Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Med Genet A ; 188(4): 1299-1306, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34970864

RESUMEN

The beta-actin gene (ACTB) encodes a ubiquitous cytoskeletal protein, essential for embryonic development in humans. De novo heterozygous missense variants in the ACTB are implicated in causing Baraitser-Winter cerebrofrontofacial syndrome (BWCFFS; MIM#243310). ACTB pathogenic variants are rarely associated with intestinal malformations. We report on a rare case of monozygotic twins presenting with proximal small bowel atresia and hydrops in one, and apple-peel bowel atresia and laryngeal dysgenesis in the other. The twin with hydrops could not be resuscitated. Intensive and surgical care was provided to the surviving twin. Rapid trio genome sequencing identified a de novo missense variant in ACTB (NM_00101.3:c.1043C>T; p.(Ser348Leu)) that guided the care plan. The identical variant subsequently was identified in the demised twin. To characterize the functional effect, the variant was recreated as a pseudoheterozygote in a haploid wild-type S. cerevisiae strain. There was an obvious growth defect of the yACT1S348L/WT pseudoheterozygote compared to a yACT1WT/WT strain when grown at 22°C but not when grown at 30°C, consistent with the yACT1 S348L variant having a functional defect that is dominant over the wild-type allele. The functional results provide supporting evidence that the Ser348Leu variant is likely to be a pathogenic variant, including being associated with intestinal malformations in BWCFFS, and can demonstrate variable expressivity within monozygotic twins.


Asunto(s)
Atresia Intestinal , Gemelos Monocigóticos , Actinas/genética , Actinas/metabolismo , Variación Biológica Poblacional , Anomalías Craneofaciales , Edema , Epilepsia , Facies , Humanos , Discapacidad Intelectual , Atresia Intestinal/diagnóstico , Atresia Intestinal/genética , Lisencefalia , Saccharomyces cerevisiae/metabolismo , Gemelos Monocigóticos/genética
3.
Genet Med ; 22(3): 511-523, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31680123

RESUMEN

PURPOSE: Fetal akinesia has multiple clinical subtypes with over 160 gene associations, but the genetic etiology is not yet completely understood. METHODS: In this study, 51 patients from 47 unrelated families were analyzed using next-generation sequencing (NGS) techniques aiming to decipher the genomic landscape of fetal akinesia (FA). RESULTS: We have identified likely pathogenic gene variants in 37 cases and report 41 novel variants. Additionally, we report putative pathogenic variants in eight cases including nine novel variants. Our work identified 14 novel disease-gene associations for fetal akinesia: ADSSL1, ASAH1, ASPM, ATP2B3, EARS2, FBLN1, PRG4, PRICKLE1, ROR2, SETBP1, SCN5A, SCN8A, and ZEB2. Furthermore, a sibling pair harbored a homozygous copy-number variant in TNNT1, an ultrarare congenital myopathy gene that has been linked to arthrogryposis via Gene Ontology analysis. CONCLUSION: Our analysis indicates that genetic defects leading to primary skeletal muscle diseases might have been underdiagnosed, especially pathogenic variants in RYR1. We discuss three novel putative fetal akinesia genes: GCN1, IQSEC3 and RYR3. Of those, IQSEC3, and RYR3 had been proposed as neuromuscular disease-associated genes recently, and our findings endorse them as FA candidate genes. By combining NGS with deep clinical phenotyping, we achieved a 73% success rate of solved cases.


Asunto(s)
Enfermedades Fetales/genética , Factores de Intercambio de Guanina Nucleótido/genética , Proteínas de Unión al ARN/genética , Canal Liberador de Calcio Receptor de Rianodina/genética , Transactivadores/genética , Adolescente , Adulto , Artrogriposis/genética , Artrogriposis/patología , Niño , Preescolar , Variaciones en el Número de Copia de ADN/genética , Femenino , Enfermedades Fetales/patología , Predisposición Genética a la Enfermedad , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Recién Nacido , Masculino , Enfermedades Musculares/genética , Enfermedades Musculares/patología , Adulto Joven
4.
Hum Mol Genet ; 26(20): 4055-4066, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-29016863

RESUMEN

Congenital cranial dysinnervation disorders (CCDDs) comprise a heterogeneous spectrum of diseases characterized by congenital, non-progressive impairment of eye, eyelid and/or facial movements including Möbius syndrome, Duane retraction syndrome, congenital ptosis, and congenital fibrosis of the extraocular muscles. Over the last 20 years, several CCDDs have been identified as neurodevelopmental disorders that are caused by mutations of genes involved in brain and cranial nerve development, e.g. KIF21A and TUBB3 that each plays a pivotal role for microtubule function. In a five-generation pedigree, we identified a heterozygous mutation of TUBB6, a gene encoding a class V tubulin which has not been linked to a human hereditary disease so far. The missense mutation (p.Phe394Ser) affects an amino acid residue highly conserved in evolution, and co-segregates with a phenotype characterized by congenital non-progressive bilateral facial palsy and congenital velopharyngeal dysfunction presenting with varying degrees of hypomimia, rhinophonia, impaired gag reflex and bilateral ptosis. Expression of the mutated protein in yeast led to an impaired viability compared to wildtype cells when exposed to the microtubule-poison benomyl. Our findings enlarge the spectrum of tubulinopathies and emphasize that mutations of TUBB6 should be considered in patients with congenital non-progressive facial palsy. Further studies are needed to verify whether this phenotype is indeed part of the CCDD spectrum.


Asunto(s)
Blefaroptosis/complicaciones , Blefaroptosis/genética , Parálisis Facial/congénito , Parálisis Facial/genética , Tubulina (Proteína)/genética , Insuficiencia Velofaríngea/congénito , Insuficiencia Velofaríngea/genética , Blefaroptosis/patología , Preescolar , Parálisis Facial/patología , Femenino , Genes Dominantes , Humanos , Masculino , Persona de Mediana Edad , Mutación , Músculos Oculomotores/patología , Linaje , Insuficiencia Velofaríngea/patología
5.
Am J Hum Genet ; 99(5): 1206-1216, 2016 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-27843126

RESUMEN

We report ten individuals of four independent consanguineous families from Turkey, India, Libya, and Pakistan with a variable clinical phenotype that comprises arthrogryposis, spontaneously resolving respiratory insufficiency at birth, muscular atrophy predominantly of the distal lower limbs, scoliosis, and mild distal sensory involvement. Using whole-exome sequencing, SNPchip-based linkage analysis, DNA microarray, and Sanger sequencing, we identified three independent homozygous frameshift mutations and a homozygous deletion of two exons in PIEZO2 that segregated in all affected individuals of the respective family. The mutations are localized in the N-terminal and central region of the gene, leading to nonsense-mediated transcript decay and consequently to lack of PIEZO2 protein. In contrast, heterozygous gain-of-function missense mutations, mainly localized at the C terminus, cause dominant distal arthrogryposis 3 (DA3), distal arthrogryposis 5 (DA5), or Marden-Walker syndrome (MWKS), which encompass contractures of hands and feet, scoliosis, ophthalmoplegia, and ptosis. PIEZO2 encodes a mechanosensitive ion channel that plays a major role in light-touch mechanosensation and has recently been identified as the principal mechanotransduction channel for proprioception. Mice ubiquitously depleted of PIEZO2 are postnatally lethal. However, individuals lacking PIEZO2 develop a not life-threatening, slowly progressive disorder, which is likely due to loss of PIEZO2 protein in afferent neurons leading to disturbed proprioception causing aberrant muscle development and function. Here we report a recessively inherited PIEZO2-related disease and demonstrate that depending on the type of mutation and the mode of inheritance, PIEZO2 causes clinically distinguishable phenotypes.


Asunto(s)
Artrogriposis/genética , Canales Iónicos/genética , Atrofia Muscular/genética , Propiocepción , Síndrome de Dificultad Respiratoria del Recién Nacido/genética , Escoliosis/genética , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Adolescente , Adulto , Alelos , Aracnodactilia/diagnóstico , Aracnodactilia/genética , Artrogriposis/diagnóstico , Blefarofimosis/diagnóstico , Blefarofimosis/genética , Niño , Preescolar , Enfermedades del Tejido Conjuntivo/diagnóstico , Enfermedades del Tejido Conjuntivo/genética , Contractura/diagnóstico , Contractura/genética , Femenino , Estudio de Asociación del Genoma Completo , Homocigoto , Humanos , India , Canales Iónicos/metabolismo , Libia , Masculino , Mecanotransducción Celular , Atrofia Muscular/diagnóstico , Mutación Missense , Oftalmoplejía/diagnóstico , Oftalmoplejía/genética , Pakistán , Linaje , Polimorfismo de Nucleótido Simple , Síndrome de Dificultad Respiratoria del Recién Nacido/diagnóstico , Escoliosis/diagnóstico , Turquía , Adulto Joven
6.
Mol Cell Probes ; 45: 89-93, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30885829

RESUMEN

Single gene (Mendelian) disorders are one of the leading causes of neonatal morbidity and mortality. However, in the setting of preterm birth phenotypic features of genetic diseases are often undifferentiated and are clinically very difficult to interpret based on the wide range of differential diagnoses. We report an extremely low birth weight infant (ELBW) born prematurely at 23 + 0 gestational weeks after twin pregnancy with a novel clinical manifestation with persistent hyperglycaemia as well as the known manifestations of disease-associated hypokinesia, renal salt wasting, and multifocal atrial tachycardia. The patient died of heart failure on the 72nd day of life. Whole exome sequencing (WES) revealed a previously well established, disease-causing heterozygous likely pathogenic variant in the Harvey rat sarcoma viral oncogene homolog (HRAS)-gene (c.35G > C, p. G12A, rs104894230), which implied the clinical diagnosis of Costello syndrome (CS; OMIM#190020.0004). The twin brother merely had complications related to preterm birth and did not show any CS symptoms. In conclusion, our case illustrated that CS should be considered in ELBW infants showing a life-threatening combination of complex cardiac arrhythmia and hypokinesia. If a syndromic disorder is suspected in the neonatal intensive care unit (NICU) setting, rapid WES is a useful, non-invasive diagnostic tool in critically ill ELBW infants.


Asunto(s)
Secuenciación del Exoma/métodos , Embarazo Gemelar/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Resultado Fatal , Femenino , Humanos , Recien Nacido con Peso al Nacer Extremadamente Bajo/sangre , Recién Nacido , Recien Nacido Prematuro/sangre , Unidades de Cuidado Intensivo Neonatal , Masculino , Polimorfismo de Nucleótido Simple , Embarazo
7.
Hum Mutat ; 39(9): 1284-1298, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29858556

RESUMEN

Spinal muscular atrophies (SMAs) are a heterogeneous group of disorders characterized by muscular atrophy, weakness, and hypotonia due to suspected lower motor neuron degeneration (LMND). In a large cohort of 3,465 individuals suspected with SMA submitted for SMN1 testing to our routine diagnostic laboratory, 48.8% carried a homozygous SMN1 deletion, 2.8% a subtle mutation, and an SMN1 deletion, whereas 48.4% remained undiagnosed. Recently, several other genes implicated in SMA/LMND have been reported. Despite several efforts to establish a diagnostic algorithm for non-5q-SMA (SMA without deletion or point mutations in SMN1 [5q13.2]), data from large-scale studies are not available. We tested the clinical utility of targeted sequencing in non-5q-SMA by developing two different gene panels. We first analyzed 30 individuals with a small panel including 62 genes associated with LMND using IonTorrent-AmpliSeq target enrichment. Then, additional 65 individuals were tested with a broader panel encompassing up to 479 genes implicated in neuromuscular diseases (NMDs) with Agilent-SureSelect target enrichment. The NMD panel provided a higher diagnostic yield (33%) than the restricted LMND panel (13%). Nondiagnosed cases were further subjected to exome or genome sequencing. Our experience supports the use of gene panels covering a broad disease spectrum for diseases that are highly heterogeneous and clinically difficult to differentiate.


Asunto(s)
Atrofia Muscular Espinal/diagnóstico , Atrofia Muscular Espinal/genética , Enfermedades Neuromusculares/diagnóstico , Patología Molecular , Adolescente , Adulto , Edad de Inicio , Niño , Preescolar , Exones/genética , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Homocigoto , Humanos , Lactante , Masculino , Persona de Mediana Edad , Atrofia Muscular Espinal/patología , Enfermedades Neuromusculares/genética , Enfermedades Neuromusculares/fisiopatología , Mutación Puntual , Eliminación de Secuencia , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Secuenciación del Exoma , Secuenciación Completa del Genoma , Adulto Joven
8.
Hum Mol Genet ; 24(9): 2594-603, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25616960

RESUMEN

Determination of variant pathogenicity represents a major challenge in the era of high-throughput sequencing. Erroneous categorization may result if variants affect genes that are in fact dispensable. We demonstrate that this also applies to rare, apparently unambiguous truncating mutations of an established disease gene. By whole-exome sequencing (WES) in a consanguineous family with congenital non-syndromic deafness, we unexpectedly identified a homozygous nonsense variant, p.Arg1066*, in AHI1, a gene associated with Joubert syndrome (JBTS), a severe recessive ciliopathy. None of four homozygotes expressed any signs of JBTS, and one of them had normal hearing, which also ruled out p.Arg1066* as the cause of deafness. Homozygosity mapping and WES in the only other reported JBTS family with a homozygous C-terminal truncation (p.Trp1088Leufs*16) confirmed AHI1 as disease gene, but based on a more N-terminal missense mutation impairing WD40-repeat formation. Morpholinos against N-terminal zebrafish Ahi1, orthologous to where human mutations cluster, produced a ciliopathy, but targeting near human p.Arg1066 and p.Trp1088 did not. Most AHI1 mutations in JBTS patients result in truncated protein lacking WD40-repeats and the SH3 domain; disease was hitherto attributed to loss of these protein interaction modules. Our findings indicate that normal development does not require the C-terminal SH3 domain. This has far-reaching implications, considering that variants like p.Glu984* identified by preconception screening ('Kingsmore panel') do not necessarily indicate JBTS carriership. Genomes of individuals with consanguineous background are enriched for homozygous variants that may unmask dispensable regions of disease genes and unrecognized false positives in diagnostic large-scale sequencing and preconception carrier screening.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Estudios de Asociación Genética , Mutación , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras del Transporte Vesicular , Animales , Encéfalo/patología , Cerebelo/anomalías , Mapeo Cromosómico , Consanguinidad , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Evolución Molecular , Exoma , Anomalías del Ojo/diagnóstico , Anomalías del Ojo/genética , Femenino , Orden Génico , Genes Recesivos , Sitios Genéticos , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Homocigoto , Humanos , Enfermedades Renales Quísticas/diagnóstico , Enfermedades Renales Quísticas/genética , Imagen por Resonancia Magnética , Masculino , Modelos Moleculares , Linaje , Conformación Proteica , Retina/anomalías , Pez Cebra/genética
9.
Am J Med Genet A ; 173(8): 2132-2138, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28574232

RESUMEN

Recently, a new syndrome with intellectual disability (ID) and dysmorphic features due to deletions or point mutations within the TBL1XR1 gene located in the chromosomal band 3q26.32 has been described (MRD41, OMIM 616944). One recurrent point mutation in the TBL1XR1 gene has been identified as the cause of Pierpont syndrome (OMIM 602342), a distinct intellectual disability syndrome with plantar lipomatosis. In addition, different de novo point mutations in the TBL1XR1 gene have been found in patients with autism spectrum disorders (ASD) and intellectual disability. Here, we report four patients from two unrelated families in whom array-CGH analysis and real-time quantitative PCR of genomic DNA revealed a TBL1XR1-microduplication. Adjacent genes were not affected. The microduplication occurred as a de novo event in one patient, whereas the other three cases occurred in two generations of a second, unrelated family. We compare and contrast the clinical findings in TBL1XR1 microdeletion, point mutation, and microduplication cases and expand the TBL1XR1-associated phenotypic spectrum. ID, hearing loss, and ASD are common features of TBL1XR1-associated diseases. Our clinical observations add to the increasing evidence of the role of TBL1XR1 in brain development, and they simultaneously demonstrate that different genetic disease mechanisms affecting TBL1XR1 can lead to similar ID phenotypes. The TBL1XR1-microduplication syndrome is an intellectual disability/learning disability syndrome with associated incomplete penetrance ASD, hearing loss, and delay of puberty. Its phenotypic overlap indicates that it is a genomic sister-disorder to the 3q26.32 microdeletion syndrome.


Asunto(s)
Trastorno del Espectro Autista/genética , Pérdida Auditiva/genética , Discapacidad Intelectual/genética , Proteínas Nucleares/genética , Receptores Citoplasmáticos y Nucleares/genética , Proteínas Represoras/genética , Adolescente , Adulto , Trastorno del Espectro Autista/fisiopatología , Niño , Cromosomas Humanos Par 3/genética , Hibridación Genómica Comparativa , Femenino , Duplicación de Gen , Genómica , Pérdida Auditiva/fisiopatología , Humanos , Discapacidad Intelectual/fisiopatología , Masculino , Maduración Sexual/genética , Hermanos
10.
Cell Mol Life Sci ; 73(10): 2089-104, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26573968

RESUMEN

Spinal muscular atrophy (SMA) is a devastating motoneuron (MN) disorder caused by homozygous loss of SMN1. Rarely, SMN1-deleted individuals are fully asymptomatic despite carrying identical SMN2 copies as their SMA III-affected siblings suggesting protection by genetic modifiers other than SMN2. High plastin 3 (PLS3) expression has previously been found in lymphoblastoid cells but not in fibroblasts of asymptomatic compared to symptomatic siblings. To find out whether PLS3 is also upregulated in MNs of asymptomatic individuals and thus a convincing SMA protective modifier, we generated induced pluripotent stem cells (iPSCs) from fibroblasts of three asymptomatic and three SMA III-affected siblings from two families and compared these to iPSCs from a SMA I patient and control individuals. MNs were differentiated from iPSC-derived small molecule neural precursor cells (smNPCs). All four genotype classes showed similar capacity to differentiate into MNs at day 8. However, SMA I-derived MN survival was significantly decreased while SMA III- and asymptomatic-derived MN survival was moderately reduced compared to controls at day 27. SMN expression levels and concomitant gem numbers broadly matched SMN2 copy number distribution; SMA I presented the lowest levels, whereas SMA III and asymptomatic showed similar levels. In contrast, PLS3 was significantly upregulated in mixed MN cultures from asymptomatic individuals pinpointing a tissue-specific regulation. Evidence for strong PLS3 accumulation in shaft and rim of growth cones in MN cultures from asymptomatic individuals implies an important role in neuromuscular synapse formation and maintenance. These findings provide strong evidence that PLS3 is a genuine SMA protective modifier.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Neuronas Motoras/patología , Atrofia Muscular Espinal/genética , Células-Madre Neurales/citología , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Regulación hacia Arriba , Biopsia , Diferenciación Celular , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Eliminación de Gen , Silenciador del Gen , Vectores Genéticos , Genotipo , Humanos , Inmunohistoquímica , Células Madre Pluripotentes Inducidas/citología , Cariotipificación , Linfocitos/citología , Masculino , Microscopía Confocal , Mutación , Linaje , Fenotipo , ARN Interferente Pequeño/metabolismo , Piel/patología
11.
Hum Mutat ; 37(7): 703-9, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27071356

RESUMEN

Biallelic loss-of-function mutations in SPG11 cause a wide spectrum of recessively inherited, neurodegenerative disorders including hereditary spastic paraplegia (HSP), amyotrophic lateral sclerosis, and Charcot-Marie-Tooth disease. By comprehensive screening of three large cohorts of HSP index patients, we identified 83 alleles with "small" mutations and 13 alleles that carry large genomic rearrangements. Including relevant data from previous studies, we estimate that copy number variants (CNVs) account for ∼19% of pathogenic SPG11 alleles. The breakpoints for all novel and some previously reported CNVs were determined by long-range PCR and sequencing. This revealed several Alu-associated recombination hotspots. We also found evidence for additional mutational mechanisms, including for a two-step event in which an Alu retrotransposition preceded the actual rearrangement. Apparently independent samples with identical breakpoints were analyzed by microsatellite PCRs. The resulting haplotypes suggested the existence of two rearrangement founder alleles. Our findings widen the spectra of mutations and mutational mechanisms in SPG11, underscore the pivotal role played by Alus, and are of high diagnostic relevance for a wide spectrum of clinical phenotypes including the most frequent form of recessive HSP.


Asunto(s)
Variaciones en el Número de Copia de ADN , Proteínas/genética , Paraplejía Espástica Hereditaria/genética , Alelos , Elementos Alu , Puntos de Rotura del Cromosoma , Cromosomas Humanos/genética , Efecto Fundador , Humanos , Mutación , Análisis de Secuencia de ADN
12.
Hum Mutat ; 37(2): 170-4, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26593283

RESUMEN

Deafblindness is part of several genetic disorders. We investigated a consanguineous Egyptian family with two siblings affected by congenital hearing loss and retinal degeneration, initially diagnosed as Usher syndrome type 1. At teenage, severe enamel dysplasia, developmental delay, and microcephaly became apparent. Genome-wide homozygosity mapping and whole-exome sequencing detected a homozygous missense mutation, c.1238G>T (p.Gly413Val), affecting a highly conserved residue of peroxisomal biogenesis factor 6, PEX6. Biochemical profiling of the siblings revealed abnormal and borderline plasma phytanic acid concentration, and cerebral imaging revealed white matter disease in both. We show that Pex6 localizes to the apical extensions of secretory ameloblasts and differentiated odontoblasts at early stages of dentin synthesis in mice, and to cilia of retinal photoreceptor cells. We propose PEX6, and possibly other peroxisomal genes, as candidate for the rare cooccurrence of deafblindness and enamel dysplasia. Our study for the first time links peroxisome biogenesis disorders to retinal ciliopathies.


Asunto(s)
Adenosina Trifosfatasas/genética , Trastornos Sordoceguera/genética , Hipoplasia del Esmalte Dental/genética , Microcefalia/genética , Mutación Missense , Degeneración Retiniana/genética , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfatasas/metabolismo , Ameloblastos/metabolismo , Ameloblastos/patología , Secuencia de Aminoácidos , Animales , Niño , Cilios/metabolismo , Cilios/patología , Consanguinidad , Trastornos Sordoceguera/metabolismo , Trastornos Sordoceguera/patología , Hipoplasia del Esmalte Dental/metabolismo , Hipoplasia del Esmalte Dental/patología , Femenino , Expresión Génica , Homocigoto , Humanos , Masculino , Ratones , Microcefalia/metabolismo , Microcefalia/patología , Datos de Secuencia Molecular , Odontoblastos/metabolismo , Odontoblastos/patología , Linaje , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Hermanos , Sustancia Blanca/metabolismo , Sustancia Blanca/patología , Adulto Joven
13.
Am J Hum Genet ; 93(1): 181-90, 2013 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-23830518

RESUMEN

Myopathies are a clinically and etiologically heterogeneous group of disorders that can range from limb girdle muscular dystrophy (LGMD) to syndromic forms with associated features including intellectual disability. Here, we report the identification of mutations in transport protein particle complex 11 (TRAPPC11) in three individuals of a consanguineous Syrian family presenting with LGMD and in five individuals of Hutterite descent presenting with myopathy, infantile hyperkinetic movements, ataxia, and intellectual disability. By using a combination of whole-exome or genome sequencing with homozygosity mapping, we identified the homozygous c.2938G>A (p.Gly980Arg) missense mutation within the gryzun domain of TRAPPC11 in the Syrian LGMD family and the homozygous c.1287+5G>A splice-site mutation resulting in a 58 amino acid in-frame deletion (p.Ala372_Ser429del) in the foie gras domain of TRAPPC11 in the Hutterite families. TRAPPC11 encodes a component of the multiprotein TRAPP complex involved in membrane trafficking. We demonstrate that both mutations impair the binding ability of TRAPPC11 to other TRAPP complex components and disrupt the Golgi apparatus architecture. Marker trafficking experiments for the p.Ala372_Ser429del deletion indicated normal ER-to-Golgi trafficking but dramatically delayed exit from the Golgi to the cell surface. Moreover, we observed alterations of the lysosomal membrane glycoproteins lysosome-associated membrane protein 1 (LAMP1) and LAMP2 as a consequence of TRAPPC11 dysfunction supporting a defect in the transport of secretory proteins as the underlying pathomechanism.


Asunto(s)
Discapacidad Intelectual/genética , Trastornos del Movimiento/genética , Enfermedades Musculares/genética , Distrofia Muscular de Cinturas/genética , Eliminación de Secuencia , Proteínas de Transporte Vesicular/metabolismo , Adolescente , Adulto , Ataxia/genética , Mapeo Cromosómico , Consanguinidad , Creatina Quinasa/sangre , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Exoma , Femenino , Aparato de Golgi/genética , Aparato de Golgi/metabolismo , Aparato de Golgi/patología , Homocigoto , Humanos , Proteína 2 de la Membrana Asociada a los Lisosomas , Proteínas de Membrana de los Lisosomas/genética , Proteínas de Membrana de los Lisosomas/metabolismo , Lisosomas/metabolismo , Masculino , Trastornos del Movimiento/patología , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Enfermedades Musculares/patología , Distrofia Muscular de Cinturas/patología , Linaje , Unión Proteica , Transporte de Proteínas , Sitios de Empalme de ARN , Siria , Proteínas de Transporte Vesicular/genética , Adulto Joven
14.
Neuropediatrics ; 47(4): 273-7, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27214504

RESUMEN

Distal arthrogryposis (DA) is a feature in genetically and clinically heterogeneous groups of disorders. Mostly myopathic and neurogenic defects have been described, but many patients remain without genetic diagnosis. We are elaborating on the clinical presentation of neonatal cases with DA who carry novel mutations in the nonselective sodium leak channel (NALCN). Two patients reported herein were remarkable for central hypertonicity in addition to DA. By trio-whole exome sequencing, two undescribed de novo mutations in NALCN were revealed. Both mutations (p.F317C and p.V595F) are located on pore-forming segments of NALCN. Dominant NALCN mutations in the pore-forming segments have been identified in similar patients, whereas recessive mutations outside the pore-forming segments result in different phenotypes. Our findings with central hypertonia broaden the phenotypic spectrum of de novo mutations in the pore-forming segments of NALCN. Recent findings of successful acetazolamide treatment in patients with channelopathies might point to potential therapies based on the ion channel similarities and the location of the mutation.


Asunto(s)
Disostosis Craneofacial/genética , Hipertonía Muscular/genética , Canales de Sodio/genética , Encéfalo/diagnóstico por imagen , Disostosis Craneofacial/complicaciones , Disostosis Craneofacial/diagnóstico , Disostosis Craneofacial/diagnóstico por imagen , Femenino , Humanos , Recién Nacido , Recien Nacido Prematuro , Canales Iónicos , Imagen por Resonancia Magnética , Proteínas de la Membrana , Hipertonía Muscular/complicaciones , Hipertonía Muscular/diagnóstico , Radiografía Torácica
15.
Hum Mol Genet ; 22(2): 398-407, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23077215

RESUMEN

Functional loss of SMN1 causes proximal spinal muscular atrophy (SMA), the most common genetic condition accounting for infant lethality. Hence, the hypomorphic copy gene SMN2 is the only resource of functional SMN protein in SMA patients and influences SMA severity in a dose-dependent manner. Consequently, current therapeutic approaches focus on SMN2. Histone deacetylase inhibitors (HDACi), such as the short chain fatty acid VPA (valproic acid), ameliorate the SMA phenotype by activating the SMN2 expression. By analyzing blood SMN2 expression in 16 VPA-treated SMA patients, about one-third of individuals were identified as positive responders presenting increased SMN2 transcript levels. In 66% of enrolled patients, a concordant response was detected in the respective fibroblasts. Most importantly, by taking the detour of reprograming SMA patients' fibroblasts, we showed that the VPA response was maintained even in GABAergic neurons derived from induced pluripotent stem cells (iPS) cells. Differential expression microarray analysis revealed a complete lack of response to VPA in non-responders, which was associated with an increased expression of the fatty acid translocase CD36. The pivotal role of CD36 as the cause of non-responsiveness was proven in various in vitro approaches. Most importantly, knockdown of CD36 in SMA fibroblasts converted non- into pos-responders. In summary, the concordant response from blood to the central nervous system (CNS) to VPA may allow selection of pos-responders prior to therapy. Increased CD36 expression accounts for VPA non-responsiveness. These findings may be essential not only for SMA but also for other diseases such as epilepsy or migraine frequently treated with VPA.


Asunto(s)
Antígenos CD36/metabolismo , Atrofia Muscular Espinal/tratamiento farmacológico , Atrofia Muscular Espinal/metabolismo , Ácido Valproico/uso terapéutico , Antígenos CD36/genética , Línea Celular , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Neuronas GABAérgicas/efectos de los fármacos , Neuronas GABAérgicas/metabolismo , Perfilación de la Expresión Génica , Humanos , Atrofia Muscular Espinal/genética , Ácido Valproico/farmacología
16.
Hum Mutat ; 35(10): 1153-62, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25044745

RESUMEN

We describe a consanguineous Iraqi family with Leber congenital amaurosis (LCA), Joubert syndrome (JBTS), and polycystic kidney disease (PKD). Targeted next-generation sequencing for excluding mutations in known LCA and JBTS genes, homozygosity mapping, and whole-exome sequencing identified a homozygous missense variant, c.317G>C (p.Arg106Pro), in POC1B, a gene essential for ciliogenesis, basal body, and centrosome integrity. In silico modeling suggested a requirement of p.Arg106 for the formation of the third WD40 repeat and a protein interaction interface. In human and mouse retina, POC1B localized to the basal body and centriole adjacent to the connecting cilium of photoreceptors and in synapses of the outer plexiform layer. Knockdown of Poc1b in zebrafish caused cystic kidneys and retinal degeneration with shortened and reduced photoreceptor connecting cilia, compatible with the human syndromic ciliopathy. A recent study describes homozygosity for p.Arg106ProPOC1B in a family with nonsyndromic cone-rod dystrophy. The phenotype associated with homozygous p.Arg106ProPOC1B may thus be highly variable, analogous to homozygous p.Leu710Ser in WDR19 causing either isolated retinitis pigmentosa or Jeune syndrome. Our study indicates that POC1B is required for retinal integrity, and we propose POC1B mutations as a probable cause for JBTS with severe PKD.


Asunto(s)
Proteínas de Ciclo Celular/genética , Enfermedades Cerebelosas/genética , Anomalías del Ojo/genética , Enfermedades Renales Quísticas/genética , Mutación , Retina/anomalías , Anomalías Múltiples , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Proteínas de Ciclo Celular/metabolismo , Enfermedades Cerebelosas/metabolismo , Enfermedades Cerebelosas/patología , Cerebelo/anomalías , Niño , Cilios/metabolismo , Cilios/ultraestructura , Anomalías del Ojo/metabolismo , Anomalías del Ojo/patología , Técnicas de Silenciamiento del Gen , Humanos , Irak , Riñón/patología , Enfermedades Renales Quísticas/metabolismo , Enfermedades Renales Quísticas/patología , Amaurosis Congénita de Leber/genética , Amaurosis Congénita de Leber/metabolismo , Masculino , Ratones , Datos de Secuencia Molecular , Linaje , Retina/metabolismo , Retina/patología , Pez Cebra
18.
Clin Transl Immunology ; 13(2): e1493, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38410820

RESUMEN

Objectives: Dominant-activating (DA) lesions in RAC2 have been reported in 18 individuals to date. Some have required haematopoietic stem cell transplantation (HSCT) for their (severe) combined immunodeficiency syndrome phenotype. We aimed to investigate clinical and cellular features of a kindred harbouring a novel variant in RAC2 p.Ile21Ser (I21S) to better understand DA lesions' phenotypic spectrum. Methods: Clinical and immunological information was collated for seven living individuals from the same kindred with RAC2 p.I21S. We evaluated neutrophil morphology, RAC2 protein expression and superoxide production using freshly isolated neutrophils stimulated with phorbol-12-myristate-13-acetate (PMA) and N-formyl-MetLeuPhe (fMLP). Results: Patient 1 (P1, aged 11, male) has a history of bacterial suppurative otitis media, viral and bacterial cutaneous infections. P1's siblings (P2, P3), mother (P4), maternal aunt (P5) and uncle (P6) have similar infection histories. P1's maternal cousin (P7) presented with Burkitt's lymphoma at age 9. All affected individuals are alive and none has required HSCT to date. They have chronic lymphopenia affecting the CD4+T and B-cell compartments. P1-3 have isolated reduction in IgM levels whereas the adults universally have normal immunoglobulins. Specific antibody responses are preserved. Affected individuals have neutrophil vacuolation, and their neutrophils have enhanced superoxide production compared to healthy controls. Conclusion: RAC2 p.I21S is an activating variant causing notable morphological and functional abnormalities similar to other reported DA mutations. This novel variant expands the broad clinical phenotypic spectrum of RAC2 DA lesions, emphasising the need to tailor clinical management according to patients' disease phenotype and severity.

20.
Brain Commun ; 5(1): fcad017, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36793789

RESUMEN

Superoxide dismutase-1 is a ubiquitously expressed antioxidant enzyme. Mutations in SOD1 can cause amyotrophic lateral sclerosis, probably via a toxic gain-of-function involving protein aggregation and prion-like mechanisms. Recently, homozygosity for loss-of-function mutations in SOD1 has been reported in patients presenting with infantile-onset motor neuron disease. We explored the bodily effects of superoxide dismutase-1 enzymatic deficiency in eight children homozygous for the p.C112Wfs*11 truncating mutation. In addition to physical and imaging examinations, we collected blood, urine and skin fibroblast samples. We used a comprehensive panel of clinically established analyses to assess organ function and analysed oxidative stress markers, antioxidant compounds, and the characteristics of the mutant Superoxide dismutase-1. From around 8 months of age, all patients exhibited progressive signs of both upper and lower motor neuron dysfunction, cerebellar, brain stem, and frontal lobe atrophy and elevated plasma neurofilament concentration indicating ongoing axonal damage. The disease progression seemed to slow down over the following years. The p.C112Wfs*11 gene product is unstable, rapidly degraded and no aggregates were found in fibroblast. Most laboratory tests indicated normal organ integrity and only a few modest deviations were found. The patients displayed anaemia with shortened survival of erythrocytes containing decreased levels of reduced glutathione. A variety of other antioxidants and oxidant damage markers were within normal range. In conclusion, non-neuronal organs in humans show a remarkable tolerance to absence of Superoxide dismutase-1 enzymatic activity. The study highlights the enigmatic specific vulnerability of the motor system to both gain-of-function mutations in SOD1 and loss of the enzyme as in the here depicted infantile superoxide dismutase-1 deficiency syndrome.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA