Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Hum Genet ; 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-39014191

RESUMEN

Camurati-Engelmann disease (CED) is an autosomal dominant bone dysplasia characterized by progressive hyperostosis of the skull base and diaphyses of the long bones. CED is further divided into two subtypes, CED1 and CED2, according to the presence or absence of TGFB1 mutations, respectively. In this study, we used exome sequencing to investigate the genetic cause of CED2 in three pedigrees and identified two de novo heterozygous mutations in TGFB2 among the three patients. Both mutations were located in the region of the gene encoding the straitjacket subdomain of the latency-associated peptide (LAP) of pro-TGF-ß2. Structural simulations of the mutant LAPs suggested that the mutations could cause significant conformational changes and lead to a reduction in TGF-ß2 inactivation. An activity assay confirmed a significant increase in TGF-ß2/SMAD signaling. In vitro osteogenic differentiation experiment using iPS cells from one of the CED2 patients showed significantly enhanced ossification, suggesting that the pathogenic mechanism of CED2 is increased activation of TGF-ß2 by loss-of-function of the LAP. These results, in combination with the difference in hyperostosis patterns between CED1 and CED2, suggest distinct functions between TGFB1 and TGFB2 in human skeletal development and homeostasis.

2.
Am J Med Genet A ; 191(5): 1164-1209, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36779427

RESUMEN

The "Nosology of genetic skeletal disorders" has undergone its 11th revision and now contains 771 entries associated with 552 genes reflecting advances in molecular delineation of new disorders thanks to advances in DNA sequencing technology. The most significant change as compared to previous versions is the adoption of the dyadic naming system, systematically associating a phenotypic entity with the gene it arises from. We consider this a significant step forward as dyadic naming is more informative and less prone to errors than the traditional use of list numberings and eponyms. Despite the adoption of dyadic naming, efforts have been made to maintain strong ties to the MIM catalog and its historical data. As with the previous versions, the list of disorders and genes in the Nosology may be useful in considering the differential diagnosis in the clinic, directing bioinformatic analysis of next-generation sequencing results, and providing a basis for novel advances in biology and medicine.

3.
Am J Hum Genet ; 104(3): 422-438, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30773277

RESUMEN

SPONASTRIME dysplasia is an autosomal-recessive spondyloepimetaphyseal dysplasia characterized by spine (spondylar) abnormalities, midface hypoplasia with a depressed nasal bridge, metaphyseal striations, and disproportionate short stature. Scoliosis, coxa vara, childhood cataracts, short dental roots, and hypogammaglobulinemia have also been reported in this disorder. Although an autosomal-recessive inheritance pattern has been hypothesized, pathogenic variants in a specific gene have not been discovered in individuals with SPONASTRIME dysplasia. Here, we identified bi-allelic variants in TONSL, which encodes the Tonsoku-like DNA repair protein, in nine subjects (from eight families) with SPONASTRIME dysplasia, and four subjects (from three families) with short stature of varied severity and spondylometaphyseal dysplasia with or without immunologic and hematologic abnormalities, but no definitive metaphyseal striations at diagnosis. The finding of early embryonic lethality in a Tonsl-/- murine model and the discovery of reduced length, spinal abnormalities, reduced numbers of neutrophils, and early lethality in a tonsl-/- zebrafish model both support the hypomorphic nature of the identified TONSL variants. Moreover, functional studies revealed increased amounts of spontaneous replication fork stalling and chromosomal aberrations, as well as fewer camptothecin (CPT)-induced RAD51 foci in subject-derived cell lines. Importantly, these cellular defects were rescued upon re-expression of wild-type (WT) TONSL; this rescue is consistent with the hypothesis that hypomorphic TONSL variants are pathogenic. Overall, our studies in humans, mice, zebrafish, and subject-derived cell lines confirm that pathogenic variants in TONSL impair DNA replication and homologous recombination-dependent repair processes, and they lead to a spectrum of skeletal dysplasia phenotypes with numerous extra-skeletal manifestations.


Asunto(s)
Inestabilidad Cromosómica , Daño del ADN , Variación Genética , Anomalías Musculoesqueléticas/patología , FN-kappa B/genética , Osteocondrodisplasias/patología , Adolescente , Adulto , Alelos , Animales , Células Cultivadas , Niño , Preescolar , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Estudios de Asociación Genética , Humanos , Ratones , Ratones Noqueados , Anomalías Musculoesqueléticas/genética , Osteocondrodisplasias/genética , Secuenciación del Exoma , Adulto Joven , Pez Cebra
4.
Hum Mutat ; 40(12): 2344-2352, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31389106

RESUMEN

Campomelic dysplasia (CD) is an autosomal dominant, perinatal lethal skeletal dysplasia characterized by a small chest and short long bones with bowing of the lower extremities. CD is the result of heterozygosity for mutations in the gene encoding the chondrogenesis master regulator, SOX9. Loss-of-function mutations have been identified in most CD cases so it has been assumed that the disease results from haploinsufficiency for SOX9. Here, we identified distal truncating SOX9 mutations in four unrelated CD cases. The mutations all leave the dimerization and DNA-binding domains intact and cultured chondrocytes from three of the four cases synthesized truncated SOX9. Relative to CD resulting from haploinsufficiency, there was decreased transactivation activity toward a major transcriptional target, COL2A1, consistent with the mutations exerting a dominant-negative effect. For one of the cases, the phenotypic consequence was a very severe form of CD, with a pronounced effect on vertebral and limb development. The data identify a novel molecular mechanism of disease in CD in which the truncated protein leads to a distinct and more significant effect on SOX9 function.


Asunto(s)
Displasia Campomélica/genética , Secuenciación del Exoma/métodos , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Displasia Campomélica/metabolismo , Células Cultivadas , Condrocitos/citología , Condrocitos/metabolismo , Colágeno Tipo II/genética , Femenino , Haploinsuficiencia , Humanos , Embarazo , Diagnóstico Prenatal , Eliminación de Secuencia
5.
Am J Med Genet A ; 179(12): 2393-2419, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31633310

RESUMEN

The application of massively parallel sequencing technology to the field of skeletal disorders has boosted the discovery of the underlying genetic defect for many of these diseases. It has also resulted in the delineation of new clinical entities and the identification of genes and pathways that had not previously been associated with skeletal disorders. These rapid advances have prompted the Nosology Committee of the International Skeletal Dysplasia Society to revise and update the last (2015) version of the Nosology and Classification of Genetic Skeletal Disorders. This newest and tenth version of the Nosology comprises 461 different diseases that are classified into 42 groups based on their clinical, radiographic, and/or molecular phenotypes. Remarkably, pathogenic variants affecting 437 different genes have been found in 425/461 (92%) of these disorders. By providing a reference list of recognized entities and their causal genes, the Nosology should help clinicians achieve accurate diagnoses for their patients and help scientists advance research in skeletal biology.


Asunto(s)
Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Enfermedades Musculoesqueléticas/diagnóstico , Enfermedades Musculoesqueléticas/genética , Alelos , Estudios de Asociación Genética/métodos , Humanos , Patrón de Herencia , Fenotipo , Guías de Práctica Clínica como Asunto
6.
PLoS Genet ; 12(9): e1006307, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27622494

RESUMEN

The acrofacial dysostoses (AFD) are a genetically heterogeneous group of inherited disorders with craniofacial and limb abnormalities. Rodriguez syndrome is a severe, usually perinatal lethal AFD, characterized by severe retrognathia, oligodactyly and lower limb abnormalities. Rodriguez syndrome has been proposed to be a severe form of Nager syndrome, a non-lethal AFD that results from mutations in SF3B4, a component of the U2 small nuclear ribonucleoprotein particle (U2 snRNP). Furthermore, a case with a phenotype intermediate between Rodriguez and Nager syndromes has been shown to have an SF3B4 mutation. We identified heterozygosity for SF3B4 mutations in Rodriguez syndrome, confirming that the phenotype is a dominant disorder that is allelic with Nager syndrome. The mutations led to reduced SF3B4 synthesis and defects in mRNA splicing, primarily exon skipping. The mutations also led to reduced expression in growth plate chondrocytes of target genes, including the DLX5, DLX6, SOX9, and SOX6 transcription factor genes, which are known to be important for skeletal development. These data provide mechanistic insight toward understanding how SF3B4 mutations lead to the skeletal abnormalities observed in the acrofacial dysostoses.


Asunto(s)
Condrocitos/metabolismo , Deformidades Congénitas de la Mano/genética , Disostosis Mandibulofacial/genética , Mutación , Factores de Empalme de ARN/genética , Empalme del ARN , Adulto , Células Cultivadas , Femenino , Deformidades Congénitas de la Mano/diagnóstico por imagen , Deformidades Congénitas de la Mano/patología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Recién Nacido , Masculino , Disostosis Mandibulofacial/diagnóstico por imagen , Disostosis Mandibulofacial/patología , Linaje , Fenotipo , Factores de Empalme de ARN/metabolismo , Factores de Transcripción SOXD/genética , Factores de Transcripción SOXD/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
PLoS Genet ; 12(3): e1005936, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27019229

RESUMEN

Spondylocarpotarsal synostosis (SCT) is an autosomal recessive disorder characterized by progressive vertebral fusions and caused by loss of function mutations in Filamin B (FLNB). FLNB acts as a signaling scaffold by linking the actin cytoskleteon to signal transduction systems, yet the disease mechanisms for SCT remain unclear. Employing a Flnb knockout mouse, we found morphologic and molecular evidence that the intervertebral discs (IVDs) of Flnb-/-mice undergo rapid and progressive degeneration during postnatal development as a result of abnormal cell fate changes in the IVD, particularly the annulus fibrosus (AF). In Flnb-/-mice, the AF cells lose their typical fibroblast-like characteristics and acquire the molecular and phenotypic signature of hypertrophic chondrocytes. This change is characterized by hallmarks of endochondral-like ossification including alterations in collagen matrix, expression of Collagen X, increased apoptosis, and inappropriate ossification of the disc tissue. We show that conversion of the AF cells into chondrocytes is coincident with upregulated TGFß signaling via Smad2/3 and BMP induced p38 signaling as well as sustained activation of canonical and noncanonical target genes p21 and Ctgf. These findings indicate that FLNB is involved in attenuation of TGFß/BMP signaling and influences AF cell fate. Furthermore, we demonstrate that the IVD disruptions in Flnb-/-mice resemble aging degenerative discs and reveal new insights into the molecular causes of vertebral fusions and disc degeneration.


Asunto(s)
Anomalías Múltiples/genética , Filaminas/genética , Degeneración del Disco Intervertebral/genética , Vértebras Lumbares/anomalías , Enfermedades Musculoesqueléticas/genética , Escoliosis/congénito , Sinostosis/genética , Vértebras Torácicas/anomalías , Factor de Crecimiento Transformador beta/genética , Anomalías Múltiples/patología , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Animales , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Condrocitos/metabolismo , Condrocitos/patología , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Modelos Animales de Enfermedad , Placa de Crecimiento/crecimiento & desarrollo , Placa de Crecimiento/patología , Humanos , Degeneración del Disco Intervertebral/patología , Vértebras Lumbares/patología , Ratones , Ratones Noqueados , Enfermedades Musculoesqueléticas/patología , Escoliosis/genética , Escoliosis/patología , Transducción de Señal , Proteínas Smad/genética , Proteínas Smad/metabolismo , Columna Vertebral/crecimiento & desarrollo , Columna Vertebral/patología , Sinostosis/patología , Vértebras Torácicas/patología
9.
Hum Mutat ; 39(1): 152-166, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29068549

RESUMEN

Defects in the biosynthesis and/or function of primary cilia cause a spectrum of disorders collectively referred to as ciliopathies. A subset of these disorders is distinguished by profound abnormalities of the skeleton that include a long narrow chest with markedly short ribs, extremely short limbs, and polydactyly. These include the perinatal lethal short-rib polydactyly syndromes (SRPS) and the less severe asphyxiating thoracic dystrophy (ATD), Ellis-van Creveld (EVC) syndrome, and cranioectodermal dysplasia (CED) phenotypes. To identify new genes and define the spectrum of mutations in the skeletal ciliopathies, we analyzed 152 unrelated families with SRPS, ATD, and EVC. Causal variants were discovered in 14 genes in 120 families, including one newly associated gene and two genes previously associated with other ciliopathies. These three genes encode components of three different ciliary complexes; FUZ, which encodes a planar cell polarity complex molecule; TRAF3IP1, which encodes an anterograde ciliary transport protein; and LBR, which encodes a nuclear membrane protein with sterol reductase activity. The results established the molecular basis of SRPS type IV, in which mutations were identified in four different ciliary genes. The data provide systematic insight regarding the genotypes associated with a large cohort of these genetically heterogeneous phenotypes and identified new ciliary components required for normal skeletal development.


Asunto(s)
Ciliopatías/diagnóstico , Ciliopatías/genética , Estudios de Asociación Genética , Variación Genética , Fenotipo , Esqueleto/anomalías , Dineínas Citoplasmáticas/genética , Marcadores Genéticos , Genotipo , Humanos , Péptidos y Proteínas de Señalización Intercelular , Mutación , Proteínas/genética , Radiografía , Secuenciación del Exoma
10.
Hum Mol Genet ; 25(18): 4012-4020, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27466190

RESUMEN

The short-rib polydactyly syndromes (SRPS) encompass a radiographically and genetically heterogeneous group of skeletal ciliopathies that are characterized by a long narrow chest, short extremities, and variable occurrence of polydactyly. Radiographic abnormalities include undermineralization of the calvarium, shortened and bowed appendicular bones, trident shaped acetabula and polydactyly. In a case of SRPS we identified compound heterozygosity for mutations in IFT52, which encodes a component of the anterograde intraflagellar transport complex. The IFT52 mutant cells synthesized a significantly reduced amount of IFT52 protein, leading to reduced synthesis of IFT74, IFT81, IFT88 and ARL13B, other key anterograde complex members. Ciliogenesis was also disrupted in the mutant cells, with a 60% reduction in the presence of cilia on mutant cells and loss of cilia length regulation for the cells with cilia. These data demonstrate that IFT52 is essential for anterograde complex integrity and for the biosynthesis and maintenance of cilia. The data identify a new locus for SRPS and show that IFT52 mutations result in a ciliopathy with primary effects on the skeleton.


Asunto(s)
Proteínas Portadoras/genética , Cilios/genética , Ciliopatías/genética , Síndrome de Costilla Pequeña y Polidactilia/genética , Cilios/metabolismo , Ciliopatías/fisiopatología , Proteínas del Citoesqueleto/genética , Flagelos/genética , Flagelos/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Complejos Multiproteicos/genética , Proteínas Musculares/genética , Mutación/genética , Síndrome de Costilla Pequeña y Polidactilia/fisiopatología , Esqueleto/crecimiento & desarrollo , Esqueleto/metabolismo , Esqueleto/patología , Proteínas Supresoras de Tumor/genética
11.
Hum Mol Genet ; 25(18): 3998-4011, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27466187

RESUMEN

The short rib polydactyly syndromes (SRPS) are a group of recessively inherited, perinatal-lethal skeletal disorders primarily characterized by short ribs, shortened long bones, varying types of polydactyly and concomitant visceral abnormalities. Mutations in several genes affecting cilia function cause SRPS, revealing a role for cilia function in skeletal development. To identify additional SRPS genes and discover novel ciliary molecules required for normal skeletogenesis, we performed exome sequencing in a cohort of patients and identified homozygosity for a missense mutation, p.E80K, in Intestinal Cell Kinase, ICK, in one SRPS family. The p.E80K mutation abolished serine/threonine kinase activity, resulting in altered ICK subcellular and ciliary localization, increased cilia length, aberrant cartilage growth plate structure, defective Hedgehog and altered ERK signalling. These data identify ICK as an SRPS-associated gene and reveal that abnormalities in signalling pathways contribute to defective skeletogenesis.


Asunto(s)
Anomalías Múltiples/genética , Proteínas Hedgehog/genética , Proteínas Serina-Treonina Quinasas/genética , Síndrome de Costilla Pequeña y Polidactilia/genética , Esqueleto/crecimiento & desarrollo , Anomalías Múltiples/fisiopatología , Cilios/genética , Cilios/patología , Exoma/genética , Femenino , Humanos , Lactante , Sistema de Señalización de MAP Quinasas , Linaje , Embarazo , Análisis de Secuencia de ADN , Síndrome de Costilla Pequeña y Polidactilia/patología , Transducción de Señal , Esqueleto/anomalías
12.
Am J Med Genet A ; 176(12): 2887-2891, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30450842

RESUMEN

Stickler syndrome is a connective tissue disorder characterized by hearing loss, ocular anomalies, palatal defects, and skeletal abnormalities. The autosomal dominant form is the most common, but autosomal recessive forms have also been described. We report the second case of autosomal recessive Stickler syndrome due to homozygosity for a loss of function mutation in COL9A3, which encodes the α3 chain of type IX procollagen. The clinical features were similar to the previously described COL9A3 Stickler syndrome family, including moderate to severe sensorineural hearing loss, high myopia, and both tibial and femoral bowing at birth. Radiographs demonstrated abnormal capital femoral epiphyses and mild irregularities of the vertebral endplates. This case further establishes the phenotype associated with mutations in this gene. We suggest that loss of the α3 chain of type IX collagen results in a Stickler syndrome phenotype similar to that of the other autosomal recessive forms caused by mutations in genes encoding the α1 and α2 chains of type IX collagen.


Asunto(s)
Artritis/diagnóstico , Artritis/genética , Colágeno Tipo IX/genética , Enfermedades del Tejido Conjuntivo/diagnóstico , Enfermedades del Tejido Conjuntivo/genética , Genes Recesivos , Pérdida Auditiva Sensorineural/diagnóstico , Pérdida Auditiva Sensorineural/genética , Mutación , Desprendimiento de Retina/diagnóstico , Desprendimiento de Retina/genética , Niño , Análisis Mutacional de ADN , Homocigoto , Humanos , Recién Nacido , Fenotipo , Radiografía , Secuenciación del Exoma
13.
BMC Genomics ; 18(1): 983, 2017 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-29262782

RESUMEN

BACKGROUND: Chondrogenesis is the earliest stage of skeletal development and is a highly dynamic process, integrating the activities and functions of transcription factors, cell signaling molecules and extracellular matrix proteins. The molecular mechanisms underlying chondrogenesis have been extensively studied and multiple key regulators of this process have been identified. However, a genome-wide overview of the gene regulatory network in chondrogenesis has not been achieved. RESULTS: In this study, employing RNA sequencing, we identified 332 protein coding genes and 34 long non-coding RNA (lncRNA) genes that are highly selectively expressed in human fetal growth plate chondrocytes. Among the protein coding genes, 32 genes were associated with 62 distinct human skeletal disorders and 153 genes were associated with skeletal defects in knockout mice, confirming their essential roles in skeletal formation. These gene products formed a comprehensive physical interaction network and participated in multiple cellular processes regulating skeletal development. The data also revealed 34 transcription factors and 11,334 distal enhancers that were uniquely active in chondrocytes, functioning as transcriptional regulators for the cartilage-selective genes. CONCLUSIONS: Our findings revealed a complex gene regulatory network controlling skeletal development whereby transcription factors, enhancers and lncRNAs participate in chondrogenesis by transcriptional regulation of key genes. Additionally, the cartilage-selective genes represent candidate genes for unsolved human skeletal disorders.


Asunto(s)
Condrocitos/metabolismo , Condrogénesis/genética , Redes Reguladoras de Genes , Placa de Crecimiento/metabolismo , Cartílago/embriología , Cartílago/metabolismo , Elementos de Facilitación Genéticos , Feto , Perfilación de la Expresión Génica , Placa de Crecimiento/citología , Humanos , Mapas de Interacción de Proteínas , ARN Largo no Codificante/metabolismo
14.
Hum Mol Genet ; 24(7): 1918-28, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25510505

RESUMEN

Osteogenesis imperfecta (OI) is a genetic disorder that results in low bone mineral density and brittle bones. Most cases result from dominant mutations in the type I procollagen genes, but mutations in a growing number of genes have been identified that produce autosomal recessive forms of the disease. Among these include mutations in the genes SERPINH1 and FKBP10, which encode the type I procollagen chaperones HSP47 and FKBP65, respectively, and predominantly produce a moderately severe form of OI. Little is known about the biochemical consequences of the mutations and how they produce OI. We have identified a new OI mutation in SERPINH1 that results in destabilization and mislocalization of HSP47 and secondarily has similar effects on FKBP65. We found evidence that HSP47 and FKBP65 act cooperatively during posttranslational maturation of type I procollagen and that FKBP65 and HSP47 but fail to properly interact in mutant HSP47 cells. These results thus reveal a common cellular pathway in cases of OI caused by HSP47 and FKBP65 deficiency.


Asunto(s)
Colágeno Tipo I/biosíntesis , Proteínas del Choque Térmico HSP47/metabolismo , Osteogénesis Imperfecta/metabolismo , Procolágeno/biosíntesis , Proteínas de Unión a Tacrolimus/metabolismo , Adulto , Secuencia de Aminoácidos , Secuencia de Bases , Preescolar , Femenino , Proteínas del Choque Térmico HSP47/química , Proteínas del Choque Térmico HSP47/genética , Humanos , Masculino , Datos de Secuencia Molecular , Osteogénesis Imperfecta/genética , Linaje , Transporte de Proteínas , Alineación de Secuencia , Proteínas de Unión a Tacrolimus/química , Proteínas de Unión a Tacrolimus/genética , Adulto Joven
15.
Am J Med Genet A ; 173(9): 2415-2421, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28742282

RESUMEN

Multiple Epiphyseal Dysplasia (MED) is a relatively mild skeletal dysplasia characterized by mild short stature, joint pain, and early-onset osteoarthropathy. Dominantly inherited mutations in COMP, MATN3, COL9A1, COL9A2, and COL9A3, and recessively inherited mutations in SLC26A2, account for the molecular basis of disease in about 80-85% of the cases. In two families with recurrent MED of an unknown molecular basis, we used exome sequencing and candidate gene analysis to identify homozygosity for recessively inherited missense mutations in CANT1, which encodes calcium-activated nucleotidase 1. The MED phenotype is thus allelic to the more severe Desbuquois dysplasia phenotype and the results identify CANT1 as a second locus for recessively inherited MED.


Asunto(s)
Genes Recesivos , Nucleotidasas/genética , Osteocondrodisplasias/genética , Adulto , Secuencia de Bases , Niño , Preescolar , Exoma/genética , Femenino , Humanos , Masculino , Mutación Missense/genética , Osteocondrodisplasias/diagnóstico por imagen , Osteocondrodisplasias/fisiopatología , Linaje , Radiografía
16.
Am J Hum Genet ; 93(5): 926-31, 2013 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-24183449

RESUMEN

Short-rib polydactyly (SRP) syndrome type III, or Verma-Naumoff syndrome, is an autosomal-recessive chondrodysplasia characterized by short ribs, a narrow thorax, short long bones, an abnormal acetabulum, and numerous extraskeletal malformations and is lethal in the perinatal period. Presently, mutations in two genes, IFT80 and DYNC2H1, have been identified as being responsible for SRP type III. Via homozygosity mapping in three affected siblings, a locus for the disease was identified on chromosome 9q34.11, and homozygosity for three missense mutations in WDR34 were found in three independent families, as well as compound heterozygosity for mutations in one family. WDR34 encodes a member of the WD repeat protein family with five WD40 domains, which acts as a TAK1-associated suppressor of the IL-1R/TLR3/TLR4-induced NF-κB activation pathway. We showed, through structural modeling, that two of the three mutations altered specific structural domains of WDR34. We found that primary cilia in WDR34 mutant fibroblasts were significantly shorter than normal and had a bulbous tip. This report expands on the pathogenesis of SRP type III and demonstrates that a regulator of the NF-κB activation pathway is involved in the pathogenesis of the skeletal ciliopathies.


Asunto(s)
Proteínas Portadoras/genética , Cilios/genética , Síndrome de Ellis-Van Creveld/genética , FN-kappa B/metabolismo , Síndrome de Costilla Pequeña y Polidactilia/genética , Transducción de Señal , Proteínas Portadoras/metabolismo , Cilios/patología , Dineínas Citoplasmáticas/genética , Síndrome de Ellis-Van Creveld/patología , Fibroblastos , Heterocigoto , Homocigoto , Humanos , Recién Nacido , Masculino , Mutación , Mutación Missense , Costillas/anomalías , Costillas/patología , Síndrome de Costilla Pequeña y Polidactilia/patología
17.
Am J Hum Genet ; 92(1): 137-43, 2013 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-23273567

RESUMEN

Opsismodysplasia is a rare, autosomal-recessive skeletal dysplasia characterized by short stature, characteristic facial features, and in some cases severe renal phosphate wasting. We used linkage analysis and whole-genome sequencing of a consanguineous trio to discover that mutations in inositol polyphosphate phosphatase-like 1 (INPPL1) cause opsismodysplasia with or without renal phosphate wasting. Evaluation of 12 families with opsismodysplasia revealed that INPPL1 mutations explain ~60% of cases overall, including both of the families in our cohort with more than one affected child and 50% of the simplex cases.


Asunto(s)
Mutación , Osteocondrodisplasias/genética , Monoéster Fosfórico Hidrolasas/genética , Niño , Preescolar , Femenino , Genoma Humano , Humanos , Lactante , Recién Nacido , Masculino , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas
18.
N Engl J Med ; 368(19): 1809-16, 2013 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-23656646

RESUMEN

This report identifies human skeletal diseases associated with mutations in WNT1. In 10 family members with dominantly inherited, early-onset osteoporosis, we identified a heterozygous missense mutation in WNT1, c.652T→G (p.Cys218Gly). In a separate family with 2 siblings affected by recessive osteogenesis imperfecta, we identified a homozygous nonsense mutation, c.884C→A, p.Ser295*. In vitro, aberrant forms of the WNT1 protein showed impaired capacity to induce canonical WNT signaling, their target genes, and mineralization. In mice, Wnt1 was clearly expressed in bone marrow, especially in B-cell lineage and hematopoietic progenitors; lineage tracing identified the expression of the gene in a subset of osteocytes, suggesting the presence of altered cross-talk in WNT signaling between the hematopoietic and osteoblastic lineage cells in these diseases.


Asunto(s)
Mutación , Osteogénesis Imperfecta/genética , Osteoporosis/genética , Proteína Wnt1/genética , Adolescente , Adulto , Edad de Inicio , Anciano , Animales , Niño , Femenino , Genes Dominantes , Genes Recesivos , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Linaje , Proteína Wnt1/metabolismo , Adulto Joven
19.
Am J Med Genet A ; 170(12): 3298-3302, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27530454

RESUMEN

Dominant mutations in TRPV4, which encodes the Transient Receptor Potential Cation Channel Subfamily V Member 4 calcium channel, result in a series of musculoskeletal disorders that include a set of peripheral neuropathies and a broad phenotypic spectrum of skeletal dysplasias. The skeletal phenotypes range from brachyolmia, in which there is scoliosis with mild short stature, through perinatal lethal metatropic dysplasia. We describe a case with phenotypic findings consistent with metatropic dysplasia, but in whom no TRPV4 mutation was detected by Sanger sequence analysis. Exome sequence analysis identified a known lethal metatropic dysplasia mutation, TRPV4L618P , which was present at lower frequency than would be expected for a heterozygous change. The affected individual was shown to be a somatic mosaic for the mutation, providing an explanation for the milder than expected phenotype. The data illustrate that high-throughput sequencing of genomic DNA can facilitate detection of mosaicism with higher sensitivity than Sanger sequence analysis and identify a new genetic mechanism for metatropic dysplasia. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Enanismo/diagnóstico , Enanismo/genética , Estudios de Asociación Genética , Mosaicismo , Mutación , Osteocondrodisplasias/diagnóstico , Osteocondrodisplasias/genética , Fenotipo , Canales Catiónicos TRPV/genética , Alelos , Análisis Mutacional de ADN , Exones , Humanos , Lactante , Imagen por Resonancia Magnética , Examen Físico , Radiografía
20.
Am J Med Genet A ; 170(10): 2652-61, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27240702

RESUMEN

Bent Bone Dysplasia-FGFR2 type is a relatively recently described bent bone phenotype with diagnostic clinical, radiographic, and molecular characteristics. Here we report on 11 individuals, including the original four patients plus seven new individuals with three longer-term survivors. The prenatal phenotype included stillbirth, bending of the femora, and a high incidence of polyhydramnios, prematurity, and perinatal death in three of 11 patients in the series. The survivors presented with characteristic radiographic findings that were observed among those with lethality, including bent bones, distinctive (moustache-shaped) small clavicles, angel-shaped metacarpals and phalanges, poor mineralization of the calvarium, and craniosynostosis. Craniofacial abnormalities, hirsutism, hepatic abnormalities, and genitourinary abnormalities were noted as well. Longer-term survivors all needed ventilator support. Heterozygosity for mutations in the gene that encodes Fibroblast Growth Factor Receptor 2 (FGFR2) was identified in the nine individuals with available DNA. Description of these patients expands the prenatal and postnatal findings of Bent Bone Dysplasia-FGFR2 type and adds to the phenotypic spectrum among all FGFR2 disorders. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Clavícula/anomalías , Falanges de los Dedos de la Mano/anomalías , Mutación , Osteocondrodisplasias/diagnóstico , Osteocondrodisplasias/genética , Fenotipo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Alelos , Sustitución de Aminoácidos , Facies , Femenino , Genotipo , Humanos , Masculino , Embarazo , Resultado del Embarazo , Diagnóstico Prenatal , Radiografía , Sistema de Registros
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA