Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 84
Filter
Add more filters

Publication year range
1.
Blood ; 138(21): 2117-2128, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34115847

ABSTRACT

Shwachman-Diamond syndrome (SDS; OMIM #260400) is caused by variants in SBDS (Shwachman-Bodian-Diamond syndrome gene), which encodes a protein that plays an important role in ribosome assembly. Recent reports suggest that recessive variants in EFL1 are also responsible for SDS. However, the precise genetic mechanism that leads to EFL1-induced SDS remains incompletely understood. Here we present 3 unrelated Korean SDS patients who carry biallelic pathogenic variants in EFL1 with biased allele frequencies, resulting from a bone marrow-specific somatic uniparental disomy in chromosome 15. The recombination events generated cells that were homozygous for the relatively milder variant, allowing for the evasion of catastrophic physiologic consequences. However, the milder EFL1 variant was still solely able to impair 80S ribosome assembly and induce SDS features in cell line and animal models. The loss of EFL1 resulted in a pronounced inhibition of terminal oligopyrimidine element-containing ribosomal protein transcript 80S assembly. Therefore, we propose a more accurate pathogenesis mechanism of EFL1 dysfunction that eventually leads to aberrant translational control and ribosomopathy.


Subject(s)
Peptide Elongation Factors/genetics , Ribonucleoprotein, U5 Small Nuclear/genetics , Shwachman-Diamond Syndrome/genetics , Uniparental Disomy/genetics , Adult , Alleles , Animals , Child , Child, Preschool , Female , Humans , Male , Mice, Inbred C57BL , Models, Molecular , Point Mutation
2.
Am J Hum Genet ; 104(3): 439-453, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30773278

ABSTRACT

SPONASTRIME dysplasia is a rare, recessive skeletal dysplasia characterized by short stature, facial dysmorphism, and aberrant radiographic findings of the spine and long bone metaphysis. No causative genetic alterations for SPONASTRIME dysplasia have yet been determined. Using whole-exome sequencing (WES), we identified bi-allelic TONSL mutations in 10 of 13 individuals with SPONASTRIME dysplasia. TONSL is a multi-domain scaffold protein that interacts with DNA replication and repair factors and which plays critical roles in resistance to replication stress and the maintenance of genome integrity. We show here that cellular defects in dermal fibroblasts from affected individuals are complemented by the expression of wild-type TONSL. In addition, in vitro cell-based assays and in silico analyses of TONSL structure support the pathogenicity of those TONSL variants. Intriguingly, a knock-in (KI) Tonsl mouse model leads to embryonic lethality, implying the physiological importance of TONSL. Overall, these findings indicate that genetic variants resulting in reduced function of TONSL cause SPONASTRIME dysplasia and highlight the importance of TONSL in embryonic development and postnatal growth.


Subject(s)
Fibroblasts/pathology , Genes, Lethal , Mutation , NF-kappa B/genetics , Osteochondrodysplasias/pathology , Adolescent , Adult , Animals , Cells, Cultured , Child , Child, Preschool , DNA Damage , Dermis/metabolism , Dermis/pathology , Female , Fibroblasts/metabolism , Humans , Infant , Infant, Newborn , Mice , Mice, Inbred C57BL , Osteochondrodysplasias/genetics , Exome Sequencing/methods , Young Adult
3.
Am J Med Genet A ; 182(2): 338-347, 2020 02.
Article in English | MEDLINE | ID: mdl-31755234

ABSTRACT

The phenotypic spectrum of Type 2 collagenopathies ranges from lethal achondrogenesis Type 2 to milder osteoarthritis with mild chondrodysplasia. All of them are monoallelic except for the two recent reports showing that biallelic variants in COL2A1 can cause spondyloepiphyseal dysplasia congenita in two children. Here we report two additional families with homozygous variants, c.4135C>T (p.Arg1379Cys) and c.3190C>T (p.Arg1133Cys) in COL2A1 resulting in two distinct skeletal dysplasia phenotypes of intermediate severity. Though all six patients from four families exhibit a spondylo-epimetaphyseal dysplasia, they demonstrate a wide variation in severity of short stature and involvement of epiphyses, metaphyses, and vertebrae. We hypothesize that the variants are likely to be hypomorphic, given the underlying mechanisms of disease causation for known heterozygous variants in COL2A1. With this report, we provide further evidence to the existence of autosomal recessive Type 2 collagenopathy.


Subject(s)
Collagen Type II/genetics , Dwarfism/genetics , Osteochondrodysplasias/congenital , Adult , Child , Child, Preschool , Dwarfism/physiopathology , Epiphyses/physiopathology , Female , Genes, Recessive , Heterozygote , Humans , Male , Mutation/genetics , Osteochondrodysplasias/genetics , Osteochondrodysplasias/physiopathology , Phenotype , Spine/physiopathology , Young Adult
4.
Am J Hum Genet ; 98(6): 1243-1248, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27236923

ABSTRACT

Spondyloepimetaphyseal dysplasias (SEMDs) comprise a heterogeneous group of autosomal-dominant and autosomal-recessive disorders. An apparent X-linked recessive (XLR) form of SEMD in a single Italian family was previously reported. We have been able to restudy this family together with a second family from Korea by segregating a severe SEMD in an X-linked pattern. Exome sequencing showed missense mutations in BGN c.439A>G (p.Lys147Glu) in the Korean family and c.776G>T (p.Gly259Val) in the Italian family; the c.439A>G (p.Lys147Glu) mutation was also identified in a further simplex SEMD case from India. Biglycan is an extracellular matrix proteoglycan that can bind transforming growth factor beta (TGF-ß) and thus regulate its free concentration. In 3-dimensional simulation, both altered residues localized to the concave arc of leucine-rich repeat domains of biglycan that interact with TGF-ß. The observation of recurrent BGN mutations in XLR SEMD individuals from different ethnic backgrounds allows us to define "XLR SEMD, BGN type" as a nosologic entity.


Subject(s)
Biglycan/genetics , Genetic Diseases, X-Linked/genetics , Mutation/genetics , Osteochondrodysplasias/genetics , Adult , Aged , Amino Acid Sequence , Biglycan/chemistry , Biglycan/metabolism , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Pedigree , Protein Binding , Protein Conformation , Sequence Homology, Amino Acid , Transforming Growth Factor beta/chemistry , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
5.
BMC Med Genet ; 20(1): 70, 2019 05 03.
Article in English | MEDLINE | ID: mdl-31053099

ABSTRACT

BACKGROUND: X-linked spondyloepiphyseal dysplasia tarda (SEDT-XL) is a skeletal disorder characterized by defective structures of vertebral bodies and/or of epiphyses of the long bones, resulting in moderately short stature and early joint degeneration. TRAPPC2 gene, which is important for collagen secretion, has been reported as causative for SEDT-XL. CASE PRESENTATION: Here, we report two variants of TRAPPC2 gene of SEDT-XL patients, a missense variant of start codon, c.1A > T, and a deletion variant, c.40delG. To understand molecular consequence of the variants, we establish an in vitro gene expression assay system and demonstrate that both mutated genes are transcribed, but are not properly translated, indicative of the pathogenic nature of those TRAPPC2 variants. CONCLUSIONS: In the current study, we provide additional experimental data showing that loss-of-function TRAPPC2 variants are probably causative for SEDT-XL phenotype. These findings further contribute to the understanding the clinical picture related to TRAPPC2 gene.


Subject(s)
Genetic Diseases, X-Linked/genetics , Membrane Transport Proteins/genetics , Osteochondrodysplasias/genetics , Transcription Factors/genetics , Adolescent , Humans , Male , Middle Aged
6.
Am J Hum Genet ; 96(2): 266-74, 2015 Feb 05.
Article in English | MEDLINE | ID: mdl-25620203

ABSTRACT

Singleton-Merten syndrome (SMS) is an autosomal-dominant multi-system disorder characterized by dental dysplasia, aortic calcification, skeletal abnormalities, glaucoma, psoriasis, and other conditions. Despite an apparent autosomal-dominant pattern of inheritance, the genetic background of SMS and information about its phenotypic heterogeneity remain unknown. Recently, we found a family affected by glaucoma, aortic calcification, and skeletal abnormalities. Unlike subjects with classic SMS, affected individuals showed normal dentition, suggesting atypical SMS. To identify genetic causes of the disease, we performed exome sequencing in this family and identified a variant (c.1118A>C [p.Glu373Ala]) of DDX58, whose protein product is also known as RIG-I. Further analysis of DDX58 in 100 individuals with congenital glaucoma identified another variant (c.803G>T [p.Cys268Phe]) in a family who harbored neither dental anomalies nor aortic calcification but who suffered from glaucoma and skeletal abnormalities. Cys268 and Glu373 residues of DDX58 belong to ATP-binding motifs I and II, respectively, and these residues are predicted to be located closer to the ADP and RNA molecules than other nonpathogenic missense variants by protein structure analysis. Functional assays revealed that DDX58 alterations confer constitutive activation and thus lead to increased interferon (IFN) activity and IFN-stimulated gene expression. In addition, when we transduced primary human trabecular meshwork cells with c.803G>T (p.Cys268Phe) and c.1118A>C (p.Glu373Ala) mutants, cytopathic effects and a significant decrease in cell number were observed. Taken together, our results demonstrate that DDX58 mutations cause atypical SMS manifesting with variable expression of glaucoma, aortic calcification, and skeletal abnormalities without dental anomalies.


Subject(s)
Aortic Diseases/genetics , DEAD-box RNA Helicases/genetics , Dental Enamel Hypoplasia/genetics , Glaucoma/genetics , Metacarpus/abnormalities , Models, Molecular , Muscular Diseases/genetics , Odontodysplasia/genetics , Osteoporosis/genetics , Vascular Calcification/genetics , Adult , Aortic Diseases/pathology , Base Sequence , Cells, Cultured , Child, Preschool , DEAD Box Protein 58 , DEAD-box RNA Helicases/chemistry , Dental Enamel Hypoplasia/pathology , Exome/genetics , Female , Genes, Dominant/genetics , Humans , Male , Metacarpus/pathology , Molecular Sequence Data , Muscular Diseases/pathology , Musculoskeletal Abnormalities/diagnostic imaging , Musculoskeletal Abnormalities/genetics , Mutation, Missense/genetics , Odontodysplasia/diagnostic imaging , Odontodysplasia/pathology , Osteoporosis/pathology , Pedigree , Polymorphism, Single Nucleotide/genetics , Radiography , Receptors, Immunologic , Sequence Analysis, DNA , Vascular Calcification/pathology
7.
Am J Med Genet A ; 173(3): 588-595, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28127940

ABSTRACT

Smith-McCort dysplasia (SMC OMIM 615222) and Dyggve-Melchior-Clausen dysplasia (DMC OMIM 223800) are allelic skeletal dysplasias caused by homozygous or compound heterozygous mutations in DYM (OMIM 607461). Both disorders share the same skeletal phenotypes characterized by spondylo-epi-metaphyseal dysplasia with distinctive lacy ilia. The difference rests on the presence or absence of intellectual disability, that is, intellectual disability in DMC and normal cognition in SMC. However, genetic heterogeneity was suspected in SMC. Recently, RAB33B (OMIM 605950) has been identified as the second gene for SMC. Nevertheless, only two affected families have been reported so far. Here we present three SMC patients with four novel pathogenic variants in RAB33B, including homozygosity for c.211C>T (p.R71*), homozygosity for c.365T>C (p.F122S), and compound heterozygosity for c.48delCGGGGCAG (p.G17Vfs*58) and c.490C>T (p.Q164*). We also summarize the clinical, radiological, and mutation profile of RAB33B after literature mining. This report ascertains the pathogenic relationship between RAB33B and SMC. © 2017 Wiley Periodicals, Inc.


Subject(s)
Mutation , Osteochondrodysplasias/diagnosis , Osteochondrodysplasias/genetics , rab GTP-Binding Proteins/genetics , Adult , Alleles , Amino Acid Sequence , Child , Consanguinity , Diagnostic Imaging , Facies , Heterozygote , Homozygote , Humans , Intracellular Signaling Peptides and Proteins , Male , Phenotype , Proteins/genetics , Sequence Analysis, DNA
8.
Am J Med Genet A ; 173(4): 1102-1108, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28328135

ABSTRACT

Since the original description of the IARS2-related cataracts, growth hormone deficiency, sensory neuropathy, sensorineural hearing loss, skeletal dysplasia syndrome (CAGSSS; OMIM 616007) in an extended consanguineous family of French-Canadian descent, no further patients have been reported. IARS2 (OMIM 612801) encodes the mitochondrial isoleucine-tRNA synthetase which belongs to the class-I aminoacyl-tRNA synthetase family, and has been implicated in CAGSSS and a form of Leigh syndrome. Here, we report on a female Danish patient with a novel homozygous IARS2 mutation, p.Gly874Arg, who presented at birth with bilateral hip dislocation and short stature. At 3 months, additional dysmorphic features were noted and at 18 months her radiographic skeletal abnormalities were suggestive of an underlying spondyloepimetaphyseal dysplasia (SEMD). Retrospective analysis of the neonatal radiographs confirmed that the skeletal changes were present at birth. It was only with time that several of the other manifestations of the CAGSSS emerged, namely, cataracts, peripheral neuropathy, and hearing loss. Growth hormone deficiency has not (yet) manifested. We present her clinical features and particularly highlight her skeletal findings, which confirm the presence of a primary SEMD skeletal dysplasia in a growing list of mitochondrial-related disorders including CAGSSS, CODAS, EVEN-PLUS, and X-linked SEMD-MR syndromes.


Subject(s)
Cataract/genetics , Genetic Diseases, X-Linked/genetics , Growth Hormone/deficiency , Hearing Loss, Sensorineural/genetics , Hereditary Sensory and Autonomic Neuropathies/genetics , Isoleucine-tRNA Ligase/genetics , Mutation , Osteochondrodysplasias/genetics , Cataract/diagnosis , Cataract/pathology , Child , Exome , Female , Gene Expression , Genetic Diseases, X-Linked/diagnosis , Genetic Diseases, X-Linked/pathology , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/pathology , Hereditary Sensory and Autonomic Neuropathies/diagnosis , Hereditary Sensory and Autonomic Neuropathies/pathology , High-Throughput Nucleotide Sequencing , Homozygote , Humans , Osteochondrodysplasias/diagnosis , Osteochondrodysplasias/pathology , Radiography , Syndrome
9.
Skeletal Radiol ; 46(9): 1297-1300, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28601949

ABSTRACT

We report transient proximal and distal femoral metaphyseal striations that have not previously been described in autosomal dominant brachyolmia. The pelvis/hip radiograph of a 13-year-old boy demonstrated bilaterally symmetrical proximal femoral metaphyseal vertical striations. Additional vertical striations were also observed at the distal femur and proximal tibia metaphysis. Radiography of the thoracolumbar spine demonstrated platyspondyly with irregular endplates and overfaced pedicles. TRPV4 mutations were confirmed in this patient. Similar proximal femoral metaphyseal vertical striations were noted in the patient's sibling. Those streaks disappeared on the follow-up radiographs, and we considered it a unique radiologic finding transiently observed in autosomal dominant brachyolmia.


Subject(s)
Osteochondrodysplasias/diagnostic imaging , Adolescent , Child , Diagnosis, Differential , Humans , Male
10.
Genet Med ; 18(6): 563-9, 2016 06.
Article in English | MEDLINE | ID: mdl-26402641

ABSTRACT

PURPOSE: The purpose of this study was to evaluate the clinical utility of targeted exome sequencing (TES) as a molecular diagnostic tool for patients with skeletal dysplasia. METHODS: A total of 185 patients either diagnosed with or suspected to have skeletal dysplasia were recruited over a period of 3 years. TES was performed for 255 genes associated with the pathogenesis of skeletal dysplasia, and candidate variants were selected using a bioinformatics analysis. All candidate variants were confirmed by Sanger sequencing, correlation with the phenotype, and a cosegregation study in the family. RESULTS: TES detected "confirmed" or "highly likely" pathogenic sequence variants in 74% (71 of 96) of cases in the assured clinical diagnosis category and 20.3% (13 of 64 cases) of cases in the uncertain clinical diagnosis category. TES successfully detected pathogenic variants in all 25 cases of previously known genotypes. The data also suggested a copy-number variation that led to a molecular diagnosis. CONCLUSION: This study demonstrates the feasibility of TES for the molecular diagnosis of skeletal dysplasia. However, further confirmation is needed for a final molecular diagnosis, including Sanger sequencing of candidate variants with suspected, poorly captured exons.Genet Med 18 6, 563-569.


Subject(s)
Exome Sequencing/methods , Musculoskeletal Abnormalities/diagnosis , Musculoskeletal Abnormalities/genetics , Pathology, Molecular , DNA Copy Number Variations/genetics , Exons/genetics , Female , Humans , Male , Musculoskeletal Abnormalities/physiopathology , Mutation , Pedigree , Phenotype
11.
J Hum Genet ; 61(6): 561-4, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26791357

ABSTRACT

SOFT syndrome (MIM614813) is an extremely rare primordial dwarfism characterized by short stature, onychodysplasia, facial dysmorphism and hypotrichosis, which is caused by biallelic mutations in the POC1A gene. Only 19 patients with mutation-confirmed SOFT syndrome have been reported to date, all of whom carried homozygous variants that were strongly associated with consanguineous marriages. We report an 8.5-year-old boy with SOFT syndrome showing primordial dwarfism, no effect of growth-hormone therapy and skeletal dysplasia. This is the first report of compound heterozygous variants in POC1A, one previously reported and the other novel. A characteristic skeletal manifestation is reported.


Subject(s)
Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Heterozygote , Mutation , Phenotype , Proteins/genetics , Skeleton/pathology , Amino Acid Substitution , Cell Cycle Proteins , Cytoskeletal Proteins , DNA Mutational Analysis , Exome , High-Throughput Nucleotide Sequencing , Humans , Infant , Male , Pedigree , Syndrome
12.
Am J Med Genet A ; 170A(2): 426-434, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26567084

ABSTRACT

The C-type natriuretic peptide (CNP)-natriuretic peptide receptor 2 (NPR2) signaling pathway plays an important role in chondrocyte development. Homozygous loss-of-function mutations of the NPR2 gene cause acromesomelic dysplasia, type Maroteaux (AMDM). The aim of this study was to identify and characterize NPR2 loss-of-function mutations in patients with AMDM. The NPR2 gene was sequenced in three Korean patients with AMDM and functional analysis of the mutated proteins was performed in vitro. Five novel NPR2 mutations were found in the three patients: two compound heterozygous mutations [c.1231T>C (Tyr411His) and c.2761C>T (Arg921X) in Patient 1 and c.1663A>T (Lys555X) and c.1711-1G>C (M571VfsX12) in Patient 3] and a homozygous mutation [c.2762G>A (Arg921Gln) in Patient 2]. Serum NT-proCNP concentration was significantly increased in each patient compared to control subjects. Cells transfected with the expression vector of each mutant except those found in Patient 3 showed a negligible or a markedly low cGMP response after treatment with CNP. HA-tagged wild-type (wt) and HA-mutant NPR2 were expressed at comparable levels: there were two bands of ∼130 and ∼120 kDa in wt and Arg921Gln, a single ∼120 kDa band in Tyr411His, and a single ∼110 kDa in the nonsense mutant. With respect to subcellular localization, Arg921Gln as well as wt-NPR2 reached the cell surface, whereas Tyr411His and Arg921X mutants did not. The Tyr411His and Arg921X NPR2 proteins were co-localized with an endoplasmic reticulum (ER) marker and failed to traffic from the ER to the Golgi apparatus. These results are consistent with deglycosylation experiments. Tyr411His and Arg921X NPR2 are complete loss-of-function mutations, whereas Arg921Gln behaves as a receptor for CNP with limited function.


Subject(s)
Bone Diseases, Developmental/genetics , Bone Diseases, Developmental/pathology , Mutation/genetics , Receptors, Atrial Natriuretic Factor/genetics , Adolescent , Child , Female , Fluorescent Antibody Technique , Genotype , HEK293 Cells , Heterozygote , Humans , Male , Pedigree , Phenotype , Prognosis
13.
Skeletal Radiol ; 45(2): 227-33, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26559670

ABSTRACT

BACKGROUND: To describe grayscale and color Doppler ultrasound features of subcutaneous intravascular papillary endothelial hyperplasia (IPEH). MATERIALS AND METHODS: The ultrasound appearances of ten histologically proven subcutaneous IPEH in ten patients (age range, 15-69 years; mean age, 38.2 years; six females, four males) were reviewed retrospectively by two musculoskeletal radiologists. Color Doppler examination and surgical excision were performed in all cases. The correlations between the ultrasound and pathological features of the lesions were done. RESULTS: All ten cases were pathologically diagnosed as pure forms of IPEH. The mean size of the lesions was 1.3 cm. The margins of the lesions were circumscribed in seven of ten patients. Three had lobular margins. The distinct internal septum-like structures were seen in seven of ten cases (70 %). The vascularity was rich in three (30 %), moderate in four (40 %), and little in three (30 %) of the ten cases. The most common vascular pattern was one or more vessels peripherally or both peripherally and centrally located in the lesion. The detectable origin vessel was noted in four of ten cases (40 %). CONCLUSIONS: Although sonographic features of subcutaneous IPEH are non-specific, they should be included in the differential diagnosis of a small, well-defined, oval or elliptical, heterogeneous, hypoechoic soft tissue mass, showing a vascular pattern of one or more vessels in the lesion and variable vascularity. The presence of the internal septum-like structures and detectable origin vessel may be help to distinguish the lesion from the other soft tissue masses.


Subject(s)
Endothelium, Vascular/diagnostic imaging , Endothelium, Vascular/pathology , Hemangioendothelioma/diagnostic imaging , Hemangioendothelioma/pathology , Adolescent , Adult , Aged , Diagnosis, Differential , Female , Humans , Male , Middle Aged , Retrospective Studies , Ultrasonography, Doppler, Color , Young Adult
14.
J Korean Med Sci ; 31(5): 735-42, 2016 May.
Article in English | MEDLINE | ID: mdl-27134495

ABSTRACT

Pachydermoperiostosis (PDP), or primary hypertrophic osteoarthropathy, is a rare genetic disease affecting both skin and bones. Both autosomal dominant with incomplete penetrance and recessive inheritance of PDP have been previously confirmed. Recently, hydroxyprostaglandin dehydrogenase (HPGD) and solute carrier organic anion transporter family member 2A1 (SLCO2A1) were reported as pathogenic genes responsible for PDP. Both genes are involved in prostaglandin E2 (PGE2) degradation. We aimed to identify responsible genes for PDP and the clinical features in Korean patients with PDP. Six affected individuals and their available healthy family members from three unrelated Korean families with PDP were studied. All of the patients displayed complete phenotypes of PDP with finger clubbing, pachydermia, and periostosis. Mutation analysis revealed a novel heterozygous mutation in the SLCO2A1 gene at nucleotide 302 causing a substitution of the amino acid isoleucine to serine at codon 101 (p.IIe101Ser) in affected individuals. We also identified known SLCO2A1 mutations, one homozygous for c.940+1G>A, and another compound heterozygous for c.940+1G>A and c.1807C>T (p.Arg603*) from two PDP families. Genetic analyses of the PDP patients showed no abnormality in the HPGD gene. Our study further supports the role of mutations in the SLCO2A1 gene in the pathogenesis of PDP and could provide additional clues to the genotype-phenotype relations of PDP.


Subject(s)
Organic Anion Transporters/genetics , Osteoarthropathy, Primary Hypertrophic/genetics , Bone and Bones/diagnostic imaging , Child, Preschool , DNA Mutational Analysis , Exons , Heterozygote , Humans , Male , Middle Aged , Osteoarthropathy, Primary Hypertrophic/diagnostic imaging , Osteoarthropathy, Primary Hypertrophic/pathology , Pedigree , Phenotype , Polymorphism, Genetic , Positron-Emission Tomography , Young Adult
15.
Hum Mutat ; 36(10): 1004-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26183434

ABSTRACT

Progressive pseudorheumatoid dysplasia (PPRD) is a rare, autosomal-recessive condition characterized by mild spondyloepiphyseal dysplasia (SED) and severe, progressive, early-onset arthritis due to WISP3 mutations. SED, Stanescu type, is a vaguely delineated autosomal-dominant dysplasia of unknown genetic etiology. Here, we report three individuals from two unrelated families with radiological features similar to PPRD and SED, Stanescu type who share the same novel COL2A1 variant and were matched following discussion at an academic conference. In the first family, we performed whole-exome sequencing on three family members, two of whom have a PPRD-like phenotype, and identified a heterozygous variant (c.619G>A, p.Gly207Arg) in both affected individuals. Independently, targeted sequencing of the COL2A1 gene in an unrelated proband with a similar phenotype identified the same heterozygous variant. We suggest that the p.Gly207Arg variant causes a distinct type II collagenopathy with features of PPRD and SED, Stanescu type.


Subject(s)
Collagen Type II/genetics , Joint Diseases/congenital , Osteochondrodysplasias/genetics , Polymorphism, Single Nucleotide , Adolescent , Adult , Child , Exome , Female , Humans , Joint Diseases/diagnostic imaging , Joint Diseases/genetics , Male , Osteochondrodysplasias/diagnostic imaging , Pedigree , Radiography , Sequence Analysis, DNA/methods
16.
Hum Mutat ; 36(2): 191-5, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25402547

ABSTRACT

Osteogenesis imperfecta (OI) comprises a heterogeneous group of disorders that are characterized by susceptibility to bone fractures, and range in severity from a subtle increase in fracture frequency to death in the perinatal period. Most patients have defects in type I collagen biosynthesis with autosomal-dominant inheritance, but many autosomal-recessive genes have been reported. We applied whole-exome sequencing to identify mutations in a Korean OI patient who had an umbilical hernia, frequent fractures, a markedly short stature, delayed motor development, scoliosis, and dislocation of the radial head, with a bowed radius and ulna. We identified two novel variants in the BMP1 gene: c.808A>G and c.1297G>T. The former variant caused a missense change p.(Met270Val) and the latter variant caused the skipping of exon 10. The hypofunctional nature of the two variants was demonstrated in a zebrafish assay.


Subject(s)
Bone Morphogenetic Protein 1/genetics , Osteogenesis Imperfecta/genetics , Amino Acid Substitution , Animals , Female , Genetic Association Studies , Heterozygote , Humans , Infant , Polymorphism, Single Nucleotide , Zebrafish
17.
Am J Hum Genet ; 91(2): 343-8, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22863190

ABSTRACT

Osteogenesis imperfecta (OI) is a heterogenous group of genetic disorders of bone fragility. OI type V is an autosomal-dominant disease characterized by calcification of the forearm interosseous membrane, radial head dislocation, a subphyseal metaphyseal radiodense line, and hyperplastic callus formation; the causative mutation involved in this disease has not been discovered yet. Using linkage analysis in a four-generation family and whole-exome sequencing, we identified a heterozygous mutation of c.-14C>T in the 5'-untranslated region of a gene encoding interferon-induced transmembrane protein 5 (IFITM5). It completely cosegregated with the disease in three families and occurred de novo in five simplex individuals. Transfection of wild-type and mutant IFITM5 constructs revealed that the mutation added five amino acids (Met-Ala-Leu-Glu-Pro) to the N terminus of IFITM5. Given that IFITM5 expression and protein localization is restricted to the skeletal tissue and IFITM5 involvement in bone formation, we conclude that this recurrent mutation would have a specific effect on IFITM5 function and thus cause OI type V.


Subject(s)
Membrane Proteins/genetics , Osteogenesis Imperfecta/genetics , 5' Untranslated Regions/genetics , Adolescent , Adult , Amino Acid Sequence , Base Sequence , Child , Exome/genetics , Female , Genetic Linkage , Humans , Male , Molecular Sequence Data , Osteogenesis Imperfecta/diagnostic imaging , Point Mutation/genetics , Radiography , Sequence Analysis, DNA
18.
Mol Genet Metab ; 115(1): 41-7, 2015 May.
Article in English | MEDLINE | ID: mdl-25892708

ABSTRACT

Mucopolysaccharidosis VI (MPS VI, Maroteaux-Lamy syndrome) is caused by deficient activity of the enzyme, N-acetylgalactosamine-4-sulfatase, resulting in impaired degradation of the glycosaminoglycan dermatan sulfate. Patients experience a range of manifestations including joint contractures, short stature, dysostosis multiplex, coarse facial features, decreased pulmonary function, cardiac abnormalities, corneal clouding and shortened life span. Recently, clinicians from institutions in the Asia-Pacific region met to discuss the occurrence and implications of delayed diagnosis and misdiagnosis of MPS VI in the patients they have managed. Eighteen patients (44% female) were diagnosed. The most common sign presented by the patients was bone deformities in 11 patients (65%). Delays to diagnosis occurred due to the lack of or distance to diagnostic facilities for four patients (31%), alternative diagnoses for two patients (15%), and misleading symptoms experienced by two patients (15%). Several patients experienced manifestations that were subtler than would be expected and were subsequently overlooked. Several cases highlighted the unique challenges associated with diagnosing MPS VI from the perspective of different specialties and provide insights into how these patients initially present, which may help to elucidate strategies to improve the diagnosis of MPS VI.


Subject(s)
Mucopolysaccharidosis VI/diagnosis , Asia , Bone and Bones/diagnostic imaging , Brain/diagnostic imaging , Delayed Diagnosis/prevention & control , Diagnosis, Differential , Diagnostic Errors/prevention & control , Female , Health Personnel/education , Humans , Male , Pacific States , Radiography , Referral and Consultation
19.
Am J Med Genet A ; 167A(5): 1033-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25728306

ABSTRACT

Chromosomal translocation of 2q37.1 just distal to the NPPC gene coding for C-type natriuretic peptide (CNP) and subsequent overproduction of CNP have been reported to cause a skeletal overgrowth syndrome. Loeys-Dietz syndrome (LDS) is one of marfanoid overgrowth syndromes, of which subtype IV is caused by haploinsufficiency of transforming growth factor beta 2 (TGFB2). We report on a girl with clinical phenotypes of overgrowth syndrome, including long and slim body habitus, macrodactyly of the big toe, scoliosis, ankle valgus deformity, coxa valga, slipped capital femoral epiphysis, and aortic root dilatation. Karyotyping revealed a balanced chromosomal translocation between 1q41 and 2q37.1, and the breakpoints could be mapped by targeted resequencing analysis. On chromosome 2q37.1, the translocation took place 200,365 bp downstream of NPPC, and serum level of the amino terminal of CNP was elevated. The contralateral site of translocation on chromosome 1q41 disrupted TGFB2 gene, presumed to cause its haploinsufficiency. This case supports the concept that NPPC is overexpressed because of the loss of a specific negative regulatory control in the normal chromosomal location, and demonstrates the effectiveness of targeted resequencing in the mapping of breakpoints.


Subject(s)
Loeys-Dietz Syndrome/genetics , Natriuretic Peptide, C-Type/biosynthesis , Translocation, Genetic/genetics , Adolescent , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 2/genetics , Female , Gene Expression Regulation , Haploinsufficiency , Humans , Karyotyping , Loeys-Dietz Syndrome/physiopathology , Natriuretic Peptide, C-Type/blood , Natriuretic Peptide, C-Type/genetics , Phenotype , Transforming Growth Factor beta2/genetics
20.
Am J Med Genet A ; 167(7): 1501-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25808063

ABSTRACT

Cerebral, ocular, dental, auricular, skeletal anomalies (CODAS) syndrome (MIM 600373) was first described and named by Shehib et al, in 1991 in a single patient. The anomalies referred to in the acronym are as follows: cerebral-developmental delay, ocular-cataracts, dental-aberrant cusp morphology and delayed eruption, auricular-malformations of the external ear, and skeletal-spondyloepiphyseal dysplasia. This distinctive constellation of anatomical findings should allow easy recognition but despite this only four apparently sporadic patients have been reported in the last 20 years indicating that the full phenotype is indeed very rare with perhaps milder or a typical presentations that are allelic but without sufficient phenotypic resemblance to permit clinical diagnosis. We performed exome sequencing in three patients (an isolated case and a brother and sister sib pair) with classical features of CODAS. Sanger sequencing was used to confirm results as well as for mutation discovery in a further four unrelated patients ascertained via their skeletal features. Compound heterozygous or homozygous mutations in LONP1 were found in all (8 separate mutations; 6 missense, 1 nonsense, 1 small in-frame deletion) thus establishing the genetic basis of CODAS and the pattern of inheritance (autosomal recessive). LONP1 encodes an enzyme of bacterial ancestry that participates in protein turnover within the mitochondrial matrix. The mutations cluster at the ATP-binding and proteolytic domains of the enzyme. Biallelic inheritance and clustering of mutations confirm dysfunction of LONP1 activity as the molecular basis of CODAS but the pathogenesis remains to be explored.


Subject(s)
ATP-Dependent Proteases/genetics , Craniofacial Abnormalities/genetics , Exome/genetics , Eye Abnormalities/genetics , Growth Disorders/genetics , Hip Dislocation, Congenital/genetics , Mitochondrial Proteins/genetics , Models, Genetic , Mutation/genetics , Osteochondrodysplasias/genetics , Tooth Abnormalities/genetics , Base Sequence , Genes, Recessive/genetics , Humans , Molecular Sequence Data , Sequence Analysis, DNA , Switzerland
SELECTION OF CITATIONS
SEARCH DETAIL