Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 234
Filter
2.
Skeletal Radiol ; 52(7): 1369-1375, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36648521

ABSTRACT

PURPOSE: To assess the prevalence of intraosseous cartilaginous lesions in patients with multiple osteochondromas based on total-body (TB) MRI examinations, used for screening purposes. SUBJECTS AND METHODS: Between 2013 and 2020, TB-MRI examinations were performed in 366 patients with proven multiple osteochondromas syndrome, to rule out malignant progression. For this study, presence, or absence of intraosseous central or eccentrical chondroid lesions, defined as lobulated lesions with low signal intensity on T1-weighted images, replacing bone marrow and high signal intensity equal to fluid on T2-weighted images in the bone marrow of the meta-diaphysis of (one of) the long bones, were recorded in the long bones as part of a TB-MRI protocol. RESULTS: In 62 patients out of the 366 MO patients (17%), one or more intraosseous chondroid lesions (either enchondroma or atypical cartilaginous tumor) were detected. The age of the patients at time of diagnosis ranged from 17 to 61 years (mean, 36). Size of the lesions varied from 4 to 69 mm (mean, 16.3 mm). The most common location was the proximal femur (n = 29), followed by the distal femur and proximal humerus (n = 18 and n = 10, respectively). In nine of the patients with an intraosseous chondroid lesion, a second and/or third TB-MRI were available during the period of evaluation (mean interval, 2.7 years between the exams). In none of these patients increase of these intraosseous lesions was noticed. CONCLUSION: Intraosseous chondroid lesions (enchondroma and ACT) appear to occur more frequently in MO patients than in the general population. TB-MRI allows to detect these, besides the identification of OC with suspicious features.


Subject(s)
Bone Neoplasms , Chondroma , Exostoses, Multiple Hereditary , Humans , Adolescent , Young Adult , Adult , Middle Aged , Exostoses, Multiple Hereditary/pathology , Bone Neoplasms/diagnostic imaging , Chondroma/diagnostic imaging , Chondroma/pathology , Epiphyses/pathology , Magnetic Resonance Imaging/methods
3.
Mol Med Rep ; 25(4)2022 04.
Article in English | MEDLINE | ID: mdl-35211766

ABSTRACT

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.


Subject(s)
Exostoses, Multiple Hereditary , N-Acetylglucosaminyltransferases/genetics , China , Exostoses, Multiple Hereditary/diagnosis , Exostoses, Multiple Hereditary/genetics , Exostoses, Multiple Hereditary/pathology , Female , Gene Deletion , Humans , Male , Mutation , Pedigree
4.
J Orthop Res ; 40(10): 2391-2401, 2022 10.
Article in English | MEDLINE | ID: mdl-34996123

ABSTRACT

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.


Subject(s)
Bone Neoplasms , Exostoses, Multiple Hereditary , Glucuronidase , N-Acetylglucosaminyltransferases , Osteochondroma , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Child , Disease Models, Animal , Exostoses, Multiple Hereditary/genetics , Exostoses, Multiple Hereditary/metabolism , Exostoses, Multiple Hereditary/pathology , Glucuronidase/genetics , Glucuronidase/metabolism , Heparitin Sulfate/genetics , Heparitin Sulfate/metabolism , Humans , Mice , Mutation , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Osteochondroma/genetics , Osteochondroma/metabolism , Osteochondroma/pathology , Retinoids , Tamoxifen , X-Ray Microtomography
5.
Medicina (Kaunas) ; 59(1)2022 Dec 31.
Article in English | MEDLINE | ID: mdl-36676722

ABSTRACT

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.


Subject(s)
Exostoses, Multiple Hereditary , Humans , Exostoses, Multiple Hereditary/genetics , Exostoses, Multiple Hereditary/pathology , Haploinsufficiency/genetics , Pakistan , Hedgehog Proteins/genetics , Mutation , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism , Heparitin Sulfate/metabolism , Amino Acids/genetics
6.
J Clin Lab Anal ; 35(9): e23968, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34403521

ABSTRACT

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.


Subject(s)
Asian People/genetics , Exome Sequencing/methods , Exostoses, Multiple Hereditary/genetics , Exostoses, Multiple Hereditary/pathology , Frameshift Mutation , N-Acetylglucosaminyltransferases/genetics , Adult , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Pedigree , Young Adult
7.
Am J Clin Pathol ; 156(4): 513-520, 2021 Sep 08.
Article in English | MEDLINE | ID: mdl-33769443

ABSTRACT

OBJECTIVES: This study investigates a unique case of multiple osteochondromas (MO) comorbid with enlarged parietal foramina and correlates the findings with the existing literature. The aim of this study is to provide a deeper understanding of anatomic variation for physicians. METHODS: A 66-year-old White male donor was examined during a routine cadaveric dissection performed by medical students in an anatomy laboratory. Detailed exploration of the skeleton and organs was performed, and photographs were taken. Tissue samples were obtained from multiple outgrowths, and histopathologic examination was done. RESULTS: Bilateral bony growths were noted rising from the long bones of the upper and lower extremities (femur, tibia, fibula, and radius). An accessory muscle was found to be associated with the left radial bony growth. Histopathologic examination was positive for osteochondroma. Inspection of the skull revealed enlarged parietal foramina. Other findings included tibiofibular synostosis, abnormally shaped vertebral bodies and ribs, and elongated styloid processes of the skull. CONCLUSIONS: In combination with the histopathologic examination, the case report and literature review elucidate a more precise clinical picture for those affected with MO or similar disorders. This report also emphasizes the necessity of further investigation of the pathogenesis of MO and Potocki-Shaffer syndrome.


Subject(s)
Chromosome Disorders/diagnosis , Encephalocele/diagnosis , Exostoses, Multiple Hereditary/diagnosis , Ossification, Heterotopic/diagnosis , Temporal Bone/abnormalities , Aged , Ankle Joint/pathology , Chromosome Deletion , Chromosome Disorders/pathology , Chromosomes, Human, Pair 11 , Encephalocele/pathology , Exostoses, Multiple Hereditary/pathology , Femur/pathology , Fibula/pathology , Humans , Male , Ossification, Heterotopic/pathology , Radius/pathology , Temporal Bone/pathology , Tibia/pathology
8.
Am J Med Genet A ; 185(3): 856-865, 2021 03.
Article in English | MEDLINE | ID: mdl-33305909

ABSTRACT

Stuve-Wiedemann syndrome (SWS; MIM 601559) is a rare autosomal recessive disease caused by mutations in the leukemia inhibitor factor receptor gene (LIFR). Common clinical and radiological findings are often observed, and high neonatal mortality occurs due to respiratory distress and hyperthermic episodes. Despite initially considered as a lethal disorder during the newborn period, in recent years, several SWS childhood survivors have been reported. We report a detailed clinical and radiological characterization of four unrelated childhood SWS molecularly confirmed patients and review 22 previously reported childhood surviving cases. We contribute to the definition of the childhood survival phenotype of SWS, emphasizing the evolving phenotype, characterized by skeletal abnormalities with typical radiological findings, distinctive dysmorphic features, and dysautonomia. Based on the typical features and clinical course, early diagnosis is possible and crucial to plan appropriate management and prevent potential complications. Genetic confirmation is advisable in order to improve genetic counseling to the patients and their families.


Subject(s)
Abnormalities, Multiple/genetics , Bone Diseases, Developmental/genetics , Exostoses, Multiple Hereditary/diagnostic imaging , Leukemia Inhibitory Factor Receptor alpha Subunit/genetics , Osteochondrodysplasias/diagnostic imaging , Bone Diseases, Developmental/diagnostic imaging , Bone Diseases, Metabolic/genetics , Child, Preschool , Consanguinity , Developmental Disabilities/genetics , Dysautonomia, Familial/genetics , Exostoses, Multiple Hereditary/genetics , Exostoses, Multiple Hereditary/pathology , Female , Genotype , Humans , Infant , Infant, Newborn , Leukemia Inhibitory Factor Receptor alpha Subunit/deficiency , Male , Muscle Hypotonia/genetics , Osteochondrodysplasias/genetics , Osteochondrodysplasias/pathology , Phenotype , Roma/genetics , Survivors
9.
J Appl Genet ; 61(4): 571-573, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32910413

ABSTRACT

Stüve-Wiedemann syndrome (SWS) is a rare genetic disorder characterized by skeletal dysplasia and severe dysautonomia, evidencing a difficult airway approach and likely increased malignant hyperthermia susceptibility. Developmental dysmorphism classically worsens with age, therefore translating in a poor prognosis. In this article, we describe a case of a 27-year-old woman diagnosed with SWS proposed for abscess drainage under dissociative anesthesia. This patient has outlived the life expectancy described for SWS, acknowledging the importance of reporting this rare adult clinical case in what SWS anesthetic management is concerned.


Subject(s)
Abnormalities, Multiple/therapy , Anesthesia/methods , Anesthetics, Dissociative/administration & dosage , Exostoses, Multiple Hereditary/therapy , Osteochondrodysplasias/therapy , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Adult , Exostoses, Multiple Hereditary/genetics , Exostoses, Multiple Hereditary/pathology , Female , Humans , Osteochondrodysplasias/genetics , Osteochondrodysplasias/pathology , Primary Dysautonomias/genetics , Primary Dysautonomias/pathology , Primary Dysautonomias/therapy
10.
Pediatr Dermatol ; 37(2): 381-382, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31975458

ABSTRACT

Stüve-Wiedemann syndrome is a rare autosomal recessive congenital primary skeletal dysplasia, characterized by small stature, bowed long bones, joint restrictions, hyperthermic episodes, dysautonomia, and respiratory and feeding difficulties, that usually leads to early mortality. Cutaneous manifestations have rarely been reported. We report the case of a girl with Stüve-Wiedemann syndrome presenting with progressive development of multiple eruptive vellus hair cysts.


Subject(s)
Epidermal Cyst/etiology , Exostoses, Multiple Hereditary/complications , Exostoses, Multiple Hereditary/pathology , Hair Diseases/etiology , Oral Ulcer/etiology , Osteochondrodysplasias/complications , Osteochondrodysplasias/pathology , Tongue Diseases/etiology , Child , Epidermal Cyst/pathology , Female , Hair Diseases/pathology , Humans , Oral Ulcer/pathology , Tongue Diseases/pathology
12.
Skeletal Radiol ; 49(1): 115-124, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31273432

ABSTRACT

OBJECTIVE: To analyze the results of annual screening using whole-body magnetic resonance imaging (WBMRI) in patients with multiple hereditary exostoses (MHE) and enchondromatosis (EC), and estimate the risk for transformation to chondrosarcoma (CS) in these disorders. MATERIALS AND METHODS: A total of 62 patients (57 with MHE and five with EC) screened during a mean follow-up period of 4.6 years (range, 1-10 years) using 253 WBMRIs (median four WBMRIs per patient, range, 1-10) were analyzed retrospectively. The time of WBMRIs was compared with dates for diagnosed CSs. A supplementary literature review was performed focusing on the risk of malignant transformation. RESULTS: Ten patients had CS before being enrolled in the screening program, nine with MHE and one with EC. Three asymptomatic CSs were detected by screening; one in a patient with EC and two in patients with MHE, one of whom had CS previously. During the screening period, there was no occurrence of CS not detected by WBMRI in the study group. Histopathologically, the CSs were predominantly grade 1 and were, except for in two patients, located at the truncus, proximal femur, and shoulder girdle. Based on the current material and literature review, the risk of CS seems to be in the range of 2-3.7% for MHE and up to 50% for EC patients. CONCLUSIONS: MRI may be used as a screening method detecting malignant transformation in MHE and EC patients, but the efficacy has to be confirmed in long-term follow-up studies including cost analysis.


Subject(s)
Bone Neoplasms/diagnostic imaging , Cell Transformation, Neoplastic/pathology , Chondrosarcoma/diagnostic imaging , Enchondromatosis/diagnostic imaging , Exostoses, Multiple Hereditary/diagnostic imaging , Magnetic Resonance Imaging/methods , Medical Audit , Whole Body Imaging , Adolescent , Adult , Aged , Bone Neoplasms/pathology , Child , Chondrosarcoma/pathology , Early Detection of Cancer , Enchondromatosis/pathology , Exostoses, Multiple Hereditary/pathology , Female , Humans , Male , Middle Aged , Young Adult
13.
Eur J Med Genet ; 63(1): 103633, 2020 Jan.
Article in English | MEDLINE | ID: mdl-30797056

ABSTRACT

Potocki-Shaffer syndrome (PSS) is a rare contiguous gene deletion syndrome caused by heterozygous deletion of 11p11.2p12. Typical features described in patients with PSS include developmental delay, intellectual disability, multiple cartilaginous exostoses, biparietal foramina, craniofacial abnormalities, and genitourinary anomalies. While hypertension has been noted in three patients with PSS, it has not been described in most patients with this syndrome. This report details the evaluation and treatment of a teenager with PSS who presented on several occasions during childhood with elevated blood pressure measurements. The renin level was elevated, likely indicating a secondary cause for the HTN. The patient's BP responded to monotherapy with Angiotensin Converting Enzyme Inhibitor (ACEI).


Subject(s)
Chromosome Disorders/genetics , Developmental Disabilities/genetics , Exostoses, Multiple Hereditary/genetics , Exostoses/genetics , Hypertension/genetics , Adolescent , Chromosome Deletion , Chromosome Disorders/blood , Chromosome Disorders/complications , Chromosome Disorders/pathology , Chromosomes, Human, Pair 11/genetics , Developmental Disabilities/blood , Developmental Disabilities/complications , Developmental Disabilities/pathology , Exostoses/blood , Exostoses/complications , Exostoses/pathology , Exostoses, Multiple Hereditary/blood , Exostoses, Multiple Hereditary/complications , Exostoses, Multiple Hereditary/pathology , Female , Heterozygote , Humans , Hypertension/blood , Hypertension/complications , Hypertension/pathology , Phenotype , Renin/blood , Sequence Deletion/genetics
14.
Medicine (Baltimore) ; 98(48): e18175, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31770268

ABSTRACT

RATIONALE: Hereditary multiple exostoses (HME) is an autosomal dominant disease that causes multiple exostoses throughout the body. It usually occurs around the metaphysis of the long bones, and when it involves the hip, symptoms arise due to deformity and the mass effect. If the lesion does not involve the joint or is not associated with arthritis, symptoms can be relieved by surgical excision of the osteochondroma. However, if secondary osteoarthritis (OA) or subluxation of the joint has progressed, joint replacement arthroplasty should be considered. PATIENT CONCERNS: A 57-year-old woman with HME visited our outpatient department with severe right hip pain. She complained of difficulty walking and severe discomfort during activities of daily living. She was short in stature and had a family history of HME. DIAGNOSIS: A physical examination revealed limited motion in the hip joint and a limb length discrepancy. Plain radiography and a computed tomography scan revealed huge osteochondromas on bilateral proximal femurs and advanced OA with subluxation of the right hip joint. INTERVENTIONS: Cementless total hip arthroplasty of the right hip joint via the modified posterolateral approach was done. OUTCOMES: The patient showed good clinical scores and functional improvement at the 2-year follow-up. LESSONS: Total hip arthroplasty for an anatomically deformed joint is technically difficult, and there are many factors to consider that can make surgeons reluctant to use this modality. However, with careful preparation, arthroplasty is a good surgical option for symptomatic and functional recovery in HME patients with hip joint involvement.


Subject(s)
Arthroplasty, Replacement, Hip/methods , Bone Neoplasms , Exostoses, Multiple Hereditary , Femur , Hip Joint , Joint Dislocations , Bone Neoplasms/complications , Bone Neoplasms/pathology , Bone Neoplasms/surgery , Exostoses, Multiple Hereditary/complications , Exostoses, Multiple Hereditary/pathology , Exostoses, Multiple Hereditary/surgery , Female , Femur/diagnostic imaging , Femur/pathology , Hip Joint/diagnostic imaging , Hip Joint/surgery , Humans , Joint Dislocations/diagnosis , Joint Dislocations/etiology , Joint Dislocations/surgery , Middle Aged , Radiography/methods , Recovery of Function , Treatment Outcome
15.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 36(8): 757-760, 2019 Aug 10.
Article in Chinese | MEDLINE | ID: mdl-31400121

ABSTRACT

OBJECTIVE: To identify pathogenic variations of EXT1 and EXT2 genes in two Chinese pedigrees affected with hereditary multiple exostosis (HME). METHODS: Genomic DNA was extracted from peripheral blood samples using a phenol-chloroform method. PCR and Sanger sequencing was conducted to amplify the exons and the flanking intronic regions of the EXT1 and EXT2 genes. RESULTS: DNA sequencing has revealed a heterozygous missense variation c.812A>G (p.Tyr271Cys) in the exon 1 of EXT1 in pedigree 1, and a heterozygous frameshift variation c.1431dup (p.Ser478Leufs*43) in the exon 6 of EXT1 in the proband from pedigree 2. Both variations have co-segregated with the disease phenotype, which was also consistent with previous report. CONCLUSION: Two heterozygous pathogenic variations underlying HME have been identified. The result has facilitated genetic counseling and prenatal diagnosis for the affected pedigrees.


Subject(s)
Exostoses, Multiple Hereditary/genetics , N-Acetylglucosaminyltransferases/genetics , Asian People , Base Sequence , DNA Mutational Analysis , Exostoses, Multiple Hereditary/pathology , Frameshift Mutation , Humans , Mutation, Missense , Pedigree
17.
Acta Vet Scand ; 61(1): 32, 2019 Jun 25.
Article in English | MEDLINE | ID: mdl-31238951

ABSTRACT

BACKGROUND: Multiple cartilaginous exostoses are a rare, benign, proliferative condition of cartilage and bone. They can be asymptomatic, or they may cause pain, lameness, paresis and even paralysis, depending on their location and size. In cases of spinal cord or nerve root compression, surgery is the treatment of choice. Therefore, an advanced imaging diagnostic work-up is indicated. Due to the unclear pathophysiology and progression of this condition, it is difficult to predict its prognosis. CASE PRESENTATION: A 9-month-old female Swiss Mountain dog was presented with a history of gait abnormalities, kyphosis and hypersensitivity consistent with a thoracolumbar myelopathy. Multiple calcified masses, most prominent at the Th7-Th9 level and the L2-L3 level, were observed. Magnetic resonance imaging of the thoracolumbar vertebral column revealed severe dorsal spinal cord compressions near the dorsal arch of the Th7-Th9 and L2-L3 vertebrae. Two of these masses were removed surgically. The successful removal of both masses was confirmed by postoperative computed tomography. The histopathological examination of the resected tissue revealed multiple cartilaginous exostoses. The first neurological and magnetic resonance follow up examination carried out 6 months postoperatively showed improvement of the clinical status. At that time, no mass regrowth was observed. The last follow up neurological examination carried out 15 months postoperatively showed gait improvement and resolution of pain. CONCLUSION: This is the first case report of multiple cartilaginous exostoses with a complete pre- and postoperative evaluation and a 15 month follow-up.


Subject(s)
Dog Diseases/diagnosis , Dog Diseases/pathology , Exostoses, Multiple Hereditary/veterinary , Spinal Cord Compression/veterinary , Spinal Cord Diseases/veterinary , Animals , Dog Diseases/surgery , Dogs , Exostoses, Multiple Hereditary/complications , Exostoses, Multiple Hereditary/diagnosis , Exostoses, Multiple Hereditary/pathology , Female , Spinal Cord Compression/etiology , Spinal Cord Compression/surgery , Spinal Cord Diseases/diagnosis , Spinal Cord Diseases/etiology , Spinal Cord Diseases/surgery , Treatment Outcome
18.
Mol Genet Genomic Med ; 7(3): e00560, 2019 03.
Article in English | MEDLINE | ID: mdl-30632316

ABSTRACT

BACKGROUND: We describe a patient presenting with pachygyria, epilepsy, developmental delay, short stature, failure to thrive, facial dysmorphisms, and multiple osteochondromas. METHODS: The patient underwent extensive genetic testing and analysis in an attempt to diagnose the cause of his condition. Clinical testing included metaphase karyotyping, array comparative genomic hybridization, direct sequencing and multiplex ligation-dependent probe amplification and trio-based exome sequencing. Subsequently, research-based whole transcriptome sequencing was conducted to determine whether it might shed light on the undiagnosed phenotype. RESULTS: Clinical exome sequencing of patient and parent samples revealed a maternally inherited splice-site variant in the doublecortin (DCX) gene that was classified as likely pathogenic and diagnostic of the patient's neurological phenotype. Clinical array comparative genome hybridization analysis revealed a 16p13.3 deletion that could not be linked to the patient phenotype based on affected genes. Further clinical testing to determine the cause of the patient's multiple osteochondromas was unrevealing despite extensive profiling of the most likely causative genes, EXT1 and EXT2, including mutation screening by direct sequence analysis and multiplex ligation-dependent probe amplification. Whole transcriptome sequencing identified a SAMD12-EXT1 fusion transcript that could have resulted from a chromosomal deletion, leading to the loss of EXT1 function. Re-review of the clinical array comparative genomic hybridization results indicated a possible unreported mosaic deletion affecting the SAMD12 and EXT1 genes that corresponded precisely to the introns predicted to be affected by a fusion-causing deletion. The existence of the mosaic deletion was subsequently confirmed clinically by an increased density copy number array and orthogonal methodologies CONCLUSIONS: While mosaic mutations and deletions of EXT1 and EXT2 have been reported in the context of multiple osteochondromas, to our knowledge, this is the first time that transcriptomics technologies have been used to diagnose a patient via fusion transcript analysis in the congenital disease setting.


Subject(s)
Exostoses, Multiple Hereditary/genetics , Gene Fusion , N-Acetylglucosaminyltransferases/genetics , Nerve Tissue Proteins/genetics , Child , Exostoses, Multiple Hereditary/pathology , Gene Deletion , Humans , Male , RNA, Messenger/genetics , Sterile Alpha Motif/genetics
19.
Int J Oncol ; 54(3): 859-868, 2019 03.
Article in English | MEDLINE | ID: mdl-30664192

ABSTRACT

The molecular mechanism of hereditary multiple exostoses (HME) remains ambiguous and a limited number of studies have investigated the pathogenic mechanism of mutations in patients with HME. In the present study, a novel heterozygous splice mutation (c.1284+2del) in exostosin glycosyltransferase 1 (EXT1) gene was identified in a three­generation family with HME. Bioinformatics and TA clone­sequencing indicated that the splice site mutation would result in exon 4 skipping. Reverse transcription­quantitative polymerase chain reaction (RT­qPCR) revealed that the expression levels of wild­type EXT1/EXT2 mRNA in patients with HME were significantly decreased, compared with normal control participants (P<0.05). Abnormal EXT1 transcript lacking exon 4 (EXT1­DEL) and full­length EXT1 mRNA (EXT1­FL) were overexpressed in 293­T cells and Cos­7 cells using lentivirus infection. RT­qPCR demonstrated that the expression level of EXT1­DEL was significantly increased, compared with EXT1­FL (17.032 vs. 6.309, respectively; P<0.05). The protein encoded by EXT1­DEL was detected by western blot analysis, and the level was increased, compared with EXT1­FL protein expression. Immunofluorescence indicated that the protein encoded by EXT1­DEL was located in the cytoplasm of Cos­7 cells, which was consistent with the localization of the EXT1­FL protein. In conclusion, the present study identified a novel splice mutation that causes exon 4 skipping during mRNA splicing and causes reduced expression of EXT1/EXT2. The mutation in EXT1­DEL generated a unique peptide that is located in the cytoplasm in vitro, and it expands the mutation spectrum and provides molecular genetic evidence for a novel pathogenic mechanism of HME.


Subject(s)
Exostoses, Multiple Hereditary/genetics , N-Acetylglucosaminyltransferases/genetics , RNA Splicing/genetics , Adult , Aged , Cell Line, Transformed , Cytoplasm/metabolism , Exostoses, Multiple Hereditary/metabolism , Exostoses, Multiple Hereditary/pathology , Female , Gene Expression , Genetic Association Studies , Genetic Testing , Humans , Male , Middle Aged , Mutation , N-Acetylglucosaminyltransferases/metabolism , RNA Splice Sites/genetics , RNA, Messenger/metabolism
20.
Eur J Hum Genet ; 27(3): 378-383, 2019 03.
Article in English | MEDLINE | ID: mdl-30487643

ABSTRACT

Potocki-Shaffer syndrome (PSS) is a contiguous gene syndrome caused by 11p11.2 deletions. PSS is clinically characterized by intellectual disability, craniofacial anomalies, enlarged parietal foramina, and multiple exostoses. PSS occasionally shows autism spectrum disorder, epilepsy, and overgrowth. Some of the clinical features are thought to be associated with haploinsufficiency of two genes in the 11p11.2 region; variants affecting the function of ALX4 cause enlarged parietal foramina and EXT2 lead to multiple exostoses. However, the remaining clinical features were still yet to be linked to specific genetic alterations. In this study, we identified de novo truncating variants in an 11p11.2 gene, PHF21A, in three cases with intellectual disability and craniofacial anomalies. Among these three cases, autism spectrum disorder was recognized in one case, epilepsy in one case, and overgrowth in two cases. This study shows that PHF21A haploinsufficiency results in intellectual disability and craniofacial anomalies and possibly contributes to susceptibility to autism spectrum disorder, epilepsy, and overgrowth, all of which are PSS features.


Subject(s)
Autism Spectrum Disorder/genetics , Chromosome Disorders/genetics , Craniofacial Abnormalities/genetics , Epilepsy/genetics , Exostoses, Multiple Hereditary/genetics , Histone Deacetylases/genetics , Intellectual Disability/genetics , Autism Spectrum Disorder/pathology , Child , Child, Preschool , Chromosome Deletion , Chromosome Disorders/pathology , Chromosomes, Human, Pair 11/genetics , Craniofacial Abnormalities/pathology , Epilepsy/pathology , Exostoses, Multiple Hereditary/pathology , Haploinsufficiency , Humans , Intellectual Disability/pathology , Male , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL