULTS: Using advanced imaging techniques and quantitative measurement, we show that genetic deletion of the highly-conserved transcription factor Grainyhead-like 3 (Grhl3) in mice (Grhl3 -/- ) leads to decreased skull size, aberrant skull morphology and premature apposition of the coronal sutures during embryogenesis. Furthermore, Grhl3 -/- mice also present with premature collagen deposition and osteoblast alignment at the sutures, and the physical interaction between the developing skull, and outermost covering of the brain (the dura mater), as well as the overlying dermis and subcutaneous tissue, appears compromised in embryos lacking Grhl3. Although Grhl3 -/- mice die at birth, we investigated skull morphology and size in adult animals lacking one Grhl3 allele (heterozygous; Grhl3 +/- ), which are viable and fertile. We found that these adult mice also present with a smaller cranial cavity, suggestive of post-natal haploinsufficiency in the context of cranial development. CONCLUSIONS: Our findings show that our Grhl3 mice present with increased apposition of the frontal and parietal bones, suggesting that Grhl3 may be involved in the developmental pathogenesis of craniosynostosis.


Subject(s)
Craniosynostoses/genetics , DNA-Binding Proteins/genetics , Frontal Bone/metabolism , Parietal Bone/metabolism , Transcription Factors/genetics , Animals , Cranial Sutures/abnormalities , Cranial Sutures/metabolism , Craniosynostoses/embryology , Craniosynostoses/metabolism , DNA-Binding Proteins/deficiency , Embryonic Development/genetics , Frontal Bone/abnormalities , Frontal Bone/diagnostic imaging , Gene Expression Regulation, Developmental , Immunohistochemistry , Mice, Knockout , Parietal Bone/abnormalities , Parietal Bone/diagnostic imaging , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Risk Factors , Skull/abnormalities , Skull/metabolism , Transcription Factors/deficiency , X-Ray Microtomography
20.
Birth Defects Res A Clin Mol Teratol ; 106(10): 803-813, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27435288

ABSTRACT

BACKGROUND: Craniosynostosis, the premature fusion of one or more of the cranial sutures, is estimated to occur in 1:1800 to 2500 births. Genetic murine models of craniosynostosis exist, but often imperfectly model human patients. Case, cohort, and surveillance studies have identified excess thyroid hormone as an agent that can either cause or exacerbate human cases of craniosynostosis. METHODS: Here we investigate the influence of in utero and in vitro exogenous thyroid hormone exposure on a murine model of craniosynostosis, Twist 1 +/-. RESULTS: By 15 days post-natal, there was evidence of coronal suture fusion in the Twist 1 +/- model, regardless of exposure. With the exception of craniofacial width, there were no significant effects of exposure; however, the Twist 1 +/- phenotype was significantly different from the wild-type control. Twist 1 +/- cranial suture cells did not respond to thyroxine treatment as measured by proliferation, osteogenic differentiation, and gene expression of osteogenic markers. However, treatment of these cells did result in modulation of thyroid associated gene expression. CONCLUSION: Our findings suggest the phenotypic effects of the genetic mutation largely outweighed the effects of thyroxine exposure in the Twist 1 +/- model. These results highlight difficultly in experimentally modeling gene-environment interactions for craniosynostotic phenotypes. Birth Defects Research (Part A) 106:803-813, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Abnormalities, Drug-Induced , Craniosynostoses , Gene-Environment Interaction , Nuclear Proteins/genetics , Phenotype , Thyroxine/adverse effects , Twist-Related Protein 1/genetics , Abnormalities, Drug-Induced/genetics , Abnormalities, Drug-Induced/metabolism , Abnormalities, Drug-Induced/pathology , Animals , Craniosynostoses/chemically induced , Craniosynostoses/genetics , Craniosynostoses/metabolism , Craniosynostoses/pathology , Disease Models, Animal , Gene Expression Regulation , Mice , Mice, Mutant Strains , Osteogenesis/drug effects , Osteogenesis/genetics , Thyroxine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL