Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Genet ; 143(1): 71-84, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38117302

RESUMEN

Coffin-Siris syndrome (CSS) is a rare multisystemic autosomal dominant disorder. Since 2012, alterations in genes of the SWI/SNF complex were identified as the molecular basis of CSS, studying largely pediatric cohorts. Therefore, there is a lack of information on the phenotype in adulthood, particularly on the clinical outcome in adulthood and associated risks. In an international collaborative effort, data from 35 individuals ≥ 18 years with a molecularly ascertained CSS diagnosis (variants in ARID1B, ARID2, SMARCA4, SMARCB1, SMARCC2, SMARCE1, SOX11, BICRA) using a comprehensive questionnaire was collected. Our results indicate that overweight and obesity are frequent in adults with CSS. Visual impairment, scoliosis, and behavioral anomalies are more prevalent than in published pediatric or mixed cohorts. Cognitive outcomes range from profound intellectual disability (ID) to low normal IQ, with most individuals having moderate ID. The present study describes the first exclusively adult cohort of CSS individuals. We were able to delineate some features of CSS that develop over time and have therefore been underrepresented in previously reported largely pediatric cohorts, and provide recommendations for follow-up.


Asunto(s)
Anomalías Múltiples , Cara/anomalías , Deformidades Congénitas de la Mano , Discapacidad Intelectual , Micrognatismo , Adulto , Humanos , Niño , Discapacidad Intelectual/genética , Discapacidad Intelectual/diagnóstico , Anomalías Múltiples/genética , Anomalías Múltiples/diagnóstico , Micrognatismo/genética , Micrognatismo/diagnóstico , Deformidades Congénitas de la Mano/genética , Cuello/anomalías , Fenotipo , ADN Helicasas/genética , Proteínas Nucleares/genética , Factores de Transcripción/genética , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN/genética
2.
Am J Hum Genet ; 107(5): 989-999, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33053334

RESUMEN

Osteogenesis imperfecta (OI) is characterized primarily by susceptibility to fractures with or without bone deformation. OI is genetically heterogeneous: over 20 genetic causes are recognized. We identified bi-allelic pathogenic KDELR2 variants as a cause of OI in four families. KDELR2 encodes KDEL endoplasmic reticulum protein retention receptor 2, which recycles ER-resident proteins with a KDEL-like peptide from the cis-Golgi to the ER through COPI retrograde transport. Analysis of patient primary fibroblasts showed intracellular decrease of HSP47 and FKBP65 along with reduced procollagen type I in culture media. Electron microscopy identified an abnormal quality of secreted collagen fibrils with increased amount of HSP47 bound to monomeric and multimeric collagen molecules. Mapping the identified KDELR2 variants onto the crystal structure of G. gallus KDELR2 indicated that these lead to an inactive receptor resulting in impaired KDELR2-mediated Golgi-ER transport. Therefore, in KDELR2-deficient individuals, OI most likely occurs because of the inability of HSP47 to bind KDELR2 and dissociate from collagen type I. Instead, HSP47 remains bound to collagen molecules extracellularly, disrupting fiber formation. This highlights the importance of intracellular recycling of ER-resident molecular chaperones for collagen type I and bone metabolism and a crucial role of HSP47 in the KDELR2-associated pathogenic mechanism leading to OI.


Asunto(s)
Huesos/metabolismo , Colágeno Tipo I/metabolismo , Proteínas del Choque Térmico HSP47/metabolismo , Osteogénesis Imperfecta/genética , Proteínas de Transporte Vesicular/metabolismo , Adulto , Alelos , Secuencia de Aminoácidos , Animales , Sitios de Unión , Huesos/patología , Pollos , Preescolar , Colágeno Tipo I/química , Colágeno Tipo I/genética , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Expresión Génica , Aparato de Golgi/metabolismo , Aparato de Golgi/patología , Proteínas del Choque Térmico HSP47/química , Proteínas del Choque Térmico HSP47/genética , Humanos , Lactante , Masculino , Osteogénesis Imperfecta/diagnóstico , Osteogénesis Imperfecta/metabolismo , Osteogénesis Imperfecta/patología , Linaje , Cultivo Primario de Células , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Transporte de Proteínas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/genética
3.
J Med Genet ; 59(7): 697-705, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34321323

RESUMEN

BACKGROUND: O'Donnell-Luria-Rodan syndrome (ODLURO) is an autosomal-dominant neurodevelopmental disorder caused by pathogenic, mostly truncating variants in KMT2E. It was first described by O'Donnell-Luria et al in 2019 in a cohort of 38 patients. Clinical features encompass macrocephaly, mild intellectual disability (ID), autism spectrum disorder (ASD) susceptibility and seizure susceptibility. METHODS: Affected individuals were ascertained at paediatric and genetic centres in various countries by diagnostic chromosome microarray or exome/genome sequencing. Patients were collected into a case cohort and were systematically phenotyped where possible. RESULTS: We report 18 additional patients from 17 families with genetically confirmed ODLURO. We identified 15 different heterozygous likely pathogenic or pathogenic sequence variants (14 novel) and two partial microdeletions of KMT2E. We confirm and refine the phenotypic spectrum of the KMT2E-related neurodevelopmental disorder, especially concerning cognitive development, with rather mild ID and macrocephaly with subtle facial features in most patients. We observe a high prevalence of ASD in our cohort (41%), while seizures are present in only two patients. We extend the phenotypic spectrum by sleep disturbances. CONCLUSION: Our study, bringing the total of known patients with ODLURO to more than 60 within 2 years of the first publication, suggests an unexpectedly high relative frequency of this syndrome worldwide. It seems likely that ODLURO, although just recently described, is among the more common single-gene aetiologies of neurodevelopmental delay and ASD. We present the second systematic case series of patients with ODLURO, further refining the mutational and phenotypic spectrum of this not-so-rare syndrome.


Asunto(s)
Trastorno del Espectro Autista , Discapacidad Intelectual , Megalencefalia , Trastornos del Neurodesarrollo , Trastorno del Espectro Autista/genética , Niño , Humanos , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/epidemiología , Discapacidad Intelectual/genética , Convulsiones/epidemiología , Convulsiones/genética , Síndrome , Secuenciación del Exoma
4.
Am J Hum Genet ; 104(4): 749-757, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30905398

RESUMEN

Over a relatively short period of time, the clinical geneticist's "toolbox" has been expanded by machine-learning algorithms for image analysis, which can be applied to the task of syndrome identification on the basis of facial photographs, but these technologies harbor potential beyond the recognition of established phenotypes. Here, we comprehensively characterized two individuals with a hitherto unknown genetic disorder caused by the same de novo mutation in LEMD2 (c.1436C>T;p.Ser479Phe), the gene which encodes the nuclear envelope protein LEM domain-containing protein 2 (LEMD2). Despite different ages and ethnic backgrounds, both individuals share a progeria-like facial phenotype and a distinct combination of physical and neurologic anomalies, such as growth retardation; hypoplastic jaws crowded with multiple supernumerary, yet unerupted, teeth; and cerebellar intention tremor. Immunofluorescence analyses of patient fibroblasts revealed mutation-induced disturbance of nuclear architecture, recapitulating previously published data in LEMD2-deficient cell lines, and additional experiments suggested mislocalization of mutant LEMD2 protein within the nuclear lamina. Computational analysis of facial features with two different deep neural networks showed phenotypic proximity to other nuclear envelopathies. One of the algorithms, when trained to recognize syndromic similarity (rather than specific syndromes) in an unsupervised approach, clustered both individuals closely together, providing hypothesis-free hints for a common genetic etiology. We show that a recurrent de novo mutation in LEMD2 causes a nuclear envelopathy whose prognosis in adolescence is relatively good in comparison to that of classical Hutchinson-Gilford progeria syndrome, and we suggest that the application of artificial intelligence to the analysis of patient images can facilitate the discovery of new genetic disorders.


Asunto(s)
Proteínas de la Membrana/genética , Mutación , Proteínas Nucleares/genética , Progeria/genética , Adolescente , Inteligencia Artificial , Línea Celular Tumoral , Núcleo Celular , Niño , Preescolar , Diagnóstico por Computador , Cara , Fibroblastos/metabolismo , Humanos , Masculino , Tamizaje Masivo/métodos , Informática Médica , Fenotipo , Pronóstico , Síndrome
5.
Am J Hum Genet ; 105(4): 836-843, 2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31564437

RESUMEN

Osteogenesis imperfecta (OI) comprises a genetically heterogeneous group of skeletal fragility diseases. Here, we report on five independent families with a progressively deforming type of OI, in whom we identified four homozygous truncation or frameshift mutations in MESD. Affected individuals had recurrent fractures and at least one had oligodontia. MESD encodes an endoplasmic reticulum (ER) chaperone protein for the canonical Wingless-related integration site (WNT) signaling receptors LRP5 and LRP6. Because complete absence of MESD causes embryonic lethality in mice, we hypothesized that the OI-associated mutations are hypomorphic alleles since these mutations occur downstream of the chaperone activity domain but upstream of ER-retention domain. This would be consistent with the clinical phenotypes of skeletal fragility and oligodontia in persons deficient for LRP5 and LRP6, respectively. When we expressed wild-type (WT) and mutant MESD in HEK293T cells, we detected WT MESD in cell lysate but not in conditioned medium, whereas the converse was true for mutant MESD. We observed that both WT and mutant MESD retained the ability to chaperone LRP5. Thus, OI-associated MESD mutations produce hypomorphic alleles whose failure to remain within the ER significantly reduces but does not completely eliminate LRP5 and LRP6 trafficking. Since these individuals have no eye abnormalities (which occur in individuals completely lacking LRP5) and have neither limb nor brain patterning defects (both of which occur in mice completely lacking LRP6), we infer that bone mass accrual and dental patterning are more sensitive to reduced canonical WNT signaling than are other developmental processes. Biologic agents that can increase LRP5 and LRP6-mediated WNT signaling could benefit individuals with MESD-associated OI.


Asunto(s)
Chaperonas Moleculares/genética , Mutación , Osteogénesis Imperfecta/genética , Animales , Femenino , Genes Recesivos , Células HEK293 , Humanos , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Masculino , Ratones , Linaje , Fenotipo , Vía de Señalización Wnt
6.
Am J Hum Genet ; 101(5): 833-843, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-29100093

RESUMEN

Gorlin-Chaudhry-Moss syndrome (GCMS) is a dysmorphic syndrome characterized by coronal craniosynostosis and severe midface hypoplasia, body and facial hypertrichosis, microphthalmia, short stature, and short distal phalanges. Variable lipoatrophy and cutis laxa are the basis for a progeroid appearance. Using exome and genome sequencing, we identified the recurrent de novo mutations c.650G>A (p.Arg217His) and c.649C>T (p.Arg217Cys) in SLC25A24 in five unrelated girls diagnosed with GCMS. Two of the girls had pronounced neonatal progeroid features and were initially diagnosed with Wiedemann-Rautenstrauch syndrome. SLC25A24 encodes a mitochondrial inner membrane ATP-Mg/Pi carrier. In fibroblasts from affected individuals, the mutated SLC25A24 showed normal stability. In contrast to control cells, the probands' cells showed mitochondrial swelling, which was exacerbated upon treatment with hydrogen peroxide (H2O2). The same effect was observed after overexpression of the mutant cDNA. Under normal culture conditions, the mitochondrial membrane potential of the probands' fibroblasts was intact, whereas ATP content in the mitochondrial matrix was lower than that in control cells. However, upon H2O2 exposure, the membrane potential was significantly elevated in cells harboring the mutated SLC25A24. No reduction of mitochondrial DNA copy number was observed. These findings demonstrate that mitochondrial dysfunction with increased sensitivity to oxidative stress is due to the SLC25A24 mutations. Our results suggest that the SLC25A24 mutations induce a gain of pathological function and link mitochondrial ATP-Mg/Pi transport to the development of skeletal and connective tissue.


Asunto(s)
Anomalías Múltiples/genética , Antiportadores/genética , Proteínas de Unión al Calcio/genética , Anomalías Craneofaciales/genética , Craneosinostosis/genética , Conducto Arterioso Permeable/genética , Hipertricosis/genética , Mitocondrias/genética , Proteínas Mitocondriales/genética , Mutación/genética , Adenosina Trifosfato/genética , Adolescente , Niño , Preescolar , Cutis Laxo/genética , ADN Mitocondrial/genética , Exoma/genética , Femenino , Retardo del Crecimiento Fetal/genética , Fibroblastos/patología , Trastornos del Crecimiento , Humanos , Peróxido de Hidrógeno/farmacología , Lactante , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/genética , Mitocondrias/efectos de los fármacos , Estrés Oxidativo/genética , Progeria/genética
7.
Mol Cell Probes ; 45: 84-88, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30914295

RESUMEN

When deciding on which genes to assess in larger Next-Generation Sequencing (NGS) datasets for the molecular genetic diagnosis of intellectual disability (ID), geneticists today have a variety of gene-phenotype databases and expert-curated gene lists available. To quantify their respective completeness, we compare an ID gene selection auto-generated from the Human Phenotype Ontology gene-phenotype association database and expert-curated ID gene lists from three reputable sources (sysID, the DDD consortium and Genomics England) and analyse some of their differences. We give examples of what we regard as genuine gaps ("missing ID genes") for each of these and conclude that a complementary or consensus approach is needed to maximise diagnostic yield in ID patients. We propose several consensus gene lists with ID-associated genes of different confidence levels.


Asunto(s)
Biología Computacional/métodos , Redes Reguladoras de Genes , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Discapacidad Intelectual/genética , Consenso , Curaduría de Datos , Bases de Datos Genéticas , Predisposición Genética a la Enfermedad , Humanos , Análisis de Secuencia de ADN
8.
J Lipid Res ; 59(8): 1529-1535, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29866657

RESUMEN

Copy-number variations (CNVs) have been studied in the context of familial hypercholesterolemia but have not yet been evaluated in patients with extreme levels of HDL cholesterol. We evaluated targeted, next-generation sequencing data from patients with very low levels of HDL cholesterol (i.e., hypoalphalipoproteinemia) with the VarSeq-CNV® caller algorithm to screen for CNVs that disrupted the ABCA1, LCAT, or APOA1 genes. In four individuals, we found three unique deletions in ABCA1: a heterozygous deletion of exon 4, a heterozygous deletion that spanned exons 8 to 31, and a heterozygous deletion of the entire ABCA1 gene. Breakpoints were identified with Sanger sequencing, and the full-gene deletion was confirmed by using exome sequencing and the Affymetrix CytoScan HD array. Previously, large-scale deletions in candidate HDL genes had not been associated with hypoalphalipoproteinemia; our findings indicate that CNVs in ABCA1 may be a previously unappreciated genetic determinant of low levels of HDL cholesterol. By coupling bioinformatic analyses with next-generation sequencing data, we can successfully assess the spectrum of genetic determinants of many dyslipidemias, including hypoalphalipoproteinemia.


Asunto(s)
Transportador 1 de Casete de Unión a ATP/deficiencia , Transportador 1 de Casete de Unión a ATP/genética , Eliminación de Gen , Hipoalfalipoproteinemias/genética , Adulto , Biología Computacional , Variaciones en el Número de Copia de ADN , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Persona de Mediana Edad
9.
Am J Hum Genet ; 96(3): 432-9, 2015 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-25683121

RESUMEN

As a result of a whole-exome sequencing study, we report three mutant alleles in SEC24D, a gene encoding a component of the COPII complex involved in protein export from the ER: the truncating mutation c.613C>T (p.Gln205(∗)) and the missense mutations c.3044C>T (p.Ser1015Phe, located in a cargo-binding pocket) and c.2933A>C (p.Gln978Pro, located in the gelsolin-like domain). Three individuals from two families affected by a similar skeletal phenotype were each compound heterozygous for two of these mutant alleles, with c.3044C>T being embedded in a 14 Mb founder haplotype shared by all three. The affected individuals were a 7-year-old boy with a phenotype most closely resembling Cole-Carpenter syndrome and two fetuses initially suspected to have a severe type of osteogenesis imperfecta. All three displayed a severely disturbed ossification of the skull and multiple fractures with prenatal onset. The 7-year-old boy had short stature and craniofacial malformations including macrocephaly, midface hypoplasia, micrognathia, frontal bossing, and down-slanting palpebral fissures. Electron and immunofluorescence microscopy of skin fibroblasts of this individual revealed that ER export of procollagen was inefficient and that ER tubules were dilated, faithfully reproducing the cellular phenotype of individuals with cranio-lentico-sutural dysplasia (CLSD). CLSD is caused by SEC23A mutations and displays a largely overlapping craniofacial phenotype, but it is not characterized by generalized bone fragility and presented with cataracts in the original family described. The cellular and morphological phenotypes we report are in concordance with the phenotypes described for the Sec24d-deficient fish mutants vbi (medaka) and bulldog (zebrafish).


Asunto(s)
Craneosinostosis/genética , Anomalías del Ojo/genética , Hidrocefalia/genética , Osteogénesis Imperfecta/genética , Proteínas de Transporte Vesicular/genética , Alelos , Animales , Huesos/patología , Niño , Retículo Endoplásmico/metabolismo , Femenino , Heterocigoto , Humanos , Masculino , Mutación Missense , Linaje , Fenotipo , Conformación Proteica , Análisis de Secuencia de ADN , Proteínas de Transporte Vesicular/metabolismo , Pez Cebra/genética
10.
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
11.
Hum Mutat ; 37(9): 847-64, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27302555

RESUMEN

Kabuki syndrome (KS) is a rare but recognizable condition that consists of a characteristic face, short stature, various organ malformations, and a variable degree of intellectual disability. Mutations in KMT2D have been identified as the main cause for KS, whereas mutations in KDM6A are a much less frequent cause. Here, we report a mutation screening in a case series of 347 unpublished patients, in which we identified 12 novel KDM6A mutations (KS type 2) and 208 mutations in KMT2D (KS type 1), 132 of them novel. Two of the KDM6A mutations were maternally inherited and nine were shown to be de novo. We give an up-to-date overview of all published mutations for the two KS genes and point out possible mutation hot spots and strategies for molecular genetic testing. We also report the clinical details for 11 patients with KS type 2, summarize the published clinical information, specifically with a focus on the less well-defined X-linked KS type 2, and comment on phenotype-genotype correlations as well as sex-specific phenotypic differences. Finally, we also discuss a possible role of KDM6A in Kabuki-like Turner syndrome and report a mutation screening of KDM6C (UTY) in male KS patients.


Asunto(s)
Anomalías Múltiples/genética , Proteínas de Unión al ADN/genética , Cara/anomalías , Enfermedades Hematológicas/genética , Histona Demetilasas/genética , Mutación , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Enfermedades Vestibulares/genética , Anomalías Múltiples/patología , Cara/patología , Femenino , Genes Ligados a X , Predisposición Genética a la Enfermedad , Enfermedades Hematológicas/patología , Humanos , Masculino , Herencia Materna , Síndrome de Noonan/genética , Análisis de Secuencia de ADN , Enfermedades Vestibulares/patología
12.
Am J Hum Genet ; 92(4): 565-74, 2013 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-23499309

RESUMEN

We report that hypofunctional alleles of WNT1 cause autosomal-recessive osteogenesis imperfecta, a congenital disorder characterized by reduced bone mass and recurrent fractures. In consanguineous families, we identified five homozygous mutations in WNT1: one frameshift mutation, two missense mutations, one splice-site mutation, and one nonsense mutation. In addition, in a family affected by dominantly inherited early-onset osteoporosis, a heterozygous WNT1 missense mutation was identified in affected individuals. Initial functional analysis revealed that altered WNT1 proteins fail to activate canonical LRP5-mediated WNT-regulated ß-catenin signaling. Furthermore, osteoblasts cultured in vitro showed enhanced Wnt1 expression with advancing differentiation, indicating a role of WNT1 in osteoblast function and bone development. Our finding that homozygous and heterozygous variants in WNT1 predispose to low-bone-mass phenotypes might advance the development of more effective therapeutic strategies for congenital forms of bone fragility, as well as for common forms of age-related osteoporosis.


Asunto(s)
Densidad Ósea/genética , Huesos/patología , Mutación/genética , Osteogénesis Imperfecta/genética , Osteoporosis/genética , Proteína Wnt1/genética , Animales , Secuencia de Bases , Células Cultivadas , Niño , Preescolar , Femenino , Heterocigoto , Humanos , Recién Nacido , Proteínas Relacionadas con Receptor de LDL/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Osteoblastos/metabolismo , Osteoblastos/patología , Osteogénesis Imperfecta/patología , Osteoporosis/patología , Linaje , Fenotipo , Embarazo
13.
Am J Hum Genet ; 91(2): 349-57, 2012 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-22863195

RESUMEN

Osteogenesis imperfecta (OI) is a clinically and genetically heterogeneous disorder associated with bone fragility and susceptibility to fractures after minimal trauma. OI type V has an autosomal-dominant pattern of inheritance and is not caused by mutations in the type I collagen genes COL1A1 and COL1A2. The most remarkable and pathognomonic feature, observed in ~65% of affected individuals, is a predisposition to develop hyperplastic callus after fractures or surgical interventions. To identify the molecular cause of OI type V, we performed whole-exome sequencing in a female with OI type V and her unaffected parents and searched for de novo mutations. We found a heterozygous de novo mutation in the 5'-untranslated region of IFITM5 (the gene encoding Interferon induced transmembrane protein 5), 14 bp upstream of the annotated translation initiation codon (c.-14C>T). Subsequently, we identified an identical heterozygous de novo mutation in a second individual with OI type V by Sanger sequencing, thereby confirming that this is the causal mutation for the phenotype. IFITM5 is a protein that is highly enriched in osteoblasts and has a putative function in bone formation and osteoblast maturation. The mutation c.-14C>T introduces an upstream start codon that is in frame with the reference open-reading frame of IFITM5 and is embedded into a stronger Kozak consensus sequence for translation initiation than the annotated start codon. In vitro, eukaryotic cells were able to recognize this start codon, and they used it instead of the reference translation initiation signal. This suggests that five amino acids (Met-Ala-Leu-Glu-Pro) are added to the N terminus and alter IFITM5 function in individuals with the mutation.


Asunto(s)
Proteínas de la Membrana/genética , Osteogénesis Imperfecta/genética , Regiones no Traducidas 5'/genética , Absorciometría de Fotón , Secuencia de Aminoácidos , Secuencia de Bases , Niño , Codón Iniciador/genética , Biología Computacional , Difosfonatos/uso terapéutico , Exoma/genética , Femenino , Humanos , Lactante , Datos de Secuencia Molecular , Osteogénesis Imperfecta/diagnóstico por imagen , Osteogénesis Imperfecta/tratamiento farmacológico , Mutación Puntual/genética , Análisis de Secuencia de ADN
14.
Am J Hum Genet ; 90(4): 661-74, 2012 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-22482805

RESUMEN

Bone morphogenetic protein 1 (BMP1) is an astacin metalloprotease with important cellular functions and diverse substrates, including extracellular-matrix proteins and antagonists of some TGFß superfamily members. Combining whole-exome sequencing and filtering for homozygous stretches of identified variants, we found a homozygous causative BMP1 mutation, c.34G>C, in a consanguineous family affected by increased bone mineral density and multiple recurrent fractures. The mutation is located within the BMP1 signal peptide and leads to impaired secretion and an alteration in posttranslational modification. We also characterize a zebrafish bone mutant harboring lesions in bmp1a, demonstrating conservation of BMP1 function in osteogenesis across species. Genetic, biochemical, and histological analyses of this mutant and a comparison to a second, similar locus reveal that Bmp1a is critically required for mature-collagen generation, downstream of osteoblast maturation, in bone. We thus define the molecular and cellular bases of BMP1-dependent osteogenesis and show the importance of this protein for bone formation and stability.


Asunto(s)
Proteína Morfogenética Ósea 1/fisiología , Osteogénesis/genética , Osteogénesis/fisiología , Animales , Secuencia de Bases , Conservadores de la Densidad Ósea/uso terapéutico , Proteína Morfogenética Ósea 1/genética , Proteína Morfogenética Ósea 1/metabolismo , Huesos/metabolismo , Diferenciación Celular , Preescolar , Colágeno/biosíntesis , Difosfonatos/uso terapéutico , Exoma , Femenino , Fracturas Óseas/tratamiento farmacológico , Fracturas Óseas/prevención & control , Sitios Genéticos , Proteínas de Choque Térmico , Humanos , Masculino , Datos de Secuencia Molecular , Mutación , Osteoblastos/efectos de los fármacos , Osteoblastos/fisiología , Osteogénesis/efectos de los fármacos , Fragmentos de Péptidos , Procesamiento Proteico-Postraduccional , Pez Cebra/genética , Pez Cebra/metabolismo
15.
Wien Med Wochenschr ; 165(13-14): 278-84, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26055811

RESUMEN

Osteogenesis imperfecta is a rare hereditary disease mostly caused by mutations impairing collagen synthesis and modification. Recently recessive forms have been described influencing differentiation and activity of osteoblasts and osteoclasts. Most prominent signs are fractures due to low traumata and deformities of long bones and vertebrae. Additional patients can be affected by dwarfism, scoliosis Dentinogenesis imperfecta, deafness and a blueish discoloration of the sclera. During childhood state of the art medical treatment are i.v. bisphosphonates to increase bone mass and to reduce fracture rate. Surgical interventions are needed to treat fractures, to correct deformities and should always be accompanied by physiotherapeutic and rehabilitative interventions.


Asunto(s)
Osteogénesis Imperfecta/fisiopatología , Osteogénesis Imperfecta/terapia , Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I , Terapia Combinada , Análisis Mutacional de ADN , Difosfonatos/administración & dosificación , Femenino , Fijación de Fractura , Fracturas Espontáneas/genética , Fracturas Espontáneas/fisiopatología , Fracturas Espontáneas/terapia , Tamización de Portadores Genéticos , Humanos , Lactante , Recién Nacido , Infusiones Intravenosas , Masculino , Osteoblastos/fisiología , Osteoclastos/fisiología , Osteogénesis Imperfecta/genética , Modalidades de Fisioterapia
16.
Am J Hum Genet ; 88(3): 362-71, 2011 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-21353196

RESUMEN

Osteogenesis imperfecta (OI) is a heterogeneous genetic disorder characterized by bone fragility and susceptibility to fractures after minimal trauma. After mutations in all known OI genes had been excluded by Sanger sequencing, we applied next-generation sequencing to analyze the exome of a single individual who has a severe form of the disease and whose parents are second cousins. A total of 26,922 variations from the human reference genome sequence were subjected to several filtering steps. In addition, we extracted the genotypes of all dbSNP130-annotated SNPs from the exome sequencing data and used these 299,494 genotypes as markers for the genome-wide identification of homozygous regions. A single homozygous truncating mutation, affecting SERPINF1 on chromosome 17p13.3, that was embedded into a homozygous stretch of 2.99 Mb remained. The mutation was also homozygous in the affected brother of the index patient. Subsequently, we identified homozygosity for two different truncating SERPINF1 mutations in two unrelated patients with OI and parental consanguinity. All four individuals with SERPINF1 mutations have severe OI. Fractures of long bones and severe vertebral compression fractures with resulting deformities were observed as early as the first year of life in these individuals. Collagen analyses with cultured dermal fibroblasts displayed no evidence for impaired collagen folding, posttranslational modification, or secretion. SERPINF1 encodes pigment epithelium-derived factor (PEDF), a secreted glycoprotein of the serpin superfamily. PEDF is a multifunctional protein and one of the strongest inhibitors of angiogenesis currently known in humans. Our data provide genetic evidence for PEDF involvement in human bone homeostasis.


Asunto(s)
Exones/genética , Proteínas del Ojo/genética , Genes Recesivos/genética , Mutación/genética , Factores de Crecimiento Nervioso/genética , Osteogénesis Imperfecta/genética , Serpinas/genética , Adolescente , Secuencia de Bases , Niño , Preescolar , Análisis Mutacional de ADN , Homocigoto , Humanos , Lactante , Datos de Secuencia Molecular , Osteogénesis Imperfecta/diagnóstico por imagen , Radiografía
17.
Ophthalmol Sci ; 4(3): 100444, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38374928

RESUMEN

Purpose: To investigate the genetic cause, clinical characteristics, and potential therapeutic targets of infantile corneal myofibromatosis. Design: Case series with genetic and functional in vitro analyses. Participants: Four individuals from 2 unrelated families with clinical signs of corneal myofibromatosis were investigated. Methods: Exome-based panel sequencing for platelet-derived growth factor receptor beta gene (PDGFRB) and notch homolog protein 3 gene (NOTCH3) was performed in the respective index patients. One clinically affected member of each family was tested for the pathogenic variant detected in the respective index by Sanger sequencing. Immunohistochemical staining on excised corneal tissue was conducted. Functional analysis of the individual PDGFRB variants was performed in vitro by luciferase reporter assays on transfected porcine aortic endothelial cells using tyrosine kinase inhibitors. Protein expression analysis of mutated PDGFRB was analyzed by Western blot. Main Outcome Measures: Sequencing data, immunohistochemical stainings, functional analysis of PDGFRB variants, and protein expression analysis. Results: We identified 2 novel, heterozygous gain-of-function variants in PDGFRB in 4 individuals from 2 unrelated families with corneal myofibromatosis. Immunohistochemistry demonstrated positivity for alpha-smooth muscle actin and ß-catenin, a low proliferation rate in Ki-67 (< 5%), marginal positivity for Desmin, and negative staining for Caldesmon and CD34. In all patients, recurrence of disease occurred after corneal surgery. When transfected in cultured cells, the PDGFRB variants conferred a constitutive activity to the receptor in the absence of its ligand and were sensitive to the tyrosine kinase inhibitor imatinib. The variants can both be classified as likely pathogenic regarding the American College of Medical Genetics and Genomics classification criteria. Conclusions: We describe 4 cases of corneal myofibromatosis caused by novel PDGFRB variants with autosomal dominant transmission. Imatinib sensitivity in vitro suggests perspectives for targeted therapy preventing recurrences in the future. Financial Disclosures: Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.

18.
Am J Hum Genet ; 86(5): 696-706, 2010 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-20381006

RESUMEN

Cenani-Lenz syndrome (CLS) is an autosomal-recessive congenital disorder affecting distal limb development. It is characterized mainly by syndactyly and/or oligodactyly and is now shown to be commonly associated with kidney anomalies. We used a homozygosity-mapping approach to map the CLS1 locus to chromosome 11p11.2-q13.1. By sequencing candidate genes, we identified recessive LRP4 mutations in 12 families with CLS. LRP4 belongs to the low-density lipoprotein (LDL) receptor-related proteins (LRPs), which are essential for various developmental processes. LRP4 is known to antagonize LRP6-mediated activation of canonical Wnt signaling, a function that is lost by the identified mutations. Our findings increase the spectrum of congenital anomalies associated with abnormal lipoprotein receptor-dependent signaling.


Asunto(s)
Enfermedades Renales/genética , Proteínas Relacionadas con Receptor de LDL/genética , Deformidades Congénitas de las Extremidades/genética , Transducción de Señal/fisiología , beta Catenina/metabolismo , Humanos , Riñón/metabolismo , Enfermedades Renales/metabolismo , Proteínas Relacionadas con Receptor de LDL/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad , Mutación , Oncogenes , Receptores de LDL/antagonistas & inhibidores , Sindactilia/genética , Sindactilia/metabolismo , Síndrome
19.
Am J Med Genet A ; 161A(9): 2158-66, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23913520

RESUMEN

Loss-of-function mutations of NSD1 and 5q35 microdeletions encompassing NSD1 are a major cause of Sotos syndrome (Sos), which is characterized by overgrowth, macrocephaly, characteristic facies, and variable intellectual disability (ID). Microduplications of 5q35.2-q35.3 including NSD1 have been reported in only five patients so far and described clinically as a reversed Sos resulting from a hypothetical gene dosage effect of NSD1. Here, we report on nine patients from five families with interstitial duplication 5q35 including NSD1 detected by molecular karyotyping. The clinical features of all 14 individuals are reviewed. Patients with microduplications including NSD1 appear to have a consistent phenotype consisting of short stature, microcephaly, learning disability or mild to moderate ID, and distinctive facial features comprising periorbital fullness, short palpebral fissures, a long nose with broad or long nasal tip, a smooth philtrum and a thin upper lip vermilion. Behavioral problems, ocular and minor hand anomalies may be associated. Based on our findings, we discuss the possible etiology and conclude that it is possible, but so far unproven, that a gene dosage effect of NSD1 may be the major cause.


Asunto(s)
Duplicación Cromosómica , Cromosomas Humanos Par 5 , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Nucleares/genética , Fenotipo , Síndrome de Sotos/diagnóstico , Síndrome de Sotos/genética , Adolescente , Adulto , Niño , Preescolar , Mapeo Cromosómico , Hibridación Genómica Comparativa , Facies , Femenino , Dosificación de Gen , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA