Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.123
Filter
1.
Clin Genet ; 106(1): 109-113, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38665048

ABSTRACT

Usmani-Riazuddin syndrome (USRISR, MIM# 619548; USRISD, MIM#619467) is a very rare genetic condition. recently associated with deleterious variants in AP1G1 (MIM* 603533). It is characterized by multisystemic involvement including intellectual disability, speech and developmental delay, behavioral anomalies, muscular tone disorders, seizures, limb defects, and unspecified facial gestalt. In this report, we describe this syndrome for the second time, in association to a novel AP1G1 variant identified in a toddler with multisystemic involvement including intellectual disability, speech and developmental delay, behavioral anomalies, arrhythmias, hearing loss, skin changes, and limb defects. Next generation sequencing (NGS) analysis through clinical exome disclosed AP1G1: c.1969C>G (p.Leu657Val), de novo, likely pathogenic variant, according to ACMG classification criteria. Proband's facial features resembled the spectrum of chromatinopathies. Clinical pictures were analyzed and a clinical overlap was supported by DeepGestalt analysis (www.face2gene.com). The system identified 6 chromatin disorders out of 30 possible diagnoses. The remaining 24 included 9 miscellaneous cryptic chromosomal abnormalities (excluded due to normal microarray study). To the best of our knowledge, this is the first description of likely distinctive facial features in a patient with Usmani-Riazuddin syndrome. Further multicentric analyses are needed for a better definition of this aspect.


Subject(s)
Intellectual Disability , Phenotype , Humans , Intellectual Disability/genetics , Intellectual Disability/pathology , Male , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , High-Throughput Nucleotide Sequencing , Mutation/genetics , Child, Preschool , Female , Developmental Disabilities/genetics , Developmental Disabilities/pathology
2.
Cell Tissue Res ; 396(2): 255-267, 2024 May.
Article in English | MEDLINE | ID: mdl-38502237

ABSTRACT

Joubert syndrome (JS) is a recessively inherited congenital ataxia characterized by hypotonia, psychomotor delay, abnormal ocular movements, intellectual disability, and a peculiar cerebellar and brainstem malformation, the "molar tooth sign." Over 40 causative genes have been reported, all encoding for proteins implicated in the structure or functioning of the primary cilium, a subcellular organelle widely present in embryonic and adult tissues. In this paper, we developed an in vitro neuronal differentiation model using patient-derived induced pluripotent stem cells (iPSCs), to evaluate possible neurodevelopmental defects in JS. To this end, iPSCs from four JS patients harboring mutations in distinct JS genes (AHI1, CPLANE1, TMEM67, and CC2D2A) were differentiated alongside healthy control cells to obtain mid-hindbrain precursors and cerebellar granule cells. Differentiation was monitored over 31 days through the detection of lineage-specific marker expression by qRT-PCR, immunofluorescence, and transcriptomics analysis. All JS patient-derived iPSCs, regardless of the mutant gene, showed a similar impairment to differentiate into mid-hindbrain and cerebellar granule cells when compared to healthy controls. In addition, analysis of primary cilium count and morphology showed notable ciliary defects in all differentiating JS patient-derived iPSCs compared to controls. These results confirm that patient-derived iPSCs are an accessible and relevant in vitro model to analyze cellular phenotypes connected to the presence of JS gene mutations in a neuronal context.


Subject(s)
Abnormalities, Multiple , Cell Differentiation , Cerebellum , Cerebellum/abnormalities , Eye Abnormalities , Induced Pluripotent Stem Cells , Kidney Diseases, Cystic , Neurons , Retina , Retina/abnormalities , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Humans , Eye Abnormalities/genetics , Eye Abnormalities/pathology , Cerebellum/pathology , Cerebellum/metabolism , Neurons/metabolism , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Retina/metabolism , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/pathology , Kidney Diseases, Cystic/metabolism , Male , Female , Mutation/genetics , Cilia/metabolism
3.
Neuropediatrics ; 55(2): 129-134, 2024 04.
Article in English | MEDLINE | ID: mdl-38365198

ABSTRACT

PGAP2 gene has been known to be the cause of "hyperphosphatasia, mental retardation syndrome-3" (HPMRS3). To date, 14 pathogenic variants in PGAP2 have been identified as the cause of this syndrome in 24 patients described in single-case reports or small clinical series with pan-ethnic distribution. We aim to present a pediatric PGAP2-mutated case, intending to further expand the clinical phenotype of the syndrome and to report our experience on a therapeutic approach to drug-resistant epilepsy.We present the clinical, neuroradiological, and genetic characterization of a Caucasian pediatric subject with biallelic pathogenic variants in the PGAP2 gene revealed by next generation sequencing analysis.We identified a subject who presented with global developmental delay and visual impairment. Brain magnetic resonance imaging showed mild hypoplasia of the inferior cerebellar vermis and corpus callosum and mild white matter reduction. Laboratory investigations detected an increase in alkaline phosphatase. At the age of 13 months, he began to present epileptic focal seizures with impaired awareness, which did not respond to various antiseizure medications. Electroencephalogram (EEG) showed progressive background activity disorganization and multifocal epileptic abnormalities. Treatment with high-dose pyridoxine showed partial benefit, but the persistence of seizures and the lack of EEG amelioration prompted us to introduce ketogenic diet treatment.Our case provides a further phenotypical expansion of HPMRS3 to include developmental and epileptic encephalopathy. Due to the limited number of patients reported so far, the full delineation of the clinical spectrum of HPMRS3 and indications for precision medicine would benefit from the description of new cases and their follow-up evaluations.


Subject(s)
Abnormalities, Multiple , Epilepsy , Intellectual Disability , Humans , Infant , Male , Abnormalities, Multiple/pathology , Brain/pathology , Epilepsy/diagnostic imaging , Epilepsy/drug therapy , Epilepsy/genetics , Intellectual Disability/genetics , Intellectual Disability/pathology , Phenotype , Seizures , Syndrome
4.
Clin Genet ; 105(6): 671-675, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38351533

ABSTRACT

The biallelic variants of the POP1 gene are associated with the anauxetic dysplasia (AAD OMIM 607095), a rare skeletal dysplasia, characterized by prenatal rhizomelic shortening of limbs and generalized joint hypermobility. Affected individuals usually have normal neurodevelopmental milestones. Here we present three cases from the same family with likely pathogenic homozygous POP1 variant and a completely novel phenotype: a girl with global developmental delay and autism, microcephaly, peculiar dysmorphic features and multiple congenital anomalies. Two subsequent pregnancies were terminated due to multiple congenital malformations. Fetal DNA samples revealed the same homozygous variant in the POP1 gene. Expression of the RMRP was reduced in the proband compared with control and slightly reduced in both heterozygous parents, carriers for this variant. To our knowledge, this is the first report of this new phenotype, associated with a novel likely pathogenic variant in POP1. Our findings expand the phenotypic spectrum of POP1-related disorders.


Subject(s)
Homozygote , Phenotype , Humans , Female , Male , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Mutation , Pedigree , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Child, Preschool , Child , Genetic Predisposition to Disease
5.
Clin Genet ; 106(1): 66-71, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38417950

ABSTRACT

Pulmonary hypoplasia, Diaphragmatic anomalies, Anophthalmia/microphthalmia, and Cardiac defects (PDAC) syndrome is a genetically heterogeneous multiple congenital malformation syndrome. Although pathogenic variants in RARB and STRA6 are established causes of PDAC, many PDAC cases remain unsolved at the molecular level. Recently, we proposed biallelic WNT7B variants as a novel etiology based on several families with typical features of PDAC syndrome albeit with variable expressivity. Here, we report three patients from two families that share a novel founder variant in WNT7B (c.739C > T; Arg247Trp). The phenotypic expression of this variant ranges from typical PDAC features to isolated genitourinary anomalies. Similar to previously reported PDAC-associated WNT7B variants, this variant was found to significantly impair WNT7B signaling activity further corroborating its proposed pathogenicity. This report adds further evidence to WNT7B-related PDAC and expands its variable expressivity.


Subject(s)
Phenotype , Wnt Proteins , Humans , Wnt Proteins/genetics , Male , Female , Anophthalmos/genetics , Anophthalmos/pathology , Microphthalmos/genetics , Microphthalmos/pathology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Founder Effect , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Pedigree , Mutation , Genetic Predisposition to Disease , Syndrome , Lung/pathology , Lung/abnormalities
6.
Genes (Basel) ; 15(2)2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38397201

ABSTRACT

The condition known as 22q11.2 deletion syndrome (MIM #188400) is a rare disease with a highly variable clinical presentation including more than 180 features; specific guidelines for screening individuals have been used to support clinical suspicion before confirmatory tests by Brazil's Craniofacial Project. Of the 2568 patients listed in the Brazilian Database on Craniofacial Anomalies, 43 individuals negative for the 22q11.2 deletion syndrome were further investigated through whole-exome sequencing. Three patients (6.7%) presented with heterozygous pathogenic variants in the KMT2A gene, including a novel variant (c.6158+1del) and two that had been previously reported (c.173dup and c.3241C>T); reverse phenotyping concluded that all three patients presented features of Wiedemann-Steiner syndrome, such as neurodevelopmental disorders and dysmorphic facial features (n = 3), hyperactivity and anxiety (n = 2), thick eyebrows and lower-limb hypertrichosis (n = 2), congenital heart disease (n = 1), short stature (n = 1), and velopharyngeal insufficiency (n = 2). Overlapping features between 22q11.2 deletion syndrome and Wiedemann-Steiner syndrome comprised neuropsychiatric disorders and dysmorphic characteristics involving the eyes and nose region; velopharyngeal insufficiency was seen in two patients and is an unreported finding in WDSTS. Therefore, we suggest that both conditions should be included in each other's differential diagnoses.


Subject(s)
Abnormalities, Multiple , Contracture , DiGeorge Syndrome , Facies , Growth Disorders , Intellectual Disability , Microcephaly , Velopharyngeal Insufficiency , Humans , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , DiGeorge Syndrome/genetics , Intellectual Disability/diagnosis , Intellectual Disability/genetics
7.
Clin Genet ; 105(6): 596-610, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38278647

ABSTRACT

Multiple congenital contractures (MCC) due to fetal akinesia manifest across a broad spectrum of diseases, ranging from mild distal arthrogryposis to lethal fetal akinesia deformation sequence. We hereby present a series of 26 fetuses displaying severe MCC phenotypes from 18 families and describe detailed prenatal ultrasound findings, postmortem clinical evaluations, and genetic investigations. Most common prenatal findings were abnormal facial profile (65%), central nervous system abnormalities (62%), polyhydramnios (50%), increased nuchal translucency (50%), and fetal hydrops (35%). Postmortem examinations unveiled additional anomalies including facial dysmorphisms, dysplastic skeletal changes, ichthyosis, multiple pterygia, and myopathy, allowing preliminary diagnosis of particular Mendelian disorders in multiple patients. Evaluation of the parents revealed maternal grip myotonia in one family. By exome sequencing and targeted testing, we identified causative variants in ACTC1, CHST14, COG6, DMPK, DOK7, HSPG2, KLHL7, KLHL40, KIAA1109, NEB, PSAT1, RAPSN, USP14, and WASHC5 in 15 families, and one patient with a plausible diagnosis associated with biallelic NEB variants. Three patients received a dual diagnosis. Pathogenic alterations in newly discovered genes or in previously known genes recently linked to new MCC phenotypes were observed in 44% of the cohort. Our results provide new insights into the clinical and molecular landscape of lethal MCC phenotypes.


Subject(s)
Arthrogryposis , Fetus , Phenotype , Humans , Female , Male , Arthrogryposis/genetics , Arthrogryposis/diagnosis , Arthrogryposis/pathology , Fetus/pathology , Exome Sequencing , Contracture/genetics , Contracture/diagnosis , Contracture/pathology , Pregnancy , Ultrasonography, Prenatal , Mutation , Abnormalities, Multiple/genetics , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/pathology
8.
Am J Med Genet A ; 194(6): e63540, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38243407

ABSTRACT

Coffin-Siris Syndrome (CSS, MIM 135900) is now a well-described genetic condition caused by pathogenic variants in the Bromocriptine activating factor (BAF) complex, including ARID1B, ARID1A, ARID2, SMARCA4, SMARCE1, SMARCB1, SOX11, SMARCC2, DPF2, and more recently, BICRA. Individuals with CSS have a spectrum of various medical challenges, most often evident at birth, including feeding difficulties, hypotonia, organ-system anomalies, and learning and developmental differences. The classic finding of fifth digit hypo- or aplasia is seen variably. ARID2, previously described, is one of the less frequently observed gene changes in CSS. Although individuals with ARID2 have been reported to have classic features of CSS including hypertrichosis, coarse facial features, short stature, and fifth digit anomalies, as with many of the other CSS genes, there appears to be a spectrum of phenotypes. We report here a cohort of 17 individuals with ARID2 variants from the Coffin-Siris/BAF clinical registry and detail their medical challenges as well as developmental progress. Feeding difficulties, hypotonia, and short stature occur often, and hip dysplasia appears to occur more often than with other genes, however more severe medical challenges such as significant brain and cardiac malformations are rarer. Individuals appear to have mild to moderate intellectual impairment and may carry additional diagnoses such as ADHD. Further phenotypic description of this gene will aid clinicians caring for individuals with this rarer form of CSS.


Subject(s)
Abnormalities, Multiple , Face/abnormalities , Hand Deformities, Congenital , Intellectual Disability , Micrognathism , Neck , Neck/abnormalities , Phenotype , Transcription Factors , Humans , Micrognathism/genetics , Micrognathism/diagnosis , Intellectual Disability/genetics , Intellectual Disability/pathology , Neck/pathology , Hand Deformities, Congenital/genetics , Hand Deformities, Congenital/diagnosis , Male , Female , Transcription Factors/genetics , Abnormalities, Multiple/genetics , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/pathology , Child , Child, Preschool , Infant , Mutation/genetics , Adolescent , DNA-Binding Proteins/genetics , Genetic Predisposition to Disease
9.
Orphanet J Rare Dis ; 19(1): 32, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38291488

ABSTRACT

Vertebral malformations (VMs) pose a significant global health problem, causing chronic pain and disability. Vertebral defects occur as isolated conditions or within the spectrum of various congenital disorders, such as Klippel-Feil syndrome, congenital scoliosis, spondylocostal dysostosis, sacral agenesis, and neural tube defects. Although both genetic abnormalities and environmental factors can contribute to abnormal vertebral development, our knowledge on molecular mechanisms of numerous VMs is still limited. Furthermore, there is a lack of resource that consolidates the current knowledge in this field. In this pioneering review, we provide a comprehensive analysis of the latest research on the molecular basis of VMs and the association of the VMs-related causative genes with bone developmental signaling pathways. Our study identifies 118 genes linked to VMs, with 98 genes involved in biological pathways crucial for the formation of the vertebral column. Overall, the review summarizes the current knowledge on VM genetics, and provides new insights into potential involvement of biological pathways in VM pathogenesis. We also present an overview of available data regarding the role of epigenetic and environmental factors in VMs. We identify areas where knowledge is lacking, such as precise molecular mechanisms in which specific genes contribute to the development of VMs. Finally, we propose future research avenues that could address knowledge gaps.


Subject(s)
Abnormalities, Multiple , Hernia, Diaphragmatic , Klippel-Feil Syndrome , Scoliosis , Humans , Spine/abnormalities , Spine/pathology , Abnormalities, Multiple/pathology , Klippel-Feil Syndrome/pathology , Hernia, Diaphragmatic/pathology
10.
Pediatr Dermatol ; 41(2): 289-291, 2024.
Article in English | MEDLINE | ID: mdl-38018302

ABSTRACT

A 6-year-old boy with multiple capillary malformations of the port-wine birthmark (PWB) type on the right leg since birth presented with a varicose vein and segmental overgrowth of the affected leg. Genetic testing on affected skin confirmed the presence of a somatic novel pathogenic HRAS 30 bp in-frame duplication/insertion in the switch II domain. This case illustrates the phenotypic overlap of different genotypes and shows that somatic HRAS pathogenic variants, especially in-frame duplications/insertions, must be added to the list of the underlying causes in capillary malformations.


Subject(s)
Abnormalities, Multiple , Capillaries/abnormalities , Port-Wine Stain , Vascular Malformations , Male , Child , Humans , Mutation , Port-Wine Stain/genetics , Vascular Malformations/genetics , Abnormalities, Multiple/pathology , Proto-Oncogene Proteins p21(ras)/genetics
11.
Pediatr Dev Pathol ; 27(2): 181-186, 2024.
Article in English | MEDLINE | ID: mdl-37981638

ABSTRACT

Coffin-Siris syndrome is an autosomal dominant disorder with neurological, cardiovascular, and gastrointestinal symptoms. Patients with Coffin-Siris syndrome typically have variable degree of developmental delay or intellectual disability, muscular hypotonia, dysmorphic facial features, sparse scalp hair, but otherwise hirsutism and fifth digit nail or distal phalanx hypoplasia or aplasia. Coffin-Siris syndrome is caused by pathogenic variants in 12 different genes including SMARCB1 and ARID1A. Pathogenic SMARCB1 gene variants cause Coffin-Siris syndrome 3 whereas pathogenic ARID1A gene variants cause Coffin-Siris syndrome 2. Here, we present two prenatal Coffin-Siris syndrome cases with autosomal dominant pathogenic variants: SMARCB1 gene c.1066_1067del, p.(Leu356AspfsTer4) variant, and a novel ARID1A gene c.1920+3_1920+6del variant. The prenatal phenotype in Coffin-Siris syndrome has been rarely described. This article widens the phenotypic spectrum of prenatal Coffin-Siris syndrome with severely hypoplastic right ventricle with VSD and truncus arteriosus type III, persisting left superior and inferior caval vein, bilateral olfactory nerve aplasia, and hypoplastic thymus. A detailed clinical description of the patients with ultrasound, MRI, and post mortem pictures of the affected fetuses showing the wide phenotypic spectrum of the disease is presented.


Subject(s)
Abnormalities, Multiple , Face/abnormalities , Hand Deformities, Congenital , Intellectual Disability , Micrognathism , Neck/abnormalities , Humans , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Intellectual Disability/pathology , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Face/pathology , Phenotype
12.
J Cell Mol Med ; 27(22): 3565-3577, 2023 11.
Article in English | MEDLINE | ID: mdl-37872881

ABSTRACT

Schinzel Giedion Syndrome (SGS) is an ultra-rare autosomal dominant Mendelian disease presenting with abnormalities spanning multiple organ systems. The most notable phenotypes involve severe developmental delay, progressive brain atrophy, and drug-resistant seizures. SGS is caused by spontaneous variants in SETBP1, which encodes for the epigenetic hub SETBP1 transcription factor (TF). SETBP1 variants causing classical SGS cluster at the degron, disrupting SETBP1 protein degradation and resulting in toxic accumulation, while those located outside cause milder atypical SGS. Due to the multisystem phenotype, we evaluated gene expression and regulatory programs altered in atypical SGS by snRNA-seq of the cerebral cortex and kidney of Setbp1S858R heterozygous mice (corresponds to the human likely pathogenic SETBP1S867R variant) compared to matched wild-type mice by constructing cell-type-specific regulatory networks. Setbp1 was differentially expressed in excitatory neurons, but known SETBP1 targets were differentially expressed and regulated in many cell types. Our findings suggest molecular drivers underlying neurodevelopmental phenotypes in classical SGS also drive atypical SGS, persist after birth, and are present in the kidney. Our results indicate SETBP1's role as an epigenetic hub leads to cell-type-specific differences in TF activity, gene targeting, and regulatory rewiring. This research provides a framework for investigating cell-type-specific variant impact on gene expression and regulation.


Subject(s)
Abnormalities, Multiple , Humans , Animals , Mice , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Kidney/pathology , Cerebral Cortex/pathology , Gene Expression
13.
J Biol Chem ; 299(9): 105154, 2023 09.
Article in English | MEDLINE | ID: mdl-37572851

ABSTRACT

Genetic germline variants of PPP2R5D (encoding: phosphoprotein phosphatase 2 regulatory protein 5D) result in PPP2R5D-related disorder (Jordan's Syndrome), which is characterized by intellectual disability, hypotonia, seizures, macrocephaly, autism spectrum disorder, and delayed motor skill development. The disorder originates from de novo single nucleotide mutations, generating missense variants that act in a dominant manner. Pathogenic mutations altering 13 different amino acids have been identified, with the E198K variant accounting for ∼40% of reported cases. However, the generation of a heterozygous E198K variant cell line to study the molecular effects of the pathogenic mutation has been challenging. Here, we use CRISPR-PRIME genomic editing to introduce a transition (c.592G>A) in a single PPP2R5D allele in HEK293 cells, generating E198K-heterozygous lines to complement existing E420K variant lines. We generate global protein and phosphorylation profiles of WT, E198K, and E420K cell lines and find unique and shared changes between variants and WT cells in kinase- and phosphatase-controlled signaling cascades. We observed ribosomal protein S6 (RPS6) hyperphosphorylation as a shared signaling alteration, indicative of increased ribosomal protein S6-kinase activity. Treatment with rapamycin or an RPS6-kinase inhibitor (LY2584702) suppressed RPS6 phosphorylation in both, suggesting upstream activation of mTORC1/p70S6K. Intriguingly, our data suggests ERK-dependent activation of mTORC1 in both E198K and E420K variant cells, with additional AKT-mediated mTORC1 activation in the E420K variant. Thus, although upstream activation of mTORC1 differs between PPP2R5D-related disorder genotypes, inhibition of mTORC1 or RPS6 kinases warrants further investigation as potential therapeutic strategies for patients.


Subject(s)
Abnormalities, Multiple , Humans , Autism Spectrum Disorder , HEK293 Cells , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Phosphorylation , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Proteomics , Ribosomal Protein S6/genetics , Ribosomal Protein S6/metabolism , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology
14.
Rev Med Liege ; 78(7-8): 407-410, 2023 Jul.
Article in French | MEDLINE | ID: mdl-37560950

ABSTRACT

Rhombencephalosynapsis (RS) is a rare cerebellar malformation characterized by the association of partial or total vermis agenesis and fusion of the cerebellar hemispheres. This malformation is often associated with supratentorial cerebral abnormalities and more rarely with extra-cranial abnormalities. The severity of the clinical manifestations and the prognosis of RS depend on the findings of the posterior fossa and the associated anomalies. This clinical case presents an isolated complete RS, documented by antenatal ultrasound, fetal and postnatal/mortem magnetic resonance imaging (MRI) and pathological examination. A RS should be suspected on antenatal morphological ultrasound in case of reduction in size of the posterior fossa and in the absence of vermis. Fetal MRI is the imaging modality of choice to confirm the diagnosis. In the event of medical termination of pregnancy, the autopsy is rarely accepted by the parents, limiting postnatal investigations. This is why postnatal/postmortem MRI can prove to be an alternative to confirm and better document this rare pathology. Finally, multidisciplinary collaboration is essential to make the diagnosis and to ensure the best care.


Le rhombencéphalosynapsis (RS) est une malformation cérébelleuse rare, caractérisée par l'association d'une agénésie partielle ou totale du vermis et d'une fusion des hémisphères cérébelleux. Cette malformation est souvent associée à des anomalies cérébrales supra-tentorielles et, plus rarement, à des anomalies extra-crâniennes. La sévérité des manifestations cliniques et le pronostic du RS dépendent des atteintes de la fosse postérieure et des anomalies associées. Ce cas clinique présente un RS complet isolé, documenté par une échographie anténatale, une imagerie par résonance magnétique (IRM) fœtale et postnatale/mortem ainsi qu'un examen anatomo-pathologique. Un RS doit être suspecté à l'échographie morphologique anténatale en cas de réduction de taille de la fosse postérieure et en l'absence de vermis. L'IRM fœtale est l'examen de choix pour confirmer le diagnostic. En cas d'interruption médicale de grossesse, l'autopsie est rarement acceptée par les parents, ce qui limite les investigations postnatales. C'est pourquoi, l'IRM postnatale/postmortem pourrait s'avérer être très utile pour confirmer et mieux documenter cette rare pathologie. Enfin, la collaboration multidisciplinaire est essentielle, d'une part, pour poser le diagnostic et, d'autre part, pour assurer la meilleure prise en charge possible.


Subject(s)
Abnormalities, Multiple , Cerebellum , Humans , Female , Pregnancy , Cerebellum/diagnostic imaging , Cerebellum/abnormalities , Cerebellum/pathology , Abnormalities, Multiple/pathology , Prenatal Diagnosis , Magnetic Resonance Imaging/methods , Prognosis
15.
Adv Exp Med Biol ; 1415: 173-182, 2023.
Article in English | MEDLINE | ID: mdl-37440031

ABSTRACT

Inherited retinal degenerations (IRDs) are a group of genetic disorders characterized by progressive dysfunction and loss of photoreceptors. IRDs are classified as non-syndromic or syndromic, depending on whether retinal degeneration manifests alone or in combination with other associated symptoms. Joubert syndrome (JBTS) is a genetically and clinically heterogeneous disorder affecting the central nervous system and other organs and tissues, including the neuroretina. To date, 39 genes have been associated with JBTS, a majority of which encode structural or functional components of the primary cilium, a specialized sensory organelle present in most post-mitotic cells, including photoreceptors. The use of whole exome and IRD panel next-generation sequencing in routine diagnostics of non-syndromic IRD cases led to the discovery of pathogenic variants in JBTS genes that cause photoreceptor loss without other syndromic features. Here, we recapitulate these findings, describing the JBTS gene defects leading to non-syndromic IRDs.


Subject(s)
Abnormalities, Multiple , Eye Abnormalities , Kidney Diseases, Cystic , Retinal Degeneration , Humans , Retina/pathology , Cerebellum/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Kidney Diseases, Cystic/genetics , Eye Abnormalities/genetics , Eye Abnormalities/pathology , Mutation , Pedigree
16.
Am J Med Genet C Semin Med Genet ; 193(2): 128-138, 2023 06.
Article in English | MEDLINE | ID: mdl-37296540

ABSTRACT

Kabuki syndrome is a recognizable Mendelian disorder characterized by the clinical constellation of childhood hypotonia, developmental delay or intellectual impairment, and characteristic dysmorphism resulting from monoallelic pathogenic variants in KMT2D or KDM6A. In the medical literature, most reported patients are children, and data is lacking on the natural history of the condition across the lifespan, with little known about adult-specific presentations and symptoms. Here, we report the results of a retrospective chart review of eight adult patients with Kabuki syndrome, seven of whom are molecularly confirmed. We use their trajectories to highlight the diagnostic challenges unique to an adult population, expand on neurodevelopmental/psychiatric phenotypes across the lifespan, and describe adult-onset medical complications, including a potential cancer risk and unusual and striking premature/accelerated aging phenotype.


Subject(s)
Abnormalities, Multiple , Hematologic Diseases , Humans , Retrospective Studies , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Face/pathology , Hematologic Diseases/diagnosis , Hematologic Diseases/genetics , Hematologic Diseases/complications , Phenotype , Mutation
17.
Prenat Diagn ; 43(6): 730-733, 2023 06.
Article in English | MEDLINE | ID: mdl-37160702

ABSTRACT

We report a case of pyruvate dehydrogenase E1 alpha subunit deficiency associated with a novel hemizygous PDHA1 variant presenting prenatally as multiple structural brain abnormalities in a male fetus. A healthy Finnish couple was initially referred to the Fetomaternal Medical Center because of suspected fetal choroid plexus cyst at 11 + 2 weeks of pregnancy. At 20 + 0 weeks, multiple abnormalities were observed with ultrasound including narrow thorax, slightly enlarged heart, hypoplastic cerebellum, absent cerebellar vermis and ventriculomegaly. Autopsy and genetic analyses were performed after the termination of pregnancy. The findings of macroscopic examination included cleft palate, abnormally overlapping position of fingers and toes and dysmorphic facial features. Neuropathological examination confirmed the absence of corpus callosum, cerebellar hypoplasia and ventriculomegaly. Nodular neuronal heterotopia was also observed. Trio exome sequencing revealed a novel hemizygous de novo variant c.1144C>T p.(Gln382*) in the PDHA1 gene, classified as likely pathogenic. We suggest that inherited metabolic disorders should be kept in mind as differential diagnoses in fetuses with structural brain abnormalities.


Subject(s)
Abnormalities, Multiple , Hydrocephalus , Pyruvate Dehydrogenase Complex Deficiency Disease , Pregnancy , Female , Humans , Male , Pyruvate Dehydrogenase Complex Deficiency Disease/genetics , Pyruvate Dehydrogenase Complex Deficiency Disease/pathology , Abnormalities, Multiple/diagnostic imaging , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Cerebellum , Fetus/diagnostic imaging , Fetus/pathology
18.
Ital J Dermatol Venerol ; 158(3): 217-223, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37166753

ABSTRACT

Keratosis pilaris (KP) is a common, hyperkeratotic skin condition characterized by small, folliculocentric papules with variable perifollicular erythema. We provide an updated review on the pathogenesis, clinical manifestations, and management of this common, and often annoying, finding. KP represents a family of follicular disorders, of which KP simplex is by far the most common. Other variants and rare subtypes include keratosis pilaris rubra, erythromelanosis follicularis faciei et colli, and the spectrum of keratosis pilaris atrophicans. Inherited mutations of the FLG gene and ABCA12 gene have been implicated etiologically. KP may be associated with ichthyosis vulgaris and palmar hyperlinearity, but less likely atopic dermatitis. Some potential differential diagnoses for KP include lichen spinulosus, phrynoderma, ichthyosis vulgaris, and trichostasis spinulosa. General cutaneous measures such as hydrating skin, avoiding long baths or showers, and using mild soaps or cleansers should be recommended. Topical keratolytic agents are first-line therapy, followed by topical retinoids and corticosteroids. Recent options include a variety of lasers and microdermabrasion if the patient is refractory to topical therapy.


Subject(s)
Abnormalities, Multiple , Darier Disease , Ichthyosis Vulgaris , Humans , Ichthyosis Vulgaris/pathology , Darier Disease/diagnosis , Darier Disease/genetics , Darier Disease/therapy , Skin , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/pathology
19.
Clin Genet ; 104(2): 245-250, 2023 08.
Article in English | MEDLINE | ID: mdl-37125481

ABSTRACT

Glycosylphosphatidylinositol-anchored proteins are involved in multiple physiological processes and the initial stage of their biosynthesis is mediated by PIGA, PIGC, PIGH, PIGP, PIGQ, PIGY, and DMP2 genes, which have been linked to a wide spectrum of phenotypes depending on the gene damaged. To date, the PIGP gene has only been related to Developmental and Epileptic Encephalopathy 55 (MIM#617599) in just seven patients. A detailed medical history was performed in two affected siblings with a multiple malformation syndrome. Genetic testing was performed using whole-exome sequencing. One patient presented dysmorphic features, congenital anomalies, hypotonia and epileptic encephalopathy as described in PIGA, PIGQ and PIGY deficiencies. The other one was a fetus with a severe malformation disorder at 17 weeks of gestation whose pregnancy was interrupted. Both were compound heterozygous of pathogenic variants in PIGP gene: NM_153682.3:c.2 T > C(p.?) and a 136 Kb deletion (GRCh37/hg19 21q22.13(chr21:38329939-38 466 066)×1) affecting the entire PIGP gene. Our results extend the clinical phenotype associated to PIGP gene and propose to include it as a novel cause of Multiple Congenital Anomalies-Hypotonia-Seizures syndrome.


Subject(s)
Abnormalities, Multiple , Epilepsy, Generalized , Epilepsy , Hexosyltransferases , Musculoskeletal Abnormalities , Humans , Seizures/genetics , Seizures/pathology , Muscle Hypotonia/genetics , Muscle Hypotonia/pathology , Mutation , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Phenotype , Membrane Proteins/genetics , Hexosyltransferases/genetics
20.
Methods Cell Biol ; 175: 235-249, 2023.
Article in English | MEDLINE | ID: mdl-36967143

ABSTRACT

Joubert syndrome (JS) is an autosomal recessive ciliopathy that mainly affects the morphogenesis of the cerebellum and brain stem. To date, mutations in at least 39 genes have been identified in JS; all these gene-encoding proteins are involved in the biogenesis of the primary cilium and centrioles. Recent studies using the mouse model carrying deleted or mutated JS-related genes exhibited cerebellar hypoplasia with a reduction in neurogenesis; however, investigating specific neuronal behaviors during their development in vivo remains challenging. Here, we describe an in vivo cerebellar electroporation technique that can be used to deliver plasmids carrying GFP and/or shRNAs into the major cerebellar cell type, granule neurons, from their progenitor state to their maturation in a spatiotemporal-specific manner. By combining this method with cerebellar immunostaining and EdU incorporation, these approaches enable the investigation of the cell-autonomous effect of JS-related genes in granule neuron progenitors, including the pathogenesis of ectopic neurons and the defects in neuronal differentiation. This approach provides information toward understanding the multifaceted roles of JS-related genes during cerebellar development in vivo.


Subject(s)
Abnormalities, Multiple , Eye Abnormalities , Kidney Diseases, Cystic , Mice , Animals , Cerebellum/metabolism , Cerebellum/pathology , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/metabolism , Kidney Diseases, Cystic/pathology , Eye Abnormalities/genetics , Eye Abnormalities/metabolism , Eye Abnormalities/pathology , Retina , Neurons/metabolism , Cell Differentiation/genetics , Proteins , Cell Proliferation/genetics , Electroporation
SELECTION OF CITATIONS
SEARCH DETAIL
...