1
|
Avsievich E, Salimgereeva D, Maluchenko A, Antysheva Z, Voloshin M, Feidorov I, Glazova O, Abramov I, Maksimov D, Kaziakhmedova S, Bodunova N, Karnaukhov N, Volchkov P, Krupinova J. Pancreatic Neuroendocrine Tumor: The Case Report of a Patient with Germline FANCD2 Mutation and Tumor Analysis Using Single-Cell RNA Sequencing. J Clin Med 2024; 13:7621. [PMID: 39768544 PMCID: PMC11728285 DOI: 10.3390/jcm13247621] [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: 10/28/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 01/16/2025] Open
Abstract
Background: Neuroendocrine neoplasms are a rare and heterogeneous group of neoplasms. Small-sized (≤2 cm) pancreatic neuroendocrine tumors (PanNETs) are of particular interest as they are often associated with aggressive behavior, with no specific prognostic or progression markers. METHODS This article describes a clinical case characterized by a progressive growth of nonfunctional PanNET requiring surgical treatment in a patient with a germline FANCD2 mutation, previously not reported in PanNETs. The patient underwent whole exome sequencing and single-cell RNA sequencing. RESULTS The patient underwent surgical treatment. We confirmed the presence of the germline mutation FANCD2 and also detected the germline mutation WNT10A. The cellular composition of the PanNET was analyzed using single-cell sequencing, and the main cell clusters were identified. We analyzed the tumor genomics, and used the data to define the effect the germline FANCD2 mutation had. CONCLUSIONS Analysis of the mutational status of patients with PanNET may provide additional data that may influence treatment tactics, refine the plan for monitoring such patients, and provide more information about the pathogenesis of PanNET. PanNET research using scRNA-seq data may help in predicting the effect of therapy on neuroendocrine cells with FANCD2 mutations.
Collapse
Affiliation(s)
- Ekaterina Avsievich
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (E.A.); (D.S.); (M.V.); (I.F.); (O.G.); (I.A.); (N.B.); (N.K.); (P.V.)
- Moscow Center for Advanced Studies, Kulakova Street 20, Moscow 123592, Russia; (A.M.); (Z.A.); (D.M.); (S.K.)
- Federal Research Center for Innovator, Emerging Biomedical and Pharmaceutical Technologies, Moscow 125315, Russia
| | - Diana Salimgereeva
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (E.A.); (D.S.); (M.V.); (I.F.); (O.G.); (I.A.); (N.B.); (N.K.); (P.V.)
| | - Alesia Maluchenko
- Moscow Center for Advanced Studies, Kulakova Street 20, Moscow 123592, Russia; (A.M.); (Z.A.); (D.M.); (S.K.)
| | - Zoia Antysheva
- Moscow Center for Advanced Studies, Kulakova Street 20, Moscow 123592, Russia; (A.M.); (Z.A.); (D.M.); (S.K.)
- Federal Research Center for Innovator, Emerging Biomedical and Pharmaceutical Technologies, Moscow 125315, Russia
| | - Mark Voloshin
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (E.A.); (D.S.); (M.V.); (I.F.); (O.G.); (I.A.); (N.B.); (N.K.); (P.V.)
| | - Ilia Feidorov
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (E.A.); (D.S.); (M.V.); (I.F.); (O.G.); (I.A.); (N.B.); (N.K.); (P.V.)
| | - Olga Glazova
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (E.A.); (D.S.); (M.V.); (I.F.); (O.G.); (I.A.); (N.B.); (N.K.); (P.V.)
- Moscow Center for Advanced Studies, Kulakova Street 20, Moscow 123592, Russia; (A.M.); (Z.A.); (D.M.); (S.K.)
| | - Ivan Abramov
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (E.A.); (D.S.); (M.V.); (I.F.); (O.G.); (I.A.); (N.B.); (N.K.); (P.V.)
- Moscow Center for Advanced Studies, Kulakova Street 20, Moscow 123592, Russia; (A.M.); (Z.A.); (D.M.); (S.K.)
- Federal Research Center for Innovator, Emerging Biomedical and Pharmaceutical Technologies, Moscow 125315, Russia
| | - Denis Maksimov
- Moscow Center for Advanced Studies, Kulakova Street 20, Moscow 123592, Russia; (A.M.); (Z.A.); (D.M.); (S.K.)
- Federal Research Center for Innovator, Emerging Biomedical and Pharmaceutical Technologies, Moscow 125315, Russia
| | - Samira Kaziakhmedova
- Moscow Center for Advanced Studies, Kulakova Street 20, Moscow 123592, Russia; (A.M.); (Z.A.); (D.M.); (S.K.)
| | - Natalia Bodunova
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (E.A.); (D.S.); (M.V.); (I.F.); (O.G.); (I.A.); (N.B.); (N.K.); (P.V.)
| | - Nikolay Karnaukhov
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (E.A.); (D.S.); (M.V.); (I.F.); (O.G.); (I.A.); (N.B.); (N.K.); (P.V.)
| | - Pavel Volchkov
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (E.A.); (D.S.); (M.V.); (I.F.); (O.G.); (I.A.); (N.B.); (N.K.); (P.V.)
- Moscow Center for Advanced Studies, Kulakova Street 20, Moscow 123592, Russia; (A.M.); (Z.A.); (D.M.); (S.K.)
- Federal Research Center for Innovator, Emerging Biomedical and Pharmaceutical Technologies, Moscow 125315, Russia
| | - Julia Krupinova
- Moscow Clinical Scientific Center N.A. A.S. Loginov, Moscow 111123, Russia; (E.A.); (D.S.); (M.V.); (I.F.); (O.G.); (I.A.); (N.B.); (N.K.); (P.V.)
- Moscow Center for Advanced Studies, Kulakova Street 20, Moscow 123592, Russia; (A.M.); (Z.A.); (D.M.); (S.K.)
- Federal Research Center for Innovator, Emerging Biomedical and Pharmaceutical Technologies, Moscow 125315, Russia
| |
Collapse
|
2
|
Eghbali A, Safdari SM, Yousefi Roozbahani M, Tavajohi K, Hosseini S. Fanconi Anemia: Challenges in Diagnosis and Management-A Case Series Report. Clin Case Rep 2024; 12:e9583. [PMID: 39559288 PMCID: PMC11570422 DOI: 10.1002/ccr3.9583] [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: 07/19/2024] [Accepted: 10/13/2024] [Indexed: 11/20/2024] Open
Abstract
Fanconi anemia (FA) is a rare inherited disorder characterized by congenital abnormalities, progressive bone marrow failure, and a predisposition to malignancies. Detecting FA can be challenging, as it involves identifying increased chromosomal sensitivity to DNA cross-linking agents and detecting causative genetic variants via genome sequencing. We report two cases of siblings with FA, both confirmed to have the FANCD2 variant through whole-exome sequencing (WES). The first patient presented with epistaxis, petechiae, ecchymosis, and lower limb edema. The second patient exhibited epistaxis, diabetes, developmental delay, and physical abnormalities. Interestingly, both patients had negative results on the initial chromosomal breakage test with mitomycin C, a commonly used diagnostic tool for FA. However, further investigation with WES revealed the presence of the FANCD2 variant, confirming the FA diagnosis. This case report highlights the challenges in diagnosing FA, particularly when initial screening tests yield negative results. Molecular genetic testing, such as WES, can provide a definitive diagnosis and guide appropriate management strategies. Early and accurate diagnosis is crucial for improving outcomes in individuals with this potentially fatal illness, as promising advancements in treatments such as hematopoietic stem cell transplantation and gene therapy offer hope for addressing FA.
Collapse
Affiliation(s)
| | - Seyed Mehrab Safdari
- Department of Hematology and Blood Transfusion, School of Allied MedicineIran University of Medical SciencesTehranIran
| | - Maedeh Yousefi Roozbahani
- Department of Hematology and Blood Transfusion, School of Allied MedicineIran University of Medical SciencesTehranIran
| | | | - Soudabeh Hosseini
- Aliasghar Children HospitalTehranIran
- Department of Hematology and Blood Transfusion, School of Allied MedicineIran University of Medical SciencesTehranIran
| |
Collapse
|
3
|
Claes KBM, Rosseel T, De Leeneer K. Dealing with Pseudogenes in Molecular Diagnostics in the Next Generation Sequencing Era. Methods Mol Biol 2021; 2324:363-381. [PMID: 34165726 DOI: 10.1007/978-1-0716-1503-4_22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Presence of pseudogenes is a dreadful issue in next generation sequencing (NGS), because their contamination can interfere with the detection of variants in the genuine gene and generate false positive and false negative variants.In this chapter we focus on issues related to the application of NGS strategies for analysis of genes with pseudogenes in a clinical setting. The degree to which a pseudogene impacts the ability to accurately detect and map variants in its parent gene depends on the degree of similarity (homology) with the parent gene itself. Hereby, target enrichment and mapping strategies are crucial factors to avoid "contaminating" pseudogene sequences. For target enrichment, we describe advantages and disadvantages of PCR- and capture-based strategies. For mapping strategies, we discuss crucial parameters that need to be considered to accurately distinguish sequences of functional genes from pseudogenic sequences. Finally, we discuss some examples of genes associated with Mendelian disorders, for which interesting NGS approaches are described to avoid interference with pseudogene sequences.
Collapse
Affiliation(s)
| | - Toon Rosseel
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Kim De Leeneer
- Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
4
|
Understanding genetic epidemiology and population disparities of inherited blood cancer syndromes from integrative analysis of population genomics datasets. PEDIATRIC HEMATOLOGY ONCOLOGY JOURNAL 2021. [DOI: 10.1016/j.phoj.2021.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
5
|
Kazemi-Sefat GE, Keramatipour M, Talebi S, Kavousi K, Sajed R, Kazemi-Sefat NA, Mousavizadeh K. The importance of CDC27 in cancer: molecular pathology and clinical aspects. Cancer Cell Int 2021; 21:160. [PMID: 33750395 PMCID: PMC7941923 DOI: 10.1186/s12935-021-01860-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/01/2021] [Indexed: 12/17/2022] Open
Abstract
Background CDC27 is one of the core components of Anaphase Promoting complex/cyclosome. The main role of this protein is defined at cellular division to control cell cycle transitions. Here we review the molecular aspects that may affect CDC27 regulation from cell cycle and mitosis to cancer pathogenesis and prognosis. Main text It has been suggested that CDC27 may play either like a tumor suppressor gene or oncogene in different neoplasms. Divergent variations in CDC27 DNA sequence and alterations in transcription of CDC27 have been detected in different solid tumors and hematological malignancies. Elevated CDC27 expression level may increase cell proliferation, invasiveness and metastasis in some malignancies. It has been proposed that CDC27 upregulation may increase stemness in cancer stem cells. On the other hand, downregulation of CDC27 may increase the cancer cell survival, decrease radiosensitivity and increase chemoresistancy. In addition, CDC27 downregulation may stimulate efferocytosis and improve tumor microenvironment. Conclusion CDC27 dysregulation, either increased or decreased activity, may aggravate neoplasms. CDC27 may be suggested as a prognostic biomarker in different malignancies. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-01860-9.
Collapse
Affiliation(s)
- Golnaz Ensieh Kazemi-Sefat
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Shahid Hemmat Highway, P.O. Box: 14665-354, Tehran, 14496-14535, Iran
| | - Mohammad Keramatipour
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Talebi
- Department of Medical Genetics, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Kaveh Kavousi
- Laboratory of Complex Biological Systems and Bioinformatics (CBB), Department of Bioinformatics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Roya Sajed
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Shahid Hemmat Highway, P.O. Box: 14665-354, Tehran, 14496-14535, Iran
| | | | - Kazem Mousavizadeh
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Shahid Hemmat Highway, P.O. Box: 14665-354, Tehran, 14496-14535, Iran. .,Cellular and Molecular Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
6
|
Skibenes ST, Clausen I, Raaschou-Jensen K. Next-generation sequencing in hypoplastic bone marrow failure: What difference does it make? Eur J Haematol 2020; 106:3-13. [PMID: 32888355 DOI: 10.1111/ejh.13513] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 12/16/2022]
Abstract
Hypoplastic bone marrow failure is a diagnostic feature of multiple haematological disorders, which also share a substantial overlap of clinical symptoms. Hence, discrimination of underlying disorders in patients presenting with hypoplastic bone marrow failure remains a major challenge in the clinic. Recent next-generation sequencing (NGS) studies have broadened our understanding of the varying molecular mechanisms and advanced diagnostics of disorders exhibiting hypoplastic bone marrow failure. In this article, we present a literature review of NGS studies of haematological disorders associated with hypoplastic bone marrow failure and highlight the relevance of NGS for improved clinical diagnostics and decision-making.
Collapse
Affiliation(s)
- Sofie T Skibenes
- Department of Hematology, Odense University Hospital, Odense, Denmark
| | - Ida Clausen
- Department of Hematology, Odense University Hospital, Odense, Denmark
| | | |
Collapse
|
7
|
Negahdari S, Zamani M, Seifi T, Sedighzadeh S, Mazaheri N, Zeighami J, Sedaghat A, Saberi A, Hamid M, Keikhaei B, Radpour R, Shariati G, Galehdari H. Identification of Three Novel Mutations in the FANCA, FANCC, and ITGA2B Genes by Whole Exome Sequencing. Int J Prev Med 2020; 11:117. [PMID: 33088445 PMCID: PMC7554563 DOI: 10.4103/ijpvm.ijpvm_462_19] [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: 12/14/2019] [Accepted: 03/27/2020] [Indexed: 11/04/2022] Open
Abstract
Background Various blood diseases are caused by mutations in the FANCA, FANCC, and ITGA2B genes. Exome sequencing is a suitable method for identifying single-gene disease and genetic heterogeneity complaints. Methods Among families who were referred to Narges Genetic and PND Laboratory in 2015-2017, five families with a history of blood diseases were analyzed using the whole exome sequencing (WES) method. Results We detected two novel mutations (c.190-2A>G and c.2840C>G) in the FANCA gene, c. 1429dupA mutation in the FANCC gene, and c.1392A>G mutation in the ITGA2B gene. The prediction of variant pathogenicity has been done using bioinformatics tools such as Mutation taster PhD-SNP and polyphen2 and were confirmed by Sanger sequencing. Conclusions WES could be as a precise tool for identifying the pathologic variants in affected patient and heterozygous carriers among families. This highly successful technique will remain at the forefront of platelet and blood genomic research.
Collapse
Affiliation(s)
| | - Mina Zamani
- Narges Genetics Diagnostic Laboratory, Ahvaz, Iran.,Department of Genetics, Faculty of Sciences, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Tahereh Seifi
- Narges Genetics Diagnostic Laboratory, Ahvaz, Iran.,Department of Genetics, Faculty of Sciences, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Sahar Sedighzadeh
- Narges Genetics Diagnostic Laboratory, Ahvaz, Iran.,Department of Genetics, Faculty of Sciences, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | | | | | - Alireza Sedaghat
- Narges Genetics Diagnostic Laboratory, Ahvaz, Iran.,Health Research Institute, Diabetes Research Center, Ahvaz Jundishapur Universityof medical Sciences, Ahvaz, Iran
| | - Alihossein Saberi
- Narges Genetics Diagnostic Laboratory, Ahvaz, Iran.,Department of Genetics, Ahvaz Jundishapur University of medical Sciences, Ahvaz, Iran
| | - Mohammad Hamid
- Narges Genetics Diagnostic Laboratory, Ahvaz, Iran.,Department of Molecular Medicine, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Bijan Keikhaei
- Health Research Institute, Research Centre of Thalassemia and Hemoglobinopathies, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ramin Radpour
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.,Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Gholamreza Shariati
- Narges Genetics Diagnostic Laboratory, Ahvaz, Iran.,Department of Genetics, Ahvaz Jundishapur University of medical Sciences, Ahvaz, Iran
| | - Hamid Galehdari
- Health Research Institute, Research Centre of Thalassemia and Hemoglobinopathies, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
8
|
Crysandt M, Brings K, Beier F, Thiede C, Brümmendorf TH, Jost E. Germ line predisposition to myeloid malignancies appearing in adulthood. Expert Rev Hematol 2018; 11:625-636. [PMID: 29958021 DOI: 10.1080/17474086.2018.1494566] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Germ line predisposition to myeloid neoplasms has been incorporated in the WHO 2016 classification of myeloid neoplasms and acute leukemia. The new category of disease is named hereditary myeloid disorder (HMD). Although most myeloid neoplasms are sporadic, germ line mutations and familial predisposition can contribute to development of chronic myeloid diseases and acute myeloid leukemia. This finding and upcoming frequent use of genome wide detection of molecular aberrations will lead to a higher detection rate of a genetic predisposition and influence treatment decisions. Hereditary predisposition is responsible for 5-10% of myeloid malignancies. Management of affected patients begins by the awareness of treating physicians of the problem and a precise work up of the patient and family members. Areas covered: This review focuses on current knowledge about germ line predisposition for myeloid neoplasms including diagnostic, prognostic, and therapeutic aspects in adult patients. Essential information for clinical routine is provided. Expert commentary: Compared to a patient without predisposition, adaptation of treatment strategy for patients with an HMD is often necessary, especially to avoid higher risk of relapse or higher toxicity during chemotherapy or transplantation. Mistakes in choice of a related donor can be omitted. Relatives at risk of developing a HMD need specific surveillance.
Collapse
Affiliation(s)
- Martina Crysandt
- a Medical Faculty, Dept. of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation , University Hospital RWTH Aachen , Aachen , Germany
| | - Kira Brings
- a Medical Faculty, Dept. of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation , University Hospital RWTH Aachen , Aachen , Germany
| | - Fabian Beier
- a Medical Faculty, Dept. of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation , University Hospital RWTH Aachen , Aachen , Germany
| | - Christian Thiede
- b Medizinische Klinik und Poliklinik I , Universitätsklinikum Carl Gustav Carus der TU Dresden , Dresden , Germany
| | - Tim H Brümmendorf
- a Medical Faculty, Dept. of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation , University Hospital RWTH Aachen , Aachen , Germany
| | - Edgar Jost
- a Medical Faculty, Dept. of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation , University Hospital RWTH Aachen , Aachen , Germany
| |
Collapse
|
9
|
Falardeau F, Camurri MV, Campeau PM. Genomic approaches to diagnose rare bone disorders. Bone 2017; 102:5-14. [PMID: 27474525 DOI: 10.1016/j.bone.2016.07.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 07/24/2016] [Indexed: 02/01/2023]
Abstract
Skeletal dysplasias are Mendelian disorders with a prevalence of approximatively 1 in every 5000 individuals and can usually be diagnosed based on clinical and radiological findings. However, given that some diseases can be caused by several different genes, and that some genes can cause a variety of different phenotypes, achieving a molecular diagnosis can be challenging. We review here different approaches, from single gene sequencing to genomic approaches using next-generation sequencing, to reach a molecular diagnosis for skeletal dysplasias. We will further describe the overall advantages and limitations of first, second and third-generation sequencing, including single gene sequencing, whole-exome and genome sequencing (WES and WGS), multiple gene panel sequencing and single molecule sequencing. We also provide a brief overview of potential future applications of emerging technologies.
Collapse
Affiliation(s)
- Félix Falardeau
- CHU Sainte-Justine Research Center, Montreal, Canada; Division of Molecular and Cellular Biology, Department of Biology, University of Sherbrooke, Sherbrooke, Canada
| | | | - Philippe M Campeau
- CHU Sainte-Justine Research Center, Montreal, Canada; Division of Medical Genetics, Department of Pediatrics, University of Montreal, Montreal, Canada.
| |
Collapse
|
10
|
Knies K, Inano S, Ramírez MJ, Ishiai M, Surrallés J, Takata M, Schindler D. Biallelic mutations in the ubiquitin ligase RFWD3 cause Fanconi anemia. J Clin Invest 2017; 127:3013-3027. [PMID: 28691929 DOI: 10.1172/jci92069] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 05/16/2017] [Indexed: 12/28/2022] Open
Abstract
The WD40-containing E3 ubiquitin ligase RFWD3 has been recently linked to the repair of DNA damage by homologous recombination (HR). Here we have shown that an RFWD3 mutation within the WD40 domain is connected to the genetic disease Fanconi anemia (FA). An individual presented with congenital abnormalities characteristic of FA. Cells from the patient carrying the compound heterozygous mutations c.205_206dupCC and c.1916T>A in RFWD3 showed increased sensitivity to DNA interstrand cross-linking agents in terms of increased chromosomal breakage, reduced survival, and cell cycle arrest in G2 phase. The cellular phenotype was mirrored in genetically engineered human and avian cells by inactivation of RFWD3 or introduction of the patient-derived missense mutation, and the phenotype was rescued by expression of wild-type RFWD3 protein. HR was disrupted in RFWD3-mutant cells as a result of impaired relocation of mutant RFWD3 to chromatin and defective physical interaction with replication protein A. Rfwd3 knockout mice appear to have increased embryonic lethality, are subfertile, show ovarian and testicular atrophy, and have a reduced lifespan resembling that of other FA mouse models. Although RFWD3 mutations have thus far been detected in a single child with FA, we propose RFWD3 as an FA gene, FANCW, supported by cellular paradigm systems and an animal model.
Collapse
Affiliation(s)
- Kerstin Knies
- Department of Human Genetics, Biozentrum, University of Wurzburg, Wurzburg, Germany
| | - Shojiro Inano
- Laboratory of DNA Damage Signaling, Department of Late Effects Studies, Radiation Biology Center, Kyoto University, Kyoto, Japan.,Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - María J Ramírez
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain.,Center for Biomedical Network Research on Rare Diseases (CIBERER), Madrid, Spain
| | - Masamichi Ishiai
- Laboratory of DNA Damage Signaling, Department of Late Effects Studies, Radiation Biology Center, Kyoto University, Kyoto, Japan
| | - Jordi Surrallés
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, Bellaterra (Barcelona), Spain.,Center for Biomedical Network Research on Rare Diseases (CIBERER), Madrid, Spain.,Genetics Department, Hospital de Sant Pau, Barcelona, Spain
| | - Minoru Takata
- Laboratory of DNA Damage Signaling, Department of Late Effects Studies, Radiation Biology Center, Kyoto University, Kyoto, Japan
| | - Detlev Schindler
- Department of Human Genetics, Biozentrum, University of Wurzburg, Wurzburg, Germany
| |
Collapse
|
11
|
Pilonetto DV, Pereira NF, Bonfim CMS, Ribeiro LL, Bitencourt MA, Kerkhoven L, Floor K, Ameziane N, Joenje H, Gille JJP, Pasquini R. A strategy for molecular diagnostics of Fanconi anemia in Brazilian patients. Mol Genet Genomic Med 2017; 5:360-372. [PMID: 28717661 PMCID: PMC5511800 DOI: 10.1002/mgg3.293] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 03/20/2017] [Accepted: 03/24/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Fanconi anemia (FA) is a predominantly autosomal recessive disease with wide genetic heterogeneity resulting from mutations in several DNA repair pathway genes. To date, 21 genetic subtypes have been identified. We aimed to identify the FA genetic subtypes in the Brazilian population and to develop a strategy for molecular diagnosis applicable to routine clinical use. METHODS We screened 255 patients from Hospital de Clínicas, Universidade Federal do Paraná for 11 common FA gene mutations. Further analysis by multiplex ligation-dependent probe amplification (MLPA) for FANCA and Sanger sequencing of all coding exons of FANCA, -C, and -G was performed in cases who harbored a single gene mutation. RESULTS We identified biallelic mutations in 128/255 patients (50.2%): 89, 11, and 28 carried FANCA,FANCC, and FANCG mutations, respectively. Of these, 71 harbored homozygous mutations, whereas 57 had compound heterozygous mutations. In 4/57 heterozygous patients, both mutations were identified by the initial screening, in 51/57 additional analyses was required for classification, and in 2/57 the second mutation remained unidentified. We found 52 different mutations of which 22 were novel. CONCLUSION The proposed method allowed genetic subtyping of 126/255 (49.4%) patients at a significantly reduced time and cost, which makes molecular diagnosis of FA Brazilian patients feasible.
Collapse
Affiliation(s)
- Daniela V. Pilonetto
- Immunogenetics LaboratoryHospital de ClínicasUniversidade Federal do ParanáCuritibaPRBrazil
| | - Noemi F. Pereira
- Immunogenetics LaboratoryHospital de ClínicasUniversidade Federal do ParanáCuritibaPRBrazil
| | - Carmem M. S. Bonfim
- Bone Marrow Transplantation ServiceHospital de ClínicasUniversidade Federal do ParanáCuritibaPRBrazil
| | - Lisandro L. Ribeiro
- Bone Marrow Transplantation ServiceHospital de ClínicasUniversidade Federal do ParanáCuritibaPRBrazil
| | - Marco A. Bitencourt
- Bone Marrow Transplantation ServiceHospital de ClínicasUniversidade Federal do ParanáCuritibaPRBrazil
| | - Lianne Kerkhoven
- Department of Clinical GeneticsVU University Medical CenterAmsterdamThe Netherlands
| | - Karijn Floor
- Department of Clinical GeneticsVU University Medical CenterAmsterdamThe Netherlands
| | - Najim Ameziane
- Department of Clinical GeneticsVU University Medical CenterAmsterdamThe Netherlands
| | - Hans Joenje
- Department of Clinical GeneticsVU University Medical CenterAmsterdamThe Netherlands
| | - Johan J. P. Gille
- Department of Clinical GeneticsVU University Medical CenterAmsterdamThe Netherlands
| | - Ricardo Pasquini
- Bone Marrow Transplantation ServiceHospital de ClínicasUniversidade Federal do ParanáCuritibaPRBrazil
| |
Collapse
|
12
|
De Leeneer K, Hellemans J, Steyaert W, Lefever S, Vereecke I, Debals E, Crombez B, Baetens M, Van Heetvelde M, Coppieters F, Vandesompele J, De Jaegher A, De Baere E, Coucke P, Claes K. Flexible, scalable, and efficient targeted resequencing on a benchtop sequencer for variant detection in clinical practice. Hum Mutat 2015; 36:379-87. [PMID: 25504618 DOI: 10.1002/humu.22739] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 12/02/2014] [Indexed: 12/30/2022]
Abstract
The release of benchtop next-generation sequencing (NGS) instruments has paved the way to implement the technology in clinical setting. The need for flexible, qualitative, and cost-efficient workflows is high. We used singleplex-PCR for highly efficient target enrichment, allowing us to reach the quality standards set in Sanger sequencing-based diagnostics. For the library preparation, a modified NexteraXT protocol was used, followed by sequencing on a MiSeq instrument. With an innovative pooling strategy, high flexibility, scalability, and cost-efficiency were obtained, independent of the availability of commercial kits. The approach was validated for ∼250 genes associated with monogenic disorders. An overall sensitivity (>99%) similar to Sanger sequencing was observed in combination with a positive predictive value of >98%. The distribution of coverage was highly uniform, guaranteeing a minimal number of gaps to be filled with alternative methods. ISO15189-accreditation was obtained for the workflow. A major asset of the singleplex PCR-based enrichment is that new targets can be easily implemented. Diagnostic laboratories have validated assays available ensuring that the proposed workflow can easily be adopted. Although our platform was optimized for constitutional variant detection of monogenic disease genes, it is now also used as a model for somatic mutation detection in acquired diseases.
Collapse
Affiliation(s)
- Kim De Leeneer
- Center for Medical Genetics Ghent, Ghent University, Ghent, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Muendlein A, Rohde BH, Gasser K, Haid A, Rauch S, Kinz E, Drexel H, Hofmann W, Schindler V, Kapoor R, Decker T, Lang AH. Evaluation of BRCA1/2 mutational status among German and Austrian women with triple-negative breast cancer. J Cancer Res Clin Oncol 2015; 141:2005-12. [PMID: 25971625 DOI: 10.1007/s00432-015-1986-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/02/2015] [Indexed: 12/27/2022]
Abstract
PURPOSE Testing for BRCA1 and BRCA2 mutations in breast cancer patients is used to identify the risk of second primary cancers and the risk of cancer in the patients' family. Women with triple-negative breast cancer (TNBC) are thought to be more likely to be BRCA1/2 mutation carriers, but most national guidelines for genetic testing, including those used in Germany and Austria, do not consider receptor triple negativity. METHODS We determined the prevalence of BRCA1 and BRCA2 mutations within a cohort of 100 unselected TNBC cases, including patients from Germany and Austria to identify those BRCA-positive patients with a masked family history and who would have been missed due to respective current national guidelines. Double-stranded Sanger sequencing of all exons of BRCA1 and BRCA2, respectively, was performed. RESULTS We identified a total of 13 deleterious mutations in BRCA1 and a total of four deleterious mutations in BRCA2. The total rate of deleterious BRCA1/2 mutation carriers was 21 % in our cohort. Six novel mutations, including two deleterious mutations, have been identified, which have not been described in public mutation databases so far. According to current German and Austrian national guidelines for genetic testing, 38.1 and 52.4 %, respectively, of BRCA1/2 mutation carriers would have been overlooked. CONCLUSIONS We conclude that the prevalence of BRCA1 and BRCA2 mutations is high in TNBC patients and that BRCA1/2 mutations are not restricted to young women or patients with a positive family history. Receptor triple negativity should therefore be considered in BRCA1/2 genetic testing guidelines.
Collapse
Affiliation(s)
- Axel Muendlein
- Vorarlberg Institute for Vascular Investigation and Treatment, Feldkirch, Austria
| | | | - Klaus Gasser
- Academic Teaching Hospital Feldkirch, Feldkirch, Austria
| | - Anton Haid
- Academic Teaching Hospital Feldkirch, Feldkirch, Austria
| | | | - Elena Kinz
- Vorarlberg Institute for Vascular Investigation and Treatment, Feldkirch, Austria
| | - Heinz Drexel
- Vorarlberg Institute for Vascular Investigation and Treatment, Feldkirch, Austria
| | | | | | | | | | - Alois H Lang
- Vorarlberg Institute for Vascular Investigation and Treatment, Feldkirch, Austria.
- Academic Teaching Hospital Feldkirch, Feldkirch, Austria.
| |
Collapse
|
14
|
Bogliolo M, Surrallés J. Fanconi anemia: a model disease for studies on human genetics and advanced therapeutics. Curr Opin Genet Dev 2015; 33:32-40. [PMID: 26254775 DOI: 10.1016/j.gde.2015.07.002] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 07/19/2015] [Accepted: 07/21/2015] [Indexed: 12/18/2022]
Abstract
Fanconi anemia (FA) is characterized by bone marrow failure, malformations, and chromosome fragility. We review the recent discovery of FA genes and efforts to develop genetic therapies for FA in the last five years. Because current data exclude FANCM as an FA gene, 15 genes remain bona fide FA genes and three (FANCO, FANCR and FANCS) cause an FA like syndrome. Monoallelic mutations in 6 FA associated genes (FANCD1, FANCJ, FANCM, FANCN, FANCO and FANCS) predispose to breast and ovarian cancer. The products of all these genes are involved in the repair of stalled DNA replication forks by unhooking DNA interstrand cross-links and promoting homologous recombination. The genetic characterization of patients with FA is essential for developing therapies, including hematopoietic stem cell transplantation from a savior sibling donor after embryo selection, gene therapy, or genome editing using genetic recombination or engineered nucleases. Newly acquired knowledge about FA promises to provide therapeutic strategies in the near future.
Collapse
Affiliation(s)
- Massimo Bogliolo
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Spain
| | - Jordi Surrallés
- Genome Instability and DNA Repair Group, Department of Genetics and Microbiology, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain; Centre for Biomedical Network Research on Rare Diseases (CIBERER), Spain.
| |
Collapse
|
15
|
Stoepker C, Ameziane N, van der Lelij P, Kooi IE, Oostra AB, Rooimans MA, van Mil SE, Brink A, Dietrich R, Balk JA, Ylstra B, Joenje H, Feller SM, Brakenhoff RH. Defects in the Fanconi Anemia Pathway and Chromatid Cohesion in Head and Neck Cancer. Cancer Res 2015; 75:3543-53. [PMID: 26122845 DOI: 10.1158/0008-5472.can-15-0528] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/19/2015] [Indexed: 11/16/2022]
Abstract
Failure to repair DNA damage or defective sister chromatid cohesion, a process essential for correct chromosome segregation, can be causative of chromosomal instability (CIN), which is a hallmark of many types of cancers. We investigated how frequent this occurs in head and neck squamous cell carcinoma (HNSCC) and whether specific mechanisms or genes could be linked to these phenotypes. The genomic instability syndrome Fanconi anemia is caused by mutations in any of at least 16 genes regulating DNA interstrand crosslink (ICL) repair. Since patients with Fanconi anemia have a high risk to develop HNSCC, we investigated whether and to which extent Fanconi anemia pathway inactivation underlies CIN in HNSCC of non-Fanconi anemia individuals. We observed ICL-induced chromosomal breakage in 9 of 17 (53%) HNSCC cell lines derived from patients without Fanconi anemia. In addition, defective sister chromatid cohesion was observed in five HNSCC cell lines. Inactivation of FANCM was responsible for chromosomal breakage in one cell line, whereas in two other cell lines, somatic mutations in PDS5A or STAG2 resulted in inadequate sister chromatid cohesion. In addition, FANCF methylation was found in one cell line by screening an additional panel of 39 HNSCC cell lines. Our data demonstrate that CIN in terms of ICL-induced chromosomal breakage and defective chromatid cohesion is frequently observed in HNSCC. Inactivation of known Fanconi anemia and chromatid cohesion genes does explain CIN in the minority of cases. These findings point to phenotypes that may be highly relevant in treatment response of HNSCC.
Collapse
Affiliation(s)
- Chantal Stoepker
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, the Netherlands
| | - Najim Ameziane
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, the Netherlands
| | - Petra van der Lelij
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, the Netherlands
| | - Irsan E Kooi
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, the Netherlands
| | - Anneke B Oostra
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, the Netherlands
| | - Martin A Rooimans
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, the Netherlands
| | - Saskia E van Mil
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, the Netherlands
| | - Arjen Brink
- Department of Otolaryngology-Head and Neck Surgery, VU University Medical Center, Amsterdam, the Netherlands
| | - Ralf Dietrich
- German Fanconi Anemia Support Group and Research Fund, Unna-Siddinghausen, Germany
| | - Jesper A Balk
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, the Netherlands
| | - Bauke Ylstra
- Department of Pathology, VU University Medical Center, Amsterdam, the Netherlands
| | - Hans Joenje
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, the Netherlands
| | - Stephan M Feller
- Biological Systems Architecture Group, Weatherall Institute of Molecular Medicine, Department of Oncology, University of Oxford, United Kingdom
| | - Ruud H Brakenhoff
- Department of Otolaryngology-Head and Neck Surgery, VU University Medical Center, Amsterdam, the Netherlands.
| |
Collapse
|
16
|
Characterization of medulloblastoma in Fanconi Anemia: a novel mutation in the BRCA2 gene and SHH molecular subgroup. Biomark Res 2015; 3:13. [PMID: 26064523 PMCID: PMC4462002 DOI: 10.1186/s40364-015-0038-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 05/26/2015] [Indexed: 11/10/2022] Open
Abstract
Fanconi Anemia (FA) is an inherited disorder characterized by the variable presence of multiple congenital somatic abnormalities, bone marrow failure and cancer susceptibility. Medulloblastoma (MB) has been described only in few cases of FA with biallelic inactivation in the tumor suppressor gene BRCA2/FANCD1 or its associated gene PALB2/FANCN. We report the case of a patient affected by Fanconi Anemia with Wilms tumor and unusual presentation of two medulloblastomas (MB1 and MB2). We identified a new pathogenetic germline BRCA2 mutation: c.2944_2944delA. Molecular analysis of MBs allowed us to define new features of MB in FA. MBs were found to belong to the Sonic Hedgehog (SHH) molecular subgroup with some differences between MB1 and MB2. We highlighted that MB in FA could share molecular aspects and hemispheric localization with sporadic adult SHH-MB. Our report provides new findings that shed new light on the genetic and molecular pathogenesis of MB in FA patients with implications in the disease management.
Collapse
|
17
|
Yohe S, Hauge A, Bunjer K, Kemmer T, Bower M, Schomaker M, Onsongo G, Wilson J, Erdmann J, Zhou Y, Deshpande A, Spears MD, Beckman K, Silverstein KAT, Thyagarajan B. Clinical validation of targeted next-generation sequencing for inherited disorders. Arch Pathol Lab Med 2015; 139:204-10. [PMID: 25611102 DOI: 10.5858/arpa.2013-0625-oa] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT Although next-generation sequencing (NGS) can revolutionize molecular diagnostics, several hurdles remain in the implementation of this technology in clinical laboratories. OBJECTIVES To validate and implement an NGS panel for genetic diagnosis of more than 100 inherited diseases, such as neurologic conditions, congenital hearing loss and eye disorders, developmental disorders, nonmalignant diseases treated by hematopoietic cell transplantation, familial cancers, connective tissue disorders, metabolic disorders, disorders of sexual development, and cardiac disorders. The diagnostic gene panels ranged from 1 to 54 genes with most of panels containing 10 genes or fewer. DESIGN We used a liquid hybridization-based, target-enrichment strategy to enrich 10 067 exons in 568 genes, followed by NGS with a HiSeq 2000 sequencing system (Illumina, San Diego, California). RESULTS We successfully sequenced 97.6% (9825 of 10 067) of the targeted exons to obtain a minimum coverage of 20× at all bases. We demonstrated 100% concordance in detecting 19 pathogenic single-nucleotide variations and 11 pathogenic insertion-deletion mutations ranging in size from 1 to 18 base pairs across 18 samples that were previously characterized by Sanger sequencing. Using 4 pairs of blinded, duplicate samples, we demonstrated a high degree of concordance (>99%) among the blinded, duplicate pairs. CONCLUSIONS We have successfully demonstrated the feasibility of using the NGS platform to multiplex genetic tests for several rare diseases and the use of cloud computing for bioinformatics analysis as a relatively low-cost solution for implementing NGS in clinical laboratories.
Collapse
Affiliation(s)
- Sophia Yohe
- From the Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis (Drs Yohe, Wilson, Zhou, Spears, and Thyagarajan); the University of Minnesota Genomics Center (Mr Hauge, Mss Bunjer and Deshpande, and Dr Beckman); the Molecular Diagnostics Laboratory, Fairview-University of Minnesota Medical Center (Ms Kemmer and Messrs Bower and Schomaker); the Division of Genetics and Metabolism, University of Minnesota (Mr Bower); and the Minnesota Supercomputing Institute, University of Minnesota (Drs Onsongo and Silverstein and Mr Erdmann)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Genetic Counseling for Fanconi Anemia: Crosslinking Disciplines. J Genet Couns 2014; 23:910-21. [DOI: 10.1007/s10897-014-9754-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 07/31/2014] [Indexed: 12/22/2022]
|
19
|
Chung NG, Kim M. Current insights into inherited bone marrow failure syndromes. KOREAN JOURNAL OF PEDIATRICS 2014; 57:337-44. [PMID: 25210520 PMCID: PMC4155177 DOI: 10.3345/kjp.2014.57.8.337] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 06/09/2014] [Indexed: 01/24/2023]
Abstract
Inherited bone marrow failure syndrome (IBMFS) encompasses a heterogeneous and complex group of genetic disorders characterized by physical malformations, insufficient blood cell production, and increased risk of malignancies. They often have substantial phenotype overlap, and therefore, genotyping is often a critical means of establishing a diagnosis. Current advances in the field of IBMFSs have identified multiple genes associated with IBMFSs and their pathways: genes involved in ribosome biogenesis, such as those associated with Diamond-Blackfan anemia and Shwachman-Diamond syndrome; genes involved in telomere maintenance, such as dyskeratosis congenita genes; genes encoding neutrophil elastase or neutrophil adhesion and mobility associated with severe congenital neutropenia; and genes involved in DNA recombination repair, such as those associated with Fanconi anemia. Early and adequate genetic diagnosis is required for proper management and follow-up in clinical practice. Recent advances using new molecular technologies, including next generation sequencing (NGS), have helped identify new candidate genes associated with the development of bone marrow failure. Targeted NGS using panels of large numbers of genes is rapidly gaining potential for use as a cost-effective diagnostic tool for the identification of mutations in newly diagnosed patients. In this review, we have described recent insights into IBMFS and how they are advancing our understanding of the disease's pathophysiology; we have also discussed the possible implications they will have in clinical practice for Korean patients.
Collapse
Affiliation(s)
- Nack-Gyun Chung
- Department of Pediatrics, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Myungshin Kim
- Department of Laboratory Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea. ; Catholic Genetic Laboratory Center, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
20
|
Abstract
Germline testing for familial predisposition to myeloid malignancies is becoming more common with the recognition of multiple familial syndromes. Currently, Clinical Laboratory Improvement Amendments-approved testing exists for the following: familial platelet disorder with propensity to acute myeloid leukemia, caused by mutations in RUNX1; familial myelodysplastic syndrome/acute myeloid leukemia with mutated GATA2; familial acute myeloid leukemia with mutated CEBPA; and the inherited bone marrow failure syndromes, including dyskeratosis congenita, a disease of abnormal telomere maintenance. With the recognition of additional families with a genetic component to their myeloid diseases, new predisposition alleles are likely to be identified. Awareness of the existence of these syndromes will facilitate proper genetic counseling, appropriate testing, and clinical management of these cases.
Collapse
Affiliation(s)
- Lucy A Godley
- Section of Hematology/Oncology and the Center for Clinical Cancer Genetics, Department of Medicine, and Comprehensive Cancer Center, The University of Chicago, Chicago, IL.
| |
Collapse
|
21
|
Shidhi PR, Nair AS, Suravajhala P. Identifying pseudogenes from hypothetical proteins for making synthetic proteins. SYSTEMS AND SYNTHETIC BIOLOGY 2014; 8:169-71. [PMID: 24799963 DOI: 10.1007/s11693-014-9148-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 04/10/2014] [Accepted: 04/11/2014] [Indexed: 11/26/2022]
Abstract
Nature selected certain regions of the genome for encoding proteins. Most of the sequences were used to encode only RNA. What happened to the remaining sections of the genome? It is possible that some sequences were retired and retained as non-functional entities called pseudogenes. Though several evolutionary prospects with functional endpoints exist, we looked at the possibility of hypothetical proteins correlating with the emergence of pseudogenes and potential of such genes to make novel synthetic molecules. In this commentary, we consider two key aspects: (1) does any correlation exist between hypothetical proteins and pseudogenes and (2)-can we make novel and functional proteins from pseudogenes?
Collapse
Affiliation(s)
- P R Shidhi
- Department of Computational Biology and Bioinformatics, University of Kerala, Kariyavattom Campus, Thiruvananthapuram, 695581 Kerala India
| | - Achuthsankar S Nair
- Department of Computational Biology and Bioinformatics, University of Kerala, Kariyavattom Campus, Thiruvananthapuram, 695581 Kerala India
| | - Prashanth Suravajhala
- Bioclues.Org, Hyderabad, India ; Bioinformatics.Org, 28 Pope Street, Hudson, MA 01749 USA
| |
Collapse
|
22
|
Vassy JL, Lautenbach DM, McLaughlin HM, Kong SW, Christensen KD, Krier J, Kohane IS, Feuerman LZ, Blumenthal-Barby J, Roberts JS, Lehmann LS, Ho CY, Ubel PA, MacRae CA, Seidman CE, Murray MF, McGuire AL, Rehm HL, Green RC. The MedSeq Project: a randomized trial of integrating whole genome sequencing into clinical medicine. Trials 2014; 15:85. [PMID: 24645908 PMCID: PMC4113228 DOI: 10.1186/1745-6215-15-85] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 02/28/2014] [Indexed: 11/28/2022] Open
Abstract
Background Whole genome sequencing (WGS) is already being used in certain clinical and research settings, but its impact on patient well-being, health-care utilization, and clinical decision-making remains largely unstudied. It is also unknown how best to communicate sequencing results to physicians and patients to improve health. We describe the design of the MedSeq Project: the first randomized trials of WGS in clinical care. Methods/Design This pair of randomized controlled trials compares WGS to standard of care in two clinical contexts: (a) disease-specific genomic medicine in a cardiomyopathy clinic and (b) general genomic medicine in primary care. We are recruiting 8 to 12 cardiologists, 8 to 12 primary care physicians, and approximately 200 of their patients. Patient participants in both the cardiology and primary care trials are randomly assigned to receive a family history assessment with or without WGS. Our laboratory delivers a genome report to physician participants that balances the needs to enhance understandability of genomic information and to convey its complexity. We provide an educational curriculum for physician participants and offer them a hotline to genetics professionals for guidance in interpreting and managing their patients’ genome reports. Using varied data sources, including surveys, semi-structured interviews, and review of clinical data, we measure the attitudes, behaviors and outcomes of physician and patient participants at multiple time points before and after the disclosure of these results. Discussion The impact of emerging sequencing technologies on patient care is unclear. We have designed a process of interpreting WGS results and delivering them to physicians in a way that anticipates how we envision genomic medicine will evolve in the near future. That is, our WGS report provides clinically relevant information while communicating the complexity and uncertainty of WGS results to physicians and, through physicians, to their patients. This project will not only illuminate the impact of integrating genomic medicine into the clinical care of patients but also inform the design of future studies. Trial registration ClinicalTrials.gov identifier
NCT01736566
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Robert C Green
- Genomes2People and Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Broad Institute and Harvard Medical School, 41 Avenue Louis Pasteur, Suite 301, 02115 Boston, MA, USA.
| | | |
Collapse
|
23
|
Claes KBM, De Leeneer K. Dealing with pseudogenes in molecular diagnostics in the next-generation sequencing era. Methods Mol Biol 2014; 1167:303-15. [PMID: 24823787 DOI: 10.1007/978-1-4939-0835-6_21] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In this chapter, we focus on issues related to the application of next-generation sequencing (NGS) strategies for the analysis of genes with pseudogenes in a clinical setting. Hereby, target enrichment and mapping strategies are crucial factors to avoid "contaminating" pseudogene sequences, which are characterized by higher mutation rates than their functional parental genes. For the target enrichment strategies, we describe advantages and disadvantages of PCR- and capture-based enrichment methodologies. For the mapping strategies, we discuss crucial parameters that need to be considered to accurately distinguish sequences of functional genes from pseudogenic sequences. Finally, we discuss some concrete examples of genes with known pseudogenes and associated with Mendelian disorders that were analyzed by NGS on various platforms and starting from different library preparations.
Collapse
|
24
|
Zheng Z, Geng J, Yao RE, Li C, Ying D, Shen Y, Ying L, Yu Y, Fu Q. Molecular defects identified by whole exome sequencing in a child with Fanconi anemia. Gene 2013; 530:295-300. [DOI: 10.1016/j.gene.2013.08.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 08/01/2013] [Accepted: 08/09/2013] [Indexed: 01/25/2023]
|
25
|
Mitchell R, Wagner JE, Hirsch B, DeFor TE, Zierhut H, MacMillan ML. Haematopoietic cell transplantation for acute leukaemia and advanced myelodysplastic syndrome in Fanconi anaemia. Br J Haematol 2013; 164:384-95. [PMID: 24172081 DOI: 10.1111/bjh.12634] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 09/30/2013] [Indexed: 01/28/2023]
Abstract
Acute leukaemia or advanced myelodysplastic syndrome (MDS ≥ 5% blasts) in Fanconi anaemia (FA) patients is associated with a poor prognosis. We report 21 FA patients with acute leukaemia or advanced MDS who underwent haematopoietic cell transplantation (HCT) at the University of Minnesota between 1988 and 2011. Six patients had biallelic BRCA2 mutations. Eight patients received pre-transplant cytoreduction, with 3 achieving complete remission. HCT donor source included human leucocyte antigen-matched sibling (n = 2) or alternative donors (n = 19). Neutrophil engraftment was 95% for the entire cohort, and the incidence of acute graft-versus-host disease was 19%. 5-year overall survival (OS) was 33%, with a relapse rate of 24%, with similar OS in patients with biallelic BRCA2 mutations. Our study supports the use of HCT in the treatment of FA patients with acute leukaemia or advanced MDS, however, the role of chemotherapy prior to HCT remains unclear for this population. FA patients with biallelic BRCA2 are unique and may benefit from higher dose chemotherapy relative to other complementation groups.
Collapse
Affiliation(s)
- Richard Mitchell
- Blood and Marrow Transplant Program, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | | | | | | | | | | |
Collapse
|
26
|
Abstract
The inherited bone marrow failure syndromes (IBMFS) are a set of clinically related yet heterogeneous disorders in which at least one hematopoietic cell lineage is significantly reduced. Many of the IBMFS have notably increased cancer risks, as well as other physical findings. Highly penetrant germline mutations in key pathways, such as DNA repair, telomere biology, or ribosomal biogenesis, are causative of Fanconi anemia (FA), dyskeratosis congenita (DC), and Diamond-Blackfan anemia (DBA), respectively. Next-generation sequencing (NGS) generally refers to high-throughput, large-scale sequencing technologies and is being used more frequently to understand disease etiology. In the IBMFS, NGS has facilitated the discovery of germline mutations that cause thrombocytopenia absent radii syndrome (TAR), a subset of DC and DBA, and other uncharacterized, but related, disorders. Panels of large numbers of genes are being used to molecularly characterize patients with IBMFS, such as FA and DBA. NGS is also accelerating the discovery of the genetic etiology of previously unclassified IBMFS. In this review, we will highlight recent studies that have employed NGS to ascertain the genetic etiology of IBMFS, namely, FA, DC, DBA, and TAR, and discuss the translational utility of these findings.
Collapse
Affiliation(s)
- Payal P. Khincha
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Children’s National Medical Center, Washington, DC
| | - Sharon A. Savage
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
27
|
Abstract
The development of novel technologies for high-throughput DNA sequencing is having a major impact on our ability to measure and define normal and pathologic variation in humans. This review discusses advances in DNA sequencing that have been applied to benign hematologic disorders, including those affecting the red blood cell, the neutrophil, and other white blood cell lineages. Relevant examples of how these approaches have been used for disease diagnosis, gene discovery, and studying complex traits are provided. High-throughput DNA sequencing technology holds significant promise for impacting clinical care. This includes development of improved disease detection and diagnosis, better understanding of disease progression and stratification of risk of disease-specific complications, and development of improved therapeutic strategies, particularly patient-specific pharmacogenomics-based therapy, with monitoring of therapy by genomic biomarkers.
Collapse
|
28
|
Nickels EM, Soodalter J, Churpek JE, Godley LA. Recognizing familial myeloid leukemia in adults. Ther Adv Hematol 2013; 4:254-69. [PMID: 23926458 DOI: 10.1177/2040620713487399] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Germline testing for familial cases of myeloid leukemia in adults is becoming more common with the recognition of multiple genetic syndromes predisposing people to bone marrow disease. Currently, Clinical Laboratory Improvement Amendments approved testing exists for several myeloid leukemia predisposition syndromes: familial platelet disorder with propensity to acute myeloid leukemia (FPD/AML), caused by mutations in RUNX1; familial AML with mutated CEBPA; familial myelodysplastic syndrome and acute leukemia with mutated GATA2; and the inherited bone marrow failure syndromes, including dyskeratosis congenita, a disease of abnormal telomere maintenance. With the recognition of additional families with a genetic component to their leukemia, new predisposition alleles will likely be identified. We highlight how to recognize and manage these cases as well as outline the characteristics of the major known syndromes. We look forward to future research increasing our understanding of the scope of inherited myeloid leukemia syndromes.
Collapse
Affiliation(s)
- Eric M Nickels
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | | | | | | |
Collapse
|