Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Nature ; 577(7788): 109-114, 2020 01.
Article in English | MEDLINE | ID: mdl-31827280

ABSTRACT

Activation of RIPK1 controls TNF-mediated apoptosis, necroptosis and inflammatory pathways1. Cleavage of human and mouse RIPK1 after residues D324 and D325, respectively, by caspase-8 separates the RIPK1 kinase domain from the intermediate and death domains. The D325A mutation in mouse RIPK1 leads to embryonic lethality during mouse development2,3. However, the functional importance of blocking caspase-8-mediated cleavage of RIPK1 on RIPK1 activation in humans is unknown. Here we identify two families with variants in RIPK1 (D324V and D324H) that lead to distinct symptoms of recurrent fevers and lymphadenopathy in an autosomal-dominant manner. Impaired cleavage of RIPK1 D324 variants by caspase-8 sensitized patients' peripheral blood mononuclear cells to RIPK1 activation, apoptosis and necroptosis induced by TNF. The patients showed strong RIPK1-dependent activation of inflammatory signalling pathways and overproduction of inflammatory cytokines and chemokines compared with unaffected controls. Furthermore, we show that expression of the RIPK1 mutants D325V or D325H in mouse embryonic fibroblasts confers not only increased sensitivity to RIPK1 activation-mediated apoptosis and necroptosis, but also induction of pro-inflammatory cytokines such as IL-6 and TNF. By contrast, patient-derived fibroblasts showed reduced expression of RIPK1 and downregulated production of reactive oxygen species, resulting in resistance to necroptosis and ferroptosis. Together, these data suggest that human non-cleavable RIPK1 variants promote activation of RIPK1, and lead to an autoinflammatory disease characterized by hypersensitivity to apoptosis and necroptosis and increased inflammatory response in peripheral blood mononuclear cells, as well as a compensatory mechanism to protect against several pro-death stimuli in fibroblasts.


Subject(s)
Caspase 8/metabolism , Hereditary Autoinflammatory Diseases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Adolescent , Adult , Amino Acid Sequence , Animals , Base Sequence , Child , Child, Preschool , Female , HEK293 Cells , Hereditary Autoinflammatory Diseases/genetics , Hereditary Autoinflammatory Diseases/pathology , Humans , Male , Mice , Mice, Knockout , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Sequence Alignment , Sequence Homology, Amino Acid
2.
Am J Hum Genet ; 108(3): 502-516, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33596411

ABSTRACT

Deletion 1p36 (del1p36) syndrome is the most common human disorder resulting from a terminal autosomal deletion. This condition is molecularly and clinically heterogeneous. Deletions involving two non-overlapping regions, known as the distal (telomeric) and proximal (centromeric) critical regions, are sufficient to cause the majority of the recurrent clinical features, although with different facial features and dysmorphisms. SPEN encodes a transcriptional repressor commonly deleted in proximal del1p36 syndrome and is located centromeric to the proximal 1p36 critical region. Here, we used clinical data from 34 individuals with truncating variants in SPEN to define a neurodevelopmental disorder presenting with features that overlap considerably with those of proximal del1p36 syndrome. The clinical profile of this disease includes developmental delay/intellectual disability, autism spectrum disorder, anxiety, aggressive behavior, attention deficit disorder, hypotonia, brain and spine anomalies, congenital heart defects, high/narrow palate, facial dysmorphisms, and obesity/increased BMI, especially in females. SPEN also emerges as a relevant gene for del1p36 syndrome by co-expression analyses. Finally, we show that haploinsufficiency of SPEN is associated with a distinctive DNA methylation episignature of the X chromosome in affected females, providing further evidence of a specific contribution of the protein to the epigenetic control of this chromosome, and a paradigm of an X chromosome-specific episignature that classifies syndromic traits. We conclude that SPEN is required for multiple developmental processes and SPEN haploinsufficiency is a major contributor to a disorder associated with deletions centromeric to the previously established 1p36 critical regions.


Subject(s)
Chromosome Disorders/genetics , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, X/genetics , DNA-Binding Proteins/genetics , RNA-Binding Proteins/genetics , Adolescent , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Child , Child, Preschool , Chromosome Deletion , Chromosome Disorders/physiopathology , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Female , Haploinsufficiency/genetics , Humans , Intellectual Disability/genetics , Intellectual Disability/physiopathology , Male , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/physiopathology , Phenotype , Young Adult
3.
Am J Med Genet A ; 185(8): 2417-2433, 2021 08.
Article in English | MEDLINE | ID: mdl-34042254

ABSTRACT

Biallelic loss-of-function variants in the thrombospondin-type laminin G domain and epilepsy-associated repeats (TSPEAR) gene have recently been associated with ectodermal dysplasia and hearing loss. The first reports describing a TSPEAR disease association identified this gene is a cause of nonsyndromic hearing loss, but subsequent reports involving additional affected families have questioned this evidence and suggested a stronger association with ectodermal dysplasia. To clarify genotype-phenotype associations for TSPEAR variants, we characterized 13 individuals with biallelic TSPEAR variants. Individuals underwent either exome sequencing or panel-based genetic testing. Nearly all of these newly reported individuals (11/13) have phenotypes that include tooth agenesis or ectodermal dysplasia, while three newly reported individuals have hearing loss. Of the individuals displaying hearing loss, all have additional variants in other hearing-loss-associated genes, specifically TMPRSS3, GJB2, and GJB6, that present competing candidates for their hearing loss phenotype. When presented alongside previous reports, the overall evidence supports the association of TSPEAR variants with ectodermal dysplasia and tooth agenesis features but creates significant doubt as to whether TSPEAR variants are a monogenic cause of hearing loss. Further functional evidence is needed to evaluate this phenotypic association.


Subject(s)
Anodontia/diagnosis , Anodontia/genetics , Ectodermal Dysplasia/diagnosis , Ectodermal Dysplasia/genetics , Genetic Variation , Phenotype , Proteins/genetics , Alleles , Amino Acid Substitution , Cohort Studies , Female , Genetic Association Studies , Genetic Loci , Humans , Male , Mutation , Pedigree , Radiography
4.
Brain ; 143(1): 55-68, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31834374

ABSTRACT

MN1 encodes a transcriptional co-regulator without homology to other proteins, previously implicated in acute myeloid leukaemia and development of the palate. Large deletions encompassing MN1 have been reported in individuals with variable neurodevelopmental anomalies and non-specific facial features. We identified a cluster of de novo truncating mutations in MN1 in a cohort of 23 individuals with strikingly similar dysmorphic facial features, especially midface hypoplasia, and intellectual disability with severe expressive language delay. Imaging revealed an atypical form of rhombencephalosynapsis, a distinctive brain malformation characterized by partial or complete loss of the cerebellar vermis with fusion of the cerebellar hemispheres, in 8/10 individuals. Rhombencephalosynapsis has no previously known definitive genetic or environmental causes. Other frequent features included perisylvian polymicrogyria, abnormal posterior clinoid processes and persistent trigeminal artery. MN1 is encoded by only two exons. All mutations, including the recurrent variant p.Arg1295* observed in 8/21 probands, fall in the terminal exon or the extreme 3' region of exon 1, and are therefore predicted to result in escape from nonsense-mediated mRNA decay. This was confirmed in fibroblasts from three individuals. We propose that the condition described here, MN1 C-terminal truncation (MCTT) syndrome, is not due to MN1 haploinsufficiency but rather is the result of dominantly acting C-terminally truncated MN1 protein. Our data show that MN1 plays a critical role in human craniofacial and brain development, and opens the door to understanding the biological mechanisms underlying rhombencephalosynapsis.


Subject(s)
Abnormalities, Multiple/genetics , Craniofacial Abnormalities/genetics , Intellectual Disability/genetics , Language Development Disorders/genetics , Nervous System Malformations/genetics , Trans-Activators/genetics , Tumor Suppressor Proteins/genetics , Abnormalities, Multiple/diagnostic imaging , Adolescent , Basilar Artery/abnormalities , Basilar Artery/diagnostic imaging , Carotid Arteries/abnormalities , Carotid Arteries/diagnostic imaging , Cerebellar Vermis/abnormalities , Cerebellar Vermis/diagnostic imaging , Cerebellum/abnormalities , Cerebellum/diagnostic imaging , Child , Child, Preschool , Cohort Studies , Comparative Genomic Hybridization , Craniofacial Abnormalities/diagnostic imaging , Female , Fibroblasts/metabolism , Humans , Imaging, Three-Dimensional , Infant , Magnetic Resonance Imaging , Male , Middle Aged , Mutation , Nervous System Malformations/diagnostic imaging , Nonsense Mediated mRNA Decay , Polymicrogyria/diagnostic imaging , Polymicrogyria/genetics , RNA-Seq , Real-Time Polymerase Chain Reaction , Syndrome , Tomography, X-Ray Computed , Exome Sequencing , Whole Genome Sequencing
5.
Nat Commun ; 11(1): 3698, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32703943

ABSTRACT

Intellectual disability (ID) is a heterogeneous clinical entity and includes an excess of males who harbor variants on the X-chromosome (XLID). We report rare FAM50A missense variants in the original Armfield XLID syndrome family localized in Xq28 and four additional unrelated males with overlapping features. Our fam50a knockout (KO) zebrafish model exhibits abnormal neurogenesis and craniofacial patterning, and in vivo complementation assays indicate that the patient-derived variants are hypomorphic. RNA sequencing analysis from fam50a KO zebrafish show dysregulation of the transcriptome, with augmented spliceosome mRNAs and depletion of transcripts involved in neurodevelopment. Zebrafish RNA-seq datasets show a preponderance of 3' alternative splicing events in fam50a KO, suggesting a role in the spliceosome C complex. These data are supported with transcriptomic signatures from cell lines derived from affected individuals and FAM50A protein-protein interaction data. In sum, Armfield XLID syndrome is a spliceosomopathy associated with aberrant mRNA processing during development.


Subject(s)
DNA-Binding Proteins/genetics , Intellectual Disability/genetics , Mental Retardation, X-Linked/genetics , Mutation/genetics , RNA-Binding Proteins/genetics , Spliceosomes/metabolism , Zebrafish Proteins/genetics , Adult , Animals , Cell Nucleus/metabolism , Child , Child, Preschool , DNA-Binding Proteins/metabolism , Family , Female , Gene Expression Regulation, Developmental , Humans , Male , Mice , Mutation, Missense/genetics , NIH 3T3 Cells , Pedigree , Phenotype , Protein Transport , RNA Splicing/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Nuclear/genetics , RNA-Binding Proteins/metabolism , Syndrome , Zebrafish/genetics , Zebrafish Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL