Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 559
Filtrar
1.
Orphanet J Rare Dis ; 19(1): 63, 2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-38351015

RESUMO

BACKGROUND: Multiple osteochondromas is genetic disorder characterized by the formation of multiple benign cartilage-capped bone tumors, named osteochondromas, during skeletal development. The most feared complication is the secondary peripheral chondrosarcoma, a malignant cartilaginous neoplasm that arises from the chondroid cap of pre-existent osteochondromas. We conducted a retrospective cohort study on patients diagnosed and followed up from 1960 to 2019 to describe the clinical and pathological features of individuals affected by peripheral chondrosarcoma in multiple osteochondromas, to evaluate follow up information and individual outcome and to compare the results with literature. Data, including age, gender, site, histological grade, cartilage cap thickness, surgical treatments, surgical margins, genotype mutational status as well as treatment details were captured from the hospital electronic health records and from Registry of Multiple Osteochondromas. In addition, a complete histological review of all hematoxylin and eosin (H&E)-stained sections has been performed by expert pathologists. RESULTS: One hundred five of the screened cases were included in the present study. The age at diagnosis of SPC ranges from 13 to 63, with median age at diagnosis of 34 years. The site most frequently affected by malignant degeneration was the pelvis (46 patients, 44%) with higher incidence in male patients (32 males vs.14 females). The second one was lower limbs (including femur, fibula, or tibia), identified in 35 patients. Histological information - available for 103 patients - showed: 59 patients with grade 1; 40 patients had a grade 2 and 4 patients had a grade 3. The most common surgical treatment was the complete resection, followed by debulking, amputation and partial resection. Most of cases did not have recurrence of the disease. Outcome in disease-free survival highlights that a worse course of the disease was associated with histological grade 2 or 3, and partial resection surgery. In most of analyzed cases (94%) a pathogenic variant was identified. CONCLUSIONS: In conclusion, the present study gives an overview of the secondary peripheral chondrosarcomas, confirming that this disease represents an impacting complication for multiple osteochondromas patients and suggests that malignant transformation can occur also in younger patient, in a not irrelevant number of cases.


Assuntos
Neoplasias Ósseas , Condrossarcoma , Exostose Múltipla Hereditária , Osteocondroma , Feminino , Humanos , Masculino , Adulto , Exostose Múltipla Hereditária/genética , Estudos Retrospectivos , Condrossarcoma/genética , Condrossarcoma/diagnóstico , Condrossarcoma/patologia , Osteocondroma/patologia , Intervalo Livre de Doença , Neoplasias Ósseas/genética , Neoplasias Ósseas/diagnóstico , Neoplasias Ósseas/patologia
2.
Am J Med Genet A ; 191(6): 1570-1575, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36869625

RESUMO

Hereditary multiple exostoses (HME), also known as hereditary multiple osteochondroma (HMO), is an autosomal dominant disorder caused by pathogenic variants in exostosin-1 or -2 (EXT1 or EXT2). It is characterized by the formation of multiple benign growing osteochondromas (exostoses) that most commonly affect the long bones; however, it may also occur throughout the body. Although many of these lesions are clinically asymptomatic, some can lead to chronic pain and skeletal deformities and interfere with adjacent neurovascular structures. Here, we report two unrelated probands that presented with a clinical and molecular diagnosis of HME with venous malformation, a clinical feature not previously reported in individuals with HME.


Assuntos
Exostose Múltipla Hereditária , Humanos , Exostose Múltipla Hereditária/diagnóstico , Exostose Múltipla Hereditária/genética , N-Acetilglucosaminiltransferases/genética , Mutação
3.
J Pediatr Orthop ; 43(5): 332-336, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36806113

RESUMO

BACKGROUND: The Pediatric Outcomes Data Collection Instrument (PODCI) is a validated quality-of-life questionnaire with 6 domains designed to provide a standardized method of measuring outcomes in pediatric musculoskeletal conditions. To our knowledge there are no reports on its use in children with multiple hereditary exostosis (MHE). QUESTIONS/PURPOSES: Most published studies on MHE patients have described the efficacy of specific surgical techniques or the specification of deformities. Little is known about the general health status of pediatric patients, the severity of pain, loss of function, and how MHE influences the activities of daily life. We aim to assess the functional levels of MHE pediatric patients with PODCI questionnaire. PATIENTS AND METHODS: As a cross-sectional study, we prospectively administered PODCI to 34 pediatric patients diagnosed with MHE and their families. The score distributions were compared with values published earlier for children and adolescents without musculoskeletal disorders using the Student and Welch t tests. Parents and adolescents' reports were compared using Wilcoxon signed rank test. Physical examination and PODCI score relation were evaluated by Spearman test. RESULTS: Children with MHE have significantly lower scores ( P <0,05) in comparison with unaffected children in all domains using the Student and Welch t test. Parents score differs from children score with statistically relevance in pain and comfort domain ( P <0,5). The Spearman test showed a negative correlation between physical examination and PODCI score with statistical significance. CONCLUSIONS: These results point towards PODCI's capacity in evaluating functional outcomes of pediatric patients with MHE. LEVEL OF EVIDENCE: Diagnostic Study, Level III.


Assuntos
Exostose Múltipla Hereditária , Doenças Musculoesqueléticas , Adolescente , Criança , Humanos , Exostose Múltipla Hereditária/genética , Estudos Transversais , Avaliação de Resultados em Cuidados de Saúde/métodos , Inquéritos e Questionários , Dor
4.
Eur J Hum Genet ; 31(2): 248-251, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36443465

RESUMO

Only a limited number of genetic diseases are diagnosable in archaeological individuals and none have had causal mutations identified in genome-wide screens. Two individuals from the Gaelic Irish Medieval burial ground of Ballyhanna, Co. Donegal, showed evidence of bone tumours consistent with the autosomal dominant condition multiple osteochondromas. Genome sequencing of the earlier individual uncovered a missense mutation in the second exon of EXT1, a specific lesion that has been identified in several modern patients. The later individual lacked this but displayed a novel frameshift mutation leading to a premature stop codon and loss of function in the same gene. These molecular confirmations of a paleopathological diagnosis within a single rural ancient context are surprisingly disjunct, given the observation of clusters of this disease in modern isolated populations and a de novo mutation rate of only 10%.


Assuntos
Exostose Múltipla Hereditária , Humanos , Exostose Múltipla Hereditária/genética , Cemitérios , N-Acetilglucosaminiltransferases/genética , Mutação , Mutação da Fase de Leitura
6.
Genes (Basel) ; 13(11)2022 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-36360300

RESUMO

Multiple Osteochondromatosis (MO, MIM 133700 & 133701), an autosomal dominant O-glycosylation disorder (EXT1/EXT2-CDG), can be associated with a reduction in skeletal growth, bony deformity, restricted joint motion, shortened stature and pathogenic variants in two tumor suppressor genes, EXT1 and EXT2. In this work, we report a cross-sectional study including 35 index patients and 20 affected family members. Clinical phenotyping of all 55 affected cases was obtained, but genetic studies were performed only in 35 indexes. Of these, a total of 40% (n = 14) had a family history of MO. Clinical severity scores were class I in 34% (n:18), class II in 24.5% (n:13) and class III in 41.5% (n:22). Pathogenic variants were identified in 83% (29/35) probands. We detected 18 (62%) in EXT1 and 11 (38%) in EXT2. Patients with EXT1 variants showed a height z-score of 1.03 SD lower than those with EXT2 variants and greater clinical severity (II-III vs. I). Interestingly, three patients showed intellectual impairment, two patients showed a dual diagnosis, one Turner Syndrome and one hypochondroplasia. This study improves knowledge of MO, reporting new pathogenic variants and forwarding the worldwide collaboration necessary to promote the inclusion of patients into future biologically based therapeutics.


Assuntos
Exostose Múltipla Hereditária , Humanos , Exostose Múltipla Hereditária/genética , Exostose Múltipla Hereditária/diagnóstico , Estudos Transversais , N-Acetilglucosaminiltransferases/genética , Mutação , Testes Genéticos
8.
Front Endocrinol (Lausanne) ; 13: 874126, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35837302

RESUMO

Background: Despite the new next-generation sequencing (NGS) molecular approaches implemented the genetic testing in clinical diagnosis, copy number variation (CNV) detection from NGS data remains difficult mainly in the absence of bioinformatics personnel (not always available among laboratory resources) and when using very small gene panels that do not meet commercial software criteria. Furthermore, not all large deletions/duplications can be detected with the Multiplex Ligation-dependent Probe Amplification (MLPA) technique due to both the limitations of the methodology and no kits available for the most of genes. Aim: We propose our experience regarding the identification of a novel large deletion in the context of a rare skeletal disease, multiple osteochondromas (MO), using and validating a user-friendly approach based on NGS coverage data, which does not require any dedicated software or specialized personnel. Methods: The pipeline uses a simple algorithm comparing the normalized coverage of each amplicon with the mean normalized coverage of the same amplicon in a group of "wild-type" samples representing the baseline. It has been validated on 11 samples, previously analyzed by MLPA, and then applied on 20 patients with MO but negative for the presence of pathogenic variants in EXT1 or EXT2 genes. Sensitivity, specificity, and accuracy were evaluated. Results: All the 11 known CNVs (exon and multi-exon deletions) have been detected with a sensitivity of 97.5%. A novel EXT2 partial exonic deletion c. (744-122)-?_804+?del -out of the MLPA target regions- has been identified. The variant was confirmed by real-time quantitative Polymerase Chain Reaction (qPCR). Conclusion: In addition to enhancing the variant detection rate in MO molecular diagnosis, this easy-to-use approach for CNV detection can be easily extended to many other diagnostic fields-especially in resource-limited settings or very small gene panels. Notably, it also allows partial-exon deletion detection.


Assuntos
Variações do Número de Cópias de DNA , Exostose Múltipla Hereditária , Éxons , Exostose Múltipla Hereditária/genética , Testes Genéticos/métodos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos
9.
Anim Genet ; 53(5): 696-699, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35719100

RESUMO

Osteochondromatosis is a benign proliferative disorder characterized by cartilage-capped bony protuberances. In humans and most mammals, variants in the EXT1 or EXT2 gene are strongly correlated with the etiology of osteochondromatosis. However, in cats, osteochondromatosis has only been associated with feline leukemia virus infection. In this study, to explore other factors involved in the etiology of feline osteochondromatosis, we examined the EXT1 and EXT2 genes in a feline leukemia virus-negative cat with osteochondromatosis. Genetic analysis revealed a heterozygous single base pair duplication in exon 6 of the EXT1 gene (XM_023248762.2:c.1468dupC), leading to a premature stop codon in the EXT1 protein. Notably, this frameshift variant is recognized as one of the most common pathogenic variants in human osteochondromatosis. Our data suggest for the first time that genetic variants can have etiologic roles in osteochondromatosis in cats, as in humans and other animals.


Assuntos
Doenças do Gato , Exostose Múltipla Hereditária , Osteocondromatose , Animais , Doenças do Gato/genética , Gatos/genética , Éxons , Exostose Múltipla Hereditária/genética , Mutação da Fase de Leitura , Humanos , Vírus da Leucemia Felina/genética , Mamíferos/genética , Osteocondromatose/genética , Osteocondromatose/patologia , Osteocondromatose/veterinária
10.
Orphanet J Rare Dis ; 17(1): 174, 2022 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-35461249

RESUMO

BACKGROUND: Stuve-Wiedemann syndrome (SWS) is a rare and severe genetic disease characterized by skeletal anomalies and dysautonomic disturbances requiring appropriate care. Peer support is mandatory to fill the lack of clinical recommendations in such rare diseases. We report a new case and provide the first systematic review of all previous published cases. OBJECTIVE: To better describe the timeline of SWS and to improve paediatric management. DATA SOURCES: SWS English publications available on Pubmed until 31/03/2021. STUDY SELECTION: Case description combining typical osteo-articular and dysautonomic involvement (with 2 items by categories required for children < 2 years and 3 items > 2 years). DATA EXTRACTION: Demographic, clinical, genetics and outcome data. RESULTS: In our cohort of 69 patients, the median age at report was 32 months. Only 46% presented antenatal signs. Mortality rate is higher during the first 2 years (42% < 2 years; 10% > 2 years) mainly due to respiratory failure, pulmonary arterial hypertension appearing to be a poor prognosis factor (mortality rate 63%). After 2 years, orthopaedic symptoms significantly increase including joint mobility restriction (81%), spinal deformations (77%) and fractures (61%). CONCLUSIONS: Natural history of SWS is marked by a high mortality rate before 2 years due to dysautonomic disturbances. A specialized multidisciplinary approach is needed to address these early mortality risks and then adapt to the specific, mainly orthopaedic, needs of patients after 2 years of age. Further research is required to provide clinical guidelines and improve pre-natal counselling.


Assuntos
Anormalidades Múltiplas , Exostose Múltipla Hereditária , Osteocondrodisplasias , Insuficiência Respiratória , Anormalidades Múltiplas/genética , Criança , Exostose Múltipla Hereditária/genética , Feminino , Humanos , Osteocondrodisplasias/genética , Gravidez
11.
Mol Genet Genomic Med ; 10(3): e1878, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35106951

RESUMO

BACKGROUND: Hereditary multiple exostosis (HME) is an autosomal dominant skeletal disorder characterized by the development of multiple cartilage-covered tumors on the external surfaces of bones (osteochondromas). Most of HME cases result from heterozygous loss-of-function mutations in EXT1 or EXT2 gene. METHODS: Clinical examination was performed to diagnose the patients: Whole exome sequencing (WES) was used to identify pathogenic mutations in the proband, which is confirmed by Sanger sequencing and co-segregation analysis: qRT-PCR was performed to identify the mRNA expression level of EXT1 in patient peripheral blood samples: minigene splicing assay was performed to mimic the splicing process of EXT1 variants in vitro. RESULTS: We evaluated the pathogenicity of EXT1 c.1056 + 1G > T in a Chinese family with HME. The clinical, phenotypic, and genetic characterization of patients in this family were described. The variant was detected by whole-exome sequencing (WES) and confirmed by Sanger sequencing. Sequencing of the RT-PCR products from the patient's blood sample identified a large deletion (94 nucleotides), which is the whole exome 2 of the EXT1 cDNA. Splicing assay indicated that the mutated minigene produced alternatively spliced transcripts, which cause a frameshift resulting in an early termination of protein expression. CONCLUSIONS: Our study establishes the pathogenesis of the splicing mutation EXT1 c.1056 + 1G > T to HME and provides scientific foundation for accurate diagnosis and precise medical intervention for HME.


Assuntos
Exostose Múltipla Hereditária , China , Exostose Múltipla Hereditária/genética , Humanos , N-Acetilglucosaminiltransferases/genética , Linhagem , Splicing de RNA
12.
Mol Med Rep ; 25(4)2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35211766

RESUMO

The aim of the present study was to report a clinical survey of hereditary multiple exostoses (HME) in a large Chinese pedigree, and the identification of a novel deletion mutation of exostosin glycosyltransferase 2 (EXT­2) gene. A patient with multiple exostoses with huge cartilage­capped tumors in scapula, knees and ankles received surgery in Department of Orthopedics (Shanghai Changhai Hospital). A total of 20 family members were recruited to the study, with seven members (five male; two female) diagnosed as HME. The family members of the patients with HME were examined, clinical data and peripheral blood samples were collected, and their DNA was sequenced. The incidence of HME in this family pedigree was 35%. Exostoses were most frequently in the tibiae with occurrence in six patients, followed by ribs, femurs, radii, fibulae, scapulae and humeri. DNA sequencing of peripheral blood revealed a novel deletion mutation, c.824­826delGCA, in exon 5 of the EXT­2 gene, which was observed in all the patients with HME, but not in the healthy family members. Several characteristics of HME in the pedigree were observed, such as susceptibility of male gender, decreased average age of onset and height and increased severity of clinical symptoms with generations.


Assuntos
Exostose Múltipla Hereditária , N-Acetilglucosaminiltransferases/genética , China , Exostose Múltipla Hereditária/diagnóstico , Exostose Múltipla Hereditária/genética , Exostose Múltipla Hereditária/patologia , Feminino , Deleção de Genes , Humanos , Masculino , Mutação , Linhagem
13.
J Orthop Res ; 40(10): 2391-2401, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-34996123

RESUMO

Hereditary multiple exostoses (HME) is a rare, pediatric disorder characterized by osteochondromas that form along growth plates and provoke significant musculoskeletal problems. HME is caused by mutations in heparan sulfate (HS)-synthesizing enzymes EXT1 or EXT2. Seemingly paradoxically, osteochondromas were found to contain excessive extracellular heparanase (Hpse) that could further reduce HS levels and exacerbate pathogenesis. To test Hpse roles, we asked whether its ablation would protect against osteochondroma formation in a conditional HME model consisting of mice bearing floxed Ext1 alleles in Agr-CreER background (Ext1f/f ;Agr-CreER mice). Mice were crossed with a new global Hpse-null (Hpse-/- ) mice to produce compound Hpse-/- ;Ext1f/f ;Agr-CreER mice. Tamoxifen injection of standard juvenile Ext1f/f ;Agr-CreER mice elicited stochastic Ext1 ablation in growth plate and perichondrium, followed by osteochondroma formation, as revealed by microcomputed tomography and histochemistry. When we examined companion conditional Ext1-deficient mice lacking Hpse also, we detected no major decreases in osteochondroma number, skeletal distribution, and overall structure by the analytical criteria above. The Ext1 mutants used here closely mimic human HME pathogenesis, but have not been previously tested for responsiveness to treatments. To exclude some innate therapeutic resistance in this stochastic model, tamoxifen-injected Ext1f/f ;Agr-CreER mice were administered daily doses of the retinoid Palovarotene, previously shown to prevent ectopic cartilage and bone formation in other mouse disease models. This treatment did inhibit osteochondroma formation compared with vehicle-treated mice. Our data indicate that heparanase is not a major factor in osteochondroma initiation and accumulation in mice. Possible roles of heparanase upregulation in disease severity in patients are discussed.


Assuntos
Neoplasias Ósseas , Exostose Múltipla Hereditária , Glucuronidase , N-Acetilglucosaminiltransferases , Osteocondroma , Animais , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Criança , Modelos Animais de Doenças , Exostose Múltipla Hereditária/genética , Exostose Múltipla Hereditária/metabolismo , Exostose Múltipla Hereditária/patologia , Glucuronidase/genética , Glucuronidase/metabolismo , Heparitina Sulfato/genética , Heparitina Sulfato/metabolismo , Humanos , Camundongos , Mutação , N-Acetilglucosaminiltransferases/genética , N-Acetilglucosaminiltransferases/metabolismo , Osteocondroma/genética , Osteocondroma/metabolismo , Osteocondroma/patologia , Retinoides , Tamoxifeno , Microtomografia por Raio-X
14.
Medicina (Kaunas) ; 59(1)2022 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-36676722

RESUMO

Background and Objectives: Hereditary multiple exostoses (HME) is a disease characterized by cartilage-capped bony protuberances at the site of growth plates of long bones. Functional mutations in the exostosin genes (EXT1 and EXT2) are reported to affect the hedgehog signalling pathways leading to multiple enchondromatosis. However, the exact role of each EXT protein in the regulation of heparan sulphate (HS) chain elongation is still an enigma. In this study, a Pakistani family with HME is investigated to find out the genetic basis of the disease. Materials and Methods: Genotyping of eight members of the family by amplifying microsatellite markers, tightly linked to the EXT1 and EXT2 genes. Results: The study revealed linkage of the HME family to the EXT1 locus 8q24.1. Sanger sequencing identified a heterozygous deletion (c.247Cdel) in exon 1 of EXT1, segregating with the disease phenotype in the family. In silico analysis predicted a shift in the frame causing an early stop codon (p.R83GfsX52). The predicted dwarf protein constituting 134 amino acids was functionally aberrant with a complete loss of the catalytic domain at the C-terminus. Interestingly, an alternative open reading frame 3 (ORF3) caused by the frame shift is predicted to encode a protein sequence, identical to the wild type and containing the catalytic domain, but lacking the first 100 amino acids of the wild-type EXT1 protein. Conclusion: Consequently, haploinsufficiency could be the cause of HME in the investigated family as the mutated copy of EXT1 is ineffective for EXT-1/2 complex formation. The predicted ORF3 protein could be of great significance in understanding several aspects of HME pathogenesis.


Assuntos
Exostose Múltipla Hereditária , Humanos , Exostose Múltipla Hereditária/genética , Exostose Múltipla Hereditária/patologia , Haploinsuficiência/genética , Paquistão , Proteínas Hedgehog/genética , Mutação , N-Acetilglucosaminiltransferases/genética , N-Acetilglucosaminiltransferases/metabolismo , Heparitina Sulfato/metabolismo , Aminoácidos/genética
15.
Am J Med Genet A ; 185(11): 3466-3475, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34477285

RESUMO

Multiple osteochondromas (MO) is a rare disorder, characterized by benign osteocartilaginous tumors (osteochondromas), arising from the perichondrium of bones. The osteochondromas increase during growth, frequently causing deformities and limitations. Our study aims to analyze the data captured by the Registry of Multiple Osteochondromas, to refine Istituto Ortopedico Rizzoli (IOR) Classification, providing a representative picture of the phenotypic manifestations throughout the lifespan. We conducted a single-institution cross-sectional study. Patients were categorized according to IOR Classification, which identifies three patients' classes on the presence/absence of deformities and/or limitations. The present dataset was compared with our previously published data, to refine the classification. Nine hundred sixty-eight patients were included: 243 children (<10 years), 136 adolescents (10-15 years), and 589 adults. Of the entire population, half patients presented at least one deformity, and one quarter reported at least one limitation. Compared with our previous study, the amount of children was more than doubled and the percentage of mild/moderate cases was notably increased, giving a better disease overview throughout the lifespan and suggesting a different cut-off for dividing Class II in subclasses. We confirmed that MO is characterized by phenotypic heterogeneity, suggesting that an early classification of the disease may offer a useful tool to follow disease pattern and evolution, to support clinical practice, and to propose timely interventions.


Assuntos
Exostose Múltipla Hereditária/genética , Osteocondroma/genética , Adolescente , Adulto , Criança , Pré-Escolar , Estudos Transversais , Exostose Múltipla Hereditária/classificação , Exostose Múltipla Hereditária/epidemiologia , Humanos , Osteocondroma/classificação , Osteocondroma/epidemiologia , Fenótipo , Adulto Jovem
16.
Biomed Res Int ; 2021: 8888948, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34409107

RESUMO

OBJECTIVES: To discuss the mutational features and their relationships with disease in a family with hereditary multiple osteochondroma (HMO) from Guangxi Province (GXBB-1 family), China. METHODS: Genomic DNA and total mRNA were extracted from peripheral blood cells of GXBB-1 family members. Whole elements of the EXT1gene and its transcript, including exons, introns, exon-intron boundaries, and coding sequence (CDS) clones, were amplified and sequenced. Allele-specific PCR was used to confirm the position and type of mutation. RESULTS: All patients from the GXBB-1 family harbored the cosegregating heterozygous c.1056+1G>A mutation located in EXT1at an exon-intron boundary. Another three single-nucleotide polymorphisms (SNPs) were also detected in the patients, including IVS2+1G>A in intron 2, c.1844 T>C [p.Pro (CCT) 614Pro (CCC)] in exon 3, and c.2534G>A [p.Glu (GAG) 844Glu (GAA)] in exon 9. The latter two SNPs were synonymous variations. CONCLUSIONS: The heterozygous c.1056+1G>A mutation cosegregated with the phenotype, indicating that it is a pathogenic mutation in the GXBB-1 family. This mutation is reported for the first time in Chinese HMO patients. IVS2+1G>A and c.2534G>A have no relationship with the occurrence of disease. However, c.1844 T>C and c.1056+1G>A are linked, and their interaction needs to be further studied. c.1844T>C is a new SNP that has not been reported internationally.


Assuntos
Povo Asiático/genética , Análise Mutacional de DNA/métodos , Exostose Múltipla Hereditária/genética , N-Acetilglucosaminiltransferases/genética , China , Feminino , Humanos , Masculino , Mutação , Linhagem , Polimorfismo de Nucleotídeo Único , Análise de Sequência de DNA
17.
J Clin Lab Anal ; 35(9): e23968, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34403521

RESUMO

BACKGROUND: Hereditary multiple exostoses (HME), also referred to as multiple osteochondromas, is an autosomal dominant skeletal disease characterized by the development of multiple overgrown benign bony tumors capped by cartilage and is associated with bone deformity, joint limitation, and short stature. Mutations in exostosin glycosyltransferase (EXT)1 and EXT2 genes, which are located on chromosomes 8q24.1 and 11p13, contribute to the pathogenesis of HME. METHODS: In the present study, a genetic analysis of a four-generation Chinese family with HME was conducted using whole-exome sequencing (WES), followed by validation using Sanger sequencing. RESULTS: A novel heterozygous frameshift mutation in exon 5 of EXT2 (c.944dupT, p.Leu316fs) was identified in all affected individuals but was not detected in any unaffected individuals. This mutation results in a frameshift that introduces a premature termination codon at position 318 (p.Leu316fs) with the ability to produce a truncated EXT2 protein that lacks the last 433 amino acids at its C-terminal to indicate a defective exostosin domain and the absence of the glycosyltransferase family 64 domain, or to lead to the degradation of mRNAs by nonsense-mediated mRNA decay, which is critical for the function of EXT2. CONCLUSION: Our results indicate that WES is effective in extending the EXT mutational spectra and is advantageous for genetic counseling and the subsequent prenatal diagnosis.


Assuntos
Povo Asiático/genética , Sequenciamento do Exoma/métodos , Exostose Múltipla Hereditária/genética , Exostose Múltipla Hereditária/patologia , Mutação da Fase de Leitura , N-Acetilglucosaminiltransferases/genética , Adulto , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Adulto Jovem
18.
Genet Test Mol Biomarkers ; 25(7): 478-485, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34280007

RESUMO

Background: Multiple osteochondroma (MO), an autosomal dominant genetic disease, is caused by heterozygous mutations in the EXT1 and EXT2 genes. Approximately 80% of pathogenic mutations are nonsense/missense mutations, small indels, and splicing mutations. Splicing mutations, particularly at the 3' and 5' splice sites, disrupt normal mRNA processing and cause exon skipping or aberrant splicing, ultimately resulting in protein truncation and loss of function. Methods: Polymerase chain reaction (PCR) and Sanger sequencing were applied to detect subtle mutations in a Chinese family with MO, the pathogenicity of a splicing variant was predicted by bioinformatics and further verified using a minigene splicing assay. Results: A novel and heterozygous splicing mutation, c.626 + 2_626 + 5delTAGG, was identified in the EXT2 gene of the proband and the father by PCR and Sanger sequencing, whereas the unaffected mother and brother had wild-type alleles at the same site. Bioinformatics predicted that the 5' splicing site of exon 3 in the EXT2 gene was destroyed due to this mutation. A hybrid minigene splicing assay (HMSA) indicated that the mutation disturbed the normal splicing of the EXT2 gene mRNA and led to a deletion of 79 bp at the 5' end of exon 3, which resulted in aberrant splicing of exon 3 and introduced an earlier stop codon in the EXT2 gene. Conclusion: A novel splicing mutation was identified that produced the MO phenotype through aberrant splicing in a Chinese family. This observation, expands our knowledge of the spectrum of molecular pathogenic mechanisms leading to aberrant mRNA splicing.


Assuntos
Exostose Múltipla Hereditária/genética , N-Acetilglucosaminiltransferases/genética , Adulto , Alelos , Povo Asiático/genética , China , Éxons/genética , Exostose Múltipla Hereditária/metabolismo , Feminino , Humanos , Íntrons/genética , Masculino , Mutação/genética , Mutação de Sentido Incorreto , N-Acetilglucosaminiltransferases/metabolismo , Linhagem , Fenótipo , Sítios de Splice de RNA/genética , Splicing de RNA/genética
19.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 38(6): 549-552, 2021 Jun 10.
Artigo em Chinês | MEDLINE | ID: mdl-34096023

RESUMO

OBJECTIVE: To explore the genetic basis for a pedigree affected with hereditary multiple osteochondroma (HMO). METHODS: Peripheral blood samples were collected from the proband and members of his pedigree with informed consent. Following extraction of genomic DNA, all coding exons and flanking intronic sequences (-10 bp) of the EXT1 and EXT2 genes were subjected to targeted capture and next generation sequencing (NGS). Suspected variant was verified by Sanger sequencing. RESULTS: A heterozygous nonsense variant (c.1911C>A) was found in exon 10 of the EXT1 gene in the proband and his affected father but not in a healthy sister and normal controls. The variant was classified as a pathogenic based on the guidelines of the American College of Medical Genetics and Genomics (PVS1+PM2+PP1). Bioinformatic analysis predicted that the c.1911C>A variant may be disease-causing via nonsense-mediated mRNA decay and anomalous splicing. CONCLUSION: The c.1911C>A variant probably underlay the disease in this pedigree. Discovery of this variant enriched the variant spectrum of HMO.


Assuntos
Exostose Múltipla Hereditária , Códon sem Sentido , Éxons/genética , Exostose Múltipla Hereditária/genética , Heterozigoto , Humanos , Linhagem
20.
Genet Test Mol Biomarkers ; 25(5): 361-367, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-34003695

RESUMO

Background: Multiple osteochondromas (MO) are an autosomal-dominant disease characterized by the growth of multiple cartilage-capped prominences in the growth plate region of the metaphysis in long and flat bones. Materials and Methods: To detect genetic mutations related to MO, a three-generation Chinese family with MO was evaluated using whole exome sequencing for mutation screening. The candidate pathogenic mutation was validated by Sanger sequencing. Results: A novel frameshift (NM_000401.3:c.1321del:p.Leu441TrpfsTer28) in exon 8 of the exotosin 2 (EXT2) gene was identified in two affected individuals. Codons 441 and 468 in the EXT2 gene are highly conserved among vertebrates as demonstrated by multiple sequence alignment. The c.1321 del C resulted in an amino acid change at codon 441, which generated a premature stop codon at position 468, causing complete loss of the glycosyltransferase domain. Conclusions: A novel frameshift mutation c.1321delC detected in the EXT2 gene may help in prenatal genetic screening and early diagnosis of MO.


Assuntos
Exostose Múltipla Hereditária/genética , N-Acetilglucosaminiltransferases/genética , Adulto , Povo Asiático/genética , Pré-Escolar , China , Códon , Éxons , Feminino , Mutação da Fase de Leitura , Predisposição Genética para Doença/genética , Genótipo , Humanos , Masculino , Mutação , N-Acetilglucosaminiltransferases/metabolismo , Linhagem , Polimorfismo de Nucleotídeo Único/genética , Fatores de Risco , Sequenciamento do Exoma
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...