Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
2.
Mol Vis ; 28: 57-69, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35693420

RESUMEN

Purpose: To investigate the molecular basis of recessively inherited congenital cataract, microcornea, and corneal opacification with or without coloboma and microphthalmia in two consanguineous families. Methods: Conventional autozygosity mapping was performed using single nucleotide polymorphism (SNP) microarrays. Whole-exome sequencing was completed on genomic DNA from one affected member of each family. Exome sequence data were also used for homozygosity mapping and copy number variation analysis. PCR and Sanger sequencing were used to confirm the identification of mutations and to screen further patients. Evolutionary conservation of protein sequences was assessed using CLUSTALW, and protein structures were modeled using PyMol. Results: In family MEP68, a novel homozygous nucleotide substitution in SIX6 was found, c.547G>C, that converts the evolutionarily conserved aspartic acid residue at the 183rd amino acid in the protein to a histidine, p.(Asp183His). This residue mapped to the third helix of the DNA-binding homeobox domain in SIX6, which interacts with the major groove of double-stranded DNA. This interaction is likely to be disrupted by the mutation. In family F1332, a novel homozygous 1034 bp deletion that encompasses the first exon of SIX6 was identified, chr14:g.60975890_60976923del. Both mutations segregated with the disease phenotype as expected for a recessive condition and were absent from publicly available variant databases. Conclusions: Our findings expand the mutation spectrum in this form of inherited eye disease and confirm that homozygous human SIX6 mutations cause a developmental spectrum of ocular phenotypes that includes not only the previously described features of microphthalmia, coloboma, and congenital cataract but also corneal abnormalities.


Asunto(s)
Catarata , Coloboma , Enfermedades de la Córnea , Anomalías del Ojo , Microftalmía , Catarata/congénito , Catarata/genética , Coloboma/genética , Enfermedades de la Córnea/genética , ADN/genética , Variaciones en el Número de Copia de ADN , Análisis Mutacional de ADN , Anomalías del Ojo/genética , Proteínas de Homeodominio/genética , Humanos , Microftalmía/genética , Mutación , Linaje , Fenotipo , Transactivadores/genética
3.
Elife ; 112022 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-35170427

RESUMEN

Primary ciliary defects cause a group of developmental conditions known as ciliopathies. Here, we provide mechanistic insight into ciliary ubiquitin processing in cells and for mouse model lacking the ciliary protein Mks1. In vivo loss of Mks1 sensitises cells to proteasomal disruption, leading to abnormal accumulation of ubiquitinated proteins. We identified UBE2E1, an E2 ubiquitin-conjugating enzyme that polyubiquitinates ß-catenin, and RNF34, an E3 ligase, as novel interactants of MKS1. UBE2E1 and MKS1 colocalised, and loss of UBE2E1 recapitulates the ciliary and Wnt signalling phenotypes observed during loss of MKS1. Levels of UBE2E1 and MKS1 are co-dependent and UBE2E1 mediates both regulatory and degradative ubiquitination of MKS1. We demonstrate that processing of phosphorylated ß-catenin occurs at the ciliary base through the functional interaction between UBE2E1 and MKS1. These observations suggest that correct ß-catenin levels are tightly regulated at the primary cilium by a ciliary-specific E2 (UBE2E1) and a regulatory substrate-adaptor (MKS1).


Asunto(s)
Ciliopatías/metabolismo , Proteínas/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Vía de Señalización Wnt , Animales , Cilios/metabolismo , Humanos , Ratones , Ratones Noqueados , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación , beta Catenina/metabolismo
4.
Neuron ; 109(2): 241-256.e9, 2021 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-33220177

RESUMEN

Autosomal-recessive cerebellar hypoplasia and ataxia constitute a group of heterogeneous brain disorders caused by disruption of several fundamental cellular processes. Here, we identified 10 families showing a neurodegenerative condition involving pontocerebellar hypoplasia with microcephaly (PCHM). Patients harbored biallelic mutations in genes encoding the spliceosome components Peptidyl-Prolyl Isomerase Like-1 (PPIL1) or Pre-RNA Processing-17 (PRP17). Mouse knockouts of either gene were lethal in early embryogenesis, whereas PPIL1 patient mutation knockin mice showed neuron-specific apoptosis. Loss of either protein affected splicing integrity, predominantly affecting short and high GC-content introns and genes involved in brain disorders. PPIL1 and PRP17 form an active isomerase-substrate interaction, but we found that isomerase activity is not critical for function. Thus, we establish disrupted splicing integrity and "major spliceosome-opathies" as a new mechanism underlying PCHM and neurodegeneration and uncover a non-enzymatic function of a spliceosomal proline isomerase.


Asunto(s)
Proteínas de Ciclo Celular/genética , Enfermedades Cerebelosas/genética , Microcefalia/genética , Mutación/genética , Isomerasa de Peptidilprolil/genética , Factores de Empalme de ARN/genética , Empalmosomas/genética , Secuencia de Aminoácidos , Animales , Proteínas de Ciclo Celular/química , Enfermedades Cerebelosas/complicaciones , Enfermedades Cerebelosas/diagnóstico por imagen , Estudios de Cohortes , Femenino , Técnicas de Inactivación de Genes/métodos , Células HEK293 , Trastornos Heredodegenerativos del Sistema Nervioso/complicaciones , Trastornos Heredodegenerativos del Sistema Nervioso/diagnóstico por imagen , Trastornos Heredodegenerativos del Sistema Nervioso/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microcefalia/complicaciones , Microcefalia/diagnóstico por imagen , Linaje , Isomerasa de Peptidilprolil/química , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Factores de Empalme de ARN/química
5.
Hum Mutat ; 40(8): 1063-1070, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31045292

RESUMEN

Microcephalic primordial dwarfism (MPD) is a group of rare single-gene disorders characterized by the extreme reduction in brain and body size from early development onwards. Proteins encoded by MPD-associated genes play important roles in fundamental cellular processes, notably genome replication and repair. Here we report the identification of four MPD individuals with biallelic variants in DNA2, which encodes an adenosine triphosphate (ATP)-dependent helicase/nuclease involved in DNA replication and repair. We demonstrate that the two intronic variants (c.1764-38_1764-37ins(53) and c.74+4A>C) found in these individuals substantially impair DNA2 transcript splicing. Additionally, we identify a missense variant (c.1963A>G), affecting a residue of the ATP-dependent helicase domain that is highly conserved between humans and yeast, with the resulting substitution (p.Thr655Ala) predicted to directly impact ATP/ADP (adenosine diphosphate) binding by DNA2. Our findings support the pathogenicity of these variants as biallelic hypomorphic mutations, establishing DNA2 as an MPD disease gene.


Asunto(s)
ADN Helicasas/genética , Enanismo/genética , Variación Genética , Microcefalia/genética , Adolescente , Alelos , ADN Helicasas/química , Femenino , Predisposición Genética a la Enfermedad , Humanos , Intrones , Masculino , Persona de Mediana Edad , Modelos Moleculares , Mutagénesis Insercional , Mutación Missense , Polimorfismo de Nucleótido Simple
6.
Nat Genet ; 51(1): 96-105, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30478443

RESUMEN

DNA methylation and Polycomb are key factors in the establishment of vertebrate cellular identity and fate. Here we report de novo missense mutations in DNMT3A, which encodes the DNA methyltransferase DNMT3A. These mutations cause microcephalic dwarfism, a hypocellular disorder of extreme global growth failure. Substitutions in the PWWP domain abrogate binding to the histone modifications H3K36me2 and H3K36me3, and alter DNA methylation in patient cells. Polycomb-associated DNA methylation valleys, hypomethylated domains encompassing developmental genes, become methylated with concomitant depletion of H3K27me3 and H3K4me3 bivalent marks. Such de novo DNA methylation occurs during differentiation of Dnmt3aW326R pluripotent cells in vitro, and is also evident in Dnmt3aW326R/+ dwarf mice. We therefore propose that the interaction of the DNMT3A PWWP domain with H3K36me2 and H3K36me3 normally limits DNA methylation of Polycomb-marked regions. Our findings implicate the interplay between DNA methylation and Polycomb at key developmental regulators as a determinant of organism size in mammals.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN/genética , Enanismo/genética , Mutación con Ganancia de Función/genética , Microcefalia/genética , Proteínas del Grupo Polycomb/genética , Animales , Línea Celular Tumoral , Células Cultivadas , ADN Metiltransferasa 3A , Metilasas de Modificación del ADN/genética , Femenino , Células HeLa , Histonas/genética , Humanos , Masculino , Ratones , Ratones Transgénicos/genética , Unión Proteica/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética
7.
Am J Hum Genet ; 103(6): 1038-1044, 2018 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-30503519

RESUMEN

During genome replication, polymerase epsilon (Pol ε) acts as the major leading-strand DNA polymerase. Here we report the identification of biallelic mutations in POLE, encoding the Pol ε catalytic subunit POLE1, in 15 individuals from 12 families. Phenotypically, these individuals had clinical features closely resembling IMAGe syndrome (intrauterine growth restriction [IUGR], metaphyseal dysplasia, adrenal hypoplasia congenita, and genitourinary anomalies in males), a disorder previously associated with gain-of-function mutations in CDKN1C. POLE1-deficient individuals also exhibited distinctive facial features and variable immune dysfunction with evidence of lymphocyte deficiency. All subjects shared the same intronic variant (c.1686+32C>G) as part of a common haplotype, in combination with different loss-of-function variants in trans. The intronic variant alters splicing, and together the biallelic mutations lead to cellular deficiency of Pol ε and delayed S-phase progression. In summary, we establish POLE as a second gene in which mutations cause IMAGe syndrome. These findings add to a growing list of disorders due to mutations in DNA replication genes that manifest growth restriction alongside adrenal dysfunction and/or immunodeficiency, consolidating these as replisome phenotypes and highlighting a need for future studies to understand the tissue-specific development roles of the encoded proteins.


Asunto(s)
Insuficiencia Suprarrenal/genética , ADN Polimerasa II/genética , Retardo del Crecimiento Fetal/genética , Mutación/genética , Osteocondrodisplasias/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Anomalías Urogenitales/genética , Adolescente , Adulto , Alelos , Niño , Preescolar , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Replicación del ADN/genética , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Fenotipo , Adulto Joven
8.
Am J Hum Genet ; 103(4): 553-567, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30290151

RESUMEN

The conserved oligomeric Golgi (COG) complex is involved in intracellular vesicular transport, and is composed of eight subunits distributed in two lobes, lobe A (COG1-4) and lobe B (COG5-8). We describe fourteen individuals with Saul-Wilson syndrome, a rare form of primordial dwarfism with characteristic facial and radiographic features. All affected subjects harbored heterozygous de novo variants in COG4, giving rise to the same recurrent amino acid substitution (p.Gly516Arg). Affected individuals' fibroblasts, whose COG4 mRNA and protein were not decreased, exhibited delayed anterograde vesicular trafficking from the ER to the Golgi and accelerated retrograde vesicular recycling from the Golgi to the ER. This altered steady-state equilibrium led to a decrease in Golgi volume, as well as morphologic abnormalities with collapse of the Golgi stacks. Despite these abnormalities of the Golgi apparatus, protein glycosylation in sera and fibroblasts from affected subjects was not notably altered, but decorin, a proteoglycan secreted into the extracellular matrix, showed altered Golgi-dependent glycosylation. In summary, we define a specific heterozygous COG4 substitution as the molecular basis of Saul-Wilson syndrome, a rare skeletal dysplasia distinct from biallelic COG4-CDG.


Asunto(s)
Síndrome del Cromosoma X Frágil/genética , Transporte de Proteínas/genética , Proteoglicanos/genética , Proteínas de Transporte Vesicular/genética , Adulto , Sustitución de Aminoácidos/genética , Animales , Animales Modificados Genéticamente/genética , Línea Celular , Niño , Preescolar , Retículo Endoplásmico/genética , Matriz Extracelular/genética , Femenino , Fibroblastos/patología , Glicosilación , Aparato de Golgi/genética , Heterocigoto , Humanos , Lactante , Masculino , Pez Cebra
10.
Am J Hum Genet ; 103(2): 221-231, 2018 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-30057030

RESUMEN

Bloom syndrome, caused by biallelic mutations in BLM, is characterized by prenatal-onset growth deficiency, short stature, an erythematous photosensitive malar rash, and increased cancer predisposition. Diagnostically, a hallmark feature is the presence of increased sister chromatid exchanges (SCEs) on cytogenetic testing. Here, we describe biallelic mutations in TOP3A in ten individuals with prenatal-onset growth restriction and microcephaly. TOP3A encodes topoisomerase III alpha (TopIIIα), which binds to BLM as part of the BTRR complex, and promotes dissolution of double Holliday junctions arising during homologous recombination. We also identify a homozygous truncating variant in RMI1, which encodes another component of the BTRR complex, in two individuals with microcephalic dwarfism. The TOP3A mutations substantially reduce cellular levels of TopIIIα, and consequently subjects' cells demonstrate elevated rates of SCE. Unresolved DNA recombination and/or replication intermediates persist into mitosis, leading to chromosome segregation defects and genome instability that most likely explain the growth restriction seen in these subjects and in Bloom syndrome. Clinical features of mitochondrial dysfunction are evident in several individuals with biallelic TOP3A mutations, consistent with the recently reported additional function of TopIIIα in mitochondrial DNA decatenation. In summary, our findings establish TOP3A mutations as an additional cause of prenatal-onset short stature with increased cytogenetic SCEs and implicate the decatenation activity of the BTRR complex in their pathogenesis.

11.
Am J Med Genet A ; 176(2): 465-469, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29265708

RESUMEN

RNU4ATAC pathogenic variants to date have been associated with microcephalic osteodysplastic primordial dwarfism, type 1 and Roifman syndrome. Both conditions are clinically distinct skeletal dysplasias with microcephalic osteodysplastic primordial dwarfism, type 1 having a more severe phenotype than Roifman syndrome. Some of the overlapping features of the two conditions include developmental delay, microcephaly, and immune deficiency. The features also overlap with Lowry Wood syndrome, another rare but well-defined skeletal dysplasia for which the genetic etiology has not been identified. Characteristic features include multiple epiphyseal dysplasia and microcephaly. Here, we describe three patients with Lowry Wood syndrome with biallelic RNU4ATAC pathogenic variants. This report expands the phenotypic spectrum for biallelic RNU4ATAC disorder causing variants and is the first to establish the genetic cause for Lowry Wood syndrome.


Asunto(s)
Cardiomiopatías/genética , Enanismo/genética , Trastornos del Crecimiento/genética , Síndromes de Inmunodeficiencia/genética , Discapacidad Intelectual/genética , Discapacidad Intelectual Ligada al Cromosoma X/genética , Microcefalia/genética , Osteocondrodisplasias/genética , ARN Nuclear Pequeño/genética , Enfermedades de la Retina/genética , Adolescente , Cardiomiopatías/fisiopatología , Preescolar , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/fisiopatología , Enanismo/fisiopatología , Femenino , Retardo del Crecimiento Fetal/genética , Retardo del Crecimiento Fetal/fisiopatología , Trastornos del Crecimiento/fisiopatología , Humanos , Síndromes de Inmunodeficiencia/fisiopatología , Discapacidad Intelectual/fisiopatología , Masculino , Discapacidad Intelectual Ligada al Cromosoma X/fisiopatología , Microcefalia/fisiopatología , Mutación , Osteocondrodisplasias/fisiopatología , Fenotipo , Enfermedades de Inmunodeficiencia Primaria , Enfermedades de la Retina/fisiopatología
12.
Am J Hum Genet ; 101(5): 856-865, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-29100095

RESUMEN

Approximately one in every 200 mammalian proteins is anchored to the cell membrane through a glycosylphosphatidylinositol (GPI) anchor. These proteins play important roles notably in neurological development and function. To date, more than 20 genes have been implicated in the biogenesis of GPI-anchored proteins. GPAA1 (glycosylphosphatidylinositol anchor attachment 1) is an essential component of the transamidase complex along with PIGK, PIGS, PIGT, and PIGU (phosphatidylinositol-glycan biosynthesis classes K, S, T, and U, respectively). This complex orchestrates the attachment of the GPI anchor to the C terminus of precursor proteins in the endoplasmic reticulum. Here, we report bi-allelic mutations in GPAA1 in ten individuals from five families. Using whole-exome sequencing, we identified two frameshift mutations (c.981_993del [p.Gln327Hisfs∗102] and c.920delG [p.Gly307Alafs∗11]), one intronic splicing mutation (c.1164+5C>T), and six missense mutations (c.152C>T [p.Ser51Leu], c.160_161delinsAA [p.Ala54Asn], c.527G>C [p.Trp176Ser], c.869T>C [p.Leu290Pro], c.872T>C [p.Leu291Pro], and c.1165G>C [p.Ala389Pro]). Most individuals presented with global developmental delay, hypotonia, early-onset seizures, cerebellar atrophy, and osteopenia. The splicing mutation was found to decrease GPAA1 mRNA. Moreover, flow-cytometry analysis of five available individual samples showed that several GPI-anchored proteins had decreased cell-surface abundance in leukocytes (FLAER, CD16, and CD59) or fibroblasts (CD73 and CD109). Transduction of fibroblasts with a lentivirus encoding the wild-type protein partially rescued the deficiency of GPI-anchored proteins. These findings highlight the role of the transamidase complex in the development and function of the cerebellum and the skeletal system.


Asunto(s)
Aciltransferasas/genética , Atrofia/genética , Enfermedades Óseas Metabólicas/genética , Discapacidades del Desarrollo/genética , Epilepsia/genética , Glicoproteínas de Membrana/genética , Mutación/genética , Adolescente , Adulto , Alelos , Cerebelo/patología , Niño , Preescolar , Exoma/genética , Femenino , Fibroblastos/patología , Glicosilfosfatidilinositoles/genética , Humanos , Masculino , Hipotonía Muscular/genética , Linaje , ARN Mensajero/genética , Convulsiones/genética
13.
Nat Genet ; 49(4): 537-549, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28191891

RESUMEN

To ensure efficient genome duplication, cells have evolved numerous factors that promote unperturbed DNA replication and protect, repair and restart damaged forks. Here we identify downstream neighbor of SON (DONSON) as a novel fork protection factor and report biallelic DONSON mutations in 29 individuals with microcephalic dwarfism. We demonstrate that DONSON is a replisome component that stabilizes forks during genome replication. Loss of DONSON leads to severe replication-associated DNA damage arising from nucleolytic cleavage of stalled replication forks. Furthermore, ATM- and Rad3-related (ATR)-dependent signaling in response to replication stress is impaired in DONSON-deficient cells, resulting in decreased checkpoint activity and the potentiation of chromosomal instability. Hypomorphic mutations in DONSON substantially reduce DONSON protein levels and impair fork stability in cells from patients, consistent with defective DNA replication underlying the disease phenotype. In summary, we have identified mutations in DONSON as a common cause of microcephalic dwarfism and established DONSON as a critical replication fork protein required for mammalian DNA replication and genome stability.


Asunto(s)
Replicación del ADN/genética , Proteínas de Unión al ADN/genética , Enanismo/genética , Inestabilidad Genómica/genética , Microcefalia/genética , Mutación/genética , Línea Celular , Daño del ADN/genética , Femenino , Humanos , Masculino
14.
Genome Biol ; 17(1): 242, 2016 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-27894351

RESUMEN

BACKGROUND: Ciliopathies are clinically diverse disorders of the primary cilium. Remarkable progress has been made in understanding the molecular basis of these genetically heterogeneous conditions; however, our knowledge of their morbid genome, pleiotropy, and variable expressivity remains incomplete. RESULTS: We applied genomic approaches on a large patient cohort of 371 affected individuals from 265 families, with phenotypes that span the entire ciliopathy spectrum. Likely causal mutations in previously described ciliopathy genes were identified in 85% (225/265) of the families, adding 32 novel alleles. Consistent with a fully penetrant model for these genes, we found no significant difference in their "mutation load" beyond the causal variants between our ciliopathy cohort and a control non-ciliopathy cohort. Genomic analysis of our cohort further identified mutations in a novel morbid gene TXNDC15, encoding a thiol isomerase, based on independent loss of function mutations in individuals with a consistent ciliopathy phenotype (Meckel-Gruber syndrome) and a functional effect of its deficiency on ciliary signaling. Our study also highlighted seven novel candidate genes (TRAPPC3, EXOC3L2, FAM98C, C17orf61, LRRCC1, NEK4, and CELSR2) some of which have established links to ciliogenesis. Finally, we show that the morbid genome of ciliopathies encompasses many founder mutations, the combined carrier frequency of which accounts for a high disease burden in the study population. CONCLUSIONS: Our study increases our understanding of the morbid genome of ciliopathies. We also provide the strongest evidence, to date, in support of the classical Mendelian inheritance of Bardet-Biedl syndrome and other ciliopathies.


Asunto(s)
Cilios/genética , Trastornos de la Motilidad Ciliar/genética , Ciliopatías/genética , Encefalocele/genética , Mutación/genética , Enfermedades Renales Poliquísticas/genética , Alelos , Cilios/patología , Trastornos de la Motilidad Ciliar/patología , Ciliopatías/patología , Análisis Mutacional de ADN , Encefalocele/patología , Estudios de Asociación Genética , Heterogeneidad Genética , Predisposición Genética a la Enfermedad , Humanos , Fenotipo , Enfermedades Renales Poliquísticas/patología , Retina/metabolismo , Retina/patología , Retinitis Pigmentosa
15.
Am J Hum Genet ; 99(4): 984-990, 2016 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-27693231

RESUMEN

Amelogenesis is the process of dental enamel formation, leading to the deposition of the hardest tissue in the human body. This process requires the intricate regulation of ion transport and controlled changes to the pH of the developing enamel matrix. The means by which the enamel organ regulates pH during amelogenesis is largely unknown. We identified rare homozygous variants in GPR68 in three families with amelogenesis imperfecta, a genetically and phenotypically heterogeneous group of inherited conditions associated with abnormal enamel formation. Each of these homozygous variants (a large in-frame deletion, a frameshift deletion, and a missense variant) were predicted to result in loss of function. GPR68 encodes a proton-sensing G-protein-coupled receptor with sensitivity in the pH range that occurs in the developing enamel matrix during amelogenesis. Immunohistochemistry of rat mandibles confirmed localization of GPR68 in the enamel organ at all stages of amelogenesis. Our data identify a role for GPR68 as a proton sensor that is required for proper enamel formation.


Asunto(s)
Amelogénesis Imperfecta/genética , Mutación , Receptores Acoplados a Proteínas G/genética , Amelogénesis/genética , Animales , Secuencia de Bases , Esmalte Dental/crecimiento & desarrollo , Esmalte Dental/patología , Femenino , Homocigoto , Humanos , Concentración de Iones de Hidrógeno , Masculino , Linaje , Ratas , Receptores Acoplados a Proteínas G/análisis
16.
Am J Hum Genet ; 98(4): 615-26, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-26996948

RESUMEN

Glycosylphosphatidylinositol (GPI) is a glycolipid that anchors >150 various proteins to the cell surface. At least 27 genes are involved in biosynthesis and transport of GPI-anchored proteins (GPI-APs). To date, mutations in 13 of these genes are known to cause inherited GPI deficiencies (IGDs), and all are inherited as recessive traits. IGDs mainly manifest as intellectual disability, epilepsy, coarse facial features, and multiple organ anomalies. These symptoms are caused by the decreased surface expression of GPI-APs or by structural abnormalities of GPI. Here, we present five affected individuals (from two consanguineous families from Egypt and Pakistan and one non-consanguineous family from Japan) who show intellectual disability, hypotonia, and early-onset seizures. We identified pathogenic variants in PIGG, a gene in the GPI pathway. In the consanguineous families, homozygous variants c.928C>T (p.Gln310(∗)) and c.2261+1G>C were found, whereas the Japanese individual was compound heterozygous for c.2005C>T (p.Arg669Cys) and a 2.4 Mb deletion involving PIGG. PIGG is the enzyme that modifies the second mannose with ethanolamine phosphate, which is removed soon after GPI is attached to the protein. Physiological significance of this transient modification has been unclear. Using B lymphoblasts from affected individuals of the Egyptian and Japanese families, we revealed that PIGG activity was almost completely abolished; however, the GPI-APs had normal surface levels and normal structure, indicating that the pathogenesis of PIGG deficiency is not yet fully understood. The discovery of pathogenic variants in PIGG expands the spectrum of IGDs and further enhances our understanding of this etiopathogenic class of intellectual disability.


Asunto(s)
Variación Genética , Glicosilfosfatidilinositoles/genética , Discapacidad Intelectual/genética , Manosiltransferasas/genética , Hipotonía Muscular/genética , Convulsiones/genética , Anomalías Múltiples/genética , Adolescente , Línea Celular Tumoral , Niño , Consanguinidad , Egipto , Técnicas de Genotipaje , Glicosilfosfatidilinositoles/metabolismo , Células HEK293 , Heterocigoto , Homocigoto , Humanos , Lactante , Japón , Mutación , Pakistán , Linaje , Adulto Joven
18.
Am J Hum Genet ; 97(6): 878-85, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26626625

RESUMEN

The neuromuscular junction (NMJ) consists of a tripartite synapse with a presynaptic nerve terminal, Schwann cells that ensheathe the terminal bouton, and a highly specialized postsynaptic membrane. Synaptic structural integrity is crucial for efficient signal transmission. Congenital myasthenic syndromes (CMSs) are a heterogeneous group of inherited disorders that result from impaired neuromuscular transmission, caused by mutations in genes encoding proteins that are involved in synaptic transmission and in forming and maintaining the structural integrity of NMJs. To identify further causes of CMSs, we performed whole-exome sequencing (WES) in families without an identified mutation in known CMS-associated genes. In two families affected by a previously undefined CMS, we identified homozygous loss-of-function mutations in COL13A1, which encodes the alpha chain of an atypical non-fibrillar collagen with a single transmembrane domain. COL13A1 localized to the human muscle motor endplate. Using CRISPR-Cas9 genome editing, modeling of the COL13A1 c.1171delG (p.Leu392Sfs(∗)71) frameshift mutation in the C2C12 cell line reduced acetylcholine receptor (AChR) clustering during myotube differentiation. This highlights the crucial role of collagen XIII in the formation and maintenance of the NMJ. Our results therefore delineate a myasthenic disorder that is caused by loss-of-function mutations in COL13A1, encoding a protein involved in organization of the NMJ, and emphasize the importance of appropriate symptomatic treatment for these individuals.


Asunto(s)
Colágeno Tipo XIII/genética , Mutación , Síndromes Miasténicos Congénitos/genética , Mioblastos/metabolismo , Unión Neuromuscular/metabolismo , Adulto , Animales , Línea Celular , Preescolar , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Colágeno Tipo XIII/metabolismo , Endonucleasas/genética , Endonucleasas/metabolismo , Exoma , Femenino , Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Homocigoto , Humanos , Masculino , Ratones , Síndromes Miasténicos Congénitos/metabolismo , Síndromes Miasténicos Congénitos/patología , Mioblastos/patología , Unión Neuromuscular/crecimiento & desarrollo , Unión Neuromuscular/patología , Linaje , Receptores Colinérgicos/genética , Receptores Colinérgicos/metabolismo , Sinapsis/genética , Sinapsis/metabolismo , Sinapsis/patología , Transmisión Sináptica
19.
J Med Genet ; 52(12): 797-803, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26424145

RESUMEN

BACKGROUND: The genetic aetiology of neurodevelopmental defects is extremely diverse, and the lack of distinctive phenotypic features means that genetic criteria are often required for accurate diagnostic classification. We aimed to identify the causative genetic lesions in two families in which eight affected individuals displayed variable learning disability, spasticity and abnormal gait. METHODS: Autosomal recessive inheritance was suggested by consanguinity in one family and by sibling recurrences with normal parents in the second. Autozygosity mapping and exome sequencing, respectively, were used to identify the causative gene. RESULTS: In both families, biallelic loss-of-function mutations in HACE1 were identified. HACE1 is an E3 ubiquitin ligase that regulates the activity of cellular GTPases, including Rac1 and members of the Rab family. In the consanguineous family, a homozygous mutation p.R219* predicted a truncated protein entirely lacking its catalytic domain. In the other family, compound heterozygosity for nonsense mutation p.R748* and a 20-nt insertion interrupting the catalytic homologous to the E6-AP carboxyl terminus (HECT) domain was present; western blot analysis of patient cells revealed an absence of detectable HACE1 protein. CONCLUSION: HACE1 mutations underlie a new autosomal recessive neurodevelopmental disorder. Previous studies have implicated HACE1 as a tumour suppressor gene; however, since cancer predisposition was not observed either in homozygous or heterozygous mutation carriers, this concept may require re-evaluation.


Asunto(s)
Trastornos del Neurodesarrollo/genética , Ubiquitina-Proteína Ligasas/deficiencia , Células Cultivadas , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Genes Recesivos , Humanos , Lactante , Masculino , Linaje , Polimorfismo de Nucleótido Simple , Síndrome , Ubiquitina-Proteína Ligasas/genética
20.
Nat Cell Biol ; 17(8): 1074-1087, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26167768

RESUMEN

Defects in primary cilium biogenesis underlie the ciliopathies, a growing group of genetic disorders. We describe a whole-genome siRNA-based reverse genetics screen for defects in biogenesis and/or maintenance of the primary cilium, obtaining a global resource. We identify 112 candidate ciliogenesis and ciliopathy genes, including 44 components of the ubiquitin-proteasome system, 12 G-protein-coupled receptors, and 3 pre-mRNA processing factors (PRPF6, PRPF8 and PRPF31) mutated in autosomal dominant retinitis pigmentosa. The PRPFs localize to the connecting cilium, and PRPF8- and PRPF31-mutated cells have ciliary defects. Combining the screen with exome sequencing data identified recessive mutations in PIBF1, also known as CEP90, and C21orf2, also known as LRRC76, as causes of the ciliopathies Joubert and Jeune syndromes. Biochemical approaches place C21orf2 within key ciliopathy-associated protein modules, offering an explanation for the skeletal and retinal involvement observed in individuals with C21orf2 variants. Our global, unbiased approaches provide insights into ciliogenesis complexity and identify roles for unanticipated pathways in human genetic disease.


Asunto(s)
Cilios/genética , Trastornos de la Motilidad Ciliar/genética , Marcadores Genéticos , Pruebas Genéticas/métodos , Genómica/métodos , Células Fotorreceptoras , Interferencia de ARN , Anomalías Múltiples , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/ultraestructura , Enfermedades Cerebelosas/genética , Cerebelo/anomalías , Cilios/metabolismo , Cilios/patología , Trastornos de la Motilidad Ciliar/metabolismo , Trastornos de la Motilidad Ciliar/patología , Proteínas del Citoesqueleto , Bases de Datos Genéticas , Síndrome de Ellis-Van Creveld/genética , Anomalías del Ojo/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Células HEK293 , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Enfermedades Renales Quísticas/genética , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Fenotipo , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/ultraestructura , Proteínas Gestacionales/genética , Proteínas Gestacionales/metabolismo , Proteínas/genética , Proteínas/metabolismo , Reproducibilidad de los Resultados , Retina/anomalías , Factores Supresores Inmunológicos/genética , Factores Supresores Inmunológicos/metabolismo , Transfección , Pez Cebra/genética , Pez Cebra/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...