Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
HGG Adv ; 5(3): 100288, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38566418

RESUMEN

Biallelic loss-of-function variants in the MUSK gene result in two allelic disorders: (1) congenital myasthenic syndrome (CMS; OMIM: 616325), a neuromuscular disorder that has a range of severity from severe neonatal-onset weakness to mild adult-onset weakness, and (2) fetal akinesia deformation sequence (OMIM: 208150), a form of pregnancy loss characterized by severe muscle weakness in the fetus. The MUSK gene codes for muscle-specific kinase (MuSK), a receptor tyrosine kinase involved in the development of the neuromuscular junction. Here, we report a case of neonatal-onset MUSK-related CMS in a patient harboring compound heterozygous deletions in the MUSK gene, including (1) a deletion of exons 2-3 leading to an in-frame MuSK protein lacking the immunoglobulin 1 (Ig1) domain and (2) a deletion of exons 7-11 leading to an out-of-frame, truncated MuSK protein. Individual domains of the MuSK protein have been elucidated structurally; however, a complete MuSK structure generated by machine learning algorithms has clear inaccuracies. We modify a predicted AlphaFold structure and integrate previously reported domain-specific structural data to suggest a MuSK protein that dimerizes in two locations (Ig1 and the transmembrane domain). We analyze known pathogenic variants in MUSK to discover domain-specific genotype-phenotype correlations; variants that lead to a loss of protein expression, disruption of the Ig1 domain, or Dok-7 binding are associated with the most severe phenotypes. A conceptual model is provided to explain the severe phenotypes seen in Ig1 variants and the poor response of our patient to pyridostigmine.

2.
Am J Med Genet A ; 191(11): 2757-2767, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37596828

RESUMEN

Oculogastrointestinal neurodevelopmental syndrome has been described in seven previously published individuals who harbor biallelic pathogenic variants in the CAPN15 gene. Biallelic missense variants have been reported to demonstrate a phenotype of eye abnormalities and developmental delay, while biallelic loss of function variants exhibit phenotypes including microcephaly and craniofacial abnormalities, cardiac and genitourinary malformations, and abnormal neurologic activity. We report six individuals from three unrelated families harboring biallelic deleterious variants in CAPN15 with phenotypes overlapping those previously described for this disorder. Of the individuals affected, four demonstrate radiographic evidence of the classical triad of Dandy-Walker malformation including hypoplastic vermis, fourth ventricle enlargement, and torcular elevation. Cerebellar anomalies have not been previously reported in association with CAPN15-related disease. Here, we present three unrelated families with findings consistent with oculogastrointestinal neurodevelopmental syndrome and cerebellar pathology including Dandy-Walker malformation. To corroborate these novel clinical findings, we present supporting data from the mouse model suggesting an important role for this protein in normal cerebellar development. Our findings add six molecularly confirmed cases to the literature and additionally establish a new association of Dandy-Walker malformation with biallelic CAPN15 variants, thereby expanding the neurologic spectrum among patients affected by CAPN15-related disease.


Asunto(s)
Vermis Cerebeloso , Síndrome de Dandy-Walker , Microcefalia , Animales , Ratones , Humanos , Síndrome de Dandy-Walker/diagnóstico , Síndrome de Dandy-Walker/genética , Cerebelo/anomalías , Microcefalia/complicaciones , Fenotipo , Calpaína/genética
3.
Cell ; 186(6): 1162-1178.e20, 2023 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-36931244

RESUMEN

Germline histone H3.3 amino acid substitutions, including H3.3G34R/V, cause severe neurodevelopmental syndromes. To understand how these mutations impact brain development, we generated H3.3G34R/V/W knock-in mice and identified strikingly distinct developmental defects for each mutation. H3.3G34R-mutants exhibited progressive microcephaly and neurodegeneration, with abnormal accumulation of disease-associated microglia and concurrent neuronal depletion. G34R severely decreased H3K36me2 on the mutant H3.3 tail, impairing recruitment of DNA methyltransferase DNMT3A and its redistribution on chromatin. These changes were concurrent with sustained expression of complement and other innate immune genes possibly through loss of non-CG (CH) methylation and silencing of neuronal gene promoters through aberrant CG methylation. Complement expression in G34R brains may lead to neuroinflammation possibly accounting for progressive neurodegeneration. Our study reveals that H3.3G34-substitutions have differential impact on the epigenome, which underlie the diverse phenotypes observed, and uncovers potential roles for H3K36me2 and DNMT3A-dependent CH-methylation in modulating synaptic pruning and neuroinflammation in post-natal brains.


Asunto(s)
ADN Metiltransferasa 3A , Histonas , Animales , Ratones , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN/genética , Metilasas de Modificación del ADN/genética , Histonas/metabolismo , Enfermedades Neuroinflamatorias
4.
Genet Med ; 23(8): 1514-1521, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33846581

RESUMEN

PURPOSE: Reports have questioned the dogma of exclusive maternal transmission of human mitochondrial DNA (mtDNA), including the recent report of an admixture of two mtDNA haplogroups in individuals from three multigeneration families. This was interpreted as being consistent with biparental transmission of mtDNA in an autosomal dominant-like mode. The authenticity and frequency of these findings are debated. METHODS: We retrospectively analyzed individuals with two mtDNA haplogroups from 2017 to 2019 and selected four families for further study. RESULTS: We identified this phenomenon in 104/27,388 (approximately 1/263) unrelated individuals. Further study revealed (1) a male with two mitochondrial haplogroups transmits only one haplogroup to some of his offspring, consistent with nuclear transmission; (2) the heteroplasmy level of paternally transmitted variants is highest in blood, lower in buccal, and absent in muscle or urine of the same individual, indicating it is inversely correlated with mtDNA content; and (3) paternally transmitted apparent large-scale mtDNA deletions/duplications are not associated with a disease phenotype. CONCLUSION: These findings strongly suggest that the observed mitochondrial haplogroup of paternal origin resulted from coamplification of rare, concatenated nuclear mtDNA segments with genuine mtDNA during testing. Evaluation of additional specimen types can help clarify the clinical significance of the observed results.


Asunto(s)
ADN Mitocondrial , Mitocondrias , ADN Mitocondrial/genética , Haplotipos , Humanos , Masculino , Mitocondrias/genética , Fenotipo , Estudios Retrospectivos
5.
Genet Med ; 23(7): 1234-1245, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33824499

RESUMEN

PURPOSE: Proline Rich 12 (PRR12) is a gene of unknown function with suspected DNA-binding activity, expressed in developing mice and human brains. Predicted loss-of-function variants in this gene are extremely rare, indicating high intolerance of haploinsufficiency. METHODS: Three individuals with intellectual disability and iris anomalies and truncating de novo PRR12 variants were described previously. We add 21 individuals with similar PRR12 variants identified via matchmaking platforms, bringing the total number to 24. RESULTS: We observed 12 frameshift, 6 nonsense, 1 splice-site, and 2 missense variants and one patient with a gross deletion involving PRR12. Three individuals had additional genetic findings, possibly confounding the phenotype. All patients had developmental impairment. Variable structural eye defects were observed in 12/24 individuals (50%) including anophthalmia, microphthalmia, colobomas, optic nerve and iris abnormalities. Additional common features included hypotonia (61%), heart defects (52%), growth failure (54%), and kidney anomalies (35%). PrediXcan analysis showed that phecodes most strongly associated with reduced predicted PRR12 expression were enriched for eye- (7/30) and kidney- (4/30) phenotypes, such as wet macular degeneration and chronic kidney disease. CONCLUSION: These findings support PRR12 haploinsufficiency as a cause for a novel disorder with a wide clinical spectrum marked chiefly by neurodevelopmental and eye abnormalities.


Asunto(s)
Haploinsuficiencia , Discapacidad Intelectual , Animales , Haploinsuficiencia/genética , Humanos , Discapacidad Intelectual/genética , Ratones , Hipotonía Muscular , Mutación Missense , Fenotipo
7.
J Pediatr Genet ; 10(1): 35-38, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33552636

RESUMEN

Cat eye syndrome (CES) is a rare genetic defect, characterized by iris colobomas, preauricular skin tags, and anal malformations. Affecting 1 in 150,000 people, this defect is caused by duplication or triplication of the proximal long (q) arm of chromosome 22. Congenital heart disease is associated with CES. One of the most common heart defects in patients with CES is total anomalous pulmonary venous return (TAPVR). In this article, we reported patients with a rare association of concomitant TAPVR and aortic arch obstruction: one with interrupted aortic arch and the other with coarctation of the aorta with an aberrant right subclavian artery.

8.
Eur J Hum Genet ; 29(2): 271-279, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32901138

RESUMEN

Trafficking protein particle (TRAPP) complexes, which include the TRAPPC4 protein, regulate membrane trafficking between lipid organelles in a process termed vesicular tethering. TRAPPC4 was recently implicated in a recessive neurodevelopmental condition in four unrelated families due to a shared c.454+3A>G splice variant. Here, we report 23 patients from 17 independent families with an early-infantile-onset neurodegenerative presentation, where we also identified the homozygous variant hg38:11:119020256 A>G (NM_016146.5:c.454+3A>G) in TRAPPC4 through exome or genome sequencing. No other clinically relevant TRAPPC4 variants were identified among any of over 10,000 patients with neurodevelopmental conditions. We found the carrier frequency of TRAPPC4 c.454+3A>G was 2.4-5.4 per 10,000 healthy individuals. Affected individuals with the homozygous TRAPPC4 c.454+3A>G variant showed profound psychomotor delay, developmental regression, early-onset epilepsy, microcephaly and progressive spastic tetraplegia. Based upon RNA sequencing, the variant resulted in partial exon 3 skipping and generation of an aberrant transcript owing to use of a downstream cryptic splice donor site, predicting a premature stop codon and nonsense mediated decay. These data confirm the pathogenicity of the TRAPPC4 c.454+3A>G variant, and refine the clinical presentation of TRAPPC4-related encephalopathy.


Asunto(s)
Homocigoto , Proteínas del Tejido Nervioso/genética , Trastornos del Neurodesarrollo/genética , Empalme del ARN , Proteínas de Transporte Vesicular/genética , Niño , Preescolar , Codón sin Sentido , Exoma , Exones , Femenino , Humanos , Masculino , Microcefalia/genética , Trastornos del Neurodesarrollo/diagnóstico por imagen , Linaje , Sitios de Empalme de ARN , Síndrome
9.
Genet Med ; 23(4): 653-660, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33299146

RESUMEN

PURPOSE: This study aims to provide a comprehensive description of the phenotypic and genotypic spectrum of SNAP25 developmental and epileptic encephalopathy (SNAP25-DEE) by reviewing newly identified and previously reported individuals. METHODS: Individuals harboring heterozygous missense or loss-of-function variants in SNAP25 were assembled through collaboration with international colleagues, matchmaking platforms, and literature review. For each individual, detailed phenotyping, classification, and structural modeling of the identified variant were performed. RESULTS: The cohort comprises 23 individuals with pathogenic or likely pathogenic de novo variants in SNAP25. Intellectual disability and early-onset epilepsy were identified as the core symptoms of SNAP25-DEE, with recurrent findings of movement disorders, cerebral visual impairment, and brain atrophy. Structural modeling for all variants predicted possible functional defects concerning SNAP25 or impaired interaction with other components of the SNARE complex. CONCLUSION: We provide a comprehensive description of SNAP25-DEE with intellectual disability and early-onset epilepsy mostly occurring before the age of two years. These core symptoms and additional recurrent phenotypes show an overlap to genes encoding other components or associated proteins of the SNARE complex such as STX1B, STXBP1, or VAMP2. Thus, these findings advance the concept of a group of neurodevelopmental disorders that may be termed "SNAREopathies."


Asunto(s)
Encefalopatías , Epilepsia , Discapacidad Intelectual , Trastornos del Neurodesarrollo , Proteína 25 Asociada a Sinaptosomas/genética , Preescolar , Epilepsia/genética , Humanos , Trastornos del Neurodesarrollo/genética , Fenotipo
10.
Am J Med Genet A ; 182(8): 1947-1951, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32445275

RESUMEN

Schinzel-Giedion syndrome (SGS; OMIM 269150) is an ultra-rare genetic disorder associated with a distinctive facial gestalt, congenital malformations, severe intellectual disability, and a progressive neurological course. The prognosis for SGS is poor, with survival beyond the first decade rare. Germline, de novo heterozygous variants in the SETBP1 gene cause SGS with the pathogenic variants associated with the SGS phenotype missense and confined to exon 4 of the gene, clustered in a four amino acid (12 bp) hotspot in the SKI homologous region of the SETBP1 protein. We report a patient with a de novo I871S variant within the SKI homologous region, which has been associated with the severe phenotype previously; but our patient has fewer features of SGS and a milder course. This is the first report of a forme-fruste phenotype in a patient with a pathogenic variant within the SGS hotspot on the SETBP1 gene and it highlights the importance of considering atypical clinical presentations in the context of severe ultra-rare genetic disorders.


Asunto(s)
Anomalías Múltiples/genética , Proteínas Portadoras/genética , Anomalías Craneofaciales/genética , Cara/anomalías , Deformidades Congénitas de la Mano/genética , Discapacidad Intelectual/genética , Uñas Malformadas/genética , Proteínas Nucleares/genética , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/patología , Adulto , Anomalías Craneofaciales/diagnóstico , Anomalías Craneofaciales/patología , Exones , Cara/patología , Femenino , Deformidades Congénitas de la Mano/diagnóstico , Deformidades Congénitas de la Mano/patología , Heterocigoto , Humanos , Lactante , Recién Nacido , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/patología , Masculino , Mutación/genética , Uñas Malformadas/diagnóstico , Uñas Malformadas/patología , Fenotipo
11.
Am J Hum Genet ; 106(6): 830-845, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32442410

RESUMEN

SOX6 belongs to a family of 20 SRY-related HMG-box-containing (SOX) genes that encode transcription factors controlling cell fate and differentiation in many developmental and adult processes. For SOX6, these processes include, but are not limited to, neurogenesis and skeletogenesis. Variants in half of the SOX genes have been shown to cause severe developmental and adult syndromes, referred to as SOXopathies. We here provide evidence that SOX6 variants also cause a SOXopathy. Using clinical and genetic data, we identify 19 individuals harboring various types of SOX6 alterations and exhibiting developmental delay and/or intellectual disability; the individuals are from 17 unrelated families. Additional, inconstant features include attention-deficit/hyperactivity disorder (ADHD), autism, mild facial dysmorphism, craniosynostosis, and multiple osteochondromas. All variants are heterozygous. Fourteen are de novo, one is inherited from a mosaic father, and four offspring from two families have a paternally inherited variant. Intragenic microdeletions, balanced structural rearrangements, frameshifts, and nonsense variants are predicted to inactivate the SOX6 variant allele. Four missense variants occur in residues and protein regions highly conserved evolutionarily. These variants are not detected in the gnomAD control cohort, and the amino acid substitutions are predicted to be damaging. Two of these variants are located in the HMG domain and abolish SOX6 transcriptional activity in vitro. No clear genotype-phenotype correlations are found. Taken together, these findings concur that SOX6 haploinsufficiency leads to a neurodevelopmental SOXopathy that often includes ADHD and abnormal skeletal and other features.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/genética , Craneosinostosis/genética , Trastornos del Neurodesarrollo/genética , Osteocondroma/genética , Factores de Transcripción SOXD/genética , Transporte Activo de Núcleo Celular , Adolescente , Secuencia de Aminoácidos , Secuencia de Bases , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Niño , Preescolar , Simulación por Computador , Femenino , Variación Estructural del Genoma/genética , Humanos , Lactante , Masculino , Mutación Missense , Trastornos del Neurodesarrollo/diagnóstico , RNA-Seq , Factores de Transcripción SOXD/química , Factores de Transcripción SOXD/metabolismo , Síndrome , Transcripción Genética , Transcriptoma , Translocación Genética/genética
12.
Genet Med ; 22(7): 1269-1275, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32366967

RESUMEN

PURPOSE: Guidelines by professional organizations for assessing variant pathogenicity include the recommendation to utilize biologically relevant transcripts; however, there is variability in transcript selection by laboratories. METHODS: We describe three patients whose genomic results were incorrect, because alternative transcripts and tissue expression patterns were not considered by the commercial laboratories. RESULTS: In individual 1, a pathogenic coding variant in a brain-expressed isoform of CKDL5 was missed twice on sequencing, because the variant was intronic in the transcripts considered in analysis. In individual 2, a microdeletion affecting KMT2C was not reported on microarray, since deletions of proximal exons in this gene are seen in healthy individuals; however, this individual had a more distal deletion involving the brain-expressed KMT2C isoform, giving her a diagnosis of Kleefstra syndrome. Individual 3 was reported to have a pathogenic variant in exon 10 of OFD1 on exome, but had no typical features of the OFD1-related disorders. Since exon 10 is spliced from the more biologically relevant transcripts of OFD1, it was determined that he did not have an OFD1 disorder. CONCLUSION: These examples illustrate the importance of considering alternative transcripts as a potential confounder when genetic results are negative or discordant with the phenotype.


Asunto(s)
Exoma , Diagnóstico Erróneo , Empalme Alternativo/genética , Exones/genética , Femenino , Humanos , Masculino , Isoformas de Proteínas/genética , Secuenciación del Exoma
13.
Hum Mutat ; 41(3): 655-667, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31705726

RESUMEN

Congenital disorders of glycosylation (CDGs) comprise a large number of inherited metabolic defects that affect the biosynthesis and attachment of glycans. CDGs manifest as a broad spectrum of disease, most often including neurodevelopmental and skeletal abnormalities and skin laxity. Two patients with biallelic CSGALNACT1 variants and a mild skeletal dysplasia have been described previously. We investigated two unrelated patients presenting with short stature with advanced bone age, facial dysmorphism, and mild language delay, in whom trio-exome sequencing identified novel biallelic CSGALNACT1 variants: compound heterozygosity for c.1294G>T (p.Asp432Tyr) and the deletion of exon 4 that includes the start codon in one patient, and homozygosity for c.791A>G (p.Asn264Ser) in the other patient. CSGALNACT1 encodes CSGalNAcT-1, a key enzyme in the biosynthesis of sulfated glycosaminoglycans chondroitin and dermatan sulfate. Biochemical studies demonstrated significantly reduced CSGalNAcT-1 activity of the novel missense variants, as reported previously for the p.Pro384Arg variant. Altered levels of chondroitin, dermatan, and heparan sulfate moieties were observed in patients' fibroblasts compared to controls. Our data indicate that biallelic loss-of-function mutations in CSGALNACT1 disturb glycosaminoglycan synthesis and cause a mild skeletal dysplasia with advanced bone age, CSGALNACT1-CDG.


Asunto(s)
Trastornos Congénitos de Glicosilación/diagnóstico , Trastornos Congénitos de Glicosilación/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Anomalías Musculoesqueléticas/diagnóstico , Anomalías Musculoesqueléticas/genética , Mutación , N-Acetilgalactosaminiltransferasas/genética , Secuencia de Aminoácidos , Huesos/anomalías , Huesos/diagnóstico por imagen , Facies , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Recién Nacido , Mutación con Pérdida de Función , Masculino , Mutación Missense , Linaje , Fenotipo
14.
Am J Hum Genet ; 104(1): 94-111, 2019 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-30609410

RESUMEN

The use of whole-exome and whole-genome sequencing has been a catalyst for a genotype-first approach to diagnostics. Under this paradigm, we have implemented systematic sequencing of neonates and young children with a suspected genetic disorder. Here, we report on two families with recessive mutations in NCAPG2 and overlapping clinical phenotypes that include severe neurodevelopmental defects, failure to thrive, ocular abnormalities, and defects in urogenital and limb morphogenesis. NCAPG2 encodes a member of the condensin II complex, necessary for the condensation of chromosomes prior to cell division. Consistent with a causal role for NCAPG2, we found abnormal chromosome condensation, augmented anaphase chromatin-bridge formation, and micronuclei in daughter cells of proband skin fibroblasts. To test the functional relevance of the discovered variants, we generated an ncapg2 zebrafish model. Morphants displayed clinically relevant phenotypes, such as renal anomalies, microcephaly, and concomitant increases in apoptosis and altered mitotic progression. These could be rescued by wild-type but not mutant human NCAPG2 mRNA and were recapitulated in CRISPR-Cas9 F0 mutants. Finally, we noted that the individual with a complex urogenital defect also harbored a heterozygous NPHP1 deletion, a common contributor to nephronophthisis. To test whether sensitization at the NPHP1 locus might contribute to a more severe renal phenotype, we co-suppressed nphp1 and ncapg2, which resulted in significantly more dysplastic renal tubules in zebrafish larvae. Together, our data suggest that impaired function of NCAPG2 results in a severe condensinopathy, and they highlight the potential utility of examining candidate pathogenic lesions beyond the primary disease locus.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN/metabolismo , Complejos Multiproteicos/metabolismo , Mutación , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/patología , Fenotipo , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Niño , Preescolar , Proteínas del Citoesqueleto , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Proteínas de la Membrana/genética , Linaje , Síndrome , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/genética
16.
Genet Med ; 21(4): 867-876, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30190611

RESUMEN

PURPOSE: Neurofibromatosis type 1 (NF1) is characterized by a highly variable clinical presentation, but almost all NF1-affected adults present with cutaneous and/or subcutaneous neurofibromas. Exceptions are individuals heterozygous for the NF1 in-frame deletion, c.2970_2972del (p.Met992del), associated with a mild phenotype without any externally visible tumors. METHODS: A total of 135 individuals from 103 unrelated families, all carrying the constitutional NF1 p.Met992del pathogenic variant and clinically assessed using the same standardized phenotypic checklist form, were included in this study. RESULTS: None of the individuals had externally visible plexiform or histopathologically confirmed cutaneous or subcutaneous neurofibromas. We did not identify any complications, such as symptomatic optic pathway gliomas (OPGs) or symptomatic spinal neurofibromas; however, 4.8% of individuals had nonoptic brain tumors, mostly low-grade and asymptomatic, and 38.8% had cognitive impairment/learning disabilities. In an individual with the NF1 constitutional c.2970_2972del and three astrocytomas, we provided proof that all were NF1-associated tumors given loss of heterozygosity at three intragenic NF1 microsatellite markers and c.2970_2972del. CONCLUSION: We demonstrate that individuals with the NF1 p.Met992del pathogenic variant have a mild NF1 phenotype lacking clinically suspected plexiform, cutaneous, or subcutaneous neurofibromas. However, learning difficulties are clearly part of the phenotypic presentation in these individuals and will require specialized care.


Asunto(s)
Discapacidades para el Aprendizaje/genética , Neurofibroma Plexiforme/genética , Neurofibromatosis 1/genética , Neurofibromina 1/genética , Adolescente , Adulto , Niño , Preescolar , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Heterocigoto , Humanos , Lactante , Discapacidades para el Aprendizaje/fisiopatología , Masculino , Mutación Missense/genética , Neurofibroma Plexiforme/fisiopatología , Neurofibromatosis 1/patología , Eliminación de Secuencia , Adulto Joven
17.
J Hum Genet ; 63(12): 1211-1222, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30250212

RESUMEN

Mitochondrial dysfunction lies behind many neurodegenerative disorders, owing largely to the intense energy requirements of most neurons. Such mitochondrial dysfunction may work through a variety of mechanisms, from direct disruption of the electron transport chain to abnormal mitochondrial biogenesis. Recently, we have identified biallelic mutations in the mitochondrial flavoprotein "ferredoxin reductase" (FDXR) gene as a novel cause of mitochondriopathy, peripheral neuropathy, and optic atrophy. In this report, we expand upon those results by describing two new cases of disease-causing FDXR variants in patients with variable severity of phenotypes, including evidence of an inflammatory response in brain autopsy. To investigate the underlying pathogenesis, we examined neurodegeneration in a mouse model. We found that Fdxr mutant mouse brain tissues share pathological changes similar to those seen in patient autopsy material, including increased astrocytes. Furthermore, we show that these abnormalities are associated with increased levels of markers for both neurodegeneration and gliosis, with the latter implying inflammation as a major factor in the pathology of Fdxr mutations. These data provide further insight into the pathogenic mechanism of FDXR-mediated central neuropathy, and suggest an avenue for mechanistic studies that will ultimately inform treatment.


Asunto(s)
Alelos , Proteínas Hierro-Azufre/genética , Mutación , Enfermedades Neurodegenerativas/genética , Oxidorreductasas/genética , Animales , Encéfalo/enzimología , Encéfalo/patología , Femenino , Humanos , Inflamación/enzimología , Inflamación/genética , Inflamación/patología , Proteínas Hierro-Azufre/metabolismo , Masculino , Ratones , Ratones Transgénicos , Enfermedades Neurodegenerativas/enzimología , Enfermedades Neurodegenerativas/patología , Oxidorreductasas/metabolismo
18.
Artículo en Inglés | MEDLINE | ID: mdl-29970384

RESUMEN

Recent evidence has implicated EFL1 in a phenotype overlapping Shwachman-Diamond syndrome (SDS), with the functional interplay between EFL1 and the previously known causative gene SBDS accounting for the similarity in clinical features. Relatively little is known about the phenotypes associated with pathogenic variants in the EFL1 gene, but the initial indication was that phenotypes may be more severe, when compared with SDS. We report a pediatric patient who presented with a metaphyseal dysplasia and was found to have biallelic variants in EFL1 on reanalysis of trio whole-exome sequencing data. The variant had not been initially reported because of the research laboratory's focus on de novo variants. Subsequent phenotyping revealed variability in her manifestations. Although her metaphyseal abnormalities were more severe than in the original reported cohort with EFL1 variants, the bone marrow abnormalities were generally mild, and there was equivocal evidence for pancreatic insufficiency. Despite the limited number of reported patients, variants in EFL1 appear to cause a broader spectrum of symptoms that overlap with those seen in SDS. Our report adds to the evidence of EFL1 being associated with an SDS-like phenotype and provides information adding to our understanding of the phenotypic variability of this disorder. Our report also highlights the value of exome data reanalysis when a diagnosis is not initially apparent.


Asunto(s)
Enfermedades de la Médula Ósea/genética , Insuficiencia Pancreática Exocrina/genética , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/fisiología , Lipomatosis/genética , Adolescente , Enfermedades de la Médula Ósea/diagnóstico , Insuficiencia Pancreática Exocrina/diagnóstico , Femenino , Variación Genética/genética , Humanos , Lipomatosis/diagnóstico , Mutación , Osteocondrodisplasias/genética , Osteocondrodisplasias/fisiopatología , Factores de Elongación de Péptidos , Fenotipo , Proteínas/genética , Ribonucleoproteína Nuclear Pequeña U5 , Síndrome de Shwachman-Diamond , Secuenciación del Exoma
19.
Hum Mol Genet ; 27(14): 2454-2465, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29726930

RESUMEN

The 17 genes of the T-box family are transcriptional regulators that are involved in all stages of embryonic development, including craniofacial, brain, heart, skeleton and immune system. Malformation syndromes have been linked to many of the T-box genes. For example, haploinsufficiency of TBX1 is responsible for many structural malformations in DiGeorge syndrome caused by a chromosome 22q11.2 deletion. We report four individuals with an overlapping spectrum of craniofacial dysmorphisms, cardiac anomalies, skeletal malformations, immune deficiency, endocrine abnormalities and developmental impairments, reminiscent of DiGeorge syndrome, who are heterozygotes for TBX2 variants. The p.R20Q variant is shared by three affected family members in an autosomal dominant manner; the fourth unrelated individual has a de novo p.R305H mutation. Bioinformatics analyses indicate that these variants are rare and predict them to be damaging. In vitro transcriptional assays in cultured cells show that both variants result in reduced transcriptional repressor activity of TBX2. We also show that the variants result in reduced protein levels of TBX2. Heterologous over-expression studies in Drosophila demonstrate that both p.R20Q and p.R305H function as partial loss-of-function alleles. Hence, these and other data suggest that TBX2 is a novel candidate gene for a new multisystem malformation disorder.


Asunto(s)
Discapacidades del Desarrollo/genética , Síndrome de DiGeorge/genética , Predisposición Genética a la Enfermedad , Proteínas de Dominio T Box/genética , Adulto , Animales , Anomalías Cardiovasculares/genética , Anomalías Cardiovasculares/fisiopatología , Sistema Cardiovascular/fisiopatología , Niño , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/fisiopatología , Discapacidades del Desarrollo/fisiopatología , Síndrome de DiGeorge/fisiopatología , Modelos Animales de Enfermedad , Drosophila melanogaster , Femenino , Regulación del Desarrollo de la Expresión Génica , Haploinsuficiencia/genética , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/fisiopatología , Humanos , Ratones , Linaje , Embarazo , Adulto Joven , Pez Cebra
20.
Epileptic Disord ; 20(1): 30-34, 2018 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-29444762

RESUMEN

Hemimegalencephaly is known to occur in Proteus syndrome, but has not been reported, to our knowledge, in the other PTEN mutation-related syndrome of Bannayan-Riley-Ruvalcaba. Here, we report a patient with Bannayan-Riley-Ruvalcaba syndrome who also had hemimegalencephaly and in whom the hemimegalencephaly was evident well before presentation of the characteristic manifestations of Bannayan-Riley-Ruvalcaba syndrome. An 11-year-old boy developed drug-resistant focal seizures on the fifth day of life. MRI revealed left hemimegalencephaly. He later showed macrocephaly, developmental delay, athetotic quadriplegic cerebral palsy, and neuromuscular scoliosis. Freckling of the penis, which is characteristic of Bannayan-Riley-Ruvalcaba syndrome, was not present at birth but was observed at 9 years of age. Gene analysis revealed a c.510 T>G PTEN mutation. This patient and his other affected family members, his father and two siblings, were started on the tumour screening procedures recommended for patients with PTEN mutations. This case highlights the importance of early screening for PTEN mutations in cases of hemimegalencephaly not otherwise explained by another disorder, even in the absence of signs of Proteus syndrome or the full manifestations of Bannayan-Riley Ruvalcaba syndrome.


Asunto(s)
Síndrome de Hamartoma Múltiple/diagnóstico , Hemimegalencefalia/diagnóstico , Fosfohidrolasa PTEN/genética , Niño , Síndrome de Hamartoma Múltiple/genética , Síndrome de Hamartoma Múltiple/patología , Síndrome de Hamartoma Múltiple/fisiopatología , Hemimegalencefalia/genética , Hemimegalencefalia/patología , Hemimegalencefalia/fisiopatología , Humanos , Masculino , Mutación , Linaje
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...