1
|
Tremosini M, Morri M, Forni C, Pedrini E, Mordenti M, Gnoli M, Di Cecco A, Moroni A, Sangiorgi L. Pain in patients with multiple inherited osteochondromas: Incidence and potential prognostic factors. A retrospective cohort study. J Bone Oncol 2025; 52:100672. [PMID: 40206490 PMCID: PMC11978362 DOI: 10.1016/j.jbo.2025.100672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
Purpose the purpose of this study was to describe the baseline characteristics, presenting phenotype and treatment interventions for patients diagnosed with multiple osteochondromas who presented with severe pain symptoms. Methods a retrospective single-centre cohort study was conducted at a Rare Skeletal Disorders Department. Pain symptomatology was measured at the first visit, pain level was reported, varying from 0, absence of pain to 10, maximum pain. Baseline characteristics, pathology phenotype using IOR classification and treatments performed/ongoing as medical, surgical and conservative therapies were collected. Results a total of 152 patients were enrolled, with a median pain score of 0 and the 25th and 75th percentiles of 0 and 4, respectively. A percentage of 25.7 % (95 % CI of 19.3-33.3) presented at the first visit with moderate/severe pain. Multiple logistic regression confirmed that age was the only factor to be significantly associated with moderate/severe pain and IOR classification was not able to provide a description of the pathology that was associated with a major pain score. Conclusion from the early stages of multiple osteochondromas diagnosis, pain symptoms must be carefully assessed. An increase in age is associated with a worsening of pain; IOR classification of the multiple osteochondromas phenotype does not currently allow an association between the various classes and pain. A re-evaluation of the classification in this light could be an important new element for clinical practice.
Collapse
Affiliation(s)
- Morena Tremosini
- Servizio di Assistenza Infermieristica Tecnica e Riabilitativa, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Mattia Morri
- Servizio di Assistenza Infermieristica Tecnica e Riabilitativa, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Cristiana Forni
- Servizio di Assistenza Infermieristica Tecnica e Riabilitativa, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Elena Pedrini
- Struttura Complessa Malattie Rare Scheletriche, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Marina Mordenti
- Struttura Complessa Malattie Rare Scheletriche, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Maria Gnoli
- Struttura Complessa Malattie Rare Scheletriche, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Alessia Di Cecco
- Servizio di Assistenza Infermieristica Tecnica e Riabilitativa, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Alice Moroni
- Struttura Complessa Malattie Rare Scheletriche, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Luca Sangiorgi
- Struttura Complessa Malattie Rare Scheletriche, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| |
Collapse
|
2
|
Peng W, Li GF, Lin GW, Cheng XX, Zuo XY, Lin QH, Liu SQ, Li DJ, Lin DC, Yin JQ, Luo CL, Zhang YY, Xie XB, Bei JX. Identification of novel germline mutations in FUT7 and EXT1 linked with hereditary multiple exostoses. Oncogene 2025; 44:835-848. [PMID: 39690272 DOI: 10.1038/s41388-024-03254-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/29/2024] [Accepted: 12/05/2024] [Indexed: 12/19/2024]
Abstract
Hereditary multiple exostoses (HME) is an autosomal dominant skeletal disorder primarily linked with mutations in Exostosin-1 (EXT1) and Exostosin-2 (EXT2) genes. However, not all HME cases can be explained by these mutations, and its pathogenic mechanisms are not fully understood. Herein, utilizing whole-exome sequencing and genetic screening with a family trio design, we identify two novel rare mutations co-segregating with HME in a Chinese family, including a nonsense mutation (c.204G>A, p.Trp68*) in EXT1 and a missense mutation (c.893T>G, p.Phe298Cys) in FUT7. Functional assays reveal that the FUT7 mutation affects the cellular localization of FUT7 protein and regulates cell proliferation. Notably, the simultaneous loss of fut7 and ext1 in a zebrafish model results in severe chondrodysplasia, indicating a functional link between FUT7 and EXT1 in chondrocyte regulation. Additionally, we unveil that FUT7 p.Phe298Cys reduces EXT1 expression through IL6/STAT3/SLUG axis at the transcription level and through ubiquitination-related proteasomal degradation at the protein level. Together, our findings not only identify novel germline mutations in FUT7 and EXT1 genes, but also highlight the critical interaction between these genes, suggesting a potential 'second-hit' mechanism over EXT1 mutations in HME pathogenesis. This insight enhances our understanding of the mechanisms underlying HME and opens new avenues for potential therapeutic interventions.
Collapse
Affiliation(s)
- Wan Peng
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, P. R. China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Blood Tranfusion Department, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, 510440, P. R. China
| | - Gao-Fei Li
- Innovation Centre of Ministry of Education for Development and Diseases, School of medicine, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Guo-Wang Lin
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xi-Xi Cheng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xiao-Yu Zuo
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Qiao-Hong Lin
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Shu-Qiang Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - De-Jun Li
- Prenatal Diagnosis Center, Reproductive Medicine Center, The First Hospital of Jilin University, Changchun, 130021, P. R. China
| | - Dao-Chao Lin
- Department of Orthopaedics, Shulan (Hangzhou) Hospital, Hangzhou, 311000, P. R. China
| | - Jun-Qiang Yin
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, P. R. China
| | - Chun-Ling Luo
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yi-Yue Zhang
- Innovation Centre of Ministry of Education for Development and Diseases, School of medicine, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Xian-Biao Xie
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, P. R. China.
| | - Jin-Xin Bei
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, 510095, P. R. China.
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
- Department of Medical Oncology, National Cancer Centre of Singapore, Singapore, Singapore.
| |
Collapse
|
3
|
Frappaolo A, Zaccagnini G, Giansanti MG. GOLPH3-mTOR Crosstalk and Glycosylation: A Molecular Driver of Cancer Progression. Cells 2025; 14:439. [PMID: 40136688 PMCID: PMC11941073 DOI: 10.3390/cells14060439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/06/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
Originally identified in proteomic-based studies of the Golgi, Golgi phosphoprotein 3 (GOLPH3) is a highly conserved protein from yeast to humans. GOLPH3 localizes to the Golgi through the interaction with phosphatidylinositol-4-phosphate and is required for Golgi architecture and vesicular trafficking. Many studies revealed that the overexpression of GOLPH3 is associated with tumor metastasis and a poor prognosis in several cancer types, including breast cancer, glioblastoma multiforme, and colon cancer. The purpose of this review article is to provide the current progress of our understanding of GOLPH3 molecular and cellular functions, which may potentially reveal therapeutic avenues to inhibit its activity. Specifically, recent papers have demonstrated that GOLPH3 protein functions as a cargo adaptor for COP I-coated intra Golgi vesicles and impinges on Golgi glycosylation pathways. In turn, GOLPH3-dependent defects have been associated with malignant phenotypes in cancer cells. Additionally, the oncogenic activity of GOLPH3 has been linked with enhanced signaling downstream of mechanistic target of rapamycin (mTOR) in several cancer types. Consistent with these data, GOLPH3 controls organ growth in Drosophila by associating with mTOR signaling proteins. Finally, compelling evidence demonstrates that GOLPH3 is essential for cytokinesis, a process required for the maintenance of genomic stability.
Collapse
Affiliation(s)
| | | | - Maria Grazia Giansanti
- Istituto di Biologia e Patologia Molecolari del CNR, c/o Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, 00185 Roma, Italy; (A.F.); (G.Z.)
| |
Collapse
|
4
|
Rueda-de-Eusebio A, Gomez-Pena S, Moreno-Casado MJ, Marquina G, Arrazola J, Crespo-Rodríguez AM. Hereditary multiple exostoses: an educational review. Insights Imaging 2025; 16:46. [PMID: 39982564 PMCID: PMC11845651 DOI: 10.1186/s13244-025-01899-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/02/2025] [Indexed: 02/22/2025] Open
Abstract
Hereditary multiple exostoses (HME), an autosomal dominant disorder with an incidence of 1:50,000 to 1:100,000, is characterised by the formation of multiple osteochondromas arising from the metaphyses of long and flat bones. These osteochondromas often present as painless palpable lumps, though some cases are symptomatic due to mechanical compression or bursitis. Diagnosis of HME is typically clinical and radiological. WHO diagnostic criteria include ≥ 2 radiological osteochondromas in the juxta-epiphyseal region of the long bones. Genetic testing is reserved for ambiguous cases. HME is associated with mutations in the EXT-1 (exostosin-1) and EXT-2 (exostosin-2) genes. Imaging techniques, including conventional radiography, CT, MRI, ultrasound, and nuclear medicine, play a crucial role in diagnosing and assessing HME, with each modality offering distinct advantages in visualising the lesions and associated complications. Common complications include skeletal deformities, fractures, bursitis, as well as neural and vascular abnormalities. Notably, there is a 10% risk of malignant transformation into secondary chondrosarcoma in HME patients, compared to only a 1% risk in those with solitary osteochondromas. Malignant transformation should be suspected in patients with new-onset pain or specific imaging features in an osteochondroma, such as growth of de cartilaginous cap. In these cases, an MRI should be performed to assess the cartilage cap thickness. Advances in imaging techniques and genetic understanding have improved the management and prognosis of HME. Follow-up is essential to rule out malignant transformation. This review summarises current knowledge on the clinical presentation, pathogenesis, imaging characteristics, complications, and treatment of HME. CRITICAL RELEVANCE STATEMENT: HME is a disorder characterised by the formation of osteochondromas arising from long and flat bones. Multi-modality imaging characteristics, clinical presentation, complications, and treatment are highlighted to familiarise the readers with this entity and offer optimal patient care. KEY POINTS: HME is characterised by multiple osteochondromas on long and flat bones. Imaging for HME includes radiography, CT, MRI, ultrasound, and nuclear medicine studies. Complications include non-malignant complications, such as bone deformities and malignant transformation. Cartilage-cap measurement with MRI or US is key to exclude malignancy. Follow-up is essential to rule out malignant transformation of the osteochondromas.
Collapse
Affiliation(s)
| | - Sara Gomez-Pena
- Department of Radiology, Hospital Clínico San Carlos, Madrid, Spain
| | - María José Moreno-Casado
- Department of Radiology, Hospital Clínico San Carlos, Madrid, Spain
- Biomedical Imaging Research Group, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
- EURACAN Referral Centre, Madrid, Spain
| | - Gloria Marquina
- EURACAN Referral Centre, Madrid, Spain
- Department of Medical Oncology, Hospital Clínico San Carlos, Madrid, Spain
- Department of Medicine, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Juan Arrazola
- Department of Radiology, Hospital Clínico San Carlos, Madrid, Spain
- Biomedical Imaging Research Group, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
- Department of Radiology and Rehabilitation, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Ana María Crespo-Rodríguez
- Department of Radiology, Hospital Clínico San Carlos, Madrid, Spain
- Biomedical Imaging Research Group, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
- EURACAN Referral Centre, Madrid, Spain
- Department of Radiology and Rehabilitation, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
| |
Collapse
|
5
|
Michaeli O, Kim SY, Mitchell SG, Jongmans MCJ, Wasserman JD, Perrino MR, Das A, MacFarland SP, Scollon SR, Greer MLC, Sobreira N, Gallinger B, Lupo PJ, Malkin D, Schneider KW, Schultz KAP, Foulkes WD, Woodward ER, Stewart DR. Update on Cancer Screening in Children with Syndromes of Bone Lesions, Hereditary Leiomyomatosis and Renal Cell Carcinoma Syndrome, and Other Rare Syndromes. Clin Cancer Res 2025; 31:457-465. [PMID: 39601780 PMCID: PMC11790369 DOI: 10.1158/1078-0432.ccr-24-2171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 10/27/2024] [Accepted: 11/26/2024] [Indexed: 11/29/2024]
Abstract
The management of children with syndromes associated with an increased risk of benign and malignant neoplasms is a complex challenge for health care professionals. The 2023 American Association for Cancer Research Childhood Cancer Predisposition Workshop provided updated consensus guidelines on cancer surveillance in these syndromes, aiming to improve early detection and intervention and reduce morbidity associated with such neoplasms. In this article, we review several of the rare conditions discussed in this workshop. Ollier disease and Maffucci syndrome are enchondromatoses (disorders featuring benign bone lesions) with up to 50% risk of malignancy, including chondrosarcoma. These patients require surveillance with baseline whole-body MRI and routine monitoring of potential malignant transformation of bony lesions. Hereditary multiple osteochondromas carry a lower risk of chondrosarcoma (<6%) but still require lifelong surveillance and baseline imaging. Related syndromes of benign bone lesions are also described. Hereditary leiomyomatosis and renal cell carcinoma syndrome, associated with fumarate hydratase pathogenic variants, is discussed in detail. Surveillance for renal cell carcinoma in pediatric age is recommended, as well as prompt intervention when a lesion is detected. Schinzel-Giedion syndrome and Rubinstein-Taybi syndrome are described for their associated malignancies and other complications, as well as expert consensus on the need for childhood cancer surveillance. Clinical recommendations, including imaging modalities and frequency of screenings, are proposed and are tailored to each syndrome's age-specific tumor risk profile. In all syndromes, patients and their families should be educated about the potential malignancy risk and advised to seek medical care for rapid growth of a mass, persistent pain, or other unexplained symptoms.
Collapse
Affiliation(s)
- Orli Michaeli
- Division of Hematology/Oncology, Schneider Children’s Medical Center of Israel, Petach Tikva, Israel
| | - Sun Young Kim
- Division of Human Genetics, Department of Pediatrics, Cincinnati Children’s Hospital, University of Cincinnati, Cincinnati, OH, United States
| | - Sarah G. Mitchell
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Department of Pediatric Hematology/Oncology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Marjolijn C. J. Jongmans
- Princess Máxima Center for Pediatric Oncology, Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jonathan D. Wasserman
- Division of Endocrinology, The Hospital for Sick Children / Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Melissa R. Perrino
- Department of Oncology, St Jude Children’s Research Hospital, Memphis, Tennessee, United States
| | - Anirban Das
- Division of Hematology/Oncology, The Hospital for Sick Children, Department of Pediatrics, University of Toronto, Toronto, Canada
| | - Suzanne P MacFarland
- Division of Oncology, The Children’s Hospital of Philadelphia, and Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Sarah R Scollon
- Department of Pediatrics, Division of Hematology/Oncology, Texas Children’s Cancer and Hematology Center, Texas Children’s Hospital, Baylor College of Medicine, Houston, Texas, United States
| | - Mary-Louise C. Greer
- Department of Diagnostic and Interventional Radiology, The Hospital for Sick Children, Department of Medical Imaging, University of Toronto, Toronto, Canada
| | - Nara Sobreira
- McKusick-Nathans Department of Genetic Medicine Johns Hopkins University, Baltimore, MD, United States
| | - Bailey Gallinger
- Clinical and Metabolic Genetics, The Hospital for Sick Children. Toronto, ON. Canada. Department of Molecular Genetics, The University of Toronto. Toronto, ON. Canada
| | - Philip J. Lupo
- Division of Hematology-Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, United States
| | - David Malkin
- Division of Hematology/Oncology, The Hospital for Sick Children, Department of Pediatrics, University of Toronto, Toronto, Canada
| | - Kami Wolfe Schneider
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Children’s Hospital Colorado, Aurora, CO
| | - Kris Ann P. Schultz
- International Pleuropulmonary Blastoma/DICER1 Registry, Cancer and Blood Disorders, Children’s Minnesota, Minneapolis, MN
| | | | - Emma R Woodward
- Faculty of Biology, Medicine and Health, University of Manchester and Manchester Centre for Genomic Medicine, Manchester, United Kingdom
| | - Douglas R. Stewart
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD; United States
| |
Collapse
|
6
|
Bobinski L, Tachtaras E, Hedström E. Minimal-invasive, image guided, 360-degree resection of ilio-lumbo-sacral ostechondroma, planned on the 3D model in a child with hereditary multiple ostechondroma (HMO). EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2025; 34:643-649. [PMID: 39738869 DOI: 10.1007/s00586-024-08625-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 12/09/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Hereditary Multiple Osteochondromas (HMO), previously known as Multiple Hereditary Exostoses (MHE), is a genetic disorder characterized by the formation of multiple, benign, exostoses (osteochondromas) growing from the metaphyseal region of long bones as well as from the axial skeleton. Lesions originating from the lumbar spine region are rare, and are most common growing from the posterior element of the vertebrae. HMO associated osteochondromas are difficult to treat due to continuous and incontrollable growth of these lesions and a lifetime risk for malignant transformation. CASE REPORT We describe a case of a 16-year old patient with known HMO who developed a giant ilio-lumbo-sacral osteochondroma. The tumor protruded into the L4-S1 intraspinal foramina with exophytic expansion to the right psoas muscle and lumbar plexus with compression of the right common iliac vein. To plan and execute the resection and minimize the risk of complications, we used a 3D printed model of the lesion with intraabdominal vessels. The patient was operated during a two-stage procedure - first by mini-open, transabdominal, navigated resection of the lesion, followed by delayed posterior, mini-invasive, navigated resection. The outcome was uneventful and there were no signs of regrowth or malignant transformation during 4 years of follow-up. CONCLUSION We describe a 360-degree surgical resection with application of a 3D printed model, navigation, and mini-invasive techniques. Our report may be useful and inspire spine surgeons to apply similar techniques to treat complex spine lesions.
Collapse
Affiliation(s)
- Lukas Bobinski
- Department of Orthopedics, Umeå University Hospital, Umeå, Sweden.
- Department of Diagnostics and Intervention, Orthopedics, Umeå University, Umeå, Sweden.
| | | | - Erik Hedström
- Department of Orthopedics, Umeå University Hospital, Umeå, Sweden
- Department of Diagnostics and Intervention, Orthopedics, Umeå University, Umeå, Sweden
| |
Collapse
|
7
|
Zhou L, Li X, Ji Z, Zhou C, Yang L, Li Y, Fu C, Gu L, Zhang S, Gao J, Yue P, Yu H. Generation of Ext1 Gene-Edited Mice Model Via Dual sgRNAs/Cas9 System and Phenotypic Analyses. Mol Biotechnol 2024:10.1007/s12033-024-01325-0. [PMID: 39565542 DOI: 10.1007/s12033-024-01325-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 11/07/2024] [Indexed: 11/21/2024]
Abstract
Hereditary multiple exostoses (HME) is an autosomal dominant skeletal disease. Genetic linkage analyses have identified that mutations in the exostosin glycosyltransferase (EXT)1 and EXT2 genes are linked to HME pathogenesis, with EXT1 mutation being the most frequent. The aim of this study was to generate a mice model with Ext1 gene editing to simulate human EXT1 mutation and investigate the genetic pathogenicity of Ext1 through phenotypic analyses. We designed a pair of dual sgRNAs targeting exon 1 of the mice Ext1 gene for precise deletion of a 46 bp DNA fragment, resulting in frameshift mutation of the Ext1 gene. The designed dual sgRNAs and Cas9 proteins were injected into mice zygotes cytoplasm. A total of 14 mice were obtained via embryo transfer, among which two genotypic chimera mice had a deletion of the 46 bp DNA fragment in exon 1 of the Ext1 gene. By hybridization and breeding, we successfully generated heterozygous mice with edited Ext1 gene (Ext+/-). Off-target effect analysis did not reveal off-target mutations in Ext+/- mice caused by the two sgRNAs used. Compared to wild-type mice, Ext+/- mice exhibited lower body weights. X-ray imaging showed hyperplastic bone near caudal vertebrae only in male Ext+/- mice, with computed tomography values approximately at 200 HU for hyperplastic bone between ribs and spine regions. Furthermore, immunohistochemical analysis revealed fewer articular chondrocytes expressing EXT1 in edited mice compared to wild-type ones. Pathological section analysis demonstrated no structural or morphological abnormalities in heart, liver, lung, or kidney tissues from Ext+/- mice. In conclusion, we successfully generated an accurate DNA deletion model for studying Ext1 using dual sgRNAs/Cas9 systems. In conclusion, we successfully generated precise DNA deletions in the Ext1 mice model using the dual sgRNAs/Cas9 system. In conclusion, we observed significant phenotypic changes in Ext+/- mice, particularly bone hyperplasia in male individuals; however, no exostosis was detected in the gene-edited mice. The introduction of a frameshift mutation into the Ext1 gene through CRISPR/Cas9 technology resulted in novel phenotypic alterations, highlighting the genetic pathogenicity of Ext1. Therefore, our Ext+/- mice serve as a valuable model for further biomedical investigations related to the Ext1 gene.
Collapse
Affiliation(s)
- Lan Zhou
- Key Laboratory of Medical Biotechnology and Translational Medicine (Guilin Medical University), Education Department of Guangxi Zhuang Autonomous Region, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin, 541199, China
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guilin Medical University, Guilin, 541199, China
- Affiliated Hospital of Panzhihua University, Panzhihua University, Panzhihua, 617000, China
| | - Xin Li
- Department of Radiology of Second Hospital Affiliated, Guilin Medical University, Guilin, 541000, China
| | - Zihan Ji
- Key Laboratory of Medical Biotechnology and Translational Medicine (Guilin Medical University), Education Department of Guangxi Zhuang Autonomous Region, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin, 541199, China
| | - Can Zhou
- Key Laboratory of Medical Biotechnology and Translational Medicine (Guilin Medical University), Education Department of Guangxi Zhuang Autonomous Region, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin, 541199, China
| | - Lingling Yang
- Key Laboratory of Medical Biotechnology and Translational Medicine (Guilin Medical University), Education Department of Guangxi Zhuang Autonomous Region, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin, 541199, China
| | - Yong Li
- Key Laboratory of Medical Biotechnology and Translational Medicine (Guilin Medical University), Education Department of Guangxi Zhuang Autonomous Region, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin, 541199, China
| | - Can Fu
- Key Laboratory of Medical Biotechnology and Translational Medicine (Guilin Medical University), Education Department of Guangxi Zhuang Autonomous Region, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin, 541199, China
| | - Lantao Gu
- Key Laboratory of Medical Biotechnology and Translational Medicine (Guilin Medical University), Education Department of Guangxi Zhuang Autonomous Region, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin, 541199, China
| | - Shun Zhang
- Key Laboratory of Medical Biotechnology and Translational Medicine (Guilin Medical University), Education Department of Guangxi Zhuang Autonomous Region, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin, 541199, China
| | - JinTao Gao
- Key Laboratory of Medical Biotechnology and Translational Medicine (Guilin Medical University), Education Department of Guangxi Zhuang Autonomous Region, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin, 541199, China
| | - Pengpeng Yue
- Key Laboratory of Medical Biotechnology and Translational Medicine (Guilin Medical University), Education Department of Guangxi Zhuang Autonomous Region, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin, 541199, China.
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guilin Medical University, Guilin, 541199, China.
| | - Honghao Yu
- Key Laboratory of Medical Biotechnology and Translational Medicine (Guilin Medical University), Education Department of Guangxi Zhuang Autonomous Region, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin, 541199, China.
- Guangxi Key Laboratory of Drug Discovery and Optimization, Guilin Medical University, Guilin, 541199, China.
| |
Collapse
|
8
|
Bartolotti I, Sobul K, Corsini S, Scognamiglio D, Moroni A, Gnoli M, Sangiorgi L, Pedrini E. A Novel Pathogenic Large Duplication in EXT1 Identified in a Family with Multiple Osteochondromas. Genes (Basel) 2024; 15:1169. [PMID: 39336760 PMCID: PMC11430977 DOI: 10.3390/genes15091169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024] Open
Abstract
Multiple osteochondromas (MO) is an autosomal dominant disorder and the most common genetic skeletal dysplasia, characterized by the growth of bone outgrowths capped by cartilage, called osteochondromas. Most MO cases are caused by mutations in the exostosin-1 (EXT1) and exostosin-2 (EXT2) genes. Only 5% of MO-causative variants are represented by single or multiple exon deletions; to date, no pathogenic large duplication has been described in the literature. In the present study, we describe the novel in-tandem intragenic duplication c.(1128_1202)_(1284+29_1344)dup involving exon 4 of EXT1 (NM_000127.2), detected in a three-generation family with MO. The variant has been detected by MLPA (multiplex ligation-dependent probe amplification) and then confirmed with qPCR (quantitative PCR). Our finding expands the spectrum of MO-causing variants describing a pathogenic large duplication, underlying the importance of quantitative analysis in patients with negative sequencing.
Collapse
Affiliation(s)
- Isabella Bartolotti
- Department of Rare Skeletal Disorders, IRCCS Istituto Ortopedico Rizzoli, 40131 Bologna, Italy
| | - Klaudia Sobul
- Department of Rare Skeletal Disorders, IRCCS Istituto Ortopedico Rizzoli, 40131 Bologna, Italy
| | - Serena Corsini
- Department of Rare Skeletal Disorders, IRCCS Istituto Ortopedico Rizzoli, 40131 Bologna, Italy
| | - Davide Scognamiglio
- Department of Rare Skeletal Disorders, IRCCS Istituto Ortopedico Rizzoli, 40131 Bologna, Italy
| | - Alice Moroni
- Department of Rare Skeletal Disorders, IRCCS Istituto Ortopedico Rizzoli, 40131 Bologna, Italy
| | - Maria Gnoli
- Department of Rare Skeletal Disorders, IRCCS Istituto Ortopedico Rizzoli, 40131 Bologna, Italy
| | - Luca Sangiorgi
- Department of Rare Skeletal Disorders, IRCCS Istituto Ortopedico Rizzoli, 40131 Bologna, Italy
| | - Elena Pedrini
- Department of Rare Skeletal Disorders, IRCCS Istituto Ortopedico Rizzoli, 40131 Bologna, Italy
| |
Collapse
|
9
|
Rošin M, Kelam N, Jurić I, Racetin A, Ogorevc M, Corre B, Čarić D, Filipović N, Vukojević K. Syndecans, Exostosins and Sulfotransferases as Potential Synovial Inflammation Moderators in Patients with Hip Osteoarthritis. Int J Mol Sci 2024; 25:4557. [PMID: 38674142 PMCID: PMC11049902 DOI: 10.3390/ijms25084557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/07/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
The gradual deterioration of articular cartilage was thought to be the central event in osteoarthritis (OA), but recent studies demonstrated the importance of low-grade synovitis in the progression of OA. The Syndecan (SDC) family of membrane proteoglycans is known to be involved in the regulation of inflammation, but there is limited evidence considering the role of syndecans in OA synovitis. Our study aimed to investigate the hip OA synovial membrane expression patterns of SDC1, SDC2 and SDC4, as well as exostosins and sulfotransferases (enzymes involved in the polymerisation and modification of syndecans' heparan sulphate chains). Synovial membrane samples of patients with OA (24) were divided into two groups according to their Krenn synovitis score severity. The immunohistochemical expressions of SDC1, SDC2, SDC4, EXT1, EXT2, NDST1 and NDST2 in synovial intima and subintima were then analysed and compared with the control group (patients with femoral neck fracture). According to our study, the immunoexpression of SDC1, NDST1 and EXT2 is significantly increased in the intimal cells of OA synovial membrane in patients with lower histological synovitis scores and SDC4 in patients with higher synovitis scores, in comparison with non-OA controls. The difference in the expression of SDC2 among the OA and non-OA groups was insignificant. SDC1, SDC4, NDST1 and EXT2 seem to be involved as inflammation moderators in low-grade OA synovitis and, therefore, should be further investigated as potential markers of disease progression and therapeutic goals.
Collapse
Affiliation(s)
- Matko Rošin
- Surgery Department, Orthopaedics and Traumatology Division, University Hospital of Split, Spinciceva 1, 21000 Split, Croatia; (M.R.); (D.Č.)
| | - Nela Kelam
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia; (N.K.); (A.R.); (M.O.); (N.F.)
| | - Ivana Jurić
- Department of Emergency Medicine, University Hospital of Split, Spinciceva 1, 21000 Split, Croatia;
| | - Anita Racetin
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia; (N.K.); (A.R.); (M.O.); (N.F.)
| | - Marin Ogorevc
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia; (N.K.); (A.R.); (M.O.); (N.F.)
| | - Brieuc Corre
- Faculty of Medicine and Health Sciences, University of Brest, 29200 Brest, France;
| | - Davor Čarić
- Surgery Department, Orthopaedics and Traumatology Division, University Hospital of Split, Spinciceva 1, 21000 Split, Croatia; (M.R.); (D.Č.)
| | - Natalija Filipović
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia; (N.K.); (A.R.); (M.O.); (N.F.)
| | - Katarina Vukojević
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia; (N.K.); (A.R.); (M.O.); (N.F.)
- Center for Translational Research in Biomedicine, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia
| |
Collapse
|
10
|
Alexandrou A, Salameh N, Papaevripidou I, Nicolaou N, Myrianthopoulos P, Ketoni A, Kousoulidou L, Anastasiou AM, Evangelidou P, Tanteles GA, Sismani C. Hereditary multiple exostoses caused by a chromosomal inversion removing part of EXT1 gene. Mol Cytogenet 2023; 16:8. [PMID: 37217936 DOI: 10.1186/s13039-023-00638-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/07/2023] [Indexed: 05/24/2023] Open
Abstract
BACKGROUND Hereditary multiple exostoses (HME) is an autosomal dominant skeletal disorder characterized by the development of multiple, circumscript and usually symmetric bony protuberances called osteochondromas. Most HME are caused by EXT1 and EXT2 loss of function mutations. Most pathogenic mutations are nonsense followed by missense mutations and deletions. CASE PRESENTATION Here we report on a patient with a rare and complex genotype resulting in a typical HME phenotype. Initial point mutation screening in EXT1 and EXT2 genes by Sanger sequencing did not reveal any pathogenic variants. The patient along with the healthy parents was subsequently referred for karyotype and array-Comparative Genomic Hybridization (CGH) analyses. Chromosomal analysis revealed two independent de novo apparently balanced rearrangements: a balanced translocation between the long arms of chromosomes 2 and 3 at breakpoints 2q22 and 3q13.2 and a pericentric inversion with breakpoints at 8p23.1q24.1. Both breakpoints were confirmed by Fluorescence In Situ Hybridization (FISH). Subsequently, array-CGH revealed a novel heterozygous deletion within the EXT1 gene at one of the inversion breakpoints, rendering the inversion unbalanced. The mode of inheritance, as well as the size of the deletion were further investigated by Quantitative Real-time PCR (qPCR), defining the deletion as de novo and of 3.1 kb in size, removing exon 10 of EXT1. The inversion in combination with the 8p23.1 deletion most likely abolishes the transcription of EXT1 downstream of exon 10 hence resulting in a truncated protein. CONCLUSIONS The identification of a rare and novel genetic cause of HME, highlights the importance of additional comprehensive investigation of patients with typical clinical manifestations, even when EXT1 and EXT2 mutation analysis is negative.
Collapse
Affiliation(s)
- Angelos Alexandrou
- Cytogenetics and Genomics Department, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371, Ayios Dometios, PO Box 23462, 1683, Nicosia, Cyprus
| | - Nicole Salameh
- Cytogenetics and Genomics Department, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371, Ayios Dometios, PO Box 23462, 1683, Nicosia, Cyprus
| | - Ioannis Papaevripidou
- Cytogenetics and Genomics Department, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371, Ayios Dometios, PO Box 23462, 1683, Nicosia, Cyprus
| | - Nayia Nicolaou
- Clinical Genetics Department, Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Panayiotis Myrianthopoulos
- Cytogenetics and Genomics Department, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371, Ayios Dometios, PO Box 23462, 1683, Nicosia, Cyprus
| | - Andria Ketoni
- Cytogenetics and Genomics Department, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371, Ayios Dometios, PO Box 23462, 1683, Nicosia, Cyprus
| | - Ludmila Kousoulidou
- Cytogenetics and Genomics Department, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371, Ayios Dometios, PO Box 23462, 1683, Nicosia, Cyprus
| | - Anna-Maria Anastasiou
- Clinical Genetics Department, Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Paola Evangelidou
- Cytogenetics and Genomics Department, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371, Ayios Dometios, PO Box 23462, 1683, Nicosia, Cyprus
| | - George A Tanteles
- Clinical Genetics Department, Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Carolina Sismani
- Cytogenetics and Genomics Department, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371, Ayios Dometios, PO Box 23462, 1683, Nicosia, Cyprus.
| |
Collapse
|
11
|
Huo X, Song L, Wang K, Wang H, Li D, Li H, Wang W, Wang Y, Chen L, Zhao Z, Wang L, Wu Z. Prognostic factors and Doxorubicin involved in malignant progression of meningioma. Sci Rep 2023; 13:5632. [PMID: 37024523 PMCID: PMC10079659 DOI: 10.1038/s41598-023-28996-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 01/27/2023] [Indexed: 04/08/2023] Open
Abstract
Meningioma was the most primary intracranial tumor, but the molecular characteristics and the treatment of malignant meningioma were still unclear. Nine malignant progression-related genes based prognostic signatures were identified by transcriptome analysis between benign meningioma and malignant meningioma. The external dataset GEO136661 and quantitative Real-time Polymerase Chain Reaction were used to verify the prognostic factors. has-miR-3605-5p, hsa-miR-664b-5p, PNRC2, BTBD8, EXTL2, SLFN13, DGKD, NSD2, and BVES were closed with malignant progression. Moreover, Doxorubicin was identified by Connectivity Map website with the differential malignant progression-related genes. CCK-8 assay, Edu assay, wound healing assay, and trans-well experiment were used to reveal that Doxorubicin could inhibit proliferation, migration and invasion of IOMM-Lee Cells.
Collapse
Affiliation(s)
- Xulei Huo
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Nansihuanxilu 119, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Lairong Song
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Nansihuanxilu 119, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Ke Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Nansihuanxilu 119, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Hongyi Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Nansihuanxilu 119, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Da Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Nansihuanxilu 119, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Huan Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Nansihuanxilu 119, Fengtai District, Beijing, 100070, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Wei Wang
- Department of Neurosurgery, Tianjin Fifth Center Hospital, Tianjin, China
| | - Yali Wang
- Department of Neuro-Oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lei Chen
- Department of Neurosurgery, Tianjin Fifth Center Hospital, Tianjin, China
| | - Zongmao Zhao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China.
| | - Liang Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Nansihuanxilu 119, Fengtai District, Beijing, 100070, China.
- China National Clinical Research Center for Neurological Diseases, Beijing, China.
| | - Zhen Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Nansihuanxilu 119, Fengtai District, Beijing, 100070, China.
- China National Clinical Research Center for Neurological Diseases, Beijing, China.
| |
Collapse
|
12
|
Premalignant Conditions of Bone. JAAOS: GLOBAL RESEARCH AND REVIEWS 2022; 6:01979360-202210000-00004. [PMID: 36227850 PMCID: PMC9575816 DOI: 10.5435/jaaosglobal-d-22-00097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 07/29/2022] [Indexed: 01/10/2023]
Abstract
Development of malignancy is a multifactorial process, and there are multitude of conditions of bone that may predispose patients to malignancy. Etiologies of malignancy include benign osseous conditions, genetic predisposition, and extrinsic conditions. New-onset pain or growth in a previously stable lesion is that should concern for malignant change and should prompt a diagnostic workup for malignancy.
Collapse
|
13
|
Mundy C, Chung J, Koyama E, Bunting S, Mahimkar R, Pacifici M. Osteochondroma formation is independent of heparanase expression as revealed in a mouse model of hereditary multiple exostoses. J Orthop Res 2022; 40:2391-2401. [PMID: 34996123 PMCID: PMC9259764 DOI: 10.1002/jor.25260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 12/20/2021] [Accepted: 01/05/2022] [Indexed: 02/04/2023]
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.
Collapse
Affiliation(s)
- Christina Mundy
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Juliet Chung
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Eiki Koyama
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | | | | | - Maurizio Pacifici
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| |
Collapse
|
14
|
Liu W, Shi X, Li Y, Qiao F, Wu Y. The identification of a novel frameshift insertion mutation in the EXT1 gene in a Chinese family with hereditary multiple exostoses. Clin Case Rep 2022; 10:e6298. [PMID: 36101782 PMCID: PMC9459098 DOI: 10.1002/ccr3.6298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 08/11/2022] [Accepted: 08/19/2022] [Indexed: 11/08/2022] Open
Abstract
To identify the pathogenic gene variation in a Chinese family with Hereditary Multiple Exostoses (HME). By examining blood-sourced DNA and clinical manifestations of the proband and his family members, the whole exome sequencing (WES) and Sanger sequencing were used to detect possibly pathogenic mutations. A novel heterozygous mutation (c.325dup) was identified in exon 1 of the exostosin 1 (EXT1) gene from the proband and the affected family members. And we found this mutation was absent in all the unaffected family members. This c.325dup mutation is in the exon 1 domain of the EXT1 gene and the change of p.C109Lfs*80 cause the early termination of protein translation. The identification of the novel frameshift insertion mutation (c.325dup) expands the mutation spectrum of HME, which provides new evidence for HME diagnosis.
Collapse
Affiliation(s)
- Wanlu Liu
- Department of Obstetrics and GynecologyTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Xinwei Shi
- Department of Obstetrics and GynecologyTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yuqi Li
- Department of Obstetrics and GynecologyTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Fuyuan Qiao
- Department of Obstetrics and GynecologyTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yuanyuan Wu
- Department of Obstetrics and GynecologyTongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
15
|
A Genotype-Phenotype Study of Multiple Hereditary Exostoses in Forty-Three Patients. J Clin Med 2022; 11:jcm11133703. [PMID: 35806987 PMCID: PMC9267182 DOI: 10.3390/jcm11133703] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/21/2022] [Accepted: 06/21/2022] [Indexed: 12/03/2022] Open
Abstract
Multiple hereditary exostoses (MHE) is a rare autosomal dominant skeletal disorder with a variety of clinical manifestations. We aimed to evaluate the general clinical phenotypic severity of MHE using our own scoring system and analyzed the risk factors associated with severe clinical phenotypes. In this study, 43 patients from 30 families were analyzed. The mutations were identified by direct sequencing of polymerase chain reaction-amplified genomic DNA or by multiplex ligation-dependent probe amplification. According to a new scoring system devised by the authors, the severity of the phenotype was assessed as mild, moderate, or severe based on the deformity of each segment, number of exostoses, leg length discrepancy, and functional limitations. Of 43 patients from 30 families, 39 patients (90.7%) and 24 families (80%) presented with EXT1 or EXT2 mutations. Patients with EXT1 mutations had a significantly worse phenotype than that of patients with EXT2 mutations or without any detectable mutation. The mean clinical score of patients with an EXT1 mutation (5.76; range, 2.0–8.0; SD = 1.60) was higher than that of patients with an EXT2 mutation (4.06; range, 2.0–7.0; SD = 1.47) or of those without any detectable mutation (4.63; range, 3.0–6.0; SD = 1.44; p = 0.005). According to our classification system, more patients with EXT1 mutations had ‘severe disease’ than those with EXT2 mutations. Deformity scores were also higher in patients with EXT1 mutations (p = 0.018). In the multivariate analysis, the deformity score was found to be associated with the ‘severe’ class (p = 0.031). In conclusion, 90.7% of patients with MHE showed EXT mutations. Our scoring system showed reliable results. We suggest that the extent of deformity is an important factor in determining the phenotype of MHE and close monitoring for the development of severe disease is recommended in patients with high deformity scores.
Collapse
|
16
|
Li J, Wang Z, Han Y, Jin C, Cheng D, Zhou YA, Zhen J. Genetic and functional analyses detect an EXT1 splicing pathogenic variant in a Chinese hereditary multiple exostosis (HME) family. Mol Genet Genomic Med 2022; 10:e1878. [PMID: 35106951 PMCID: PMC8922959 DOI: 10.1002/mgg3.1878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 12/19/2021] [Accepted: 01/06/2022] [Indexed: 11/22/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Jianwei Li
- The Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Zhiqiang Wang
- Lvliang People's Hospital, Shanxi Medical University, Lvliang, China
| | - Yaxin Han
- The First Hospital, Shanxi Medical University, Taiyuan, China
| | - Chengfang Jin
- Lvliang People's Hospital, Shanxi Medical University, Lvliang, China
| | - Dalin Cheng
- Lvliang People's Hospital, Shanxi Medical University, Lvliang, China
| | - Yong-An Zhou
- The Second Hospital, Shanxi Medical University, Taiyuan, China
| | - Junping Zhen
- The Second Hospital, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
17
|
Miwa S, Yamamoto N, Hayashi K, Takeuchi A, Igarashi K, Tsuchiya H. Therapeutic Targets and Emerging Treatments in Advanced Chondrosarcoma. Int J Mol Sci 2022; 23:ijms23031096. [PMID: 35163019 PMCID: PMC8834928 DOI: 10.3390/ijms23031096] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/16/2022] [Accepted: 01/18/2022] [Indexed: 12/25/2022] Open
Abstract
Due to resistance to standard anticancer agents, it is difficult to control the disease progression in patients with metastatic or unresectable chondrosarcoma. Novel therapeutic approaches, such as molecule-targeting drugs and immunotherapy, are required to improve clinical outcomes in patients with advanced chondrosarcoma. Recent studies have suggested several promising biomarkers and therapeutic targets for chondrosarcoma, including IDH1/2 and COL2A1. Several molecule-targeting agents and immunotherapies have shown favorable antitumor activity in clinical studies in patients with advanced chondrosarcomas. This review summarizes recent basic studies on biomarkers and molecular targets and recent clinical studies on the treatment of chondrosarcomas.
Collapse
|
18
|
Corsini S, Pedrini E, Patavino C, Gnoli M, Lanza M, Sangiorgi L. An Easy-to-Use Approach to Detect CNV From Targeted NGS Data: Identification of a Novel Pathogenic Variant in MO Disease. Front Endocrinol (Lausanne) 2022; 13:874126. [PMID: 35837302 PMCID: PMC9273874 DOI: 10.3389/fendo.2022.874126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/25/2022] [Indexed: 11/17/2022] Open
Abstract
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.
Collapse
|
19
|
Oppelaar JJ, Rorije NMG, Olde Engberink RHG, Chahid Y, van Vlies N, Verberne HJ, van den Born BJH, Vogt L. Perturbed body fluid distribution and osmoregulation in response to high salt intake in patients with hereditary multiple exostoses. Mol Genet Metab Rep 2021; 29:100797. [PMID: 34815940 PMCID: PMC8591465 DOI: 10.1016/j.ymgmr.2021.100797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 08/24/2021] [Indexed: 11/26/2022] Open
Abstract
Background Hereditary Multiple Exostoses (HME) is a rare autosomal disorder characterized by the presence of multiple exostoses (osteochondromas) caused by a heterozygous loss of function mutation in EXT1 or EXT2; genes involved in heparan sulfate (HS) chain elongation. Considering that HS and other glycosaminoglycans play an important role in sodium and water homeostasis, we hypothesized that HME patients have perturbed whole body volume regulation and osmolality in response to high sodium conditions. Methods We performed a randomized cross-over study in 7 male HME patients and 12 healthy controls, matched for age, BMI, blood pressure and renal function. All subjects followed both an 8-day low sodium diet (LSD, <50 mmol/d) and high sodium diet (HSD, >200 mmol/d) in randomized order. After each diet, blood and urine samples were collected. Body fluid compartment measurements were performed by using the distribution curve of iohexol and 125I-albumin. Results In HME patients, HSD resulted in significant increase of intracellular fluid volume (ICFV) (1.2 L, p = 0.01). In this group, solute-mediated water clearance was significantly lower after HSD, and no changes in interstitial fluid volume (IFV), plasma sodium, and effective osmolality were observed. In healthy controls, HSD did not influence ICFV, but expanded IFV (1.8 L, p = 0.058) and increased plasma sodium and effective osmolality. Conclusion HME patients show altered body fluid distribution and osmoregulation after HSD compared to controls. Our results might indicate reduced interstitial sodium accumulation capacity in HME, leading to ICFV increase. Therefore, this study provides additional support that HS is crucial for maintaining constancy of the internal environment.
Collapse
Key Words
- BMI, Body mass index
- BP, Blood pressure
- ECFV, Extracellular fluid volume
- EXT1/EXT2, Extosin-1 / Extosin-2
- GAG, Glycosaminoglycan
- Glycosaminoglycans
- HME, Hereditary Multiple Exostoses
- HSD, High sodium diet
- Heparan sulfate
- Hereditary Multiple Exostoses
- ICFV, Intracellular fluid volume
- IFV, Interstital fluid volume
- LSD, Low sodium diet
- Osmoregulation
- PV, Plasma volume
- Sodium
- TBW, Total body water
- Water balance
Collapse
Affiliation(s)
- Jetta J Oppelaar
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam, the Netherlands
| | - Nienke M G Rorije
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam, the Netherlands
| | - Rik H G Olde Engberink
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam, the Netherlands
| | - Youssef Chahid
- Amsterdam UMC, University of Amsterdam, Department of Radiology and Nuclear Medicine, Meibergdreef 9, Amsterdam, the Netherlands
| | - Naomi van Vlies
- Amsterdam UMC, University of Amsterdam, Laboratory Genetic Metabolic Diseases, Meibergdreef 9, Amsterdam, the Netherlands
| | - Hein J Verberne
- Amsterdam UMC, University of Amsterdam, Department of Radiology and Nuclear Medicine, Meibergdreef 9, Amsterdam, the Netherlands
| | - Bert-Jan H van den Born
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Vascular Medicine, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam, the Netherlands
| | - Liffert Vogt
- Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Section of Nephrology, Amsterdam Cardiovascular Sciences, Meibergdreef 9, Amsterdam, the Netherlands
| |
Collapse
|
20
|
Kodama M, Shimura H, Tien JC, Newberg JY, Kodama T, Wei Z, Rangel R, Yoshihara K, Kuruma A, Nakae A, Hashimoto K, Sawada K, Kimura T, Jenkins NA, Copeland NG. Sleeping Beauty Transposon Mutagenesis Identifies Genes Driving the Initiation and Metastasis of Uterine Leiomyosarcoma. Cancer Res 2021; 81:5413-5424. [PMID: 34475109 DOI: 10.1158/0008-5472.can-21-0356] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 07/29/2021] [Accepted: 09/01/2021] [Indexed: 11/16/2022]
Abstract
Uterine leiomyosarcoma (ULMS) is a malignancy, which arises from the uterine smooth muscle. Because of its rarity, aggressive nature, and extremely poor prognosis, the molecular mechanisms driving ULMS remain elusive. To identify candidate cancer genes (CCG) driving ULMS, we conducted an in vivo Sleeping Beauty (SB) transposon mutagenesis screen in uterine myometrium-specific, PTEN knockout, KRAS mutant (PTEN KO/KRAS) mice. ULMS quickly developed in SB PTEN KO/KRAS mice, but not in PTEN KO/KRAS mice, demonstrating the critical importance of SB mutagenesis for driving ULMS in this model. Subsequent sequencing of SB insertion sites in these tumors identified 19 ULMS CCGs that were significantly enriched in known cancer genes. Among them, Zfp217 and Sfmbt2 functioned at early stages of tumor initiation and appeared to be oncogenes. Expression of ZNF217, the human homolog of ZFP217, was shown to be elevated in human ULMS compared with paired normal uterine smooth muscle, where it negatively correlated with patient prognosis. Inhibition of ZNF217 suppressed, whereas overexpression induced, proliferation, survival, migration, and stemness of human ULMS. In a second ex vivo ULMS SB metastasis screen, three CCGs were identified that may drive ULMS metastasis to the lung. One of these CCGs, Nrd1 (NRDC in humans), showed stronger expression in human metastatic tumors compared with primary ULMS and negatively associated with patient survival. NRDC knockdown impaired migration and adhesion without affecting cell proliferation, whereas overexpression had the opposite effect. Together, these results reveal novel mechanism driving ULMS tumorigenesis and metastasis and identify ZNF217 and NRDC as potential targets for ULMS therapy. SIGNIFICANCE: An in vivo Sleeping Beauty transposon mutagenesis screen identifies candidate cancer genes that drive initiation and progression of uterine leiomyosarcoma and may serve as therapeutic targets.
Collapse
Affiliation(s)
- Michiko Kodama
- Cancer Research Program, Houston Methodist Research Institute, Houston, Texas. .,Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroko Shimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Jean C Tien
- Cancer Research Program, Houston Methodist Research Institute, Houston, Texas.,Department of Pathology, Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Justin Y Newberg
- Cancer Research Program, Houston Methodist Research Institute, Houston, Texas
| | - Takahiro Kodama
- Cancer Research Program, Houston Methodist Research Institute, Houston, Texas.,Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Zhubo Wei
- Cancer Research Program, Houston Methodist Research Institute, Houston, Texas.,Center for Genomic and Precision Medicine, Institute of Biosciences and Technology, Texas A&M University, Houston, Texas
| | - Roberto Rangel
- Cancer Research Program, Houston Methodist Research Institute, Houston, Texas.,Department of Head and Neck Surgery, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kosuke Yoshihara
- Department of Obstetrics and Gynecology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Airi Kuruma
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Aya Nakae
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kae Hashimoto
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kenjiro Sawada
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tadashi Kimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Nancy A Jenkins
- Cancer Research Program, Houston Methodist Research Institute, Houston, Texas.,Genetics Department, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Neal G Copeland
- Cancer Research Program, Houston Methodist Research Institute, Houston, Texas. .,Genetics Department, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
21
|
An update on the imaging of diaphyseal aclasis. Skeletal Radiol 2021; 50:1941-1962. [PMID: 33791832 DOI: 10.1007/s00256-021-03770-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/23/2021] [Accepted: 03/23/2021] [Indexed: 02/02/2023]
Abstract
Solitary osteochondromas are common, benign hyaline cartilage-capped exostoses that primarily arise from the metaphyses of long and flat bones. Diaphyseal aclasis is an autosomal dominant condition resulting from EXT1 or EXT2 gene mutations and is characterized by multifocal osteochondromas. These can result in a wide spectrum of complications, such as skeletal deformity, neurological and vascular complications, adventitial bursa formation, fracture, and rarely malignant transformation to peripheral chondrosarcoma. In this review, we outline in detail the multimodality imaging features of DA and its associated complications.
Collapse
|
22
|
Mordenti M, Gnoli M, Boarini M, Trisolino G, Evangelista A, Pedrini E, Corsini S, Tremosini M, Staals EL, Antonioli D, Stilli S, Donati DM, Sangiorgi L. The Rizzoli Multiple Osteochondromas Classification revised: describing the phenotype to improve clinical practice. Am J Med Genet A 2021; 185:3466-3475. [PMID: 34477285 PMCID: PMC9293117 DOI: 10.1002/ajmg.a.62470] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 07/29/2021] [Accepted: 08/05/2021] [Indexed: 11/07/2022]
Abstract
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.
Collapse
Affiliation(s)
- Marina Mordenti
- Department of Rare Skeletal DisordersIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Maria Gnoli
- Department of Rare Skeletal DisordersIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Manila Boarini
- Department of Rare Skeletal DisordersIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Giovanni Trisolino
- Unit of Pediatric Orthopedics and Traumatology, IRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Andrea Evangelista
- Department of Rare Skeletal DisordersIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Elena Pedrini
- Department of Rare Skeletal DisordersIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Serena Corsini
- Department of Rare Skeletal DisordersIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Morena Tremosini
- Department of Rare Skeletal DisordersIRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Eric L. Staals
- Department of Third Orthopedic and Traumatologic Clinic prevalently Oncologic, IRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Diego Antonioli
- Unit of Pediatric Orthopedics and Traumatology, IRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Stefano Stilli
- Unit of Pediatric Orthopedics and Traumatology, IRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Davide M. Donati
- Department of Third Orthopedic and Traumatologic Clinic prevalently Oncologic, IRCCS Istituto Ortopedico RizzoliBolognaItaly
| | - Luca Sangiorgi
- Department of Rare Skeletal DisordersIRCCS Istituto Ortopedico RizzoliBolognaItaly
| |
Collapse
|
23
|
Yang M, Xie H, Xu B, Xiang Q, Wang H, Hu T, Liu S. Identification of a novel EXT2 frameshift mutation in a family with hereditary multiple exostoses by whole-exome sequencing. J Clin Lab Anal 2021; 35:e23968. [PMID: 34403521 PMCID: PMC8418499 DOI: 10.1002/jcla.23968] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 08/04/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023] Open
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.
Collapse
Affiliation(s)
- Mei Yang
- Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Hanbing Xie
- Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Bocheng Xu
- Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Qinqin Xiang
- Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - He Wang
- Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Ting Hu
- Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Shanling Liu
- Department of Obstetrics & Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
24
|
Tong Y, Zhang Y, Luo J, Hong Z, Chen X, Bi Q. Identification of Novel Mutations in the EXT1 and EXT2 Genes of Chinese Patients with Hereditary Multiple Osteochondromas. Genet Test Mol Biomarkers 2021; 25:145-151. [PMID: 33596140 DOI: 10.1089/gtmb.2020.0098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Aim: To detect mutations in the EXT1 and EXT2 genes in four Chinese families with hereditary multiple osteochondromas (HMO). HMO is an autosomal dominant disorder characterized by the overgrowth of multiple cartilage-capped bones in the metaphysis of long bones and flat bones. Methods: Polymerase chain reaction-based amplification followed by DNA sequencing of the complete coding sequences of EXT1 and EXT2 was performed for four Chinese families with HMO. Results: The mutant allele was found in six patients: three mutations were found in EXT1 and two in EXT2. A novel frameshift mutation, which generates a premature stop codon at codon 586 and causes partial loss of the glycosyltransferase domain, was detected in exon 9 of EXT1 (F579Yfs*8). We hypothesize that F579Yfs*8 is a pathogenic mutation. Two novel missense mutations (G339S and V545D) were found in EXT1. The variant c.1634T>A (V545D) is apparently benign. In addition we found a novel deletion mutation in EXT2, c.856_864 del TTCCTCCTG, which results in the deletion of 286Phe, 287Leu, and 288Leu, that is likely pathogenic. Finally, we identified a likely benign variant in exon 13 of EXT2. c.2035-41T>C (rs3740878). Conclusions: We found three novel, potentially pathogenic mutations in EXT1 and EXT2, including a novel frameshift mutation. More importantly, our study results have expanded the spectrum of EXT mutations conducive to the genetic diagnosis and counseling of patients with HMO.
Collapse
Affiliation(s)
- Yu Tong
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yin Zhang
- Graduate Department, Bengbu Medical College, Bengbu, China
| | - Junchao Luo
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zheping Hong
- Department of Orthopedic Surgery, Zhejiang Provincial People's Hospital and People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Xinji Chen
- Department of Orthopedic Surgery, Zhejiang Provincial People's Hospital and People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Qing Bi
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Orthopedic Surgery, Zhejiang Provincial People's Hospital and People's Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
25
|
Guo X, Chen S, Lin M, Pan Y, Liu N, Shi T. A Novel Intronic Splicing Mutation in the EXT2 Gene of a Chinese Family with Multiple Osteochondroma. Genet Test Mol Biomarkers 2021; 25:478-485. [PMID: 34280007 DOI: 10.1089/gtmb.2021.0030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Xiaoyan Guo
- Department of Laboratory Medicine, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou, P.R. China
| | - Shunyou Chen
- Department of Orthopedics, and Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou, P.R. China
| | - Mingrui Lin
- Intensive Care Unit, The Affiliated People's Hospital of Fujian Traditional Medical University, Fuzhou, P.R. China
| | - Yuancheng Pan
- Department of Orthopedics, and Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou, P.R. China
| | - Nannan Liu
- Department of Orthopedics Institute, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou, P.R. China
| | - Tengfei Shi
- Department of Laboratory Medicine, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou, P.R. China
| |
Collapse
|
26
|
Tong Y, Luo J, Zhang Y, Hong Z, Cao L, Chen X, Chen J, Bi Q. Novel Mutations in Chinese Patients with Multiple Osteochondromas Identified Using Whole Exome Sequencing. Genet Test Mol Biomarkers 2021; 25:361-367. [PMID: 34003695 DOI: 10.1089/gtmb.2020.0317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
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.
Collapse
Affiliation(s)
- Yu Tong
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Department of Orthopedic Surgery, People's Hospital of Hangzhou Medical College, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Junchao Luo
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yin Zhang
- Graduate Department, Bengbu Medical College, Bengbu, China
| | - Zheping Hong
- Department of Orthopedic Surgery, People's Hospital of Hangzhou Medical College, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Li Cao
- Department of Orthopedic Surgery, People's Hospital of Hangzhou Medical College, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Xinji Chen
- Department of Orthopedic Surgery, People's Hospital of Hangzhou Medical College, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Jihang Chen
- Department of Orthopedic Surgery, People's Hospital of Hangzhou Medical College, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Qing Bi
- The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.,Department of Orthopedic Surgery, People's Hospital of Hangzhou Medical College, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
27
|
Ismailoglu T. A Family with Hereditary Multiple Exostoses. EURASIAN JOURNAL OF FAMILY MEDICINE 2021. [DOI: 10.33880/ejfm.2021100105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Hereditary multiple exostoses is a rare autosomal dominant genetic disorder characterized by multiple exostoses (osteochondromas), mostly diagnosed in childhood. It may manifest with a wide spectrum from asymptomatic to skeletal deformities or neurovascular complications. Pain and/or swelling are often the first symptoms for patients to consult a doctor. Although no medical treatment is currently available, lesions can be removed with surgical excision in case of aesthetic anxiety or complications such as deformities or rarely, malign transformation. In this article, three individuals from the same family with hereditary multiple exostoses are described who were evaluated within the core competencies of family medicine.
Keywords: exostoses, multiple hereditary, osteochondroma, scoliosis
Collapse
|
28
|
Al-Zayed Z, Al-Rijjal RA, Al-Ghofaili L, BinEssa HA, Pant R, Alrabiah A, Al-Hussainan T, Zou M, Meyer BF, Shi Y. Mutation spectrum of EXT1 and EXT2 in the Saudi patients with hereditary multiple exostoses. Orphanet J Rare Dis 2021; 16:100. [PMID: 33632255 PMCID: PMC7905910 DOI: 10.1186/s13023-021-01738-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 02/11/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hereditary Multiple Exostoses (HME), also known as Multiple Osteochondromas (MO) is a rare genetic disorder characterized by multiple benign cartilaginous bone tumors, which are caused by mutations in the genes for exostosin glycosyltransferase 1 (EXT1) and exostosin glycosyltransferase 2 (EXT2). The genetic defects have not been studied in the Saudi patients. AIM OF STUDY We investigated mutation spectrum of EXT1 and EXT2 in 22 patients from 17 unrelated families. METHODS Genomic DNA was extracted from peripheral leucocytes. The coding regions and intron-exon boundaries of both EXT1 and EXT2 genes were screened for mutations by PCR-sequencing analysis. Gross deletions were analyzed by MLPA analysis. RESULTS EXT1 mutations were detected in 6 families (35%) and 3 were novel mutations: c.739G > T (p. E247*), c.1319delG (p.R440Lfs*4), and c.1786delA (p.S596Afs*25). EXT2 mutations were detected in 7 families (41%) and 3 were novel mutations: c.541delG (p.D181Ifs*89), c.583delG (p.G195Vfs*75), and a gross deletion of approximately 10 kb including promoter and exon 1. Five patients from different families had no family history and carried de novo mutations (29%, 5/17). No EXT1 and EXT2 mutations were found in the remaining four families. In total, EXT1 and EXT2 mutations were found in 77% (13/17) of Saudi HME patients. CONCLUSION EXT1 and EXT2 mutations contribute significantly to the pathogenesis of HME in the Saudi population. In contrast to high mutation rate in EXT 1 (65%) and low mutation rate in EXT2 (25%) in other populations, the frequency of EXT2 mutations are much higher (41%) and comparable to that of EXT1 among Saudi patients. De novo mutations are also common and the six novel EXT1/EXT2 mutations further expands the mutation spectrum of HME.
Collapse
Affiliation(s)
- Zayed Al-Zayed
- Department of Orthopedics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Roua A Al-Rijjal
- Department of Genetics, MBC 3, Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh, 11211, Saudi Arabia
| | | | - Huda A BinEssa
- Department of Genetics, MBC 3, Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh, 11211, Saudi Arabia
| | - Rajeev Pant
- Department of Orthopedics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Anwar Alrabiah
- Department of Orthopedics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Thamer Al-Hussainan
- Department of Orthopedics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Minjing Zou
- Department of Genetics, MBC 3, Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh, 11211, Saudi Arabia
| | - Brian F Meyer
- Department of Genetics, MBC 3, Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh, 11211, Saudi Arabia
| | - Yufei Shi
- Department of Genetics, MBC 3, Centre for Genomic Medicine, King Faisal Specialist Hospital and Research Center, P.O. Box 3354, Riyadh, 11211, Saudi Arabia.
| |
Collapse
|
29
|
Mohaidat Z, Bodoor K, Almomani R, Alorjani M, Awwad MA, Bany-Khalaf A, Al-Batayneh K. Hereditary multiple osteochondromas in Jordanian patients: Mutational and immunohistochemical analysis of EXT1 and EXT2 genes. Oncol Lett 2020; 21:151. [PMID: 33552269 PMCID: PMC7798038 DOI: 10.3892/ol.2020.12412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 11/26/2020] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the molecular characteristics of hereditary multiple osteochondromas (HMO) in a subset of Jordanian patients with a focus on the genetic variants of exostosin (EXT1)/(EXT2) and their protein expression. Patients with HMO and their family members were included. Recorded clinical characteristics included age, sex, tumors number and location, joint deformities and associated functional limitations. Mutational analysis of EXT1 and EXT2 exonic regions was performed. Immunohistochemical staining for EXT1 and EXT2 was performed manually using two different commercially available rabbit anti-human EXT1 and EXT2 antibodies. A total of 16 patients with HMO from nine unrelated families were included, with a mean age of 13.9 years. A total of 75% (12/16) of the patients were male and (69%) (11/16) had a mild disease (class I). EXT mutation analysis revealed only EXT1 gene mutations in 13 patients. Seven variants were detected, among which three were novel: c.1019G>A, p. (Arg340His), c.962+1G>A and c.1469del, p. (Leu490Argfs*9). Of the 16 patients, 3 did not harbor any mutations for either EXT1 or EXT2. Immunohistochemical examination revealed decreased expression of EXT1 protein in all patients with EXT1 mutation. Surprisingly, EXT2 protein was not detected in these patients, although none had EXT2 mutations. The majority of Jordanian patients with HMO, who may represent an ethnic group that is infrequently investigated, were males and had a mild clinical disease course; whereas most patients with EXT1 gene mutations were not necessarily associated with a severe clinical disease course. The role of EXT2 gene remains a subject of debate, since patients with EXT1 mutations alone did not express the non-mutated EXT2 gene.
Collapse
Affiliation(s)
- Ziyad Mohaidat
- Orthopedic Division, Special Surgery Department, Faculty of Medicine, Jordan University of Science and Technology, King Abdullah University Hospital, Irbid 22110, Jordan
| | - Khaldon Bodoor
- Department of Applied Biology, Faculty of Science, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Rowida Almomani
- Department of Laboratory Medical Sciences, Faculty of Science, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mohammed Alorjani
- Department of Pathology, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Mohammad-Akram Awwad
- Department of Clinical Sciences, Faculty of Medicine, Yarmouk University, Irbid 21110, Jordan
| | - Audai Bany-Khalaf
- Orthopedic Division, Special Surgery Department, Faculty of Medicine, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Khalid Al-Batayneh
- Department of Biology, Faculty of Sciences, Yarmouk University, Irbid 21110, Jordan
| |
Collapse
|
30
|
Wang Y, Zhong L, Xu Y, Ding L, Ji Y, Schutz S, Férec C, Cooper DN, Xu C, Chen JM, Luo Y. EXT1 and EXT2 Variants in 22 Chinese Families With Multiple Osteochondromas: Seven New Variants and Potentiation of Preimplantation Genetic Testing and Prenatal Diagnosis. Front Genet 2020; 11:607838. [PMID: 33414810 PMCID: PMC7783290 DOI: 10.3389/fgene.2020.607838] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/02/2020] [Indexed: 11/13/2022] Open
Abstract
Multiple osteochondromas (MO), the most common type of benign bone tumor, is an autosomal dominant skeletal disorder characterized by multiple cartilage-capped bony protuberances. In most cases, EXT1 and EXT2, which encode glycosyltransferases involved in the biosynthesis of heparan sulfate, are the genes responsible. Here we describe the clinical, phenotypic and genetic characterization of MO in 22 unrelated Chinese families involving a total of 60 patients. Variant detection was performed by means of a battery of different techniques including Sanger sequencing and whole-exome sequencing (WES). The pathogenicity of the missense and splicing variants was explored by means of in silico prediction algorithms. Sixteen unique pathogenic variants, including 10 in the EXT1 gene and 6 in the EXT2 gene, were identified in 18 (82%) of the 22 families. Fourteen (88%) of the 16 variants were predicted to give rise to truncated proteins whereas the remaining two were missense. Seven variants were newly described here, further expanding the spectrum of MO-causing variants in the EXT1 and EXT2 genes. More importantly, the identification of causative variants allowed us to provide genetic counseling to 8 MO patients in terms either of preimplantation genetic testing (PGT) or prenatal diagnosis, thereby preventing the reoccurrence of MO in the corresponding families. This study is the first to report the successful implementation of PGT in MO families and describes the largest number of subjects undergoing prenatal diagnosis to date.
Collapse
Affiliation(s)
- Ye Wang
- Fetal Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liangying Zhong
- Department of Laboratory Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yan Xu
- Reproductive Medicine Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Reproductive Medicine, Guangzhou, China
| | - Lei Ding
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuanjun Ji
- Fetal Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Sacha Schutz
- Inserm, Univ Brest, EFS, UMR 1078, GGB, Brest, France
- CHRU Brest, Brest, France
| | - Claude Férec
- Inserm, Univ Brest, EFS, UMR 1078, GGB, Brest, France
- CHRU Brest, Brest, France
| | - David N. Cooper
- School of Medicine, Institute of Medical Genetics, Cardiff University, Cardiff, United Kingdom
| | - Caixia Xu
- Research Center for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jian-Min Chen
- Inserm, Univ Brest, EFS, UMR 1078, GGB, Brest, France
| | - Yanmin Luo
- Fetal Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
31
|
The Close Relationship between the Golgi Trafficking Machinery and Protein Glycosylation. Cells 2020; 9:cells9122652. [PMID: 33321764 PMCID: PMC7764369 DOI: 10.3390/cells9122652] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
Glycosylation is the most common post-translational modification of proteins; it mediates their correct folding and stability, as well as their transport through the secretory transport. Changes in N- and O-linked glycans have been associated with multiple pathological conditions including congenital disorders of glycosylation, inflammatory diseases and cancer. Glycoprotein glycosylation at the Golgi involves the coordinated action of hundreds of glycosyltransferases and glycosidases, which are maintained at the correct location through retrograde vesicle trafficking between Golgi cisternae. In this review, we describe the molecular machinery involved in vesicle trafficking and tethering at the Golgi apparatus and the effects of mutations in the context of glycan biosynthesis and human diseases.
Collapse
|
32
|
Trombetta A, Migliarino V, Faletra F, Barbi E, Tornese G. An unusual diagnosis for an usual test. Ital J Pediatr 2020; 46:81. [PMID: 32522262 PMCID: PMC7285577 DOI: 10.1186/s13052-020-00846-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/03/2020] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Hereditary multiple osteochondromas (HMO) is a genetic condition characterized by the presence of multiple osteochondromas, usually at the lateral side of the most active growth plate of a long bone. These lesions may persist, be asymptomatic during childhood, and may increase in number and size until growth plates close. Therefore, diagnosis of HMO in children and young people can be challenging; while short stature can be more evident at the onset of puberty, asymptomatic ostheocondromas can progress into different degrees of orthopedic deformity. Moreover, multiple complications may arise due to the presence of osteochondromas, including tendon and compression muscle pain, neurovascular disorders, obstetric problems, scoliosis and malignant transformation into secondary peripheral chondrosarcoma in adulthood.
Case presentation
We report the case of a girl admitted to our Institute for growth delay. While laboratory tests, including growth hormone stimulation test, were normal, left hand X-ray revealed multiple osteochondromas, suggestive for HMO. The genetic test for EXT1 and EXT2 genes confirmed the radiological diagnosis, with a mutation inherited from the mother who displayed the same radiological abnormalities along with recurrent limb pain episodes.
Conclusions
HMO is a genetic condition whose diagnosis can be challenging, especially in females. Every pediatricians should consider a skeletal dysplasia in case of unexplained growth delay and a skeletal survey might be fundamental in reaching a diagnosis.
Collapse
|
33
|
Hassan N, Greve B, Espinoza-Sánchez NA, Götte M. Cell-surface heparan sulfate proteoglycans as multifunctional integrators of signaling in cancer. Cell Signal 2020; 77:109822. [PMID: 33152440 DOI: 10.1016/j.cellsig.2020.109822] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/28/2020] [Accepted: 10/29/2020] [Indexed: 12/15/2022]
Abstract
Proteoglycans (PGs) represent a large proportion of the components that constitute the extracellular matrix (ECM). They are a diverse group of glycoproteins characterized by a covalent link to a specific glycosaminoglycan type. As part of the ECM, heparan sulfate (HS)PGs participate in both physiological and pathological processes including cell recruitment during inflammation and the promotion of cell proliferation, adhesion and motility during development, angiogenesis, wound repair and tumor progression. A key function of HSPGs is their ability to modulate the expression and function of cytokines, chemokines, growth factors, morphogens, and adhesion molecules. This is due to their capacity to act as ligands or co-receptors for various signal-transducing receptors, affecting pathways such as FGF, VEGF, chemokines, integrins, Wnt, notch, IL-6/JAK-STAT3, and NF-κB. The activation of those pathways has been implicated in the induction, progression, and malignancy of a tumor. For many years, the study of signaling has allowed for designing specific drugs targeting these pathways for cancer treatment, with very positive results. Likewise, HSPGs have become the subject of cancer research and are increasingly recognized as important therapeutic targets. Although they have been studied in a variety of preclinical and experimental models, their mechanism of action in malignancy still needs to be more clearly defined. In this review, we discuss the role of cell-surface HSPGs as pleiotropic modulators of signaling in cancer and identify them as promising markers and targets for cancer treatment.
Collapse
Affiliation(s)
- Nourhan Hassan
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany; Biotechnology Program, Department of Chemistry, Faculty of Science, Cairo University, Egypt
| | - Burkhard Greve
- Department of Radiotherapy-Radiooncology, Münster University Hospital, Albert-Schweitzer-Campus 1, A1, 48149 Münster, Germany
| | - Nancy A Espinoza-Sánchez
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany; Department of Radiotherapy-Radiooncology, Münster University Hospital, Albert-Schweitzer-Campus 1, A1, 48149 Münster, Germany.
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Münster, Germany.
| |
Collapse
|
34
|
Kawashima K, Ogawa H, Komura S, Ishihara T, Yamaguchi Y, Akiyama H, Matsumoto K. Heparan sulfate deficiency leads to hypertrophic chondrocytes by increasing bone morphogenetic protein signaling. Osteoarthritis Cartilage 2020; 28:1459-1470. [PMID: 32818603 PMCID: PMC7606622 DOI: 10.1016/j.joca.2020.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 08/03/2020] [Accepted: 08/11/2020] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Exostosin-1 (EXT1) and EXT2 are the major genetic etiologies of multiple hereditary exostoses and are essential for heparan sulfate (HS) biosynthesis. Previous studies investigating HS in several mouse models of multiple hereditary exostoses have reported that aberrant bone morphogenetic protein (BMP) signaling promotes osteochondroma formation in Ext1-deficient mice. This study examined the mechanism underlying the effects of HS deficiency on BMP/Smad signaling in articular cartilage in a cartilage-specific Ext-/- mouse model. METHOD We generated mice with a conditional Ext1 knockout in cartilage tissue (Ext1-cKO mice) using Prg4-Cre transgenic mice. Structural cartilage alterations were histologically evaluated and phospho-Smad1/5/9 (pSmad1/5/9) expression in mouse chondrocytes was analyzed. The effect of pharmacological intervention of BMP signaling using a specific inhibitor was assessed in the articular cartilage of Ext1-cKO mice. RESULTS Hypertrophic chondrocytes were significantly more abundant (P = 0.021) and cartilage thickness was greater in Ext1-cKO mice at 3 months postnatal than in control littermates (P = 0.036 for femur; and P < 0.001 for tibia). However, osteoarthritis did not spontaneously occur before the 1-year follow-up. matrix metalloproteinase (MMP)-13 and adamalysin-like metalloproteinases with thrombospondin motifs(ADAMTS)-5 were upregulated in hypertrophic chondrocytes of transgenic mice. Immunostaining and western blotting revealed that pSmad1/5/9-positive chondrocytes were more abundant in the articular cartilage of Ext1-cKO mice than in control littermates. Furthermore, the BMP inhibitor significantly decreased the number of hypertrophic chondrocytes in Ext1-cKO mice (P = 0.007). CONCLUSIONS HS deficiency in articular chondrocytes causes chondrocyte hypertrophy, wherein upregulated BMP/Smad signaling partially contributes to this phenotype. HS might play an important role in maintaining the cartilaginous matrix by regulating BMP signaling.
Collapse
Affiliation(s)
- K. Kawashima
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Japan
| | - H. Ogawa
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Japan
| | - S. Komura
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Japan
| | - T. Ishihara
- Innovative and Clinical Research Promotion Center, Gifu University Hospital, 1-1 Yanagido, Gifu, Japan
| | - Y. Yamaguchi
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - H. Akiyama
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Japan
| | - K. Matsumoto
- Department of Orthopaedic Surgery, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, Japan,Address correspondence and reprint requests to: K. Matsumoto, Department of Orthopedic Surgery, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu, 501-1194, Japan. Tel.: 81-58-230-6333; Fax: 81-58-230-6334. (K. Matsumoto)
| |
Collapse
|
35
|
Shen Y, Zhang L, Chen B, Dong L, Wang Y, Wang S. Novel deletion and 2397 G>T mutations of the EXT1 gene identified in two Chinese pedigrees with hereditary multiple exostoses using exon sequencing. Transl Pediatr 2020; 9:619-628. [PMID: 33209724 PMCID: PMC7658772 DOI: 10.21037/tp-20-191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Hereditary multiple exostoses (HME), a rare genetic pediatric disorder, has a peculiar pathogenic mechanism. The results of previous studies have shown that HME is associated with mutations of the EXT1 and EXT2 genes at a molecular genetics level. In our study, two families who received therapy in the Department of Orthopedics of Shanghai Children's Hospital between June, 2017 and November, 2018 were recruited, and a mutational analysis of the EXT1 genes was conducted to further elucidating the relationship between HME and EXT1. METHODS Venous blood samples were collected from individuals with HME and their families. Exon sequencing and RT-PCR were performed to comprehensively analyze 11 exons of the EXT1 gene. RESULTS The deletion of exon 7 and the 2397 G>T mutation in exon 7 caused deletion mutation and nonsense mutation only in the HME patients. The mutations in exon 7 were tested and verified by Sanger sequencing. RT-PCR showed that the mRNA expression of EXT1 was significantly decreased in the mutation samples compared with the normal samples, which exerted a great influence on the function of EXT1. CONCLUSIONS This study identified new mutation sites for the pathogenesis of HME and further clarified the relationship between HME and EXT1.
Collapse
Affiliation(s)
- Yang Shen
- Department of Orthopedics, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Lei Zhang
- The First Maternity and Infant Hospital Affiliated to Tongji University, Shanghai, China
| | - Bosong Chen
- Department of Orthopedics, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Liangchao Dong
- Department of Orthopedics, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yicheng Wang
- Department of Orthopedics, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Sun Wang
- Department of Orthopedics, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
36
|
Mordenti M, Shih F, Boarini M, Pedrini E, Gnoli M, Antonioli D, Tremosini M, Sangiorgi L. The natural history of multiple osteochondromas in a large Italian cohort of pediatric patients. Bone 2020; 139:115499. [PMID: 32592948 DOI: 10.1016/j.bone.2020.115499] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 06/04/2020] [Accepted: 06/14/2020] [Indexed: 12/17/2022]
Abstract
IMPORTANCE Multiple osteochondromas is a rare hereditary skeletal disorder, characterized by bony protrusions arising from growth plates on long bones during skeletal development. The disorder frequently leads to diminished stature, deformities and functional limitations. Understanding of the natural history of multiple osteochondromas and its evolution in children and adolescents is limited. OBJECTIVE To provide valuable information on the natural history of multiple osteochondromas, to inform recommendations for treatment and prevent impairments caused by osteochondromas. DESIGN This retrospective cohort study in children with multiple osteochondromas includes longitudinal data collected from first to last follow-up visit for patient demographics, and over 36 months for disease evolution. SETTING Data were collected from the Registry of Multiple Osteochondromas, which includes data from circa 1200 patients with multiple osteochondromas treated from 2003 to 2017 at IRCCS Istituto Ortopedico Rizzoli in Bologna. PARTICIPANTS Patients ≤18 years with multiple osteochondromas, who provided written informed consent and had data for ≥1 12-month follow-up visit. MAIN OUTCOME(S) AND MEASUREMENT(S) Demographics, clinical features, incidence of surgeries, and disease evolution (progression or regression) were assessed. Results were summarized using descriptive statistics, annual rates of new clinical features and surgeries, and Kaplan-Meier estimates. Patient height was evaluated following Italian growth charts. RESULTS 158 patients were included in these analyses. Throughout follow-up, 80.4% of patients developed new osteochondromas, 57.6% developed new deformities, 23.4% developed new functional limitation(s). New osteochondroma(s) were developed by 28.5% patients by Month 12, 39.9% at Month 24, 50% at Month 36. Most new osteochondromas were detected in the younger population; patients aged 0-4 years underwent a significantly higher number of lesions within 12, 24 and 36 months of follow-up. The overall incidence of patients with ≥1 new deformity within 12 months was 17.7%, with incidences decreasing with increasing age (p = .023). In addition, the analyses on height highlight that 13 years is a cut off age for slow growth of the stature (p < .0005). At last follow-up visit, 46.2% of patients had disease progression, while regression (spontaneous and surgical) occurred in 7.6% (p = .007). CONCLUSIONS AND RELEVANCE This natural history study reports the main set of clinically relevant data for patients with multiple osteochondromas during skeletal development, providing insight for patient management and development of therapeutic interventions.
Collapse
Affiliation(s)
- Marina Mordenti
- Department of Medical Genetics and Rare Orthopedic Diseases and CLIBI Laboratory, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.
| | | | - Manila Boarini
- Department of Medical Genetics and Rare Orthopedic Diseases and CLIBI Laboratory, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.
| | - Elena Pedrini
- Department of Medical Genetics and Rare Orthopedic Diseases, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.
| | - Maria Gnoli
- Department of Medical Genetics and Rare Orthopedic Diseases, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.
| | - Diego Antonioli
- Ward of Pediatric Orthopedics and Traumatology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.
| | - Morena Tremosini
- Department of Medical Genetics and Rare Orthopedic Diseases, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.
| | - Luca Sangiorgi
- Department of Medical Genetics and Rare Orthopedic Diseases and CLIBI Laboratory, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.
| |
Collapse
|
37
|
A Novel Prognostic Index Based on the Analysis of Glycolysis-Related Genes in Head and Neck Squamous Cell Carcinomas. JOURNAL OF ONCOLOGY 2020; 2020:7353874. [PMID: 33029143 PMCID: PMC7532401 DOI: 10.1155/2020/7353874] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 08/29/2020] [Indexed: 12/24/2022]
Abstract
Aims The preferential dependence on glycolysis as a pathway of energy metabolism is a hallmark of cancer cells. However, the prognostic significance of glycolysis-related genes in head and neck squamous cell carcinoma (HNSCC) remains obscure. The purpose of this study was to identify glycolysis-related genes of prognostic value in HNSCC. Results Transcriptional and clinical data of 544 HNSCC samples were obtained from The Cancer Genome Atlas (TCGA) dataset. By gene set enrichment analysis (GSEA) and by employing a univariate and subsequently a stepwise multivariate Cox proportional regression model, eight glycolysis-related genes of prognostic significance in HNSCC (KIF2A, JMJD8, HMMR, STC2, HK1, EXT2, GPR8, and STC1) were identified. The patients were clustered into two groups (high and low risk) based on the expression of these genes. High-risk patients had significantly a shorter overall survival than low-risk patients. Furthermore, a new prognostic indicator based on selected glycolysis-related genes was developed by multivariate Cox analysis that proved to be a better predictor of patient outcome compared to other clinical factors. Conclusion Our findings provide new insights into the role of glycolysis in HNSCC. The identified genes predict the patient prognosis and might substantially contribute to the development of individualized treatments.
Collapse
|
38
|
Li Y, Lin X, Zhu M, Li J, Yuan Z, Xu H. Whole‑exome sequencing identifies a novel mutation of SLC20A2 (c.C1849T) as a possible cause of hereditary multiple exostoses in a Chinese family. Mol Med Rep 2020; 22:2469-2477. [PMID: 32705272 PMCID: PMC7411400 DOI: 10.3892/mmr.2020.11298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 06/04/2020] [Indexed: 12/11/2022] Open
Abstract
Although the main causative genes for hereditary multiple exostoses (HME) are exostosin (EXT)‑1 and EXT‑2, there are numerous patients with HME without EXT‑1 and EXT‑2 mutations. The present study aimed to identify novel candidate genes for the development of HME in patients without EXT‑1 and EXT‑2 mutations. Whole‑exome sequencing was performed in a Chinese family with HME and without EXT‑1 and EXT‑2 mutations, followed by a combined bioinformatics pipeline including annotation and filtering processes to identify candidate variants. Candidate variants were then validated using Sanger sequencing. A total of 1,830 original variants were revealed to be heterozygous mutations in three patients with HME which were not present in healthy controls. Two mutations [c.C1849T in solute carrier family 20 member 2 (SLC20A2) and c.G506A in leucine zipper and EF‑hand containing transmembrane protein 1 (LETM1)] were identified as possible causative variants for HME through a bioinformatics filtering procedure and harmful prediction. Sanger sequencing results confirmed these two mutations in all patients with HME. A mutation in SLC20A2 (c.C1849T) led to a change in an amino acid (p.R617C), which may be involved in the development of HME by inducing metabolic disorders of phosphate and abnormal proliferation and differentiation in chondrocytes. In conclusion, the present study revealed two mutations [SLC20A2 (c.C1849T) and LETM1 (c.G506A) in a Chinese family with HME. The mutation in SLC20A2 (c.C1849T)] was more likely to be involved in the development of HME.
Collapse
Affiliation(s)
- Yiqiang Li
- Department of Pediatric Orthopedics, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510623, P.R. China
| | - Xuemei Lin
- Department of Pediatric Orthopedics, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510623, P.R. China
| | - Mingwei Zhu
- Department of Pediatric Orthopedics, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510623, P.R. China
| | - Jingchun Li
- Department of Pediatric Orthopedics, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510623, P.R. China
| | - Zhe Yuan
- Department of Pediatric Orthopedics, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510623, P.R. China
| | - Hongwen Xu
- Department of Pediatric Orthopedics, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510623, P.R. China
| |
Collapse
|
39
|
Kim DH, Lee HS, Mun YH, Koh S, Park JS, Lee SM, Kang NW, Lee MY, Cho CW, Kim DD, Lee JY. An overview of chondrosarcoma with a focus on nanoscale therapeutics. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2020. [DOI: 10.1007/s40005-020-00492-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
40
|
Liang C, Wang YJ, Wei YX, Dong Y, Zhang ZC. Identification of Novel EXT Mutations in Patients with Hereditary Multiple Exostoses Using Whole-Exome Sequencing. Orthop Surg 2020; 12:990-996. [PMID: 32293802 PMCID: PMC7307237 DOI: 10.1111/os.12660] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 02/11/2020] [Accepted: 02/19/2020] [Indexed: 12/16/2022] Open
Abstract
Objective To find novel potential gene mutations other than EXT1 and EXT2 mutations, to expand the mutational spectrum of EXT and to explore the correlation between clinical outcome and genotype in patients with hereditary multiple exostoses (HME). Methods The study recruited seven families diagnosed with multiple osteochondromas (MO). Family histories and clinical information were collected in detail through comprehensive physical and image examination. Patients with deformities and functional limitations were classified as “severe” and the remaining without functional limitations were classified as “mild,” in accordance with previous study. Whole‐exome sequencing (WES) was performed on a total of 13 affected individuals, 1 available unaffected relative, and 10 healthy unrelated individuals. Sanger sequencing was used to validate the screened mutations. Finally, the structural change in protein caused by pathogenic mutations was analyzed using information from the relevant database online and we attempted to correlate clinical phenotype with genotype in patients with HME. Results Other than EXT1 and EXT2, no novel potential gene mutations were found through WES. We identified nine heterozygous mutations in EXT1 or EXT2. Of these mutations, four have not been reported previously. These are c.996delT in exon 2 of EXT1 (family 1), c.544C > T in exon 3 of EXT2 (family 2), c.1171C > T in exon 7 of EXT2 (family 5), and c.823–824delAA in exon 5 of EXT1 (family 7). The other five mutations have already been reported in previous works. It was surprising that we found two mutation sites, in exon 2 and exon 5, respectively, of EXT1 in 1 patient diagnosed with MO, when his father had two mutation sites, in exon 6 and exon 5, respectively, of EXT1 and EXT2 (family 4). In addition, 1 patient showed degeneration, while his father only exhibited slight symptoms (family 7). In our study, among 51 affected patients in seven families, the sex ratio (male vs female) was 58.9% (n = 30) vs 41.2% (n = 21). Male patients seemed to show more severe symptoms compared to females, but because the sample was small, we did not obtain statistically significance results. Conclusion Whole‐exome sequencing to screen pathogenic gene mutations was applied successfully. Although no third‐gene mutation associated with HME was found, a total of nine mutations across EXT1 and EXT2 were identified, four of which are novel. Our results expand the mutational spectrum of EXT and can be used in genetic counseling and prenatal diagnosis for patients with MO.
Collapse
Affiliation(s)
- Chao Liang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yong-Jie Wang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yu-Xuan Wei
- Department of Orthopaedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Sciences and Peking Union Medical College, Shenzhen, China
| | - Yang Dong
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zhi-Chang Zhang
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
41
|
Abstract
Multiple hereditary exostoses (MHE) and enchondromatosis are rare multifocal benign disorders usually causing skeletal deformities appearing already in childhood. MHE is a dominant autosomal inherited disorder characterized by multiple osteochondromas (exostoses) growing outward from the metaphyses of long bones as well as from flat bones. They may cause reduced joint motion and pain due to tendon, muscle, and nerve compression. Enchondromatosis (or Ollier's disease) is a noninherited disorder characterized by the presence of multiple intraosseous enchondromas located asymmetrically in the skeleton and with a wide variation regarding location, size, and number ranging from the involvement of a single hand to the involvement of the entire skeleton. It can occur together with soft-tissue hemangiomas in Maffucci's syndrome. Clinical problems caused by the enchondromas are mainly related to skeletal deformities causing malalignment and restricted motion of joint. In both disorders, there is a risk of malignant transformation as well as secondary degenerative joint changes.
Collapse
Affiliation(s)
- Anne Grethe Jurik
- Department of Radiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 35, Entrance C, location C118/ reference point C109, 8200, Aarhus N, Denmark.
| |
Collapse
|
42
|
Fusco C, Nardella G, Fischetto R, Copetti M, Petracca A, Annunziata F, Augello B, D'Asdia MC, Petrucci S, Mattina T, Rella A, Cassina M, Bengala M, Biagini T, Causio FA, Caldarini C, Brancati F, De Luca A, Guarnieri V, Micale L, D'Agruma L, Castori M. Mutational spectrum and clinical signatures in 114 families with hereditary multiple osteochondromas: insights into molecular properties of selected exostosin variants. Hum Mol Genet 2020; 28:2133-2142. [PMID: 30806661 DOI: 10.1093/hmg/ddz046] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/22/2019] [Accepted: 02/22/2019] [Indexed: 01/05/2023] Open
Abstract
Hereditary multiple osteochondromas (HMO) is a rare autosomal dominant skeletal disorder, caused by heterozygous variants in either EXT1 or EXT2, which encode proteins involved in the biogenesis of heparan sulphate. Pathogenesis and genotype-phenotype correlations remain poorly understood. We studied 114 HMO families (158 affected individuals) with causative EXT1 or EXT2 variants identified by Sanger sequencing, or multiplex ligation-dependent probe amplification and qPCR. Eighty-seven disease-causative variants (55 novel and 32 known) were identified including frameshift (42%), nonsense (32%), missense (11%), splicing (10%) variants and genomic rearrangements (5%). Informative clinical features were available for 42 EXT1 and 27 EXT2 subjects. Osteochondromas were more frequent in EXT1 as compared to EXT2 patients. Anatomical distribution of lesions showed significant differences based on causative gene. Microscopy analysis for selected EXT1 and EXT2 variants verified that EXT1 and EXT2 mutants failed to co-localize each other and loss Golgi localization by surrounding the nucleus and/or assuming a diffuse intracellular distribution. In a cell viability study, cells expressing EXT1 and EXT2 mutants proliferated more slowly than cells expressing wild-type proteins. This confirms the physiological relevance of EXT1 and EXT2 Golgi co-localization and the key role of these proteins in the cell cycle. Taken together, our data expand genotype-phenotype correlations, offer further insights in the pathogenesis of HMO and open the path to future therapies.
Collapse
Affiliation(s)
- Carmela Fusco
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Grazia Nardella
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.,Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Rita Fischetto
- Unit of Metabolic Diseases and Medical Genetics, University Hospital, P.O. Giovanni XXIII Hospital, Bari, Italy
| | - Massimiliano Copetti
- Unit of Biostatistics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Antonio Petracca
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Francesca Annunziata
- Unit of Molecular Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Bartolomeo Augello
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Maria Cecilia D'Asdia
- Unit of Molecular Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Simona Petrucci
- Unit of Molecular Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy.,Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Teresa Mattina
- Unit of Medical Genetics, University of Catania, Catania, Italy
| | - Annalisa Rella
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Matteo Cassina
- Unit of Clinical Genetics, Department of Women's and Children's Health, University of Padua, Padua, Italy
| | - Mario Bengala
- Dipartimento di Oncoematologia, U.O.C Laboratorio di Genetica Medica, Fondazione Policlinico di Tor Vergata, Rome, Italy
| | - Tommaso Biagini
- Unit of Bioinformatics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Francesco Andrea Causio
- Unit of Metabolic Diseases and Medical Genetics, University Hospital, P.O. Giovanni XXIII Hospital, Bari, Italy
| | - Camilla Caldarini
- Division of Orthopedics and Traumatology, Azienda Socio Sanitaria Territoriale Gaetano Pini, Milan, Italy
| | - Francesco Brancati
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy.,Laboratory of Molecular and Cell Biology, Istituto Dermopatico dell'Immacolata (IDI) IRCCS, Rome, Italy
| | - Alessandro De Luca
- Unit of Molecular Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Vito Guarnieri
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Lucia Micale
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Leonardo D'Agruma
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Marco Castori
- Division of Medical Genetics, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| |
Collapse
|
43
|
Yudistira A, Fujiwara Y, Sukmajaya WP, Pandiangan RAH, Abduh M. Multiple osteochondromas of the cervical spine, a potential cause of radiculopathy in the elderly: A case report and review of literature. Int J Surg Case Rep 2020; 69:13-19. [PMID: 32248010 PMCID: PMC7132051 DOI: 10.1016/j.ijscr.2020.03.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 03/03/2020] [Accepted: 03/11/2020] [Indexed: 11/26/2022] Open
Abstract
Osteochondroma is the most common type of bone tumor. It rarely arises on the cervical spine. It rarely occurs in the elderly. Although rare, it may be considered as a differential diagnosis of radiculopathy among the elderly.
Introduction Osteochondroma is the most prevalent type of bone tumour, often arising in the long bones. Most are found in the third decade of life, and rarely in the axial skeleton of elderly patients. This case report aims to highlight the possibility of this differential diagnosis in an elderly patient with symptoms of radiculopathy, aside from degenerative cause. Presentation of case A 76-years-old woman presented to the authors’ hospital with complaints of pain and numbness of the left suboccipital and preauricular region. There was hypoesthesia of left C2 and C3 dermatome without any signs of myelopathy. CT-scan and MRI showed an expansile bone lesion from the posterior arch of C1 and lamina of C2. The patient underwent laminectomy of C1 and hemilaminectomy of C2. Postoperative histopathologic examination showed the features of osteochondroma. Radiologic follow-up after nine months showed no sign of recurrence. After eighteen months, the patient was ambulatory with complaints of fingers numbness and moderate neck pain. Discussion Osteochondroma usually arises in long bone metaphysis. This lesion normally ceases to grow with growth plate closure, but other findings suggest it may continue to grow beyond skeletal maturity. About 29.5% of all osteochondroma of the spine would cause radiculopathy, and 27% would cause myelopathy. The cervical spine osteochondroma is usually treated by en bloc resection through posterior approach. Conclusion Osteochondroma of the cervical spine in the elderly is rare. However, this diagnosis could be considered as a possible cause of radiculopathy in this age group aside from other degenerative causes.
Collapse
Affiliation(s)
- Andhika Yudistira
- Department of Orthopaedics and Traumatology, Saiful Anwar General Hospital-Universitas Brawijaya, Malang, Indonesia.
| | - Yasushi Fujiwara
- Department of Orthopaedic Surgery, Asa Citizens' Hospital, Hiroshima City, Japan
| | - William Putera Sukmajaya
- Department of Orthopaedics and Traumatology, Saiful Anwar General Hospital-Universitas Brawijaya, Malang, Indonesia
| | - Ray Asaf Hexa Pandiangan
- Department of Orthopaedics and Traumatology, Saiful Anwar General Hospital-Universitas Brawijaya, Malang, Indonesia
| | - Muhammad Abduh
- Department of Orthopaedics and Traumatology, Saiful Anwar General Hospital-Universitas Brawijaya, Malang, Indonesia
| |
Collapse
|
44
|
Heparanase: A Potential Therapeutic Target in Sarcomas. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:405-431. [PMID: 32274719 DOI: 10.1007/978-3-030-34521-1_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sarcomas comprise a heterogeneous group of rare malignancies of mesenchymal origin including more than 70 subtypes. They may arise in muscle, bone, cartilage and other connective tissues. Their high histological and genetic heterogeneity makes diagnosis and treatment very challenging. Deregulation of heparanase has been found in several sarcoma subtypes and high expression levels have been correlated with poor prognosis in Ewing's sarcoma and osteosarcoma. Altered expression of specific heparan sulfate proteoglycans and heparan sulfate biosynthetic enzymes has also been observed. Advances in molecular pathogenesis of sarcomas have evidenced the critical role of several heparan sulfate binding growth factors and receptor tyrosine kinases, highly interconnected with the microenvironment, in sustaining tumor growth and progression. Interference with heparanase/heparan sulfate functions represents a potential therapeutic approach in sarcoma. In this chapter, we summarize the current knowledge about the biological significance of heparanase expression and its potential as a therapeutic target in subtypes of both soft tissue and bone sarcomas. Particular emphasis is given to the involvement of heparan sulfate proteoglycans and their synthesizing and modifying enzymes in bone physiology and disorders leading up to the pathobiology of bone sarcomas. The chapter also describes the cooperation between exostin loss-of-function and heparanase upregulation in hereditary Multiple Osteochondroma syndrome as a paradigmatic example of constitutive alteration of the heparanase/heparan sulfate proteoglycan system which may contribute to progression to malignant secondary chondrosarcoma. Preclinical evidence of the role of heparanase as a promising therapeutic target in various sarcoma subtypes is finally resumed.
Collapse
|
45
|
D'Arienzo A, Andreani L, Sacchetti F, Colangeli S, Capanna R. Hereditary Multiple Exostoses: Current Insights. Orthop Res Rev 2019; 11:199-211. [PMID: 31853203 PMCID: PMC6916679 DOI: 10.2147/orr.s183979] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 10/11/2019] [Indexed: 12/31/2022] Open
Abstract
Hereditary multiple exostoses (HME), also called hereditary multiple osteochondromas, is a rare genetic disorder characterized by multiple osteochondromas that grow near the growth plates of bones such as the ribs, pelvis, vertebrae and especially long bones. The disease presents with various clinical manifestations including chronic pain syndromes, restricted range of motion, limb deformity, short stature, scoliosis and neurovascular alteration. Malignant transformation of exostosis is rarely seen. The disease has no medical treatment and surgery is only recommended in symptomatic exostoses or in cases where a malignant transformation is suspected. HME is mainly caused by mutations and functional loss of the EXT1 and EXT2 genes which encode glycosyltransferases, an enzyme family involved in heparan sulfate (HS) synthesis. However, the peculiar molecular mechanism that leads to the structural changes of the cartilage and to osteochondroma formation is still being studied. Basic science studies have recently shown new insights about altering the molecular and cellular mechanism caused by HS deficiency. Pediatricians, geneticists and orthopedic surgeons play an important role in the study and treatment of this severe pathology. Despite the recent significant advances, we still need novel insights to better specify the role of HS in signal transduction. The purpose of this review was to analyze the most relevant aspects of HME from the literature review, give readers an important tool to understand its clinical features and metabolic-pathogenetic mechanism, and to identify an effective treatment method. We focused on the aspects of the disease related to clinical management and surgical treatment in order to give up-to-date information that could be useful for following best clinical practice.
Collapse
Affiliation(s)
- Antonio D'Arienzo
- Department of Translational Research on New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Lorenzo Andreani
- Department of Translational Research on New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Federico Sacchetti
- Department of Translational Research on New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Simone Colangeli
- Department of Translational Research on New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Rodolfo Capanna
- Department of Translational Research on New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| |
Collapse
|
46
|
Recent Insights into Long Bone Development: Central Role of Hedgehog Signaling Pathway in Regulating Growth Plate. Int J Mol Sci 2019; 20:ijms20235840. [PMID: 31757091 PMCID: PMC6928971 DOI: 10.3390/ijms20235840] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 11/17/2019] [Accepted: 11/18/2019] [Indexed: 12/30/2022] Open
Abstract
The longitudinal growth of long bone, regulated by an epiphyseal cartilaginous component known as the “growth plate”, is generated by epiphyseal chondrocytes. The growth plate provides a continuous supply of chondrocytes for endochondral ossification, a sequential bone replacement of cartilaginous tissue, and any failure in this process causes a wide range of skeletal disorders. Therefore, the cellular and molecular characteristics of the growth plate are of interest to many researchers. Hedgehog (Hh), well known as a mitogen and morphogen during development, is one of the best known regulatory signals in the developmental regulation of the growth plate. Numerous animal studies have revealed that signaling through the Hh pathway plays multiple roles in regulating the proliferation, differentiation, and maintenance of growth plate chondrocytes throughout the skeletal growth period. Furthermore, over the past few years, a growing body of evidence has emerged demonstrating that a limited number of growth plate chondrocytes transdifferentiate directly into the full osteogenic and multiple mesenchymal lineages during postnatal bone development and reside in the bone marrow until late adulthood. Current studies with the genetic fate mapping approach have shown that the commitment of growth plate chondrocytes into the skeletal lineage occurs under the influence of epiphyseal chondrocyte-derived Hh signals during endochondral bone formation. Here, we discuss the valuable observations on the role of the Hh signaling pathway in the growth plate based on mouse genetic studies, with some emphasis on recent advances.
Collapse
|
47
|
Oliver GR, Tang X, Schultz-Rogers LE, Vidal-Folch N, Jenkinson WG, Schwab TL, Gaonkar K, Cousin MA, Nair A, Basu S, Chanana P, Oglesbee D, Klee EW. A tailored approach to fusion transcript identification increases diagnosis of rare inherited disease. PLoS One 2019; 14:e0223337. [PMID: 31577830 PMCID: PMC6774566 DOI: 10.1371/journal.pone.0223337] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 09/18/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND RNA sequencing has been proposed as a means of increasing diagnostic rates in studies of undiagnosed rare inherited disease. Recent studies have reported diagnostic improvements in the range of 7.5-35% by profiling splicing, gene expression quantification and allele specific expression. To-date however, no study has systematically assessed the presence of gene-fusion transcripts in cases of germline disease. Fusion transcripts are routinely identified in cancer studies and are increasingly recognized as having diagnostic, prognostic or therapeutic relevance. Isolated reports exist of fusion transcripts being detected in cases of developmental and neurological phenotypes, and thus, systematic application of fusion detection to germline conditions may further increase diagnostic rates. However, current fusion detection methods are unsuited to the investigation of germline disease due to performance biases arising from their development using tumor, cell-line or in-silico data. METHODS We describe a tailored approach to fusion candidate identification and prioritization in a cohort of 47 undiagnosed, suspected inherited disease patients. We modify an existing fusion transcript detection algorithm by eliminating its cell line-derived filtering steps, and instead, prioritize candidates using a custom workflow that integrates genomic and transcriptomic sequence alignment, biological and technical annotations, customized categorization logic, and phenotypic prioritization. RESULTS We demonstrate that our approach to fusion transcript identification and prioritization detects genuine fusion events excluded by standard analyses and efficiently removes phenotypically unimportant candidates and false positive events, resulting in a reduced candidate list enriched for events with potential phenotypic relevance. We describe the successful genetic resolution of two previously undiagnosed disease cases through the detection of pathogenic fusion transcripts. Furthermore, we report the experimental validation of five additional cases of fusion transcripts with potential phenotypic relevance. CONCLUSIONS The approach we describe can be implemented to enable the detection of phenotypically relevant fusion transcripts in studies of rare inherited disease. Fusion transcript detection has the potential to increase diagnostic rates in rare inherited disease and should be included in RNA-based analytical pipelines aimed at genetic diagnosis.
Collapse
Affiliation(s)
- Gavin R. Oliver
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Xiaojia Tang
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Laura E. Schultz-Rogers
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Noemi Vidal-Folch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - W. Garrett Jenkinson
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Tanya L. Schwab
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Krutika Gaonkar
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Margot A. Cousin
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Asha Nair
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Shubham Basu
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Pritha Chanana
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Devin Oglesbee
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Medical Genetics, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Eric W. Klee
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota, United States of America
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
48
|
Fatigue and pain in children and adults with multiple osteochondromas in Norway, a cross-sectional study. Int J Orthop Trauma Nurs 2019; 34:28-35. [DOI: 10.1016/j.ijotn.2019.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 12/18/2018] [Accepted: 02/04/2019] [Indexed: 11/16/2022]
|
49
|
Paganini C, Costantini R, Superti-Furga A, Rossi A. Bone and connective tissue disorders caused by defects in glycosaminoglycan biosynthesis: a panoramic view. FEBS J 2019; 286:3008-3032. [PMID: 31286677 DOI: 10.1111/febs.14984] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 05/22/2019] [Accepted: 07/04/2019] [Indexed: 02/06/2023]
Abstract
Glycosaminoglycans (GAGs) are a heterogeneous family of linear polysaccharides that constitute the carbohydrate moiety covalently attached to the protein core of proteoglycans, macromolecules present on the cell surface and in the extracellular matrix. Several genetic disorders of bone and connective tissue are caused by mutations in genes encoding for glycosyltransferases, sulfotransferases and transporters that are responsible for the synthesis of sulfated GAGs. Phenotypically, these disorders all reflect alterations in crucial biological functions of GAGs in the development, growth and homoeostasis of cartilage and bone. To date, up to 27 different skeletal phenotypes have been linked to mutations in 23 genes encoding for proteins involved in GAG biosynthesis. This review focuses on recent genetic, molecular and biochemical studies of bone and connective tissue disorders caused by GAG synthesis defects. These insights and future research in the field will provide a deeper understanding of the molecular pathogenesis of these disorders and will pave the way for developing common therapeutic strategies that might be targeted to a range of individual phenotypes.
Collapse
Affiliation(s)
- Chiara Paganini
- Department of Molecular Medicine, Unit of Biochemistry, University of Pavia, Italy
| | - Rossella Costantini
- Department of Molecular Medicine, Unit of Biochemistry, University of Pavia, Italy
| | - Andrea Superti-Furga
- Division of Genetic Medicine, Lausanne University Hospital, University of Lausanne, Switzerland
| | - Antonio Rossi
- Department of Molecular Medicine, Unit of Biochemistry, University of Pavia, Italy
| |
Collapse
|
50
|
Erickson LA, Inwards CY. Multiple Hereditary Osteochondromas. Mayo Clin Proc 2019; 94:1388-1389. [PMID: 31272585 DOI: 10.1016/j.mayocp.2019.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 05/09/2019] [Indexed: 11/27/2022]
Affiliation(s)
- Lori A Erickson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Carrie Y Inwards
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| |
Collapse
|