Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Clin Genet ; 105(1): 87-91, 2024 01.
Article in English | MEDLINE | ID: mdl-37619988

ABSTRACT

Skeletal ciliopathies are a heterogenous group of congenital disorders characterized by multiple internal abnormalities, and distinct radiographic presentation. Pathogenic variants in at least 30 cilia genes are known to cause skeletal ciliopathies. Here we report a fetus with an atypical skeletal ciliopathy phenotype and compound heterozygous variants in the RAB34 gene. The affected fetus had multiple malformations, including posterior neck edema, micrognathia, low-set and small ears, auricular hypoplasia, cleft lip and palate, short extremities, and a combination of rarely occurring pre- and postaxial polydactyly. Genome sequencing identified compound heterozygous variants in the RAB34 gene: maternal c.254T>C, p.(Ile85Thr), and paternal c.691C>T, p.(Arg231*) variants. Only the paternal variant was present in the unaffected sibling. Evidence in the literature indicated that Rab34-/- mice displayed a ciliopathy phenotype with cleft palate and polydactyly. These features were consistent with malformations detected in our patient supporting the pathogenicity of the identified RAB34 variants. Overall, this case report further expands genetic landscape of human ciliopathy syndromes and suggests RAB34 as a candidate gene for skeletal ciliopathies.


Subject(s)
Abnormalities, Multiple , Ciliopathies , Cleft Lip , Cleft Palate , Polydactyly , Humans , Animals , Mice , Cleft Palate/diagnostic imaging , Cleft Palate/genetics , Ciliopathies/diagnostic imaging , Ciliopathies/genetics , Ciliopathies/pathology , Polydactyly/genetics , Abnormalities, Multiple/genetics , Syndrome , rab GTP-Binding Proteins/genetics
2.
Am J Med Genet A ; 191(7): 1929-1934, 2023 07.
Article in English | MEDLINE | ID: mdl-37017437

ABSTRACT

Seckel syndrome is an ultrarare autosomal recessive genetically heterogenous condition characterized by intrauterine and postnatal growth restriction, proportionate severe short stature, severe microcephaly, intellectual disability, and distinctive facial features including a prominent nose. Up to now, 40 patients with molecularly confirmed Seckel syndrome have been reported with biallelic variants in nine genes: ATR, CENPJ, CEP63, CEP152, DNA2, NIN, NSMCE2, RBBP8, and TRAIP. Homozygosity for nonsense variant (c.129G>A, p.43*) in CEP63 was described in three cousins with microcephaly, short stature, mild to moderate intellectual disability and diagnoses of Seckel syndrome. Here, we report a second family with three siblings who are compound heterozygous for loss-of-function variants in CEP63, c.1125T>G, p.(Tyr375*) and c.595del, p.(Glu199Asnfs*11). All siblings present with microcephaly, prominent nose, and intellectual disability but only one has severe short stature. Two siblings have aggressive behavior, a feature previously not reported in Seckel syndrome. This report adds two novel truncating variants in CEP63 and extends the clinical knowledge on CEP63-related conditions.


Subject(s)
Dwarfism , Intellectual Disability , Microcephaly , Humans , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Microcephaly/diagnosis , Microcephaly/genetics , Dwarfism/diagnosis , Dwarfism/genetics , Facies , Phenotype , Ligases/genetics , Cell Cycle Proteins/genetics
3.
Radiographics ; 43(5): e220067, 2023 05.
Article in English | MEDLINE | ID: mdl-37053103

ABSTRACT

Skeletal dysplasias are a heterogeneous collection of genetic disorders characterized by bone and cartilage abnormalities, and they encompass over 400 disorders. These disorders are rare individually, but collectively they are common (approximate incidence of one in 5000 births). Radiologists occasionally encounter skeletal dysplasias in daily practice. In the 1980s, Professor Juergen Spranger proposed a concept suitable for the diagnosis of skeletal dysplasias termed bone dysplasia families. He stated that (a) different bone dysplasias that share a similar skeletal pattern can be grouped into a "family," (b) the final diagnosis is feasible through the provisional recognition of a pattern followed by a more careful analysis, and (c) families of bone dysplasias may be the result of similar pathogenetic mechanisms. The prototypes of bone dysplasia families include dysostosis multiplex family, achondroplasia family, spondyloepiphyseal dysplasia congenita family, and Larsen syndrome-otopalatodigital syndrome family. Since Spranger's proposal, the concept of bone dysplasia families, along with advancing genetic techniques, has been validated and further expanded. Today, this molecularly proven concept enables a simple stepwise approach to be applied to the radiologic diagnosis of skeletal dysplasias. The first step is the categorization of a given case into a family based on pattern recognition, and the second step is more meticulous observation, such as identification of different severities of the same pattern or subtle but distinctive findings. Since major skeletal dysplasias are limited in number, radiologists can be familiar with the representative patterns of these disorders. The authors describe a stepwise radiologic approach to diagnosing major skeletal dysplasia families and review the clinical and genetic features of these disorders. Published under a CC BY 4.0 license. Quiz questions for this article are available through the Online Learning Center. Online supplemental material and the slide presentation from the RSNA Annual Meeting are available for this article.


Subject(s)
Bone Diseases, Developmental , Hand Deformities, Congenital , Osteochondrodysplasias , Male , Humans , Osteochondrodysplasias/diagnostic imaging , Osteochondrodysplasias/genetics , Bone Diseases, Developmental/diagnostic imaging , Bone Diseases, Developmental/genetics , Radiography
4.
Hum Mutat ; 43(11): 1567-1575, 2022 11.
Article in English | MEDLINE | ID: mdl-35842787

ABSTRACT

Prader-Willi syndrome (PWS; MIM# 176270) is a neurodevelopmental disorder caused by the loss of expression of paternally imprinted genes within the PWS region located on 15q11.2. It is usually caused by either maternal uniparental disomy of chromosome 15 (UPD15) or 15q11.2 recurrent deletion(s). Here, we report a healthy carrier of a balanced X;15 translocation and her two daughters, both with the karyotype 45,X,der(X)t(X;15)(p22;q11.2),-15. Both daughters display symptoms consistent with haploinsufficiency of the SHOX gene and PWS. We explored the architecture of the derivative chromosomes and investigated effects on gene expression in patient-derived neural cells. First, a multiplex ligation-dependent probe amplification methylation assay was used to determine the methylation status of the PWS-region revealing maternal UPD15 in daughter 2, explaining her clinical symptoms. Next, short read whole genome sequencing and 10X genomics linked read sequencing was used to pinpoint the exact breakpoints of the translocation. Finally, we performed transcriptome sequencing on neuroepithelial stem cells from the mother and from daughter 1 and observed biallelic expression of genes in the PWS region (including SNRPN) in daughter 1. In summary, our multi-omics analysis highlights two different PWS mechanisms in one family and provide an example of how structural variation can affect imprinting through long-range interactions.


Subject(s)
DNA Methylation , Prader-Willi Syndrome , Chromosomes, Human, Pair 15/genetics , Female , Genomic Imprinting , Humans , Prader-Willi Syndrome/genetics , Translocation, Genetic , Uniparental Disomy/genetics , snRNP Core Proteins/genetics
5.
Genet Med ; 24(11): 2296-2307, 2022 11.
Article in English | MEDLINE | ID: mdl-36066546

ABSTRACT

PURPOSE: Individuals with intellectual disability (ID) and/or neurodevelopment disorders (NDDs) are currently investigated with several different approaches in clinical genetic diagnostics. METHODS: We compared the results from 3 diagnostic pipelines in patients with ID/NDD: genome sequencing (GS) first (N = 100), GS as a secondary test (N = 129), or chromosomal microarray (CMA) with or without FMR1 analysis (N = 421). RESULTS: The diagnostic yield was 35% (GS-first), 26% (GS as a secondary test), and 11% (CMA/FMR1). Notably, the age of diagnosis was delayed by 1 year when GS was performed as a secondary test and the cost per diagnosed individual was 36% lower with GS first than with CMA/FMR1. Furthermore, 91% of those with a negative result after CMA/FMR1 analysis (338 individuals) have not yet been referred for additional genetic testing and remain undiagnosed. CONCLUSION: Our findings strongly suggest that genome analysis outperforms other testing strategies and should replace traditional CMA and FMR1 analysis as a first-line genetic test in individuals with ID/NDD. GS is a sensitive, time- and cost-effective method that results in a confirmed molecular diagnosis in 35% of all referred patients.


Subject(s)
Intellectual Disability , Neurodevelopmental Disorders , Child , Humans , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Developmental Disabilities/genetics , Genetic Testing/methods , Microarray Analysis , Neurodevelopmental Disorders/genetics , Fragile X Mental Retardation Protein/genetics
6.
Am J Hum Genet ; 103(4): 553-567, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30290151

ABSTRACT

The conserved oligomeric Golgi (COG) complex is involved in intracellular vesicular transport, and is composed of eight subunits distributed in two lobes, lobe A (COG1-4) and lobe B (COG5-8). We describe fourteen individuals with Saul-Wilson syndrome, a rare form of primordial dwarfism with characteristic facial and radiographic features. All affected subjects harbored heterozygous de novo variants in COG4, giving rise to the same recurrent amino acid substitution (p.Gly516Arg). Affected individuals' fibroblasts, whose COG4 mRNA and protein were not decreased, exhibited delayed anterograde vesicular trafficking from the ER to the Golgi and accelerated retrograde vesicular recycling from the Golgi to the ER. This altered steady-state equilibrium led to a decrease in Golgi volume, as well as morphologic abnormalities with collapse of the Golgi stacks. Despite these abnormalities of the Golgi apparatus, protein glycosylation in sera and fibroblasts from affected subjects was not notably altered, but decorin, a proteoglycan secreted into the extracellular matrix, showed altered Golgi-dependent glycosylation. In summary, we define a specific heterozygous COG4 substitution as the molecular basis of Saul-Wilson syndrome, a rare skeletal dysplasia distinct from biallelic COG4-CDG.


Subject(s)
Fragile X Syndrome/genetics , Protein Transport/genetics , Proteoglycans/genetics , Vesicular Transport Proteins/genetics , Adult , Amino Acid Substitution/genetics , Animals , Animals, Genetically Modified/genetics , Cell Line , Child , Child, Preschool , Endoplasmic Reticulum/genetics , Extracellular Matrix/genetics , Female , Fibroblasts/pathology , Glycosylation , Golgi Apparatus/genetics , Heterozygote , Humans , Infant , Male , Zebrafish
7.
J Hum Genet ; 66(10): 995-1008, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33875766

ABSTRACT

Skeletal ciliopathies are a heterogenous group of disorders with overlapping clinical and radiographic features including bone dysplasia and internal abnormalities. To date, pathogenic variants in at least 30 genes, coding for different structural cilia proteins, are reported to cause skeletal ciliopathies. Here, we summarize genetic and phenotypic features of 34 affected individuals from 29 families with skeletal ciliopathies. Molecular diagnostic testing was performed using massively parallel sequencing (MPS) in combination with copy number variant (CNV) analyses and in silico filtering for variants in known skeletal ciliopathy genes. We identified biallelic disease-causing variants in seven genes: DYNC2H1, KIAA0753, WDR19, C2CD3, TTC21B, EVC, and EVC2. Four variants located in non-canonical splice sites of DYNC2H1, EVC, and KIAA0753 led to aberrant splicing that was shown by sequencing of cDNA. Furthermore, CNV analyses showed an intragenic deletion of DYNC2H1 in one individual and a 6.7 Mb de novo deletion on chromosome 1q24q25 in another. In five unsolved cases, MPS was performed in family setting. In one proband we identified a de novo variant in PRKACA and in another we found a homozygous intragenic deletion of IFT74, removing the first coding exon and leading to expression of a shorter message predicted to result in loss of 40 amino acids at the N-terminus. These findings establish IFT74 as a new skeletal ciliopathy gene. In conclusion, combined single nucleotide variant, CNV and cDNA analyses lead to a high yield of genetic diagnoses (90%) in a cohort of patients with skeletal ciliopathies.


Subject(s)
Bone Diseases, Developmental/genetics , Ciliopathies/genetics , Genetic Predisposition to Disease , Protein Isoforms/genetics , Adult , Aged , Bone Diseases, Developmental/epidemiology , Bone Diseases, Developmental/pathology , Ciliopathies/epidemiology , Ciliopathies/pathology , Cytoplasmic Dyneins/genetics , Cytoskeletal Proteins/genetics , Female , Genome, Human/genetics , High-Throughput Nucleotide Sequencing , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Male , Membrane Proteins/genetics , Microtubule-Associated Proteins/genetics , Middle Aged , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Whole Genome Sequencing
8.
Am J Med Genet A ; 185(2): 517-527, 2021 02.
Article in English | MEDLINE | ID: mdl-33398909

ABSTRACT

Bone dysplasias (osteochondrodysplasias) are a large group of conditions associated with short stature, skeletal disproportion, and radiographic abnormalities of skeletal elements. Nearly all are genetic in origin. We report a series of seven children with similar findings of chondrodysplasia and growth failure following early hematopoietic stem cell transplantation (HSCT) for pediatric non-oncologic disease: hemophagocytic lymphohistiocytosis or HLH (five children, three with biallelic HLH-associated variants [in PRF1 and UNC13D] and one with HLH secondary to visceral Leishmaniasis), one child with severe combined immunodeficiency and one with Omenn syndrome (both children had biallelic RAG1 pathogenic variants). All children had normal growth and no sign of chondrodysplasia at birth and prior to their primary disease. After HSCT, all children developed growth failure, with standard deviation scores for height at or below -3. Radiographically, all children had changes in the spine, metaphyses and epiphyses, compatible with a spondyloepimetaphyseal dysplasia. Genomic sequencing failed to detect pathogenic variants in genes associated with osteochondrodysplasias. We propose that such chondrodysplasia with growth failure is a novel, rare, but clinically important complication following early HSCT for non-oncologic pediatric diseases. The pathogenesis is unknown but could possibly involve loss or perturbation of the cartilage-bone stem cell population.


Subject(s)
Hematopoietic Stem Cell Transplantation/adverse effects , Lymphohistiocytosis, Hemophagocytic/genetics , Osteochondrodysplasias/genetics , Child , Child, Preschool , Female , Humans , Lymphohistiocytosis, Hemophagocytic/complications , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/therapy , Male , Membrane Proteins/genetics , Osteochondrodysplasias/complications , Osteochondrodysplasias/diagnosis , Osteochondrodysplasias/therapy , Perforin/genetics , Treatment Outcome
9.
Radiographics ; 41(1): 192-209, 2021.
Article in English | MEDLINE | ID: mdl-33186059

ABSTRACT

Type II collagen is a major component of the cartilage matrix. Pathogenic variants (ie, disease-causing aberrations) in the type II collagen gene (COL2A1) lead to an abnormal structure of type II collagen, causing a large group of skeletal dysplasias termed type II collagenopathies. Because type II collagen is also located in the vitreous body of the eyes and inner ears, type II collagenopathies are commonly associated with vitreoretinal degeneration and hearing impairment. Type II collagenopathies can be radiologically divided into two major groups: the spondyloepiphyseal dysplasia congenita (SEDC) group and the Kniest-Stickler group. The SEDC group is characterized by delayed ossification of the juxtatruncal bones, including pear-shaped vertebrae. These collagenopathies comprise achondrogenesis type 2, hypochondrogenesis, SEDC, and other uncommon subtypes. The Kniest-Stickler group is characterized by disordered tubular bone growth that leads to "dumbbell" deformities. It comprises Kniest dysplasia and Stickler dysplasia type 1, whose radiographic manifestations overlap with those of type XI collagenopathies (a group of disorders due to abnormal type XI collagen) such as Stickler dysplasia types 2 and 3. This phenotypic overlap is caused by type II and type XI collagen molecules sharing part of the same connective tissues. The authors describe the diagnostic pathways to type II and type XI collagenopathies and the associated differential diagnoses. In addition, they review the clinical features and genetic bases of these conditions, which radiologists should know to participate in multidisciplinary care and translational research. Online supplemental material is available for this article. ©RSNA, 2020.


Subject(s)
Achondroplasia , Collagen Diseases , Hyaline Membrane Disease , Osteochondrodysplasias , Cartilage , Collagen Diseases/diagnostic imaging , Humans , Infant, Newborn , Osteochondrodysplasias/diagnostic imaging
10.
Genet Med ; 22(5): 857-866, 2020 05.
Article in English | MEDLINE | ID: mdl-31949312

ABSTRACT

PURPOSE: Four patients with Saul-Wilson syndrome were reported between 1982 and 1994, but no additional individuals were described until 2018, when the molecular etiology of the disease was elucidated. Hence, the clinical phenotype of the disease remains poorly defined. We address this shortcoming by providing a detailed characterization of its phenotype. METHODS: Retrospective chart reviews were performed and primary radiographs assessed for all 14 individuals. Four individuals underwent detailed ophthalmologic examination by the same physician. Two individuals underwent gynecologic evaluation. Z-scores for height, weight, head circumference and body mass index were calculated at different ages. RESULTS: All patients exhibited short stature, with sharp decline from the mean within the first months of life, and a final height Z-score between -4 and -8.5 standard deviations. The facial and radiographic features evolved over time. Intermittent neutropenia was frequently observed. Novel findings included elevation of liver transaminases, skeletal fragility, rod-cone dystrophy, and cystic macular changes. CONCLUSIONS: Saul-Wilson syndrome presents a remarkably uniform phenotype, and the comprehensive description of our cohort allows for improved understanding of the long-term morbidity of the condition, establishment of follow-up recommendations for affected individuals, and documentation of the natural history into adulthood for comparison with treated patients, when therapeutics become available.


Subject(s)
Dwarfism , Adult , Female , Humans , Phenotype , Retrospective Studies
11.
Hum Mutat ; 39(10): 1456-1467, 2018 10.
Article in English | MEDLINE | ID: mdl-30080953

ABSTRACT

Skeletal dysplasias are a diverse group of rare Mendelian disorders with clinical and genetic heterogeneity. Here, we used targeted copy number variant (CNV) screening and identified intragenic exonic duplications, formed through Alu-Alu fusion events, in two individuals with skeletal dysplasia and negative exome sequencing results. First, we detected a homozygous tandem duplication of exon 9 and 10 in IFT81 in a boy with Jeune syndrome, or short-rib thoracic dysplasia (SRTD) (MIM# 208500). Western blot analysis did not detect any wild-type IFT81 protein in fibroblasts from the patient with the IFT81 duplication, but only a shorter isoform of IFT81 that was also present in the normal control samples. Complementary zebrafish studies suggested that loss of full-length IFT81 protein but expression of a shorter form of IFT81 protein affects the phenotype while being compatible with life. Second, a de novo tandem duplication of exons 2 to 5 in MATN3 was identified in a girl with multiple epiphyseal dysplasia (MED) type 5 (MIM# 607078). Our data highlights the importance of detection and careful characterization of intragenic duplication CNVs, presenting them as a novel and very rare genetic mechanism in IFT81-related Jeune syndrome and MATN3-related MED.


Subject(s)
Alu Elements , Gene Duplication , Genetic Association Studies , Muscle Proteins/genetics , Osteochondrodysplasias/diagnosis , Osteochondrodysplasias/genetics , Adolescent , Animals , Child , Comparative Genomic Hybridization , DNA Copy Number Variations , Ellis-Van Creveld Syndrome/diagnosis , Ellis-Van Creveld Syndrome/genetics , Female , Homozygote , Humans , Male , Matrilin Proteins/genetics , Pedigree , Phenotype , Radiography , Whole Genome Sequencing , Zebrafish
12.
Am J Med Genet A ; 176(8): 1723-1734, 2018 08.
Article in English | MEDLINE | ID: mdl-30070757

ABSTRACT

As growth references for achondroplasia are limited to reports from United States, Japan, Argentina, and Australia, the aim of this study was to construct growth references for height, weight, head circumference, and body mass index (BMI) from a European cohort of children with achondroplasia and to discuss the development of these anthropometric variables. A mix of cross-sectional and longitudinal, retrospective, and prospective data from 466 children with achondroplasia and 4,375 measuring occasions were modeled with generalized additive model for location, scale and shape (GAMLSS) to sex-specific references for ages 0 to 20 years. Loss in height position, that is, reduction in height standard deviation scores, occurred mainly during first 2 years of life while pubertal growth seemed normal if related to adult height. Adult height was 132 cm in boys and 124 cm in girls with a variability comparable to that of the general population and seems to be remarkably similar in most studies of children with achondroplasia. BMI had a syndrome-specific development that was not comparable to BMI development in the general population. Weight and BMI might be misleading when evaluating, for example, metabolic health in achondroplasia. Head circumference reached adult head size earlier than in the general population. Increased tempo of head circumference growth necessitates thus close clinical follow-up during first postnatal years.

13.
Am J Med Genet A ; 176(9): 1819-1829, 2018 09.
Article in English | MEDLINE | ID: mdl-30152086

ABSTRACT

The aims of this study was to construct references for sitting height, leg length, arm span, relative sitting height (sitting height/height), and foot length and to discuss the development for these anthropometric variables in achondroplasia. Sex-specific references covering ±2 SD are presented for ages 2-20 years. Legs and arms in achondroplasia are already at 2 years of age considerably shorter than in the general population and this deviation increases with age. At adult ages, legs are almost 50% shorter than in the general population and arm span roughly 35% shorter. As sitting height is only mildly affected, relative sitting height position develops far beyond normal ranges. Foot length is also not as affected as limbs.


Subject(s)
Achondroplasia/diagnosis , Body Weights and Measures , Growth and Development , Adolescent , Adult , Arm/growth & development , Body Height , Child , Child, Preschool , Female , Foot/growth & development , Growth Charts , Humans , Male , Phenotype , Sitting Position , Young Adult
14.
Am J Hum Genet ; 95(3): 285-93, 2014 Sep 04.
Article in English | MEDLINE | ID: mdl-25152457

ABSTRACT

Neu-Laxova syndrome (NLS) is a rare autosomal-recessive disorder characterized by a recognizable pattern of severe malformations leading to prenatal or early postnatal lethality. Homozygous mutations in PHGDH, a gene involved in the first and limiting step in L-serine biosynthesis, were recently identified as the cause of the disease in three families. By studying a cohort of 12 unrelated families affected by NLS, we provide evidence that NLS is genetically heterogeneous and can be caused by mutations in all three genes encoding enzymes of the L-serine biosynthesis pathway. Consistent with recently reported findings, we could identify PHGDH missense mutations in three unrelated families of our cohort. Furthermore, we mapped an overlapping homozygous chromosome 9 region containing PSAT1 in four consanguineous families. This gene encodes phosphoserine aminotransferase, the enzyme for the second step in L-serine biosynthesis. We identified six families with three different missense and frameshift PSAT1 mutations fully segregating with the disease. In another family, we discovered a homozygous frameshift mutation in PSPH, the gene encoding phosphoserine phosphatase, which catalyzes the last step of L-serine biosynthesis. Interestingly, all three identified genes have been previously implicated in serine-deficiency disorders, characterized by variable neurological manifestations. Our findings expand our understanding of NLS as a disorder of the L-serine biosynthesis pathway and suggest that NLS represents the severe end of serine-deficiency disorders, demonstrating that certain complex syndromes characterized by early lethality could indeed be the extreme end of the phenotypic spectrum of already known disorders.


Subject(s)
Abnormalities, Multiple/genetics , Brain Diseases/genetics , Fetal Growth Retardation/genetics , Ichthyosis/genetics , Limb Deformities, Congenital/genetics , Microcephaly/genetics , Mutation/genetics , Phosphoglycerate Dehydrogenase/genetics , Phosphoric Monoester Hydrolases/genetics , Serine/biosynthesis , Transaminases/genetics , Abnormalities, Multiple/metabolism , Amino Acid Sequence , Brain Diseases/metabolism , Consanguinity , Family , Female , Fetal Growth Retardation/metabolism , Homozygote , Humans , Ichthyosis/metabolism , Limb Deformities, Congenital/metabolism , Male , Microcephaly/metabolism , Molecular Sequence Data , Phosphoglycerate Dehydrogenase/chemistry , Phosphoglycerate Dehydrogenase/deficiency , Phosphoric Monoester Hydrolases/chemistry , Phosphoric Monoester Hydrolases/deficiency , Protein Conformation , Sequence Homology, Amino Acid , Serine/chemistry , Transaminases/chemistry , Transaminases/deficiency
15.
J Hum Genet ; 62(4): 503-506, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28123176

ABSTRACT

Axial spondylometaphyseal dysplasia (axial SMD) is a unique form of SMD characterized by dysplasia of axial skeleton and retinal dystrophy. Recently, C21orf2 has been identified as the first disease gene for axial SMD; however, the presence of genetic heterogeneity is known. In this study, we identified NEK1 as the second disease gene for axial SMD. By whole-exome sequencing in a patient with axial SMD, we identified compound heterozygous mutations of NEK1, c.3107C>G (p.S1036*) and c.3830A>C (p.D1277A), which co-segregated in the family. NEK1 mutations have previously been found in three types of short rib thoracic dystrophy, which have no retinal dystrophy. The skeletal phenotype of our patient was milder than those of previously reported cases with NEK1 mutations and those with axial SMD harboring C21orf2 mutations. Phenotypes associated with NEK1 mutations are variable and the phenotype-genotype corelation in skeletal ciliopathies is challenging.


Subject(s)
Mutation , NIMA-Related Kinase 1/genetics , Osteochondrodysplasias/genetics , Child , Cytoskeletal Proteins , Genetic Association Studies , Humans , Male , Pedigree , Proteins/genetics
16.
Am J Hum Genet ; 92(6): 990-5, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23684011

ABSTRACT

Kenny-Caffey syndrome (KCS) and the similar but more severe osteocraniostenosis (OCS) are genetic conditions characterized by impaired skeletal development with small and dense bones, short stature, and primary hypoparathyroidism with hypocalcemia. We studied five individuals with KCS and five with OCS and found that all of them had heterozygous mutations in FAM111A. One mutation was identified in four unrelated individuals with KCS, and another one was identified in two unrelated individuals with OCS; all occurred de novo. Thus, OCS and KCS are allelic disorders of different severity. FAM111A codes for a 611 amino acid protein with homology to trypsin-like peptidases. Although FAM111A has been found to bind to the large T-antigen of SV40 and restrict viral replication, its native function is unknown. Molecular modeling of FAM111A shows that residues affected by KCS and OCS mutations do not map close to the active site but are clustered on a segment of the protein and are at, or close to, its outer surface, suggesting that the pathogenesis involves the interaction with as yet unidentified partner proteins rather than impaired catalysis. FAM111A appears to be crucial to a pathway that governs parathyroid hormone production, calcium homeostasis, and skeletal development and growth.


Subject(s)
Abnormalities, Multiple/genetics , Bone Diseases, Developmental/genetics , Craniofacial Abnormalities/genetics , Dwarfism/genetics , Hyperostosis, Cortical, Congenital/genetics , Hypocalcemia/genetics , Hypoparathyroidism/genetics , Receptors, Virus/genetics , Abnormalities, Multiple/diagnostic imaging , Abnormalities, Multiple/mortality , Abnormalities, Multiple/pathology , Adolescent , Adult , Bone Diseases, Developmental/mortality , Bone Diseases, Developmental/pathology , Child , Craniofacial Abnormalities/mortality , Craniofacial Abnormalities/pathology , Dwarfism/diagnostic imaging , Dwarfism/mortality , Genetic Association Studies , Heterozygote , Humans , Hyperostosis, Cortical, Congenital/diagnostic imaging , Hyperostosis, Cortical, Congenital/mortality , Hypocalcemia/diagnostic imaging , Hypocalcemia/mortality , Hypoparathyroidism/diagnostic imaging , Hypoparathyroidism/mortality , Infant , Infant, Newborn , Male , Mutation, Missense , Parathyroid Hormone/deficiency , Radiography
17.
Skeletal Radiol ; 45(11): 1557-60, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27544198

ABSTRACT

Brachyolmia (BO) is a heterogeneous group of skeletal dysplasias with skeletal changes limited to the spine or with minimal extraspinal features. BO is currently classified into types 1, 2, 3, and 4. BO types 1 and 4 are autosomal recessive conditions caused by PAPSS2 mutations, which may be merged together as an autosomal recessive BO (AR-BO). The clinical and radiological signs of AR-BO in late childhood have already been reported; however, the early manifestations and their age-dependent evolution have not been well documented. We report an affected boy with AR-BO, whose skeletal abnormalities were detected in utero and who was followed until 10 years of age. Prenatal ultrasound showed bowing of the legs. In infancy, radiographs showed moderate platyspondyly and dumbbell deformity of the tubular bones. Gradually, the platyspondyly became more pronounced, while the bowing of the legs and dumbbell deformities of the tubular bones diminished with age. In late childhood, the overall findings were consistent with known features of AR-BO. Genetic testing confirmed the diagnosis. Being aware of the initial skeletal changes may facilitate early diagnosis of PAPSS2-related skeletal dysplasias.


Subject(s)
Aging/pathology , Osteochondrodysplasias/diagnostic imaging , Osteochondrodysplasias/pathology , Radiography, Thoracic/methods , Child , Child, Preschool , Diagnosis, Differential , Disease Progression , Humans , Infant , Male
18.
J Med Genet ; 51(1): 45-54, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24203977

ABSTRACT

BACKGROUND: Point mutations in PDE4D have been recently linked to acrodysostosis, an autosomal dominant disorder with skeletal dysplasia, severe brachydactyly, midfacial hypoplasia and intellectual disability. The purpose of the present study was to investigate clinical and cellular implications of different types of mutations in the PDE4D gene. METHODS: We studied five acrodysostosis patients and three patients with gene dose imbalances involving PDE4D clinically and by whole exome sequencing, Sanger sequencing and array comparative hybridisation. To evaluate the functional consequences of the PDE4D changes, we used overexpression of mutated human PDE4D message and morpholino-based suppression of pde4d in zebrafish. RESULTS: We identified three novel and two previously described PDE4D point mutations in the acrodysostosis patients and two deletions and one duplication involving PDE4D in three patients suffering from an intellectual disability syndrome with low body mass index, long fingers, toes and arms, prominent nose and small chin. When comparing symptoms in patients with missense mutations and gene dose imbalances involving PDE4D, a mirror phenotype was observed. By comparing overexpression of human mutated transcripts with pde4d knockdown in zebrafish embryos, we could successfully assay the pathogenicity of the mutations. CONCLUSIONS: Our findings indicate that haploinsufficiency of PDE4D results in a novel intellectual disability syndrome, the 5q12.1-haploinsufficiency syndrome, with several opposing features compared with acrodysostosis that is caused by dominant negative mutations. In addition, our results expand the spectrum of PDE4D mutations underlying acrodysostosis and indicate that, in contrast to previous reports, patients with PDE4D mutations may have significant hormone resistance with consequent endocrine abnormalities.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Developmental Disabilities/diagnosis , Developmental Disabilities/genetics , Mutation , Phenotype , Animals , Comparative Genomic Hybridization , Dysostoses/diagnosis , Dysostoses/genetics , Facies , Female , Gene Deletion , Gene Expression , Gene Order , Genetic Association Studies , Humans , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Male , Osteochondrodysplasias/diagnosis , Osteochondrodysplasias/genetics , Point Mutation , Zebrafish/genetics
19.
Am J Med Genet A ; 164A(7): 1635-41, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24677493

ABSTRACT

Autosomal dominant brachyolmia (Type 3, OMIM #113500) belongs to a group of skeletal dysplasias caused by mutations in the transient receptor potential cation channel, subfamily V, member 4 (TRPV4) gene, encoding a Ca++-permeable, non-selective cation channel. The disorder is characterized by disproportionate short stature with short trunk, scoliosis and platyspondyly. The phenotypic variability and long-term natural course remain inadequately characterized. The purpose of this study was to describe a large Swedish family with brachyolmia type 3 due to a heterozygous TRPV4 mutation c.1847G>A (p.R616Q) in 11 individuals. The mutation has previously been detected in another family with autosomal dominant brachyolmia [Rock et al., 2008]. Review of hospital records and patient assessments indicated that clinical symptoms of brachyolmia became evident by school age with chronic pain in the spine and hips; radiographic changes were evident earlier. Growth was not affected during early childhood but deteriorated with age in some patients due to increasing spinal involvement. Affected individuals had a wide range of subjective symptoms with chronic pain in the extremities and the spine, and paresthesias. Our findings indicate that autosomal dominant brachyolmia may be associated with significant long-term morbidity, as seen in this family.


Subject(s)
Genetic Association Studies , Mutation , Osteochondrodysplasias/diagnosis , Osteochondrodysplasias/genetics , Phenotype , TRPV Cation Channels/genetics , Adult , Child, Preschool , Female , Genotype , Humans , Male , Pedigree , Radiography , Spine/diagnostic imaging , Spine/pathology
20.
Am J Med Genet A ; 161A(10): 2554-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23956106

ABSTRACT

Achondrogenesis 1A (ACG1A; OMIM 200600) is an autosomal recessive perinatally lethal skeletal dysplasia comprising intrauterine growth failure, micromelia, minor facial anomalies, deficient ossification of the skull, absent or extremely defective spinal ossification, short beaded ribs, and short deformed long bones with a stellate appearance. ACG1A is caused by mutations in the TRIP11 gene, resulting in deficiency of the Golgi microtubule associated protein 210. In this study we describe dizygotic twins with a clinical and radiological phenotype of ACG1A who were homozygous for a novel nonsense mutation in the TRIP11 gene. In addition, another patient with a milder manifestation, not readily distinguishable from those of other lethal skeletal dysplasias, was found to be a compound heterozygote for a nonsense mutation and a deletion of the 3' end of the TRIP11 gene. We conclude that mutations of the TRIP11 gene may encompass a wider phenotypic range than previously recognized.


Subject(s)
Achondroplasia/diagnosis , Phenotype , Achondroplasia/genetics , Cytoskeletal Proteins , Diagnostic Imaging , Female , Genotype , Humans , Infant, Newborn , Male , Mutation , Nuclear Proteins/genetics , Twins, Dizygotic
SELECTION OF CITATIONS
SEARCH DETAIL