Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.001
Filter
1.
Proc Natl Acad Sci U S A ; 121(39): e2405523121, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39292749

ABSTRACT

Pathogenic variants in SF3B4, a component of the U2 snRNP complex important for branchpoint sequence recognition and splicing, are responsible for the acrofacial disorders Nager and Rodriguez Syndrome, also known as SF3B4-related syndromes. Patients exhibit malformations in the head, face, limbs, vertebrae as well as the heart. To uncover the etiology of craniofacial malformations found in SF3B4-related syndromes, mutant mouse lines with homozygous deletion of Sf3b4 in neural crest cells (NCC) were generated. Like in human patients, these embryos had craniofacial and cardiac malformations with variable expressivity and penetrance. The severity and survival of Sf3b4 NCC mutants was modified by the level of Sf3b4 in neighboring non-NCC. RNA sequencing analysis of heads of embryos prior to morphological abnormalities revealed significant changes in expression of genes forming the NCC regulatory network, as well as an increase in exon skipping. Additionally, several key histone modifiers involved in craniofacial and cardiac development showed increased exon skipping. Increased exon skipping was also associated with use of a more proximal branch point, as well as an enrichment in thymidine bases in the 50 bp around the branch points. We propose that decrease in Sf3b4 causes changes in the expression and splicing of transcripts required for proper craniofacial and cardiac development, leading to abnormalities.


Subject(s)
Craniofacial Abnormalities , Disease Models, Animal , Heart Defects, Congenital , Neural Crest , RNA Splicing Factors , Animals , Mice , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Neural Crest/metabolism , Neural Crest/pathology , Neural Crest/embryology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/etiology , Heart Defects, Congenital/pathology , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/pathology , Craniofacial Abnormalities/etiology , RNA Splicing , Exons/genetics , Humans
2.
JCI Insight ; 9(16)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39171526

ABSTRACT

Craniofacial dysmorphisms are among the most common birth defects. Proteasome mutations frequently result in craniofacial dysmorphisms, including lower jaw malformations; however, the underlying mechanisms are unknown. Here, we used a zebrafish proteasome subunit ß 1 (psmb1) mutant to define the cellular mechanisms underlying proteasome mutation-induced craniofacial dysmorphisms. psmb1 mutants exhibited a flattened ceratohyal and smaller Meckel's and palatoquadrate cartilages. Ceratohyal flattening was a result of failed chondrocyte convergent extension, accompanied by reduced numbers of chondrocytes in the lower jaw due to defects in chondrocyte differentiation. Morphogenesis of craniofacial muscles and tendons was similarly perturbed. psmb1 mutants lacked the hyohyal muscles, and craniofacial tendons were shortened and disorganized. We additionally identified a critical period for proteasome function in craniofacial development, specifically during chondrocyte and muscle differentiation. psmb1 overexpression in sox10+ cells of mutant embryos rescued both cartilage and tendon phenotypes but induced only a partial rescue of the muscle phenotype, indicating that psmb1 was required in both tissue-autonomous and nonautonomous fashions during craniofacial development. Overall, our work demonstrates that psmb1 is required for craniofacial cartilage, tendon, and muscle differentiation and morphogenesis.


Subject(s)
Cartilage , Chondrocytes , Morphogenesis , Proteasome Endopeptidase Complex , Zebrafish Proteins , Zebrafish , Animals , Zebrafish/genetics , Morphogenesis/genetics , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/genetics , Chondrocytes/metabolism , Cartilage/metabolism , Cartilage/embryology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/pathology , Tendons/embryology , Tendons/metabolism , Tendons/abnormalities , Tendons/pathology , Cell Differentiation/genetics , Mutation , Gene Expression Regulation, Developmental , Chondrogenesis/genetics
3.
Eur J Med Genet ; 71: 104966, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39147273

ABSTRACT

OBJECTIVE: to report the first case series of Brazilian children diagnosed with Kleefstra syndrome, present a possible phenotype expansion to the syndrome and to raise physicians' awareness for this rare disease. RESULTS: seven patients with confirmed KS were evaluated, including 5 males and 2 females. Abnormal prenatal findings were observed in 4 patients. Most patients were born at term, with normal birth measurements. All patients had neurodevelopmental delay and 6 evolved with intellectual disability. Hearing loss was present in 57.1% of patients and 28.7% had congenital heart disease. In males, cryptorchidism was present in 75%. Despite the facial dysmorphisms, only 2 out of 7 patients had a pre-test clinical suspicion of KS. One specific patient presented bilateral agenesis of the semicircular canals, a very rare ear manifestation in Kleefstra syndrome, representing a possible phenotype expansion of the syndrome. CONCLUSION: this report aims to promote awareness among physicians evaluating patients in a context of neurodevelopmental delay or congenital malformations, especially congenital heart defects. We also highlight a possible phenotype expansion of the syndrome, with a case of semicircular anomaly, not reported in this syndrome so far.


Subject(s)
Chromosome Deletion , Craniofacial Abnormalities , Intellectual Disability , Phenotype , Semicircular Canals , Humans , Male , Female , Intellectual Disability/genetics , Intellectual Disability/pathology , Brazil , Craniofacial Abnormalities/pathology , Craniofacial Abnormalities/genetics , Child , Child, Preschool , Semicircular Canals/abnormalities , Semicircular Canals/pathology , Heart Defects, Congenital/pathology , Heart Defects, Congenital/genetics , Chromosomes, Human, Pair 9/genetics , Infant
4.
Dis Model Mech ; 17(6)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38967226

ABSTRACT

Robinow syndrome is a rare disease caused by variants of seven WNT pathway genes. Craniofacial features include widening of the nasal bridge and jaw hypoplasia. We used the chicken embryo to test whether two missense human FZD2 variants (1301G>T, p.Gly434Val; 425C>T, p.Pro142Lys) were sufficient to change frontonasal mass development. In vivo, the overexpression of retroviruses with wild-type or variant human FZD2 inhibited upper beak ossification. In primary cultures, wild-type and variant human FZD2 significantly inhibited chondrogenesis, with the 425C>T variant significantly decreasing activity of a SOX9 luciferase reporter compared to that for the wild type or 1301G>T. Both variants also increased nuclear shuttling of ß-catenin (CTNNB1) and increased the expression of TWIST1, which are inhibitory to chondrogenesis. In canonical WNT luciferase assays using frontonasal mass cells, the variants had dominant-negative effects on wild-type FZD2. In non-canonical assays, the 425C>T variant failed to activate the reporter above control levels and was unresponsive to exogenous WNT5A. This is the first single amino acid change to selectively alter ligand binding in a FZD receptor. Therefore, FZD2 missense variants are pathogenic and could lead to the altered craniofacial morphogenesis seen in Robinow syndrome.


Subject(s)
Chondrogenesis , Craniofacial Abnormalities , Frizzled Receptors , Animals , Chick Embryo , Humans , Beak , beta Catenin/metabolism , Cell Nucleus/metabolism , Chondrogenesis/genetics , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/pathology , Dwarfism , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , Limb Deformities, Congenital , Skull/pathology , Skull/embryology , Twist-Related Protein 1/metabolism , Twist-Related Protein 1/genetics , Urogenital Abnormalities , Wnt Signaling Pathway
5.
Eur J Med Genet ; 71: 104964, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39069253

ABSTRACT

Weiss-Kruszka Syndrome (WSKA) is caused by pathogenic variants in ZNF462 representing a rare autosomal dominant congenital anomaly syndrome. It is characterized by global developmental delay, hypotonia, feeding difficulties, and craniofacial abnormalities, documented in fewer than 30 patients. ZNF462, located on chromosome 9p31.2, is a transcription factor and has an important role during embryonic development and chromatin remodelling. Here, we report three new patients with WSKA, Through whole exome sequencing (WES) analysis, we identified two novel variants in three patients, two of whom are siblings. These variants (c.3078dup, p.Val1027Cysfs5 and c.4792A > T p.Lys1598*) in the ZNF462 gene are likely resulting in haploinsufficiency. Our patients help to further delineate the phenotype, genotype and potential therapeutic management strategies for WSKA. Since we report a second WSKA patient with an autoimmune disease further clinical and functional studies are needed to elucidate the association between this chromatin remodelling disorder and the development of autoimmune problems. In the future, collaborative efforts are encouraged to develop an episignature for WSKA, given the gene's function and associated patient phenotypes. This new technology has the potential to provide valuable insights into the disorder.


Subject(s)
DNA-Binding Proteins , Nerve Tissue Proteins , Transcription Factors , Child , Child, Preschool , Female , Humans , Infant , Male , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/pathology , Developmental Disabilities/genetics , Developmental Disabilities/pathology , DNA-Binding Proteins/genetics , Exome Sequencing , Haploinsufficiency , Mutation , Phenotype , Syndrome , Transcription Factors/genetics , Nerve Tissue Proteins/genetics
6.
Clin Genet ; 106(2): 209-213, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38693682

ABSTRACT

SATB2-associated syndrome (SAS, glass syndrome, OMIM#612313) is a neurodevelopmental autosomal dominant disorder with frequent craniofacial abnormalities including palatal and dental anomalies. To assess the role of Satb2 in craniofacial development, we analyzed mutant mice at different stages of development. Here, we show that Satb2 is broadly expressed in early embryonic mouse development including the mesenchyme of the second and third arches. Satb2-/- mutant mice exhibit microglossia, a shortened lower jaw, smaller trigeminal ganglia, and larger thyroids. We correlate these findings with the detailed clinical phenotype of four individuals with SAS and remarkable craniofacial phenotypes with one requiring mandibular distraction in childhood. We conclude that the mouse and patient data presented support less well-described phenotypic aspects of SAS including mandibular morphology and thyroid anatomical/functional issues.


Subject(s)
Branchial Region , Matrix Attachment Region Binding Proteins , Phenotype , Transcription Factors , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism , Animals , Humans , Mice , Transcription Factors/genetics , Branchial Region/abnormalities , Branchial Region/pathology , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/pathology , Female , Male , Mice, Knockout , Syndrome , Mandible/abnormalities , Mandible/pathology
7.
Am J Med Genet A ; 194(8): e63615, 2024 08.
Article in English | MEDLINE | ID: mdl-38563316

ABSTRACT

The Society for Craniofacial Genetics and Developmental Biology (SCGDB) held its 46th Annual Meeting at Cincinnati Children's Hospital Medical Center in Cincinnati, Ohio on October 10th-12th, 2023. On the first day of the meeting, Drs. Sally Moody and Justin Cotney were each honored with the SCGDB Distinguished Scientist Awards for their exceptional contributions to the field of craniofacial biology. The following two days of the meeting featured five sessions that highlighted new discoveries in signaling and genomic mechanisms regulating craniofacial development, human genetics, translational and regenerative approaches, and clinical management of craniofacial differences. Interactive workshops on spatial transcriptomics and scientific communication, as well as a poster session facilitated meaningful interactions among the 122 attendees representing diverse career stages and research backgrounds in developmental biology and genetics, strengthened the SCGDB community.


Subject(s)
Craniofacial Abnormalities , Developmental Biology , Humans , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/pathology
8.
Genet Med ; 26(7): 101126, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38529886

ABSTRACT

PURPOSE: DISP1 encodes a transmembrane protein that regulates the secretion of the morphogen, Sonic hedgehog, a deficiency of which is a major cause of holoprosencephaly (HPE). This disorder covers a spectrum of brain and midline craniofacial malformations. The objective of the present study was to better delineate the clinical phenotypes associated with division transporter dispatched-1 (DISP1) variants. METHODS: This study was based on the identification of at least 1 pathogenic variant of the DISP1 gene in individuals for whom detailed clinical data were available. RESULTS: A total of 23 DISP1 variants were identified in heterozygous, compound heterozygous or homozygous states in 25 individuals with midline craniofacial defects. Most cases were minor forms of HPE, with craniofacial features such as orofacial cleft, solitary median maxillary central incisor, and congenital nasal pyriform aperture stenosis. These individuals had either monoallelic loss-of-function variants or biallelic missense variants in DISP1. In individuals with severe HPE, the DISP1 variants were commonly found associated with a variant in another HPE-linked gene (ie, oligogenic inheritance). CONCLUSION: The genetic findings we have acquired demonstrate a significant involvement of DISP1 variants in the phenotypic spectrum of midline defects. This underlines its importance as a crucial element in the efficient secretion of Sonic hedgehog. We also demonstrated that the very rare solitary median maxillary central incisor and congenital nasal pyriform aperture stenosis combination is part of the DISP1-related phenotype. The present study highlights the clinical risks to be flagged up during genetic counseling after the discovery of a pathogenic DISP1 variant.


Subject(s)
Alleles , Holoprosencephaly , Phenotype , Adolescent , Child , Child, Preschool , Female , Humans , Infant , Male , Anodontia , Cleft Lip/genetics , Cleft Lip/pathology , Cleft Palate/genetics , Cleft Palate/pathology , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/pathology , Heterozygote , Holoprosencephaly/genetics , Holoprosencephaly/pathology , Homozygote , Incisor/abnormalities , Membrane Proteins/genetics , Mutation, Missense/genetics
9.
Clin Genet ; 105(6): 655-660, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38384171

ABSTRACT

Precise regulation of gene expression is important for correct neurodevelopment. 9q34.3 deletions affecting the EHMT1 gene result in a syndromic neurodevelopmental disorder named Kleefstra syndrome. In contrast, duplications of the 9q34.3 locus encompassing EHMT1 have been suggested to cause developmental disorders, but only limited information has been available. We have identified 15 individuals from 10 unrelated families, with 9q34.3 duplications <1.5 Mb in size, encompassing EHMT1 entirely. Clinical features included mild developmental delay, mild intellectual disability or learning problems, autism spectrum disorder, and behavior problems. The individuals did not consistently display dysmorphic features, congenital anomalies, or growth abnormalities. DNA methylation analysis revealed a weak DNAm profile for the cases with 9q34.3 duplication encompassing EHMT1, which could segregate the majority of the affected cases from controls. This study shows that individuals with 9q34.3 duplications including EHMT1 gene present with mild non-syndromic neurodevelopmental disorders and DNA methylation changes different from Kleefstra syndrome.


Subject(s)
Chromosome Deletion , Chromosome Duplication , Chromosomes, Human, Pair 9 , DNA Methylation , Heart Defects, Congenital , Histone-Lysine N-Methyltransferase , Intellectual Disability , Neurodevelopmental Disorders , Humans , DNA Methylation/genetics , Chromosomes, Human, Pair 9/genetics , Male , Female , Intellectual Disability/genetics , Intellectual Disability/pathology , Chromosome Duplication/genetics , Child , Child, Preschool , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/pathology , Adolescent , Phenotype
10.
Eur Arch Otorhinolaryngol ; 280(12): 5539-5546, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37505262

ABSTRACT

PURPOSE: To investigate the anatomical relationships between the structures adjacent to the cartilaginous portion of the ear canal in children with Work type I congenital branchial cleft anomalies (CFBCAs) and to develop new classifications and surgical strategies. METHODS: Retrospective analysis was performed on 50 children with Work type I CFBCAs admitted between December 2018 and December 2022. RESULTS: Among the 50 children, total parotidectomy was performed on 49 sides. In 44 cases (88%), the main body of the lesion was closely associated with the cartilage of the inferior ear canal wall. Among these cases, the lesions in 40 cases occurred within the space enclosed by the dorsal inferior wall cartilage, mastoid process, and parotid gland, while in the remaining four cases, the lesions were located between the anterior inferior wall cartilage and parotid gland. Based on the preoperative imaging observations, clinical manifestations, and intraoperative findings, the cases were classified into 6 subtypes (a to f) including 21 cases (42%) of Type Ia (inferior wall of EAC), 7 cases (14%) of Type Ib (bottom wall of EAC), 12 cases (24%) of Type Ic (posterior-inferior wall of EAC), 4 cases (8%) of Type Id (anterior-inferior wall of EAC), 4 cases (8%) of Type Ie (anterior ear wall of EAC), and 2 cases (4%) of Type If (isolated from parotid). CONCLUSION: Surgical intervention is the only treatment for first branchial cleft anomalies and a comprehensive understanding of the classifications will help with the precise localisation and excision of the lesions.


Subject(s)
Craniofacial Abnormalities , Pharyngeal Diseases , Child , Humans , Retrospective Studies , Craniofacial Abnormalities/pathology , Pharyngeal Diseases/surgery , Ear Canal/surgery , Branchial Region/diagnostic imaging , Branchial Region/surgery , Branchial Region/abnormalities
11.
Am J Hum Genet ; 110(5): 809-825, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37075751

ABSTRACT

Heterozygous pathogenic variants in POLR1A, which encodes the largest subunit of RNA Polymerase I, were previously identified as the cause of acrofacial dysostosis, Cincinnati-type. The predominant phenotypes observed in the cohort of 3 individuals were craniofacial anomalies reminiscent of Treacher Collins syndrome. We subsequently identified 17 additional individuals with 12 unique heterozygous variants in POLR1A and observed numerous additional phenotypes including neurodevelopmental abnormalities and structural cardiac defects, in combination with highly prevalent craniofacial anomalies and variable limb defects. To understand the pathogenesis of this pleiotropy, we modeled an allelic series of POLR1A variants in vitro and in vivo. In vitro assessments demonstrate variable effects of individual pathogenic variants on ribosomal RNA synthesis and nucleolar morphology, which supports the possibility of variant-specific phenotypic effects in affected individuals. To further explore variant-specific effects in vivo, we used CRISPR-Cas9 gene editing to recapitulate two human variants in mice. Additionally, spatiotemporal requirements for Polr1a in developmental lineages contributing to congenital anomalies in affected individuals were examined via conditional mutagenesis in neural crest cells (face and heart), the second heart field (cardiac outflow tract and right ventricle), and forebrain precursors in mice. Consistent with its ubiquitous role in the essential function of ribosome biogenesis, we observed that loss of Polr1a in any of these lineages causes cell-autonomous apoptosis resulting in embryonic malformations. Altogether, our work greatly expands the phenotype of human POLR1A-related disorders and demonstrates variant-specific effects that provide insights into the underlying pathogenesis of ribosomopathies.


Subject(s)
Craniofacial Abnormalities , Mandibulofacial Dysostosis , Humans , Mice , Animals , Mandibulofacial Dysostosis/genetics , Apoptosis , Mutagenesis , Ribosomes/genetics , Phenotype , Neural Crest/pathology , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/pathology
12.
Genesis ; 61(1-2): e23509, 2023 03.
Article in English | MEDLINE | ID: mdl-36622051

ABSTRACT

Craniofacial anomalies (CFAs) are a diverse group of disorders affecting the shapes of the face and the head. Malformation of the cranial base in humans leads CFAs, such as midfacial hypoplasia and craniosynostosis. These patients have significant burdens associated with breathing, speaking, and chewing. Invasive surgical intervention is the current primary option to correct these structural deficiencies. Understanding molecular cellular mechanism for craniofacial development would provide novel therapeutic options for CFAs. In this study, we found that enhanced bone morphogenetic protein (BMP) signaling in cranial neural crest cells (NCCs) (P0-Cre;caBmpr1a mice) causes premature fusion of intersphenoid synchondrosis (ISS) resulting in leading to short snouts and hypertelorism. Histological analyses revealed reduction of proliferation and higher cell death in ISS at postnatal day 3. We demonstrated to prevent the premature fusion of ISS in P0-Cre;caBmpr1a mice by injecting a p53 inhibitor Pifithrin-α to the pregnant mother from E15.5 to E18.5, resulting in rescue from short snouts and hypertelorism. We further demonstrated to prevent premature fusion of cranial sutures in P0-Cre;caBmpr1a mice by injecting Pifithrin-α through E8.5 to E18.5. These results suggested that enhanced BMP-p53-induced cell death in cranial NCCs causes premature fusion of ISS and sutures in time-dependent manner.


Subject(s)
Craniofacial Abnormalities , Skull Base , Bone Morphogenetic Proteins/metabolism , Neural Crest/metabolism , Neural Crest/pathology , Cell Proliferation , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Male , Female , Animals , Mice , Animals, Newborn , Signal Transduction , Apoptosis , Chondrocytes/metabolism , Smad Proteins/metabolism , Protein Binding , Craniofacial Abnormalities/metabolism , Craniofacial Abnormalities/pathology , Pregnancy , Skull Base/abnormalities , Skull Base/metabolism , Skull Base/pathology , Hypertelorism/metabolism , Hypertelorism/pathology
13.
Article in Chinese | MEDLINE | ID: mdl-36597371

ABSTRACT

Objective:To investigate the effect and influencing factors of individualized operation for congenital preauricular fistula in children. Methods:The clinical data of 98 cases (109 ears) of congenital preauricular fistula treated in Department of Otolaryngology,Fuzhou Children's Hospital of Fujian Medical University from July 2016 to December 2020 were retrospectively analyzed. According to the characteristics and infection of preauricular fistula,they were divided into common type and variant type,static period of inflammation and period of infection.Individual surgical methods such as classical fistula resection, double fusiform incision and fistula location resection were used respectively.The efficacy,complication and influencing factors of different surgical methods were analyzed. Results:The operation time of classical fistula resection was shorter, and the difference was statistically significant(t = -2.905 and-3.005 respectively, all P<0.05). According to the stages and types of fistulas, the selection of individualized surgical methods had achieved good results. There was no significant difference in incision complications and fistula recurrence among different surgical methods (all P>0.05). Conclusion:Once infection occurs in congenital preauricular fistula, surgical resection should be performed as soon as possible after infection control, or as early as possible after infection maximum control if infection cannot completely subside. Surgical incision design should be individualized, complete resection of fistulas and lesions, minimally invasive and aesthetic.


Subject(s)
Craniofacial Abnormalities , Fistula , Child , Humans , Retrospective Studies , Fistula/surgery , Ear/pathology , Craniofacial Abnormalities/pathology
14.
Nat Cardiovasc Res ; 2(12): 1190-1203, 2023 Dec.
Article in English | MEDLINE | ID: mdl-39183978

ABSTRACT

Maternal diabetes mellitus is among the most frequent environmental contributors to congenital birth defects, including heart defects and craniofacial anomalies, yet the cell types affected and mechanisms of disruption are largely unknown. Using multi-modal single cell analyses, here we show that maternal diabetes affects the epigenomic landscape of specific subsets of cardiac and craniofacial progenitors during embryogenesis. A previously unrecognized cardiac progenitor subpopulation expressing the homeodomain-containing protein ALX3 showed prominent chromatin accessibility changes and acquired a more posterior identity. Similarly, a subpopulation of neural crest-derived cells in the second pharyngeal arch, which contributes to craniofacial structures, displayed abnormalities in the epigenetic landscape and axial patterning defects. Chromatin accessibility changes in both populations were associated with increased retinoic acid signaling, known to establish anterior-posterior identity. This work highlights how an environmental insult can have highly selective epigenomic consequences on discrete cell types leading to developmental patterning defects.


Subject(s)
Epigenesis, Genetic , Gene Expression Regulation, Developmental , Pregnancy in Diabetics , Single-Cell Analysis , Female , Animals , Pregnancy , Pregnancy in Diabetics/genetics , Pregnancy in Diabetics/metabolism , Transcriptome , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Epigenomics , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Neural Crest/metabolism , Neural Crest/pathology , Disease Models, Animal , Signal Transduction/genetics , Tretinoin/metabolism , Mice , Gene Expression Profiling , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/pathology
15.
J Neural Transm (Vienna) ; 129(11): 1387-1391, 2022 11.
Article in English | MEDLINE | ID: mdl-36205783

ABSTRACT

Symptoms of obsessive-compulsive disorder (OCD) may rarely occur in the context of genetic syndromes. So far, an association between obsessive-compulsive symptoms (OCS) and ACTG1-associated Baraitser-Winter cerebrofrontofacial syndrome has not been described as yet. A thoroughly phenotyped patient with OCS and ACTG1-associated Baraitser-Winter cerebrofrontofacial syndrome is presented. The 25-year-old male patient was admitted to in-patient psychiatric care due to OCD. A whole-exome sequencing analysis was initiated as the patient also showed an autistic personality structure, below average intelligence measures, craniofacial dysmorphia signs, sensorineural hearing loss, and sinus cavernoma as well as subtle cardiac and ophthalmological alterations. The diagnosis of Baraitser-Winter cerebrofrontofacial syndrome type 2 was confirmed by the detection of a heterozygous likely pathogenic variant in the ACTG1 gene [c.1003C > T; p.(Arg335Cys), ACMG class 4]. The automated analysis of magnetic resonance imaging (MRI) revealed changes in the orbitofrontal, parietal, and occipital cortex of both sides and in the right mesiotemporal cortex. Electroencephalography (EEG) revealed intermittent rhythmic delta activity in the occipital and right temporal areas. Right mesiotemporal MRI and EEG alterations could be caused by a small brain parenchymal defect with hemosiderin deposits after a cavernomectomy. This paradigmatic case provides evidence of syndromic OCS in ACTG1-associated Baraitser-Winter cerebrofrontofacial syndrome. The MRI findings are compatible with a dysfunction of the cortico-striato-thalamo-cortical loops involved in OCD. If a common pathophysiology is confirmed in future studies, corresponding patients with Baraitser-Winter cerebrofrontofacial syndrome type 2 should be screened for OCS. The association may also contribute to a better understanding of OCD pathophysiology.


Subject(s)
Craniofacial Abnormalities , Obsessive-Compulsive Disorder , Abnormalities, Multiple , Actins , Adult , Craniofacial Abnormalities/diagnosis , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/pathology , Epilepsy , Facies , Hemosiderin , Humans , Intellectual Disability , Lissencephaly , Male , Obsessive-Compulsive Disorder/diagnosis , Obsessive-Compulsive Disorder/genetics
16.
Eur J Med Genet ; 65(4): 104455, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35182808

ABSTRACT

Rare deletions and duplications on the long arm of Chromosome 21 have previously been reported in many patients with craniofacial and developmental phenotypes. However, this Down Syndrome Critical Region (DSCR) contains multiple genes, making identifying a single causative gene difficult. Here, we report a case of a boy with bicoronal craniosynostosis, facial dysmorphism, developmental delay, and intellectual impairment who was found by whole genome sequencing to have a homozygous missense mutation in the Single-Minded Homolog 2 (SIM2) gene (c.461 A > G, p.Tyr154Cys) within the DSCR. SIM2 encodes an essential bHLH and PAS domain transcription factor expressed during fetal brain development and acts as a master regulator of neurogenesis. This variant is globally very rare, segregates in the family, and is predicted to be highly deleterious by in silico analysis, 3D molecular modeling of protein structure, and functional analysis of zebrafish models. Zebrafish expressing the human SIM2p.Y154C variant displayed a progressed microcephaly-like phenotype and head shape abnormalities. When combined with careful phenotyping of the patient vis-à-vis previously reported cases harboring structural variants in this critical 21q22 region, the data support a pathogenic role of SIM2 in this complex syndrome and demonstrates the utility of next-generation sequencing in prioritizing genes in contiguous deletions/duplications syndromes and diagnosing microarray-negative patients in the craniofacial clinic.


Subject(s)
Craniofacial Abnormalities , Down Syndrome , Intellectual Disability , Microcephaly , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/pathology , Homozygote , Humans , Intellectual Disability/genetics , Intellectual Disability/pathology , Microcephaly/genetics , Phenotype , Zebrafish/genetics
17.
Rev. ABENO ; 21(1): 1544, dez. 2021. ilus
Article in English | BBO - Dentistry | ID: biblio-1370622

ABSTRACT

Almost 30% of oral cleftsare associated with other structural abnormalities.However, little is known on orofacial characteristics related tothese cases since they are not systematically reported. To close this gap, we developed a collaborative learning approach supported by an interprofessional team aiming to systematicallydescribe oral findings and impactthe training of future professionals that hopefully will incorporate these descriptionsintotheir clinical practice. The methodological proposal consisted of small group sessions focusing on a particular syndrome or group of syndromes followed by examiningpatients with those conditions. Twenty cases were examined and studied over one semester andaset of conditions to be identified in the orofacial regionwasdefined. Here, we present a guideline that we suggest that dentists and dental institutions use. We also present the advantages of using collaborative learning as a tool in the training of the clinician (AU).


Quase 30% das fissuras orais estão associadas a outras anormalidades estruturais. No entanto, pouco se sabe sobre as características orofaciais relacionadas a esses casos, uma vez que não são relatados de forma sistemática. Para fechar essa lacuna, desenvolvemos uma abordagem de aprendizagem colaborativa apoiada por uma equipe interprofissional com o objetivo de descrever sistematicamente os achados orais e impactar o treinamento de futuros profissionais que, esperançosamente, irão incorporar essas descrições em sua prática clínica. A proposta metodológica consistia em sessões de pequenos grupos enfocando uma determinada síndrome ou grupo de síndromes seguidas de exame de pacientes com essas condições. Vinte casos foram examinados e estudados ao longo de um semestre e foi definido um conjunto de condições a serem identificadas na região orofacial. Aqui, apresentamos uma diretriz que sugerimos que os dentistas e instituições odontológicas utilizem. Também apresentamos as vantagens de usar a aprendizagem colaborativa como uma ferramenta no treinamento do clínico (AU).


Subject(s)
Humans , Orofaciodigital Syndromes/pathology , Cleft Palate/diagnosis , Craniofacial Abnormalities/pathology , Dentists , Education, Dental/methods , Interdisciplinary Placement/methods , Cleft Lip/pathology , Focus Groups/methods , Qualitative Research , Learning
18.
Development ; 148(21)2021 11 01.
Article in English | MEDLINE | ID: mdl-34739029

ABSTRACT

Genome editing simplifies the generation of new animal models for congenital disorders. However, the detailed and unbiased phenotypic assessment of altered embryonic development remains a challenge. Here, we explore how deep learning (U-Net) can automate segmentation tasks in various imaging modalities, and we quantify phenotypes of altered renal, neural and craniofacial development in Xenopus embryos in comparison with normal variability. We demonstrate the utility of this approach in embryos with polycystic kidneys (pkd1 and pkd2) and craniofacial dysmorphia (six1). We highlight how in toto light-sheet microscopy facilitates accurate reconstruction of brain and craniofacial structures within X. tropicalis embryos upon dyrk1a and six1 loss of function or treatment with retinoic acid inhibitors. These tools increase the sensitivity and throughput of evaluating developmental malformations caused by chemical or genetic disruption. Furthermore, we provide a library of pre-trained networks and detailed instructions for applying deep learning to the reader's own datasets. We demonstrate the versatility, precision and scalability of deep neural network phenotyping on embryonic disease models. By combining light-sheet microscopy and deep learning, we provide a framework for higher-throughput characterization of embryonic model organisms. This article has an associated 'The people behind the papers' interview.


Subject(s)
Deep Learning , Embryonic Development/genetics , Phenotype , Animals , Craniofacial Abnormalities/embryology , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/pathology , Disease Models, Animal , Image Processing, Computer-Assisted , Mice , Microscopy , Mutation , Neural Networks, Computer , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Polycystic Kidney Diseases/embryology , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/pathology , Xenopus Proteins/genetics , Xenopus laevis
19.
Mol Genet Genomic Med ; 9(11): e1645, 2021 11.
Article in English | MEDLINE | ID: mdl-34582124

ABSTRACT

BACKGROUND: Terminal deletions of the long arm of chromosome 7 are well known and frequently associated with syndromic holoprosencephaly due to the involvement of the SHH (aliases HHG1, SMMCI, TPT, TPTPS, and MCOPCB5) gene region. However, interstitial deletions including CNTNAP2 (aliases Caspr2, KIAA0868, and NRXN4) and excluding the SHH region are less common. METHODS: We report the clinical and molecular characterization associated with pure 7q35 and 7q35q36.1 deletion in two unrelated patients as detected by oligonucleotide-based array-CGH analysis. RESULTS: The common clinical features were abnormal maternal serum screening during first-trimester pregnancy, low occipitofrontal circumference at birth, hypotonia, abnormal feet, developmental delay, impaired language development, generalized seizures, hyperactive behavior, friendly personality, and cranio-facial dysmorphism. Both deletions occurred de novo and sequencing of CNTNAP2, a candidate gene for epilepsy and autism showed absence of mutation on the contralateral allele. CONCLUSION: Combined haploinsufficiency of GALNTL5 (alias GalNAc-T5L), CUL1, SSPO (aliases SCO-spondin, KIAA0543, and FLJ36112), AOC1 (alias DAO), RHEB, and especially KMT2C (alias KIAA1506 and HALR) with monoallelic disruption of CNTNAP2 may explain neurologic abnormalities, hypotonia, and exostoses. Haploinsufficiency of PRKAG2 (aliases AAKG, AAKG2, H91620p, WPWS, and CMH6) and KCNH2 (aliases Kv11.1, HERG, and erg1) genes may be responsible of long QT syndrome observed for one patient.


Subject(s)
Chromosome Deletion , Chromosome Disorders/genetics , Chromosomes, Human, Pair 7/genetics , Craniofacial Abnormalities/genetics , DNA-Binding Proteins/genetics , Developmental Disabilities/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Child, Preschool , Chromosome Disorders/pathology , Craniofacial Abnormalities/pathology , Developmental Disabilities/pathology , Haploinsufficiency , Humans , Male , Noninvasive Prenatal Testing , Phenotype
20.
Int J Mol Sci ; 22(17)2021 Aug 27.
Article in English | MEDLINE | ID: mdl-34502207

ABSTRACT

The complexity of skeletal pathologies makes use of in vivo models essential to elucidate the pathogenesis of the diseases; nevertheless, chondrocyte and osteoblast cell lines provide relevant information on the underlying disease mechanisms. Due to the limitations of primary chondrocytes, immortalized cells represent a unique tool to overcome this problem since they grow very easily for several passages. However, in the immortalization procedure the cells might lose the original phenotype; thus, these cell lines should be deeply characterized before their use. We immortalized primary chondrocytes from a Cant1 knock-out mouse, an animal model of Desbuquois dysplasia type 1, with a plasmid expressing the SV40 large and small T antigen. This cell line, based on morphological and biochemical parameters, showed preservation of the chondrocyte phenotype. In addition reduced proteoglycan synthesis and oversulfation of glycosaminoglycan chains were demonstrated, as already observed in primary chondrocytes from the Cant1 knock-out mouse. In conclusion, immortalized Cant1 knock-out chondrocytes maintained the disease phenotype observed in primary cells validating the in vitro model and providing an additional tool to further study the proteoglycan biosynthesis defect. The same approach might be extended to other cartilage disorders.


Subject(s)
Acid Anhydride Hydrolases/physiology , Chondrocytes/pathology , Craniofacial Abnormalities/pathology , Dwarfism/pathology , Glycosaminoglycans/metabolism , Joint Instability/pathology , Ossification, Heterotopic/pathology , Phenotype , Polydactyly/pathology , Animals , Cell Line, Transformed , Chondrocytes/metabolism , Craniofacial Abnormalities/etiology , Craniofacial Abnormalities/metabolism , Dwarfism/etiology , Dwarfism/metabolism , Joint Instability/etiology , Joint Instability/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Ossification, Heterotopic/etiology , Ossification, Heterotopic/metabolism , Polydactyly/etiology , Polydactyly/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL