Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Pediatr Gastroenterol Nutr ; 65(1): e5-e15, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28644354

RESUMEN

OBJECTIVE: Monogenic defects in the interleukin-10 (IL-10) pathway are extremely rare and cause infantile-onset inflammatory bowel disease (IBD)-like pathology. Understanding how immune responses are dysregulated in monogenic IBD-like diseases can provide valuable insight in "classical" IBD pathogenesis. Here, we studied long-term immune cell development and function in an adolescent IL-10 receptor (IL10RA)-deficient patient who presented in infancy with severe colitis and fistulizing perianal disease and is currently treated with immune suppressants. METHODS: Biomaterial was collected from the IL10RA-deficient patient, pediatric patients with IBD, and healthy controls. The frequency and phenotype of immune cells were determined in peripheral blood and intestinal biopsies by flow cytometry and immunohistochemistry. Functional changes in monocyte-derived dendritic cells and T cells were assessed by in vitro activation assays. RESULTS: The IL10RA-deficient immune system developed normally with respect to numbers and phenotype of circulating immune cells. Despite normal co-stimulatory molecule expression, bacterial lipopolysaccharide-stimulated monocyte-derived dendritic cells from the IL10RA-deficient patient released increased amounts of tumor necrosis factor α compared to healthy controls. Upon T-cell receptor ligation, IL10RA-deficient peripheral blood mononuclear cells released increased amounts of T-cell cytokines interferon γ and IL-17 agreeing with high numbers of T-bet and IL-17 cells in intestinal biopsies taken at disease onset. In vitro, the immunosuppressive drug thalidomide used to treat the patient's decreased peripheral blood mononuclear cell-derived tumor necrosis factor production. CONCLUSIONS: With time and during immunosuppressive treatment the IL10RA-deficient immune system develops relatively normally. Upon activation, IL-10 is crucial for controlling excessive inflammatory cytokine release by dendritic cells and preventing interferon γ and IL-17-mediated T-cell responses.


Asunto(s)
Inmunidad Adaptativa/fisiología , Células Dendríticas/metabolismo , Inmunidad Innata/fisiología , Enfermedades Inflamatorias del Intestino/inmunología , Subunidad alfa del Receptor de Interleucina-10/deficiencia , Subgrupos de Linfocitos T/metabolismo , Adolescente , Adulto , Biomarcadores/metabolismo , Estudios de Casos y Controles , Niño , Preescolar , Codón sin Sentido , Femenino , Mutación del Sistema de Lectura , Marcadores Genéticos , Humanos , Lactante , Enfermedades Inflamatorias del Intestino/genética , Subunidad alfa del Receptor de Interleucina-10/genética , Masculino , Persona de Mediana Edad
2.
Genet Med ; 17(11): 843-53, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25719457

RESUMEN

Two proα1(IV) chains, encoded by COL4A1, form trimers that contain, in addition, a proα2(IV) chain encoded by COL4A2 and are the major component of the basement membrane in many tissues. Since 2005, COL4A1 mutations have been known as an autosomal dominant cause of hereditary porencephaly. COL4A1 and COL4A2 mutations have been reported with a broader spectrum of cerebrovascular, renal, ophthalmological, cardiac, and muscular abnormalities, indicated as "COL4A1 mutation-related disorders." Genetic counseling is challenging because of broad phenotypic variation and reduced penetrance. At the Erasmus University Medical Center, diagnostic DNA analysis of both COL4A1 and COL4A2 in 183 index patients was performed between 2005 and 2013. In total, 21 COL4A1 and 3 COL4A2 mutations were identified, mostly in children with porencephaly or other patterns of parenchymal hemorrhage, with a high de novo mutation rate of 40% (10/24). The observations in 13 novel families harboring either COL4A1 or COL4A2 mutations prompted us to review the clinical spectrum. We observed recognizable phenotypic patterns and propose a screening protocol at diagnosis. Our data underscore the importance of COL4A1 and COL4A2 mutations in cerebrovascular disease, also in sporadic patients. Follow-up data on symptomatic and asymptomatic mutation carriers are needed for prognosis and appropriate surveillance.


Asunto(s)
Colágeno Tipo IV/genética , Estudios de Asociación Genética , Mutación , Fenotipo , Alelos , Segmento Anterior del Ojo/anomalías , Encéfalo/patología , Hemorragia Cerebral/diagnóstico , Hemorragia Cerebral/genética , Estudios de Cohortes , Anomalías del Ojo/diagnóstico , Anomalías del Ojo/genética , Enfermedades Hereditarias del Ojo , Familia , Orden Génico , Sitios Genéticos , Genotipo , Humanos , Leucomalacia Periventricular/diagnóstico , Leucomalacia Periventricular/genética , Imagen por Resonancia Magnética/métodos , Linaje , Porencefalia/diagnóstico , Porencefalia/genética
3.
Am J Med Genet A ; 164A(1): 113-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24243761

RESUMEN

So far only mutations in the filamin A gene (FLNA) have been identified as causing familial mitral valve prolapse (MVP). Previous studies have linked dysregulation of the transforming growth factor beta (TGF-ß) cytokine family to MVP. We investigated whether mutations in the TGF-ß receptors genes type I (TGFBR1) and II (TGFBR2) underlie isolated familial MVP cases. Eight families with isolated familial MVP were evaluated clinically and genetically. Ventricular arrhythmias were present in five of the eight families and sudden cardiac death occurred in six patients. Tissue obtained during mitral valve surgery or autopsy was available for histological examination in six cases; all demonstrated myxomatous degeneration. A previously described FLNA missense mutation (p.G288R) was identified in one large family, but no mutations were discovered in TGFBR1 or TGFBR2. An FLNA missense mutation was identified in one family but we found no TGFBR1 or TGFBR2 mutations. Our results suggest that TGFBR1 and TGFBR2 mutations do not play a major role in isolated myxomatous valve dystrophy. Screening for FLNA mutations is recommended in familial myxomatous valvular dystrophy, particularly if X-linked inheritance is suspected.


Asunto(s)
Filaminas/genética , Prolapso de la Válvula Mitral/diagnóstico , Prolapso de la Válvula Mitral/genética , Proteínas Serina-Treonina Quinasas/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Adolescente , Adulto , Femenino , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Válvula Mitral/patología , Mutación , Linaje , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Adulto Joven
4.
Am J Med Genet A ; 161A(9): 2376-84, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23873601

RESUMEN

Mutations in FLNA (Filamin A, OMIM 300017) cause X-linked periventricular nodular heterotopia (XL-PNH). XL-PNH-associated mutations are considered lethal in hemizygous males. However, a few males with unusual mutations (including distal truncating and hypomorphic missense mutations), and somatic mosaicism have been reported to survive past infancy. Two brothers had an atypical presentation with failure to thrive and distinct facial appearance including hypertelorism. Evaluations of these brothers and their affected cousin showed systemic involvement including severe intestinal malfunction, malrotation, congenital short bowel, PNH, pyloric stenosis, wandering spleen, patent ductus arteriosus, atrial septal defect, inguinal hernia, and vesicoureteral reflux. The unanticipated finding of PNH led to FLNA testing and subsequent identification of a novel no-stop FLNA mutation (c.7941_7942delCT, p.(*2648Serext*100)). Western blotting and qRT-PCR of patients' fibroblasts showed diminished levels of protein and mRNA. This FLNA mutation, the most distal reported so far, causes in females classical XL-PNH, but in males an unusual, multi-organ phenotype, providing a unique insight into the FLNA-associated phenotypes.


Asunto(s)
Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Filaminas/genética , Mutación Missense , Secuencia de Bases , Encéfalo/patología , Facies , Femenino , Genotipo , Humanos , Lactante , Imagen por Resonancia Magnética , Masculino , Linaje , Heterotopia Nodular Periventricular/diagnóstico , Heterotopia Nodular Periventricular/genética , Fenotipo , Radiografía , Bazo/diagnóstico por imagen , Bazo/patología
6.
Am J Med Genet A ; 158A(6): 1472-6, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22585566

RESUMEN

Mutations in the ARX gene, at Xp22.3, cause several disorders, including infantile spasms, X-linked lissencephaly with abnormal genitalia (XLAG), callosal agenesis and isolated intellectual disability. Genotype/phenotype studies suggested that polyalanine tract expansion is associated with non-malformative phenotypes, while missense and nonsense mutations cause cerebral malformations, however, patients with structural normal brain and missense mutations have been reported. We report on a male patient born with cleft lip and palate who presented with infantile spasms and hemiplegia. MRI showed agenesis of corpus callosum (ACC), an interhemispheric cyst, periventricular nodular heterotopia (PVNH), and extensive left frontal polymicrogyria (PMG). Sequencing of the ARX gene in the patient identified a six basepair insertion (c.335ins6, exon 2). The insertion leads to a two-residue expansion of the first polyalanine tract and was described previously in a family with non-syndromic X-linked mental retardation. To our knowledge, ARX mutation causing PMG and PVNH is unique, but the spasms and ACC are common in ARX mutations. Clinicians should be aware of the broad clinical range of ARX mutations, and further studies are necessary to investigate the association with PMG and PVNH and to identify possible modifying factors.


Asunto(s)
Proteínas de Homeodominio/genética , Malformaciones del Desarrollo Cortical/genética , Mutación , Heterotopia Nodular Periventricular/genética , Factores de Transcripción/genética , Facies , Heterocigoto , Humanos , Lactante , Cariotipo , Masculino , Malformaciones del Desarrollo Cortical/diagnóstico , Mutagénesis Insercional , Neuroimagen , Heterotopia Nodular Periventricular/diagnóstico
7.
Ann Neurol ; 68(6): 876-87, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21061399

RESUMEN

OBJECTIVE: Mucopolysaccharidosis (MPS) IIIA (Sanfilippo syndrome type A) is a lysosomal storage disorder caused by deficiency of the enzyme sulfamidase. Information on the natural course of MPS IIIA is scarce, but is much needed in view of emerging therapies. METHODS: Clinical history and molecular defects of all 110 MPS IIIA patients identified by enzymatic studies in the Netherlands were collected and included in this study. RESULTS: First clinical signs, mainly consisting of delayed speech development and behavioral problems, were noted between the ages of 1 and 6 years. Other symptoms included sleeping and hearing problems, recurrent upper airway infections, diarrhea, and epilepsy. The clinical course varied remarkably and could be correlated with the molecular defects. The frequent pathogenic mutations p.R245H, p.Q380R, p.S66W, and c.1080delC were associated with the classical severe phenotype. Patients compound heterozygous for the p.S298P mutation in combination with 1 of the mutations associated with the classical severe phenotype had a significantly longer preservation of psychomotor functions and a longer survival. Two patients homozygous for the p.S298P mutation, and 4 patients from 3 families heterozygous for 3 missense variants not reported previously (p.T421R, p.P180L, and p.L12Q), showed a remarkably attenuated phenotype. INTERPRETATION: We report the natural history and mutational analysis in a large unbiased cohort of MPS IIIA patients. We demonstrate that the clinical spectrum of MPS IIIA is much broader than previously reported. A significant genotype-phenotype correlation was established in this cohort.


Asunto(s)
Estudios de Asociación Genética , Genotipo , Fenotipo , Adolescente , Adulto , Síntomas Conductuales/etiología , Células Cultivadas , Niño , Preescolar , Estudios de Cohortes , Análisis Mutacional de ADN , Epilepsia/etiología , Femenino , Fibroblastos , Trastornos de la Audición/etiología , Humanos , Hidrolasas/genética , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Mucopolisacaridosis III/complicaciones , Mucopolisacaridosis III/enzimología , Mucopolisacaridosis III/genética , Mucopolisacaridosis III/patología , Mutación/genética , Embarazo , Análisis de Regresión , Índice de Severidad de la Enfermedad , Piel/patología , Trastornos del Sueño-Vigilia/etiología , Trastornos de la Visión/etiología , Adulto Joven
8.
Dev Med Child Neurol ; 53(5): 417-21, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21410694

RESUMEN

AIM: To evaluate survival, clinical, and genetic characteristics of all patients with classic or type 1 lissencephaly born between 1972 and 1990 in the Netherlands, who at the time were enrolled in an observational study. METHOD: We re-evaluated 24 patients (11 males, 13 females) for long-term follow-up and survival information. RESULTS: Mean length of follow-up was 14 years (SD 9 y 8 mo). Eleven patients were alive at follow-up. All patients showed severe intellectual disability, intractable epilepsy, and complete dependency on care. Life expectancy was related to the severity of the lissencephaly on neuroimaging. Molecular analysis of the LIS1 gene was not possible at the time of the original study and was now requested by eight parents. This revealed a pathogenic nonsense mutation or deletion in seven patients. INTERPRETATION: Our study provides information about the long-term course of lissencephaly and the relationship between lissencephaly severity and prognosis. It also shows that renewed attention to genetic counselling remains valued by families of patients with a severe congenital neurological disease.


Asunto(s)
Encéfalo/anomalías , Encéfalo/patología , Lisencefalias Clásicas y Heterotopias Subcorticales en Banda/mortalidad , Lisencefalias Clásicas y Heterotopias Subcorticales en Banda/patología , Adulto , Lisencefalias Clásicas y Heterotopias Subcorticales en Banda/diagnóstico por imagen , Femenino , Humanos , Estudios Longitudinales , Masculino , Análisis de Supervivencia , Tomografía Computarizada por Rayos X/métodos , Adulto Joven
9.
Hum Mutat ; 31(1): 52-9, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19830808

RESUMEN

Crigler-Najjar syndrome (CN), caused by deficiency of UGT isoform 1A1 (UGT1A1), is characterized by severe unconjugated hyperbilirubinemia. In this study we have analyzed 19 CN patients diagnosed in The Netherlands (18) and in Belgium (1), and have identified 14 different UGT1A1 mutations, four of which are novel. Two mutations were present in several unrelated patients, suggesting the presence of two founder effects in The Netherlands. In addition, we show linkage of the UGT1A1 *28 promoter polymorphism (rs5719145insTA) to three structural mutations. Functional studies of partial active UGT1A1 mutants are limited. Therefore, we performed in vitro studies to determine the functional activity of seven missense mutants identified in this study and of three reported previously. In addition to bilirubin, we also determined their activity toward eight other UGT1A1 substrates. We demonstrate that five mutants have residual activity that, depending on the substrate, varies from not detectable to 94% of wild-type UGT1A1 activity. The identification of four novel pathogenic mutations and the analysis of residual activity of 10 UGT1A1 missense mutants are useful for clinical diagnosis, and provides new insights in enzyme activity, whereas the identification of two founder mutations will speed up genetic counseling for newly identified CN patients in The Netherlands.


Asunto(s)
Alelos , Síndrome de Crigler-Najjar/genética , Glucuronosiltransferasa/genética , Glucuronosiltransferasa/metabolismo , Mutación Missense , Adolescente , Adulto , Animales , Bélgica , Bilirrubina/metabolismo , Células Cultivadas , Síndrome de Crigler-Najjar/patología , Femenino , Regulación Enzimológica de la Expresión Génica , Genotipo , Humanos , Hiperbilirrubinemia/genética , Insectos , Masculino , Persona de Mediana Edad , Países Bajos , Fenotipo , Adulto Joven
10.
Hum Mutat ; 31(5): E1348-60, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20232353

RESUMEN

Mucopolysaccharidosis III D (Sanfilippo disease type D, MPS IIID) is a rare autosomal recessive lysosomal storage disorder previously described in only 20 patients. MPS IIID is caused by a deficiency of N-acetylglucosamine-6-sulphate sulphatase (GNS), one of the enzymes required for the degradation of heparan sulphate. So far only seven mutations in the GNS gene have been reported. The clinical phenotype of 12 new MPS IIID patients from 10 families was studied. Mutation analysis of GNS was performed in 16 patients (14 index cases). Clinical signs and symptoms of the MPS IIID patients appeared to be similar to previously described patients with MPS III. Early development was normal with onset of behavioral problems around the age of 4 years, followed by developmental stagnation, deterioration of verbal communication and subsequent deterioration of motor functions. Sequence analysis of the coding regions of the gene encoding GNS (GNS) resulted in the identification of 15 novel mutations: 3 missense mutations, 1 nonsense mutation, 4 splice site mutations, 3 frame shift mutations, 3 large deletions and 1 in-frame small deletion. They include the first missense mutations and a relatively high proportion of large rearrangements, which warrants the inclusion of quantitative techniques in routine mutation screening of the GNS gene.


Asunto(s)
Mucopolisacaridosis III/genética , Sulfatasas/deficiencia , Sulfatasas/genética , Adolescente , Adulto , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Humanos , Masculino , Mutación/genética , Fenotipo , Adulto Joven
11.
J Inherit Metab Dis ; 33(6): 759-67, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20852935

RESUMEN

Mucopolysaccharidosis type IIIB (MPS IIIB, Sanfilippo syndrome type B) is a lysosomal storage disorder caused by deficiency of the enzyme N-acetyl-α-D-glucosaminidase (NAGLU). Information on the natural course of MPS IIIB is scarce but much needed in view of emerging therapies. To improve knowledge on the natural course, data on all 52 MPS IIIB patients ever identified by enzymatic studies in the Netherlands were gathered. Clinical data on 44 patients could be retrieved. Only a small number (n = 9; 21%) presented with a classical MPS III phenotype; all other patients showed a much more attenuated course of the disease characterized by a significantly slower regression of intellectual and motor abilities. The majority of patients lived well into adulthood. First signs of the disease, usually mild developmental delay, were observed at a median age of 4 years. Subsequently, patients showed a slowing and eventually a stagnation of development. Patients with the attenuated phenotype had a stable intellectual disability for many years. Molecular analysis was performed in 24 index patients. The missense changes p.R643C, p.S612G, p.E634K, and p.L497V were exclusively found in patients with the attenuated phenotype. MPS IIIB comprises a remarkably wide spectrum of disease severity, and an unselected cohort including all Dutch patients showed a large proportion (79%) with an attenuated phenotype. MPS IIIB must be considered in patients with a developmental delay, even in the absence of a progressive decline in intellectual abilities. A key feature, necessitating metabolic studies, is the coexistence of behavioral problems.


Asunto(s)
Fenotipo , Algoritmos , Causas de Muerte , Células Cultivadas , Niño , Preescolar , Parto Obstétrico/métodos , Parto Obstétrico/estadística & datos numéricos , Diagnóstico Diferencial , Femenino , Humanos , Lactante , Masculino , Mucopolisacaridosis III/clasificación , Mucopolisacaridosis III/diagnóstico , Mucopolisacaridosis III/epidemiología , Mucopolisacaridosis III/mortalidad , Embarazo , Sistema de Registros
12.
Eur J Hum Genet ; 27(6): 919-927, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30737479

RESUMEN

Analyses in our diagnostic DNA laboratory include genes involved in autosomal recessive (AR) lysosomal storage disorders such as glycogenosis type II (Pompe disease) and mucopolysaccharidosis type I (MPSI, Hurler disease). We encountered 4 cases with apparent homozygosity for a disease-causing sequence variant that could be traced to one parent only. In addition, in a young child with cardiomyopathy, in the absence of other symptoms, a diagnosis of Pompe disease was considered. Remarkably, he presented with different enzymatic and genotypic features between leukocytes and skin fibroblasts. All cases were examined with microsatellite markers and SNP genotyping arrays. We identified one case of total uniparental disomy (UPD) of chromosome 17 leading to Pompe disease and three cases of segmental uniparental isodisomy (UPiD) causing Hurler-(4p) or Pompe disease (17q). One Pompe patient with unusual combinations of features was shown to have a mosaic segmental UPiD of chromosome 17q. The chromosome 17 UPD cases amount to 11% of our diagnostic cohort of homozygous Pompe patients (plus one case of pseudoheterozygosity) where segregation analysis was possible. We conclude that inclusion of parental DNA is mandatory for reliable DNA diagnostics. Mild or unusual phenotypes of AR diseases should alert physicians to the possibility of mosaic segmental UPiD. SNP genotyping arrays are used in diagnostic workup of patients with developmental delay. Our results show that even small Regions of Homozygosity that include telomeric areas are worth reporting, regardless of the imprinting status of the chromosome, as they might indicate segmental UPiD.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Mucopolisacaridosis I/genética , Polimorfismo de Nucleótido Simple , Disomía Uniparental , Adolescente , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino
13.
Eur J Hum Genet ; 16(8): 875-9, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18301443

RESUMEN

We discuss four cases of acid alpha-glucosidase deficiency (EC, 3.2.1.3/20) without evident symptoms of Pompe disease (OMIM No 232300) in individuals of Asian descent. In three cases, the deficiency was associated with homozygosity for the sequence variant c.[1726G>A; 2065G>A] in the acid alpha-glucosidase gene (GAA) translating into p.[G576S; E689K]. One of these cases was a patient with profound muscular atrophy, another had cardio-myopathy and the third had no symptoms. The fourth case, the mother of a child with Pompe disease, was compound heterozygote for the GAA sequence variants c.[1726G>A; 2065G>A]/c.2338G>A (p.W746X) and had no symptoms either. Further investigations revealed that c.[1726A; 2065A] is a common GAA allele in the Japanese and Chinese populations. Our limited study predicts that approximately 4% of individuals in these populations are homozygote c.[1726A; 2065A]. The height of this figure in contrast to the rarity of Pompe disease in Asian populations and the clinical history of the cases described in this paper virtually exclude that homozygosity for c.[1726A; 2065A] causes Pompe disease. As c.[1726A; 2065A] homozygotes have been observed with similarly low acid alpha-glucosidase activity as some patients with Pompe disease, we caution they may present as false positives in newborn screening programs especially in Asian populations.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Polimorfismo Genético , alfa-Glucosidasas/genética , Adolescente , Adulto , Células Cultivadas , Niño , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo II/epidemiología , Homocigoto , Humanos , Leucocitos/citología , Leucocitos/metabolismo , Linfocitos/citología , Linfocitos/metabolismo , Masculino , Músculos/citología , Músculos/metabolismo
14.
Arch Neurol ; 65(3): 358-66, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18332248

RESUMEN

BACKGROUND: Malformations of cortical development (MCDs) are a major source of handicap. Much progress in understanding the genetic causes has been made recently. The number of affected children in whom a molecularly confirmed diagnosis can be made is unclear. OBJECTIVE: To evaluate the etiology of MCDs in children and the effect of a combined radiological, clinical, and syndrome classification. DESIGN: A case series of 113 children with a radiological diagnosis of MCD from January 1, 1992, to January 1, 2006. SETTING: The Erasmus Medical Center-Sophia Children's Hospital, a secondary and tertiary referral center. PATIENTS: Patients with MCD underwent a complete radiological, clinical, and neurological assessment and testing for known genes involved in the pathogenesis of MCD as appropriate for their phenotype. RESULTS: We established an etiological diagnosis in 45 of 113 cases (40%). For 21 patients (19%), this included molecular and/or genetic confirmation (Miller-Dieker syndrome; LIS1, DCX, FLNA, EIF2AK3, or KIAA1279 mutations; or an inborn error of metabolism). In 17 (15%), a syndrome with an unknown genetic defect was diagnosed. In 7 patients (6%), we found evidence of a gestational insult. Of the remaining 68 patients, 34 probably have a yet-unknown genetic disorder based on the presence of multiple congenital anomalies (15 patients), a family history with multiple affected persons (12 patients), or consanguineous parents (7 patients). CONCLUSIONS: In our cohort, combining diagnostic molecular testing with clinical, radiological, and genetic classification; syndrome identification; and family study provided a diagnosis in 40% of the cases of MCD. This contributes to the possibility of prenatal diagnosis and improved patient treatment and disease management.


Asunto(s)
Corteza Cerebral/diagnóstico por imagen , Corteza Cerebral/patología , Malformaciones del Sistema Nervioso/patología , Adolescente , Niño , Preescolar , Estudios de Cohortes , Estudios de Evaluación como Asunto , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Malformaciones del Desarrollo Cortical/genética , Malformaciones del Desarrollo Cortical/patología , Malformaciones del Sistema Nervioso/clasificación , Malformaciones del Sistema Nervioso/etiología , Malformaciones del Sistema Nervioso/genética , Fenotipo , Radiografía , Estudios Retrospectivos
15.
Hum Mutat ; 27(7): 654-66, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16683254

RESUMEN

Rapid and reliable identification of deleterious changes in the breast cancer genes BRCA1 and BRCA2 has become one of the major issues in most DNA services laboratories. To rapidly detect all possible changes within the coding and splice site determining sequences of the breast cancer genes, we established a semiautomated denaturing gradient gel electrophoresis (DGGE) mutation scanning system. All exons of both genes are covered by the DGGE scan, comprising 120 amplicons. We use a semiautomated approach, amplifying all individual amplicons with the same PCR program, after which the amplicons are pooled. DGGE is performed using three slightly different gel conditions. Validation was performed using DNA samples with known sequence variants in 107 of the 120 amplicons; all variants were detected. This DGGE mutation scanning, in combination with a PCR test for two Dutch founder deletions in BRCA1 was then applied in 431 families in which 52 deleterious changes and 70 unclassified variants were found. Fifteen unclassified variants were not reported before. The system was easily adopted by five other laboratories, where in another 3,593 families both exons 11 were analyzed by the protein truncation test (PTT) and the remaining exons by DGGE. In total, a deleterious change (nonsense, frameshift, splice-site mutation, or large deletion) was found in 661 families (16.4%), 462 in BRCA1 (11.5%), 197 in BRCA2 (4.9%), and in two index cases a deleterious change in both BRCA1 and BRCA2 was identified. Eleven deleterious changes in BRCA1 and 36 in BRCA2 had not been reported before. In conclusion, this DGGE mutation screening method for BRCA1 and BRCA2 is proven to be highly sensitive and is easy to adopt, which makes screening of large numbers of patients feasible. The results of screening of BRCA1 and BRCA2 in more than 4,000 families present a valuable overview of mutations in the Dutch population.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Análisis Mutacional de ADN/métodos , Electroforesis en Gel de Poliacrilamida , Genes BRCA1 , Genes BRCA2 , Pruebas Genéticas/métodos , Neoplasias Ováricas/diagnóstico , Instituciones de Atención Ambulatoria , Femenino , Efecto Fundador , Humanos , Masculino , Países Bajos
16.
Eur J Hum Genet ; 13(1): 59-68, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15483652

RESUMEN

Tuberous sclerosis is an autosomal dominant human disorder caused by inactivating mutations to either the TSC1 or TSC2 tumour suppressor gene. Hamartin and tuberin, the TSC1 and TSC2 gene products, interact and the tuberin-hamartin complex inhibits cell growth by antagonising signal transduction to downstream effectors of the mammalian target of rapamycin (mTOR) through the small GTPase rheb. Previously, we showed that pathogenic tuberin amino-acid substitutions disrupt the tuberin-hamartin complex. Here, we investigate how these mutations affect the role of tuberin in the control of signal transduction through mTOR. Our data indicate that specific amino-acid substitutions have distinct effects on tuberin function.


Asunto(s)
Sustitución de Aminoácidos/fisiología , Quimiocinas CC/fisiología , Proteínas Represoras/fisiología , Transducción de Señal , Proteínas Supresoras de Tumor/fisiología , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Animales , Quimiocina CCL26 , Fibroblastos/metabolismo , Humanos , Ratones , Ratones Noqueados , Proteínas de Unión al GTP Monoméricas/metabolismo , Mutagénesis Sitio-Dirigida , Mutación Missense , Neuropéptidos/metabolismo , Fosforilación , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Homóloga de Ras Enriquecida en el Cerebro , Proteínas Quinasas S6 Ribosómicas/metabolismo , Serina-Treonina Quinasas TOR , Esclerosis Tuberosa/genética , Esclerosis Tuberosa/metabolismo , Proteína 1 del Complejo de la Esclerosis Tuberosa , Proteína 2 del Complejo de la Esclerosis Tuberosa
17.
Eur J Hum Genet ; 23(9): 1142-50, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25626705

RESUMEN

Implementation of next-generation DNA sequencing (NGS) technology into routine diagnostic genome care requires strategic choices. Instead of theoretical discussions on the consequences of such choices, we compared NGS-based diagnostic practices in eight clinical genetic centers in the Netherlands, based on genetic testing of nine pre-selected patients with cardiomyopathy. We highlight critical implementation choices, including the specific contributions of laboratory and medical specialists, bioinformaticians and researchers to diagnostic genome care, and how these affect interpretation and reporting of variants. Reported pathogenic mutations were consistent for all but one patient. Of the two centers that were inconsistent in their diagnosis, one reported to have found 'no causal variant', thereby underdiagnosing this patient. The other provided an alternative diagnosis, identifying another variant as causal than the other centers. Ethical and legal analysis showed that informed consent procedures in all centers were generally adequate for diagnostic NGS applications that target a limited set of genes, but not for exome- and genome-based diagnosis. We propose changes to further improve and align these procedures, taking into account the blurring boundary between diagnostics and research, and specific counseling options for exome- and genome-based diagnostics. We conclude that alternative diagnoses may infer a certain level of 'greediness' to come to a positive diagnosis in interpreting sequencing results. Moreover, there is an increasing interdependence of clinic, diagnostics and research departments for comprehensive diagnostic genome care. Therefore, we invite clinical geneticists, physicians, researchers, bioinformatics experts and patients to reconsider their role and position in future diagnostic genome care.


Asunto(s)
Cardiomiopatías/diagnóstico , Pruebas Genéticas/normas , Genoma Humano , Secuenciación de Nucleótidos de Alto Rendimiento/normas , Mutación , Proteínas de Unión al Calcio/genética , Miosinas Cardíacas/genética , Cardiomiopatías/genética , Proteínas Portadoras/genética , Exoma , Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento/instrumentación , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Consentimiento Informado/legislación & jurisprudencia , Ensayos de Aptitud de Laboratorios/estadística & datos numéricos , Quinasas Quinasa Quinasa PAM/genética , Cadenas Pesadas de Miosina/genética , Países Bajos , Proteínas Serina-Treonina Quinasas
18.
Am J Med Genet ; 114(6): 673-8, 2002 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-12210286

RESUMEN

A recent genome-wide scan showed strong evidence for a major locus for common syndromes of idiopathic generalized epilepsy (IGE) at the marker D18S474 on chromosome 18q21.1 (LOD score 4.5/5.2 multipoint/two-point). The present replication study tested the presence of an IGE locus in the chromosomal region 18q21.1. Our linkage study included 130 multiplex families of probands with common IGE syndromes. Eleven microsatellite polymorphisms encompassing a candidate region of 30 cM on either side of the marker D18S474 were genotyped. The two-point homogeneity LOD score for D18S474 showed strong evidence against linkage at the original linkage peak (Z = -18.86 at theta(m = f) = 0.05), assuming a recessive mode of inheritance with 50% penetrance. Multipoint parametric heterogeneity LOD scores < -2 were obtained along the candidate region when proportions of linked families greater than 35% were assumed under recessive inheritance. Furthermore, non-parametric multipoint linkage analyses showed no hint of linkage throughout the candidate region (P > 0.19). Accordingly, we failed to support evidence for a major IGE locus in the chromosomal region 18p11-18q23. If there is a susceptibility locus for IGE in this region then the size of the effect or the proportion of linked families is too small to detect linkage in the investigated family sample.


Asunto(s)
Cromosomas Humanos Par 18/genética , Epilepsia Generalizada/genética , Predisposición Genética a la Enfermedad , Adolescente , Adulto , Edad de Inicio , Niño , Mapeo Cromosómico , Susceptibilidad a Enfermedades , Epilepsias Mioclónicas/genética , Femenino , Ligamiento Genético , Marcadores Genéticos , Humanos , Escala de Lod , Masculino , Repeticiones de Microsatélite/genética , Modelos Genéticos , Núcleo Familiar , Polimorfismo Genético
19.
Eur J Paediatr Neurol ; 17(6): 666-70, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23755938

RESUMEN

BACKGROUND: The BIG2 protein, coded by ARFGEF2 indirectly assists neuronal proliferation and migration during cortical development. Mutations in ARFGEF2 have been reported as a rare cause of periventricular heterotopia. METHODS: The presence of periventricular heterotopia, acquired microcephaly and suspected recessive inheritance led to mutation analysis of ARFGEF2 in two affected siblings and their healthy consanguineous parents, after mutations in FLNA had been ruled out. RESULTS: A homozygous c.242_249delins7 (p.Pro81fs) mutation in exon 3 of ARFGEF2 was identified in the siblings. The alteration is a combination of 2 missense mutations (c.242C > A and c.247G > T) and a frameshift mutation (c.249delA) resulting in a premature stop codon. The clinical phenotype was characterized by dystonic quadriplegia, marked developmental delay, obstructive cardiomyopathy, recurrent infections and feeding difficulties. Degenerative features included early regression, acquired microcephaly and cerebral atrophy. Brain MRI revealed bilateral periventricular heterotopia, small corpus callosum, cerebral and hippocampal atrophy and hyperintensity in the putamen. CONCLUSION: Mutations in ARFGEF2 can be anticipated based on characteristic clinical and imaging features.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/genética , Mutación Missense/genética , Heterotopia Nodular Periventricular/genética , Animales , Encéfalo/patología , Análisis Mutacional de ADN , Salud de la Familia , Humanos , Lactante , Imagen por Resonancia Magnética , Masculino , Fenotipo
20.
Orphanet J Rare Dis ; 8: 51, 2013 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-23557332

RESUMEN

BACKGROUND: Mucopolysaccharidosis type VI (Maroteaux-Lamy syndrome; MPS VI) is an autosomal recessive lysosomal storage disorder in which deficiency of N-acetylgalactosamine 4-sulfatase (arylsulfatase B; ARSB) leads to the storage of glycosaminoglycans (GAGs) in connective tissue. The genotype-phenotype correlation has been addressed in several publications but the picture is not complete. Since 2007, enzyme-replacement therapy (ERT) has been available for patients with MPS VI in the Netherlands. The purpose of our study was to learn more about the genotype-phenotype correlations in MPS VI and the antibody response to ERT with galsulfase (recombinant human arylsulfatase B). METHODS: We identified ARSB mutations in 12 patients and used site-directed mutagenesis to study their effect. Antibody levels to galsulfase were measured using ELISA and a semi-quantitative immunoprecipitation method. We assessed the in vitro inhibitory effect of antibodies on galsulfase uptake and their effect on clinical outcome. RESULTS: Five patients had a rapidly progressive phenotype and seven a slowly progressive phenotype. In total 9 pathogenic mutations were identified including 4 novel mutations (N301K, V332G, A237D, and c.1142 + 2 T > C) together composing 8 pathogenic genotypes. Most mutations appeared not to affect the synthesis of ARSB (66 kD precursor), but to hamper its maturation (43 kD ARSB). Disease severity was correlated with urinary GAG excretion. All patients developed antibodies to galsulfase within 26 weeks of treatment. It was demonstrated that these antibodies can inhibit the uptake of galsulfase in vitro. CONCLUSIONS: The clinical phenotypes and the observed defects in the biosynthesis of ARSB show that some of the mutations that we identified are clearly more severe than others. Patients receiving galsulfase as enzyme-replacement therapy can develop antibodies towards the therapeutic protein. Though most titers are modest, they can exceed a level at which they potentially affect the clinical outcome of enzyme-replacement therapy.


Asunto(s)
Formación de Anticuerpos/inmunología , Mucopolisacaridosis VI/genética , Mucopolisacaridosis VI/patología , N-Acetilgalactosamina-4-Sulfatasa/inmunología , Adolescente , Niño , Preescolar , Ensayo de Inmunoadsorción Enzimática , Femenino , Genotipo , Humanos , Inmunoprecipitación , Lactante , Masculino , Mucopolisacaridosis VI/inmunología , Mutagénesis Sitio-Dirigida , Fenotipo , Proteínas Recombinantes/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA