Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Genes (Basel) ; 12(4)2021 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-33916386

RESUMEN

Spondylocarpotarsal synostosis syndrome (SCT) is characterized by vertebral fusions, a disproportionately short stature, and synostosis of carpal and tarsal bones. Pathogenic variants in FLNB, MYH3, and possibly in RFLNA, have been reported to be responsible for this condition. Here, we present two unrelated individuals presenting with features typical of SCT in which Sanger sequencing combined with whole genome sequencing identified novel, homozygous intragenic deletions in FLNB (c.1346-1372_1941+389del and c.3127-353_4223-1836del). Both deletions remove several consecutive exons and are predicted to result in a frameshift. To our knowledge, this is the first time that large structural variants in FLNB have been reported in SCT, and thus our findings add to the classes of variation that can lead to this disorder. These cases highlight the need for copy number sensitive methods to be utilized in order to be comprehensive in the search for a molecular diagnosis in individuals with a clinical diagnosis of SCT.


Asunto(s)
Anomalías Múltiples/etiología , Filaminas/genética , Eliminación de Gen , Vértebras Lumbares/anomalías , Enfermedades Musculoesqueléticas/etiología , Mutación , Escoliosis/congénito , Sinostosis/etiología , Vértebras Torácicas/anomalías , Anomalías Múltiples/patología , Adulto , Niño , Femenino , Humanos , Vértebras Lumbares/patología , Masculino , Enfermedades Musculoesqueléticas/patología , Linaje , Escoliosis/etiología , Escoliosis/patología , Síndrome , Sinostosis/patología , Vértebras Torácicas/patología
2.
J Clin Endocrinol Metab ; 105(3)2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31970420

RESUMEN

CONTEXT: The WNT/ß-catenin pathway is central to the pathogenesis of various human diseases including those affecting bone development and tumor progression. OBJECTIVE: To evaluate the role of a gain-of-function variant in CTNNB1 in a child with a sclerosing bone dysplasia and an adrenocortical adenoma. DESIGN: Whole exome sequencing with corroborative biochemical analyses. PATIENTS: We recruited a child with a sclerosing bone dysplasia and an adrenocortical adenoma together with her unaffected parents. INTERVENTION: Whole exome sequencing and performance of immunoblotting and luciferase-based assays to assess the cellular consequences of a de novo variant in CTNNB1. MAIN OUTCOME MEASURE(S)/RESULT: A de novo variant in CTNNB1 (c.131C>T; p.[Pro44Leu]) was identified in a patient with a sclerosing bone dysplasia and an adrenocortical adenoma. A luciferase-based transcriptional assay of WNT signaling activity verified that the activity of ß-catenin was increased in the cells transfected with a CTNNB1p.Pro44Leu construct (P = 4.00 × 10-5). The ß-catenin p.Pro44Leu variant was also associated with a decrease in phosphorylation at Ser45 and Ser33/Ser37/Thr41 in comparison to a wild-type (WT) CTNNB1 construct (P = 2.16 × 10-3, P = 9.34 × 10-8 respectively). CONCLUSION: Increased ß-catenin activity associated with a de novo gain-of-function CTNNB1 variant is associated with osteosclerotic phenotype and adrenocortical neoplasia.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/patología , Carcinoma Corticosuprarrenal/patología , Enfermedades del Desarrollo Óseo/patología , Mutación , beta Catenina/genética , Neoplasias de la Corteza Suprarrenal/genética , Carcinoma Corticosuprarrenal/genética , Enfermedades del Desarrollo Óseo/genética , Femenino , Humanos , Recién Nacido , Masculino , Linaje , Fenotipo , Pronóstico , Secuenciación del Exoma
3.
Eur J Hum Genet ; 28(4): 445-452, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31792352

RESUMEN

Connective tissue disorders are a spectrum of diseases that affect the integrity of tissues including skin, vasculature, and joints. They are often caused by variants that disrupt genes encoding components of extracellular matrix (ECM). The fibulin glycoproteins are ECM proteins important for integrity of tissues including dermis, retina, fascia, and vasculature. The fibulin family consists of seven members (fibulins-1 to -7) and is defined by a fibulin-type domain at the C-terminus. The family is associated with human diseases, for instance a variant in FBLN1, encoding fibulin-1, is associated with synpolydactyly, while one in EFEMP1, encoding fibulin-3, causes Doyne honeycomb degeneration of the retina. Loss-of-function of fibulins-4 and -5 causes cutis laxa, while variants in fibulins-5 and -6 are associated with age-related macular degeneration. Of note, EFEMP1 is not currently associated with any connective tissue disorder. Here we show biallelic loss-of-function variants in EFEMP1 in an individual with multiple and recurrent abdominal and thoracic herniae, myopia, hypermobile joints, scoliosis, and thin translucent skin. Fibroblasts from this individual express significantly lower EFEMP1 transcript than age-matched control cells. A skin biopsy, visualised using light microscopy, showed normal structure and abundance of elastic fibres. The phenotype of this individual is remarkably similar to the Efemp1 knockout mouse model that displays multiple herniae with premature aging and scoliosis. We conclude that loss of EFEMP1 function in this individual is the cause of a connective tissue disorder with a novel combination of phenotypic features, and can perhaps explain similar, previously reported cases in the literature.


Asunto(s)
Enfermedades del Tejido Conjuntivo/genética , Proteínas de la Matriz Extracelular/genética , Mutación con Pérdida de Función , Fenotipo , Adulto , Alelos , Células Cultivadas , Enfermedades del Tejido Conjuntivo/patología , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Masculino
4.
Front Genet ; 10: 800, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31616463

RESUMEN

The etiology of dental anomalies is multifactorial; and genetic and environmental factors that affect the dental lamina have been implicated. We investigated two families of European ancestry in which males were affected by taurodontism, microdontia and dens invaginatus. In both families, males were related to each other via unaffected females. A linkage analysis was conducted in a New Zealand family, followed by exome sequencing and focused analysis of the X-chromosome. In a US family, exome sequencing of the X-chromosome was followed by Sanger sequencing to conduct segregation analyses. We identified two independent missense variants in KIF4A that segregate in affected males and female carriers. The variant in a New Zealand family (p.Asp371His) predicts the substitution of a residue in the motor domain of the protein while the one in a US family (p.Arg771Lys) predicts the substitution of a residue in the domain that interacts with Protein Regulator of Cytokinesis 1 (PRC1). We demonstrated that the gene is expressed in the developing tooth bud during development, and that the p.Arg771Lys variant influences cell migration in an in vitro assay. These data implicate missense variations in KIF4A in a pathogenic mechanism that causes taurodontism, microdontia and dens invaginatus phenotypes.

6.
Cell Rep ; 25(10): 2729-2741.e6, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30517861

RESUMEN

The mammalian neocortex has undergone remarkable changes through evolution. A consequence of such rapid evolutionary events could be a trade-off that has rendered the brain susceptible to certain neurodevelopmental and neuropsychiatric conditions. We analyzed the exomes of 65 patients with the structural brain malformation periventricular nodular heterotopia (PH). De novo coding variants were observed in excess in genes defining a transcriptomic signature of basal radial glia, a cell type linked to brain evolution. In addition, we located two variants in human isoforms of two genes that have no ortholog in mice. Modulating the levels of one of these isoforms for the gene PLEKHG6 demonstrated its role in regulating neuroprogenitor differentiation and neuronal migration via RhoA, with phenotypic recapitulation of PH in human cerebral organoids. This suggests that this PLEKHG6 isoform is an example of a primate-specific genomic element supporting brain development.


Asunto(s)
Movimiento Celular , Factores de Intercambio de Guanina Nucleótido/metabolismo , Neurogénesis , Neuronas/citología , Neuronas/metabolismo , Alelos , Animales , Secuencia de Bases , Encéfalo/embriología , Encéfalo/metabolismo , Exoma/genética , Regulación de la Expresión Génica , Genoma , Factores de Intercambio de Guanina Nucleótido/genética , Células HEK293 , Humanos , Recién Nacido , Masculino , Ratones Endogámicos C57BL , Células-Madre Neurales/metabolismo , Neuroglía/metabolismo , Organoides/embriología , Primates , Isoformas de Proteínas/metabolismo , Especificidad de la Especie , Proteína de Unión al GTP rhoA/metabolismo
7.
PLoS Genet ; 14(6): e1007399, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29912901

RESUMEN

Wilms tumour is a childhood tumour that arises as a consequence of somatic and rare germline mutations, the characterisation of which has refined our understanding of nephrogenesis and carcinogenesis. Here we report that germline loss of function mutations in TRIM28 predispose children to Wilms tumour. Loss of function of this transcriptional co-repressor, which has a role in nephrogenesis, has not previously been associated with cancer. Inactivation of TRIM28, either germline or somatic, occurred through inactivating mutations, loss of heterozygosity or epigenetic silencing. TRIM28-mutated tumours had a monomorphic epithelial histology that is uncommon for Wilms tumour. Critically, these tumours were negative for TRIM28 immunohistochemical staining whereas the epithelial component in normal tissue and other Wilms tumours stained positively. These data, together with a characteristic gene expression profile, suggest that inactivation of TRIM28 provides the molecular basis for defining a previously described subtype of Wilms tumour, that has early age of onset and excellent prognosis.


Asunto(s)
Mutación de Línea Germinal , Neoplasias Renales/genética , Mutación con Pérdida de Función , Recurrencia Local de Neoplasia/genética , Proteína 28 que Contiene Motivos Tripartito/genética , Tumor de Wilms/genética , Adulto , Biomarcadores de Tumor/genética , Epigénesis Genética , Femenino , Perfilación de la Expresión Génica , Humanos , Riñón/patología , Neoplasias Renales/epidemiología , Neoplasias Renales/patología , Masculino , Recurrencia Local de Neoplasia/epidemiología , Recurrencia Local de Neoplasia/patología , Pronóstico , Urotelio/patología , Secuenciación del Exoma , Tumor de Wilms/epidemiología , Tumor de Wilms/patología , Adulto Joven
8.
Am J Hum Genet ; 102(6): 1115-1125, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29805041

RESUMEN

Spondylocarpotarsal synostosis syndrome (SCTS) is characterized by intervertebral fusions and fusion of the carpal and tarsal bones. Biallelic mutations in FLNB cause this condition in some families, whereas monoallelic variants in MYH3, encoding embryonic heavy chain myosin 3, have been implicated in dominantly inherited forms of the disorder. Here, five individuals without FLNB mutations from three families were hypothesized to be affected by recessive SCTS on account of sibling recurrence of the phenotype. Initial whole-exome sequencing (WES) showed that all five were heterozygous for one of two independent splice-site variants in MYH3. Despite evidence indicating that three of the five individuals shared two allelic haplotypes encompassing MYH3, no second variant could be located in the WES datasets. Subsequent genome sequencing of these three individuals demonstrated a variant altering a 5' UTR splice donor site (rs557849165 in MYH3) not represented by exome-capture platforms. When the cohort was expanded to 16 SCTS-affected individuals without FLNB mutations, nine had truncating mutations transmitted by unaffected parents, and six inherited the rs557849165 variant in trans, an observation at odds with the population allele frequency for this variant. The rs557849165 variant disrupts splicing in the 5' UTR but is still permissive of MYH3 translational initiation, albeit with reduced efficiency. Although some MYH3 variants cause dominant SCTS, these data indicate that others (notably truncating variants) do not, except in the context of compound heterozygosity for a second hypomorphic allele. These observations make genetic diagnosis challenging in the context of simplex presentations of the disorder.


Asunto(s)
Anomalías Múltiples/genética , Genes Recesivos , Vértebras Lumbares/anomalías , Enfermedades Musculoesqueléticas/genética , Mutación/genética , Cadenas Pesadas de Miosina/genética , Escoliosis/congénito , Sinostosis/genética , Vértebras Torácicas/anomalías , Alelos , Mapeo Cromosómico , Femenino , Filaminas/genética , Haplotipos/genética , Heterocigoto , Humanos , Masculino , Linaje , Fenotipo , Empalme del ARN/genética , Escoliosis/genética , Síndrome , Secuenciación del Exoma
9.
Front Cell Neurosci ; 12: 57, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29593499

RESUMEN

Disorders of neuronal mispositioning during brain development are phenotypically heterogeneous and their genetic causes remain largely unknown. Here, we report biallelic variants in a Hippo signaling factor-MOB2-in a patient with one such disorder, periventricular nodular heterotopia (PH). Genetic and cellular analysis of both variants confirmed them to be loss-of-function with enhanced sensitivity to transcript degradation via nonsense mediated decay (NMD) or increased protein turnover via the proteasome. Knockdown of Mob2 within the developing mouse cortex demonstrated its role in neuronal positioning. Cilia positioning and number within migrating neurons was also impaired with comparable defects detected following a reduction in levels of an upstream modulator of Mob2 function, Dchs1, a previously identified locus associated with PH. Moreover, reduced Mob2 expression increased phosphorylation of Filamin A, an actin cross-linking protein frequently mutated in cases of this disorder. These results reveal a key role for Mob2 in correct neuronal positioning within the developing cortex and outline a new candidate locus for PH development.

10.
Hum Mutat ; 39(1): 103-113, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29024177

RESUMEN

Loss-of-function mutations in the X-linked gene FLNA can lead to abnormal neuronal migration, vascular and cardiac defects, and congenital intestinal pseudo-obstruction (CIPO), the latter characterized by anomalous intestinal smooth muscle layering. Survival in male hemizygotes for such mutations is dependent on retention of residual FLNA function but it is unclear why a subgroup of males with mutations in the 5' end of the gene can present with CIPO alone. Here, we demonstrate evidence for the presence of two FLNA isoforms differing by 28 residues at the N-terminus initiated at ATG+1 and ATG+82 . A male with CIPO (c.18_19del) exclusively expressed FLNA ATG+82 , implicating the longer protein isoform (ATG+1 ) in smooth muscle development. In contrast, mutations leading to reduction of both isoforms are associated with compound phenotypes affecting the brain, heart, and intestine. RNA-seq data revealed three distinct transcription start sites, two of which produce a protein isoform utilizing ATG+1 while the third utilizes ATG+82 . Transcripts sponsoring translational initiation at ATG+1 predominate in intestinal smooth muscle, and are more abundant compared with the level measured in fibroblasts. Together these observations describe a new mechanism of tissue-specific regulation of FLNA that could reflect the differing mechanical requirements of these cell types during development.


Asunto(s)
Filaminas/genética , Estudios de Asociación Genética , Heterogeneidad Genética , Mutación con Pérdida de Función , Fenotipo , Transcripción Genética , Adolescente , Encéfalo/anomalías , Encéfalo/diagnóstico por imagen , Niño , Secuencia Conservada , Análisis Mutacional de ADN , Femenino , Filaminas/química , Filaminas/metabolismo , Tracto Gastrointestinal/metabolismo , Expresión Génica , Humanos , Imagen por Resonancia Magnética , Masculino , Músculo Liso/metabolismo , Isoformas de Proteínas , Adulto Joven
11.
Am J Hum Genet ; 101(1): 139-148, 2017 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-28686853

RESUMEN

We report 15 individuals with de novo pathogenic variants in WDR26. Eleven of the individuals carry loss-of-function mutations, and four harbor missense substitutions. These 15 individuals comprise ten females and five males, and all have intellectual disability with delayed speech, a history of febrile and/or non-febrile seizures, and a wide-based, spastic, and/or stiff-legged gait. These subjects share a set of common facial features that include a prominent maxilla and upper lip that readily reveal the upper gingiva, widely spaced teeth, and a broad nasal tip. Together, these features comprise a recognizable facial phenotype. We compared these features with those of chromosome 1q41q42 microdeletion syndrome, which typically contains WDR26, and noted that clinical features are consistent between the two subsets, suggesting that haploinsufficiency of WDR26 contributes to the pathology of 1q41q42 microdeletion syndrome. Consistent with this, WDR26 loss-of-function single-nucleotide mutations identified in these subjects lead to nonsense-mediated decay with subsequent reduction of RNA expression and protein levels. We derived a structural model of WDR26 and note that missense variants identified in these individuals localize to highly conserved residues of this WD-40-repeat-containing protein. Given that WDR26 mutations have been identified in ∼1 in 2,000 of subjects in our clinical cohorts and that WDR26 might be poorly annotated in exome variant-interpretation pipelines, we would anticipate that this disorder could be more common than currently appreciated.


Asunto(s)
Facies , Marcha/genética , Haploinsuficiencia/genética , Discapacidad Intelectual/genética , Proteínas/genética , Convulsiones/genética , Proteínas Adaptadoras Transductoras de Señales , Secuencia de Aminoácidos , Secuencia de Bases , Preescolar , Deleción Cromosómica , Femenino , Crecimiento y Desarrollo/genética , Humanos , Discapacidad Intelectual/complicaciones , Masculino , Mutación/genética , Proteínas/química , Estabilidad del ARN/genética , Convulsiones/complicaciones , Síndrome
12.
Nat Commun ; 8: 16077, 2017 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-28681861

RESUMEN

Multinucleate cellular syncytial formation is a hallmark of skeletal muscle differentiation. Myomaker, encoded by Mymk (Tmem8c), is a well-conserved plasma membrane protein required for myoblast fusion to form multinucleated myotubes in mouse, chick, and zebrafish. Here, we report that autosomal recessive mutations in MYMK (OMIM 615345) cause Carey-Fineman-Ziter syndrome in humans (CFZS; OMIM 254940) by reducing but not eliminating MYMK function. We characterize MYMK-CFZS as a congenital myopathy with marked facial weakness and additional clinical and pathologic features that distinguish it from other congenital neuromuscular syndromes. We show that a heterologous cell fusion assay in vitro and allelic complementation experiments in mymk knockdown and mymkinsT/insT zebrafish in vivo can differentiate between MYMK wild type, hypomorphic and null alleles. Collectively, these data establish that MYMK activity is necessary for normal muscle development and maintenance in humans, and expand the spectrum of congenital myopathies to include cell-cell fusion deficits.


Asunto(s)
Proteínas de la Membrana/genética , Síndrome de Mobius/genética , Morfogénesis/genética , Proteínas Musculares/genética , Músculo Esquelético/metabolismo , Enfermedades Musculares/genética , Mutación , Mioblastos/metabolismo , Síndrome de Pierre Robin/genética , Proteínas de Pez Cebra/genética , Adulto , Secuencia de Aminoácidos , Animales , Fusión Celular , Niño , Modelos Animales de Enfermedad , Embrión no Mamífero , Femenino , Expresión Génica , Genes Recesivos , Prueba de Complementación Genética , Humanos , Lactante , Masculino , Proteínas de la Membrana/deficiencia , Síndrome de Mobius/metabolismo , Síndrome de Mobius/patología , Proteínas Musculares/deficiencia , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/patología , Enfermedades Musculares/metabolismo , Enfermedades Musculares/patología , Mioblastos/patología , Linaje , Síndrome de Pierre Robin/metabolismo , Síndrome de Pierre Robin/patología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Pez Cebra , Proteínas de Pez Cebra/deficiencia
13.
Am J Med Genet A ; 173(7): 1739-1746, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28498505

RESUMEN

Frontometaphyseal dysplasia (FMD) is caused by gain-of-function mutations in the X-linked gene FLNA in approximately 50% of patients. Recently we characterized an autosomal dominant form of FMD (AD-FMD) caused by mutations in MAP3K7, which accounts for the condition in the majority of patients who lack a FLNA mutation. We previously also described a patient with a de novo variant in TAB2, which we hypothesized was causative of another form of AD-FMD. In this study, a cohort of 20 individuals with AD-FMD is clinically evaluated. This cohort consists of 15 individuals with the recently described, recurrent mutation (c.1454C>T) in MAP3K7, as well as three individuals with missense mutations that result in substitutions in the N-terminal kinase domain of TGFß-activated kinase 1 (TAK1), encoded by MAP3K7. Additionally, two individuals have missense variants in the gene TAB2, which encodes a protein with a close functional relationship to TAK1, TAK1-associated binding protein 2 (TAB2). Although the X-linked and autosomal dominant forms of FMD are very similar, there are distinctions to be made between the two conditions. Individuals with AD-FMD have characteristic facial features, and are more likely to be deaf, have scoliosis and cervical fusions, and have a cleft palate. Furthermore, there are features only found in AD-FMD in our review of the literature including valgus deformity of the feet and predisposition to keloid scarring. Finally, intellectual disability is present in a small number of subjects with AD-FMD but has not been described in association with X-linked FMD.

14.
Am J Hum Genet ; 99(2): 392-406, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27426733

RESUMEN

Frontometaphyseal dysplasia (FMD) is a progressive sclerosing skeletal dysplasia affecting the long bones and skull. The cause of FMD in some individuals is gain-of-function mutations in FLNA, although how these mutations result in a hyperostotic phenotype remains unknown. Approximately one half of individuals with FMD have no identified mutation in FLNA and are phenotypically very similar to individuals with FLNA mutations, except for an increased tendency to form keloid scars. Using whole-exome sequencing and targeted Sanger sequencing in 19 FMD-affected individuals with no identifiable FLNA mutation, we identified mutations in two genes-MAP3K7, encoding transforming growth factor ß (TGF-ß)-activated kinase (TAK1), and TAB2, encoding TAK1-associated binding protein 2 (TAB2). Four mutations were found in MAP3K7, including one highly recurrent (n = 15) de novo mutation (c.1454C>T [ p.Pro485Leu]) proximal to the coiled-coil domain of TAK1 and three missense mutations affecting the kinase domain (c.208G>C [p.Glu70Gln], c.299T>A [p.Val100Glu], and c.502G>C [p.Gly168Arg]). Notably, the subjects with the latter three mutations had a milder FMD phenotype. An additional de novo mutation was found in TAB2 (c.1705G>A, p.Glu569Lys). The recurrent mutation does not destabilize TAK1, or impair its ability to homodimerize or bind TAB2, but it does increase TAK1 autophosphorylation and alter the activity of more than one signaling pathway regulated by the TAK1 kinase complex. These findings show that dysregulation of the TAK1 complex produces a close phenocopy of FMD caused by FLNA mutations. Furthermore, they suggest that the pathogenesis of some of the filaminopathies caused by FLNA mutations might be mediated by misregulation of signaling coordinated through the TAK1 signaling complex.


Asunto(s)
Frente/anomalías , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Mutación/genética , Osteocondrodisplasias/genética , Transducción de Señal/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Femenino , Filaminas/genética , Humanos , Sistema de Señalización de MAP Quinasas/genética , Masculino , FN-kappa B/metabolismo , Osteocondrodisplasias/metabolismo , Fosforilación , Unión Proteica , Multimerización de Proteína
15.
Am J Hum Genet ; 97(6): 837-47, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26637977

RESUMEN

The periosteum contributes to bone repair and maintenance of cortical bone mass. In contrast to the understanding of bone development within the epiphyseal growth plate, factors that regulate periosteal osteogenesis have not been studied as intensively. Osteofibrous dysplasia (OFD) is a congenital disorder of osteogenesis and is typically sporadic and characterized by radiolucent lesions affecting the cortical bone immediately under the periosteum of the tibia and fibula. We identified germline mutations in MET, encoding a receptor tyrosine kinase, that segregate with an autosomal-dominant form of OFD in three families and a mutation in a fourth affected subject from a simplex family and with bilateral disease. Mutations identified in all families with dominant inheritance and in the one simplex subject with bilateral disease abolished the splice inclusion of exon 14 in MET transcripts, which resulted in a MET receptor (MET(Δ14)) lacking a cytoplasmic juxtamembrane domain. Splice exclusion of this domain occurs during normal embryonic development, and forced induction of this exon-exclusion event retarded osteoblastic differentiation in vitro and inhibited bone-matrix mineralization. In an additional subject with unilateral OFD, we identified a somatic MET mutation, also affecting exon 14, that substituted a tyrosine residue critical for MET receptor turnover and, as in the case of the MET(Δ14) mutations, had a stabilizing effect on the mature protein. Taken together, these data show that aberrant MET regulation via the juxtamembrane domain subverts core MET receptor functions that regulate osteogenesis within cortical diaphyseal bone.


Asunto(s)
Enfermedades del Desarrollo Óseo/genética , Exones , Mutación de Línea Germinal , Osteogénesis/genética , Periostio/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Adulto , Secuencia de Bases , Enfermedades del Desarrollo Óseo/metabolismo , Enfermedades del Desarrollo Óseo/patología , Diferenciación Celular , Niño , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes Dominantes , Humanos , Masculino , Persona de Mediana Edad , Datos de Secuencia Molecular , Osteoblastos/metabolismo , Osteoblastos/patología , Linaje , Periostio/crecimiento & desarrollo , Periostio/patología , Cultivo Primario de Células , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-met/metabolismo , Empalme del ARN
16.
Am J Hum Genet ; 96(4): 623-30, 2015 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-25817014

RESUMEN

Robinow syndrome (RS) is a phenotypically and genetically heterogeneous condition that can be caused by mutations in genes encoding components of the non-canonical Wnt signaling pathway. In contrast, germline mutations that act to increase canonical Wnt signaling lead to distinctive osteosclerotic phenotypes. Here, we identified de novo frameshift mutations in DVL1, a mediator of both canonical and non-canonical Wnt signaling, as the cause of RS-OS, an RS subtype involving osteosclerosis, in three unrelated individuals. The mutations all delete the DVL1 C terminus and replace it, in each instance, with a novel, highly basic sequence. We showed the presence of mutant transcript in fibroblasts from one individual with RS-OS and demonstrated unimpaired protein stability with transfected GFP-tagged constructs bearing a frameshift mutation. In vitro TOPFlash assays, in apparent contradiction to the osteosclerotic phenotype, revealed that the mutant allele was less active than the wild-type allele in the canonical Wnt signaling pathway. However, when the mutant and wild-type alleles were co-expressed, canonical Wnt activity was 2-fold higher than that in the wild-type construct alone. This work establishes that DVL1 mutations cause a specific RS subtype, RS-OS, and that the osteosclerosis associated with this subtype might be the result of an interaction between the wild-type and mutant alleles and thus lead to elevated canonical Wnt signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/patología , Enanismo/genética , Enanismo/patología , Mutación del Sistema de Lectura/genética , Deformidades Congénitas de las Extremidades/genética , Deformidades Congénitas de las Extremidades/patología , Osteosclerosis/genética , Fosfoproteínas/genética , Anomalías Urogenitales/genética , Anomalías Urogenitales/patología , Vía de Señalización Wnt/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Dishevelled , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Fosfoproteínas/metabolismo , Fosforilación
17.
Mol Vis ; 15: 1700-8, 2009 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-19710953

RESUMEN

PURPOSE: With advances in phenotyping tools and availability of molecular characterization, an increasing number of phenotypically and genotypically diverse inherited corneal dystrophies are described. We aimed to determine the underlying causative genetic mechanism in a three-generation pedigree affected with a unique anterior membrane corneal dystrophy characterized by early onset recurrent corneal erosions, small discrete focal opacities at the level of Bowman layer and anterior stroma, anterior stromal flecks, and prominent corneal nerves. METHODS: Twenty affected and unaffected members of a three-generation family were examined and extensively clinically characterized including corneal topography and in vivo confocal microscopy, and biological specimens were collected for DNA extraction. Mutational analysis of two corneal genes (TGFBI [Transforming Growth factor-beta induced] and ZEB1 [zinc finger E box-binding homeobox 1]) was undertaken, in addition to testing with the Asper Corneal Dystrophy gene chip (Asper Ophthalmics, Tartu, Estonia). Subsequent Genotyping To 11 Known Corneal Gene Loci (COL8A2 [Collagen, Type VIII, Alpha-2], TACSTD2 [Tumor-Associated Calcium Signal Transducer 2], PIP5K3 [Phosphatidylinositol-3-Phosphate 5-Kinase, Type III], GSN [Gelsolin], KERA [Keratocan], VSX1 [Visual System Homeobox Gene 1], COL6A1 [Collagen, Type VI, Alpha-1], MMP9 [Matrix Metalloproteinase 9], KRT3 [Keratin 3]), and two putative loci, 3p14-q13 and 15q22.33-24) was undertaken using polymorphic markers, and haplotypes constructed. Multipoint linkage analysis was performed to generate LOD scores and produce LOD plots across the candidate intervals. RESULTS: No pathogenic sequence variations were detected in TGFBI or ZEB1 of the proband nor on the Asper Corneal Dystrophy gene chip (302 mutations in 12 genes). Multipoint linkage analysis of 11 known corneal genes and loci generated negative LOD plots and was able to exclude all genes tested including PIP5K3. CONCLUSIONS: Exclusion of linkage to candidate corneal loci combined with an absence of pathogenic mutations in known corneal genes in this pedigree suggest a different genetic causative mechanism in this dystrophy than the previously documented corneal genes. This unique phenotype of an anterior membrane dystrophy may therefore provide an opportunity to identify a new gene responsible for corneal disease.


Asunto(s)
Distrofias Hereditarias de la Córnea/genética , Distrofias Hereditarias de la Córnea/patología , Genes Dominantes , Linaje , Adolescente , Adulto , Anciano , Niño , Preescolar , Córnea/patología , Demografía , Femenino , Humanos , Escala de Lod , Masculino , Membranas/patología , Microscopía Confocal , Persona de Mediana Edad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...