51
|
Songdej N, Rao AK. Inherited platelet dysfunction and hematopoietic transcription factor mutations. Platelets 2017; 28:20-26. [PMID: 27463948 PMCID: PMC5628047 DOI: 10.1080/09537104.2016.1203400] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/16/2016] [Accepted: 05/30/2016] [Indexed: 01/19/2023]
Abstract
Transcription factors (TFs) are proteins that bind to specific DNA sequences and regulate expression of genes. The molecular and genetic mechanisms in most patients with inherited platelet dysfunction are unknown. There is now increasing evidence that mutations in hematopoietic TFs are an important underlying cause for the defects in platelet production, morphology, and function. The hematopoietic TFs implicated in the patients with impaired platelet function include Runt related TF 1 (RUNX1), Fli-1 proto-oncogene, ETS TF (FLI1), GATA-binding protein 1 (GATA1), and growth factor independent 1B transcriptional repressor (GFI1B). These TFs act in a combinatorial manner to bind sequence-specific DNA within a promoter region to regulate lineage-specific gene expression, either as activators or as repressors. TF mutations induce rippling downstream effects by simultaneously altering the expression of multiple genes. Mutations involving these TFs affect diverse aspects of megakaryocyte biology and platelet production and function, culminating in thrombocytopenia, platelet dysfunction, and associated clinical features. Mutations in TFs may occur more frequently in the patients with inherited platelet dysfunction than generally appreciated. This review focuses on the alterations in hematopoietic TFs in the pathobiology of inherited platelet dysfunction.
Collapse
Affiliation(s)
- Natthapol Songdej
- a Sol Sherry Thrombosis Research Center, and Hematology Section, Department of Medicine , Lewis Katz School of Medicine at Temple University , Philadelphia , PA , USA
| | - A Koneti Rao
- a Sol Sherry Thrombosis Research Center, and Hematology Section, Department of Medicine , Lewis Katz School of Medicine at Temple University , Philadelphia , PA , USA
| |
Collapse
|
52
|
Koeffler HP, Leong G. Preleukemia: one name, many meanings. Leukemia 2016; 31:534-542. [PMID: 27899806 PMCID: PMC5339433 DOI: 10.1038/leu.2016.364] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 11/11/2016] [Accepted: 11/14/2016] [Indexed: 12/15/2022]
Abstract
Definition of preleukemia has evolved. It was first used to describe the myelodysplastic syndrome (MDS) with a propensity to progress to acute myeloid leukemia (AML). Individuals with germline mutations of either RUNX1, CEBPA, or GATA2 can also be called as preleukemic because they have a markedly increased incidence of evolution into AML. Also, alkylating chemotherapy or radiation can cause MDS/preleukemia, which nearly always progress to AML. More recently, investigators noted that AML patients who achieved complete morphological remission after chemotherapy often have clonal hematopoiesis predominantly marked by either DNMT3A, TET2 or IDH1/2 mutations, which were also present at diagnosis of AML. This preleukemic clone represents involvement of an early hematopoietic stem cells, which is resistant to standard therapy. The same clonal hematopoietic mutations have been identified in older ‘normal' individuals who have a modest increased risk of developing frank AML. These individuals have occasionally been said, probably inappropriately, to have a preleukemia clone. Our evolving understanding of the term preleukemia has occurred by advancing technology including studies of X chromosome inactivation, cytogenetics and more recently deep nucleotide sequencing.
Collapse
Affiliation(s)
- H P Koeffler
- Department of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.,Cancer Science Institute of Singapore, National University of Singapore, Singapore.,National University Cancer Institute of Singapore, National University Hospital, Singapore
| | - G Leong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| |
Collapse
|
53
|
Melazzini F, Palombo F, Balduini A, De Rocco D, Marconi C, Noris P, Gnan C, Pippucci T, Bozzi V, Faleschini M, Barozzi S, Doubek M, Di Buduo CA, Kozubik KS, Radova L, Loffredo G, Pospisilova S, Alfano C, Seri M, Balduini CL, Pecci A, Savoia A. Clinical and pathogenic features of ETV6-related thrombocytopenia with predisposition to acute lymphoblastic leukemia. Haematologica 2016; 101:1333-1342. [PMID: 27365488 PMCID: PMC5394865 DOI: 10.3324/haematol.2016.147496] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 06/29/2016] [Indexed: 11/09/2022] Open
Abstract
ETV6-related thrombocytopenia is an autosomal dominant thrombocytopenia that has been recently identified in a few families and has been suspected to predispose to hematologic malignancies. To gain further information on this disorder, we searched for ETV6 mutations in the 130 families with inherited thrombocytopenia of unknown origin from our cohort of 274 consecutive pedigrees with familial thrombocytopenia. We identified 20 patients with ETV6-related thrombocytopenia from seven pedigrees. They have five different ETV6 variants, including three novel mutations affecting the highly conserved E26 transformation-specific domain. The relative frequency of ETV6-related thrombocytopenia was 2.6% in the whole case series and 4.6% among the families with known forms of inherited thrombocytopenia. The degree of thrombocytopenia and bleeding tendency of the patients with ETV6-related thrombocytopenia were mild, but four subjects developed B-cell acute lymphoblastic leukemia during childhood, resulting in a significantly higher incidence of this condition compared to that in the general population. Clinical and laboratory findings did not identify any particular defects that could lead to the suspicion of this disorder from the routine diagnostic workup. However, at variance with most inherited thrombocytopenias, platelets were not enlarged. In vitro studies revealed that the maturation of the patients' megakaryocytes was defective and that the patients have impaired proplatelet formation. Moreover, platelets from patients with ETV6-related thrombocytopenia have reduced ability to spread on fibrinogen. Since the dominant thrombocytopenias due to mutations in RUNX1 and ANKRD26 are also characterized by normal platelet size and predispose to hematologic malignancies, we suggest that screening for ETV6, RUNX1 and ANKRD26 mutations should be performed in all subjects with autosomal dominant thrombocytopenia and normal platelet size.
Collapse
Affiliation(s)
- Federica Melazzini
- Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation and University of Pavia, Italy
| | - Flavia Palombo
- Department of Medical and Surgical Science, Policlinico Sant'Orsola Malpighi and University of Bologna, Italy
| | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Italy
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Daniela De Rocco
- Department of Medical, Surgical and Health Sciences, IRCCS Burlo Garofolo and University of Trieste, Italy
| | - Caterina Marconi
- Department of Medical and Surgical Science, Policlinico Sant'Orsola Malpighi and University of Bologna, Italy
| | - Patrizia Noris
- Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation and University of Pavia, Italy
| | - Chiara Gnan
- Department of Medical, Surgical and Health Sciences, IRCCS Burlo Garofolo and University of Trieste, Italy
| | - Tommaso Pippucci
- Department of Medical and Surgical Science, Policlinico Sant'Orsola Malpighi and University of Bologna, Italy
| | - Valeria Bozzi
- Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation and University of Pavia, Italy
| | - Michela Faleschini
- Department of Medical, Surgical and Health Sciences, IRCCS Burlo Garofolo and University of Trieste, Italy
| | - Serena Barozzi
- Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation and University of Pavia, Italy
| | - Michael Doubek
- University Hospital and Masaryk University, Brno, Czech Republic
| | | | - Katerina Stano Kozubik
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Lenka Radova
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Giuseppe Loffredo
- Department of Oncology, Azienda "Santobono-Pausilipon", Pausilipon Hospital, Napoli, Italy
| | - Sarka Pospisilova
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | | | - Marco Seri
- Department of Medical and Surgical Science, Policlinico Sant'Orsola Malpighi and University of Bologna, Italy
| | - Carlo L Balduini
- Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation and University of Pavia, Italy
| | - Alessandro Pecci
- Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation and University of Pavia, Italy
| | - Anna Savoia
- Department of Medical, Surgical and Health Sciences, IRCCS Burlo Garofolo and University of Trieste, Italy
| |
Collapse
|
54
|
Visconte V, Przychodzen B, Han Y, Nawrocki ST, Thota S, Kelly KR, Patel BJ, Hirsch C, Advani AS, Carraway HE, Sekeres MA, Maciejewski JP, Carew JS. Complete mutational spectrum of the autophagy interactome: a novel class of tumor suppressor genes in myeloid neoplasms. Leukemia 2016; 31:505-510. [PMID: 27773925 PMCID: PMC5844476 DOI: 10.1038/leu.2016.295] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- V Visconte
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - B Przychodzen
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Y Han
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - S T Nawrocki
- Department of Medicine, Division of Translational and Regenerative Medicine, University of Arizona Cancer Center, Tucson, AZ, USA
| | - S Thota
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - K R Kelly
- Department of Medicine, USC Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | - B J Patel
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - C Hirsch
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - A S Advani
- Leukemia Program, Department of Hematology/Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - H E Carraway
- Leukemia Program, Department of Hematology/Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - M A Sekeres
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA.,Leukemia Program, Department of Hematology/Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - J P Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - J S Carew
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA.,Department of Medicine, Division of Translational and Regenerative Medicine, University of Arizona Cancer Center, Tucson, AZ, USA
| |
Collapse
|
55
|
Bodei L, Modlin IM, Luster M, Forrer F, Cremonesi M, Hicks RJ, Ezziddin S, Kidd M, Chiti A. Myeloid neoplasms after chemotherapy and PRRT: myth and reality. Endocr Relat Cancer 2016; 23:C1-7. [PMID: 27353035 DOI: 10.1530/erc-16-0258] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 06/27/2016] [Indexed: 12/14/2022]
Abstract
Peptide receptor radionuclide therapy (PRRT) with (90)Y-octreotide or (177)Lu-octreotate is an effective treatment for inoperable or metastatic neuroendocrine tumors (NETs), particularly well-differentiated gastroenteropancreatic or bronchopulmonary NETs. PRRT is generally extremely well tolerated, with modest toxicity to target organs, kidney and bone marrow. Nevertheless, a priori concerns regarding long-term effects lead clinicians such as Brieau and coworkers, in this ERC issue, to ascribe to the combination of alkylating agents and PRRT the apparently high occurrence (n=4) of myeloproliferative events (therapy-related myeloid neoplasms (t-MNs)) in a small cohort of 20 progressive, advanced digestive NETs treated with PRRT after chemotherapy. Anecdotal reports of myelotoxic events should be placed in the correct perspective of larger series, where the reported incidence of these events is ~2%, with the aim of promoting a balanced awareness of the issue and unbiased and reasonable overall conclusions. For a comprehensive definition of the issue, we provide an evaluation of the occurrence of t-MN in patients treated with various myelotoxic treatments.
Collapse
Affiliation(s)
- Lisa Bodei
- Department of RadiologyMemorial Sloan Kettering Cancer Center, New York, New York, USA LuGenIum Consortium for Independent ResearchMilan, Rotterdam, Bad Berka, London
| | - Irvin M Modlin
- LuGenIum Consortium for Independent ResearchMilan, Rotterdam, Bad Berka, London Emeritus Professor Gastroenterological SurgeryYale University, School of Medicine, New Haven, Connecticut, USA
| | - Markus Luster
- Department of Nuclear MedicineUniversity Hospital Marburg, Marburg, Germany
| | - Flavio Forrer
- Nuclear MedicineCantonal Hospital, St. Gallen, Switzerland
| | - Marta Cremonesi
- Division of Health PhysicsEuropean Institute of Oncology, Milan, Italy
| | - Rodney J Hicks
- Centre for Cancer ImagingThe Peter MacCallum Cancer Centre, St Andrew's Place, East Melbourne, Australia
| | - Samer Ezziddin
- Department of Nuclear MedicineSaarland University Hospital, Homburg, Saarland, Germany
| | - Mark Kidd
- Wren LaboratoriesBranford, Connecticut, USA
| | | |
Collapse
|
56
|
How I diagnose and manage individuals at risk for inherited myeloid malignancies. Blood 2016; 128:1800-1813. [PMID: 27471235 DOI: 10.1182/blood-2016-05-670240] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 07/15/2016] [Indexed: 01/24/2023] Open
Abstract
Although inherited hematopoietic malignancies have been reported clinically since the early twentieth century, the molecular basis for these diseases has only recently begun to be elucidated. Growing utilization of next-generation sequencing technologies has facilitated the rapid discovery of an increasing number of recognizable heritable hematopoietic malignancy syndromes while also deepening the field's understanding of the molecular mechanisms that underlie these syndromes. Because individuals with inherited hematopoietic malignancies continue to be underdiagnosed and are increasingly likely to be encountered in clinical practice, clinicians need to have a high index of suspicion and be aware of the described syndromes. Here, we present the methods we use to identify, test, and manage individuals and families suspected of having a hereditary myeloid malignancy syndrome. Finally, we address the areas of ongoing research in the field and encourage clinicians and researchers to contribute and collaborate.
Collapse
|
57
|
Daly ME. Transcription factor defects causing platelet disorders. Blood Rev 2016; 31:1-10. [PMID: 27450272 DOI: 10.1016/j.blre.2016.07.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 06/10/2016] [Accepted: 07/12/2016] [Indexed: 01/19/2023]
Abstract
Recent years have seen increasing recognition of a subgroup of inherited platelet function disorders which are due to defects in transcription factors that are required to regulate megakaryopoiesis and platelet production. Thus, germline mutations in the genes encoding the haematopoietic transcription factors RUNX1, GATA-1, FLI1, GFI1b and ETV6 have been associated with both quantitative and qualitative platelet abnormalities, and variable bleeding symptoms in the affected patients. Some of the transcription factor defects are also associated with an increased predisposition to haematologic malignancies (RUNX1, ETV6), abnormal erythropoiesis (GATA-1, GFI1b, ETV6) and immune dysfunction (FLI1). The persistence of MYH10 expression in platelets is a surrogate marker for FLI1 and RUNX1 defects. Characterisation of the transcription factor defects that give rise to platelet function disorders, and of the genes that are differentially regulated as a result, are yielding insights into the roles of these genes in platelet formation and function.
Collapse
Affiliation(s)
- Martina E Daly
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Beech Hill Road, Sheffield, S10 2RX, UK.
| |
Collapse
|
58
|
Mutations of myelodysplastic syndromes (MDS): An update. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2016; 769:47-62. [DOI: 10.1016/j.mrrev.2016.04.009] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/11/2016] [Indexed: 01/08/2023]
|
59
|
Balduini CL, Melazzini F, Pecci A. Inherited thrombocytopenias-recent advances in clinical and molecular aspects. Platelets 2016; 28:3-13. [PMID: 27161842 DOI: 10.3109/09537104.2016.1171835] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Since the beginning of the century, our knowledge of inherited thrombocytopenias greatly advanced, and we presently know 30 forms with well-defined genetic defects. This great advancement changed our view of these disorders, as we realized that most patients have only mild thrombocytopenia with inconspicuous bleeding or no bleeding tendency at all. However, better knowledge of inherited thrombocytopenias also revealed that some of the most prevalent forms expose to the risk of acquiring during infancy or adulthood additional disorders that endanger the life of patients much more than hemorrhages. Thus, inherited thrombocytopenias are complex disorders with quite different clinical features and prognosis. Identification of novel genes whose mutations result in low platelet count greatly advanced also our knowledge of the megakaryocyte biology and proved beyond any doubt that the defective proteins play an essential role in platelet biogenesis or survival in humans. Based on the study of inherited thrombocytopenias, we better understood the sequence of molecular events regulating megakaryocyte differentiation, maturation, and platelet release. Since nearly 50% of patients have as yet unidentified genetic or molecular mechanisms underlying their inherited thrombocytopenia, further studies are expected to reveal new clinical entities and new molecular mechanisms of platelet production.
Collapse
Affiliation(s)
- Carlo L Balduini
- a Department of Medicine , IRCCS Policlinico San Matteo Foundation - University of Pavia , Pavia , Italy
| | - Federica Melazzini
- a Department of Medicine , IRCCS Policlinico San Matteo Foundation - University of Pavia , Pavia , Italy
| | - Alessandro Pecci
- a Department of Medicine , IRCCS Policlinico San Matteo Foundation - University of Pavia , Pavia , Italy
| |
Collapse
|
60
|
Yoshimi A, Toya T, Nannya Y, Takaoka K, Kirito K, Ito E, Nakajima H, Hayashi Y, Takahashi T, Moriya-Saito A, Suzuki K, Harada H, Komatsu N, Usuki K, Ichikawa M, Kurokawa M. Spectrum of clinical and genetic features of patients with inherited platelet disorder with suspected predisposition to hematological malignancies: a nationwide survey in Japan. Ann Oncol 2016; 27:887-95. [DOI: 10.1093/annonc/mdw066] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Accepted: 02/08/2016] [Indexed: 11/13/2022] Open
|
61
|
Latger-Cannard V, Philippe C, Bouquet A, Baccini V, Alessi MC, Ankri A, Bauters A, Bayart S, Cornillet-Lefebvre P, Daliphard S, Mozziconacci MJ, Renneville A, Ballerini P, Leverger G, Sobol H, Jonveaux P, Preudhomme C, Nurden P, Lecompte T, Favier R. Haematological spectrum and genotype-phenotype correlations in nine unrelated families with RUNX1 mutations from the French network on inherited platelet disorders. Orphanet J Rare Dis 2016; 11:49. [PMID: 27112265 PMCID: PMC4845427 DOI: 10.1186/s13023-016-0432-0] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 04/18/2016] [Indexed: 11/20/2022] Open
Abstract
Background Less than 50 patients with FPD/AML (OMIM 601309) have been reported as of today and there may an underestimation. The purpose of this study was to describe the natural history, the haematological features and the genotype-phenotype correlations of this entity in order to, first, screen it better and earlier, before leukaemia occurrence and secondly to optimize appropriate monitoring and treatment, in particular when familial stem cell transplantation is considered. Methods We have investigated 41 carriers of RUNX1 alteration belonging to nine unrelated French families with FPD/AML and two syndromic patients, registered in the French network on rare platelet disorders from 2005 to 2015. Results Five missense, one non-sense, three frameshift mutations and two large deletions involving several genes including RUNX1 were evidenced. The history of familial leukaemia was suggestive of FPD/AML in seven pedigrees, whereas an autosomal dominant pattern of lifelong thrombocytopenia was the clinical presentation of two. Additional syndromic features characterized two large sporadic deletions. Bleeding tendency was mild and thrombocytopenia moderate (>50 x109/L), with normal platelet volume. A functional platelet defect consistent with a δ-granule release defect was found in ten patients regardless of the type of RUNX1 alteration. The incidence of haematological malignancies was higher when the mutated RUNX1 allele was likely to cause a dominant negative effect (19/34) in comparison with loss of function alleles (3/9). A normal platelet count does not rule out the diagnosis of FPD/AML, since the platelet count was found normal for three mutated subjects, a feature that has a direct impact in the search for a related donor in case of allogeneic haematopoietic stem cell transplantation. Conclusions Platelet dysfunction suggestive of defective δ-granule release could be of values for the diagnosis of FPD/AML particularly when the clinical presentation is an autosomal dominant thrombocytopenia with normal platelet size in the absence of familial malignancies. The genotype-phenotype correlations might be helpful in genetic counselling and appropriate optimal therapeutic management.
Collapse
Affiliation(s)
- Veronique Latger-Cannard
- Service d'Hématologie Biologique, Centre Hospitalier Universitaire de Nancy, Nancy, France.,Centre de Compétence Nord-Est des Pathologies Plaquettaires (CCPP), Nancy, France
| | - Christophe Philippe
- Laboratoire de Génétique, Centre Hospitalier Universitaire de Nancy, Nancy, France
| | - Alexandre Bouquet
- Service d'Hématologie Biologique, Centre de Biologie Pathologie, Centre Hospitalier Régional Universitaire de Lille, Lille, France
| | - Veronique Baccini
- Laboratoire d'Hématologie, Hôpital La Timone, Marseille, France.,Centre de Référence des Pathologies Plaquettaires (CRPP), Hôpital La Timone, Marseille, France
| | - Marie-Christine Alessi
- Laboratoire d'Hématologie, Hôpital La Timone, Marseille, France.,Centre de Référence des Pathologies Plaquettaires (CRPP), Hôpital La Timone, Marseille, France
| | - Annick Ankri
- Assistance Publique-Hôpitaux de Paris, Laboratoire d'Hématologie, La Pitié Salpetrière, Paris, France
| | - Anne Bauters
- Centre de Compétence Nord-Est des Pathologies Plaquettaires (CCPP), Nancy, France.,Service d'Hématologie Biologique, Centre de Biologie Pathologie, Centre Hospitalier Régional Universitaire de Lille, Lille, France
| | - Sophie Bayart
- Centre Régional de Traitement des Hémophiles, Centre Hospitalier Universitaire de Rennes, Rennes, France
| | | | - Sylvie Daliphard
- Laboratoire d'Hématologie, Centre Hospitalier Universitaire Robert Debré, Reims, France
| | - Marie-Joelle Mozziconacci
- Département de Biopathologie, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille, Marseille, France
| | - Aline Renneville
- Service d'Hématologie Biologique, Centre de Biologie Pathologie, Centre Hospitalier Régional Universitaire de Lille, Lille, France
| | - Paola Ballerini
- Assistance Publique-Hôpitaux de Paris, Département d'Hématologie, Hôpital Armand Trousseau, Paris, France.,Centre de Référence des Pathologies Plaquettaires (CRPP), Hôpital La Timone, Marseille, France
| | - Guy Leverger
- Assistance Publique-Hôpitaux de Paris, Département d'Hématologie, Hôpital Armand Trousseau, Paris, France.,Centre de Référence des Pathologies Plaquettaires (CRPP), Hôpital La Timone, Marseille, France
| | - Hagay Sobol
- Département de Biopathologie, Institut Paoli-Calmettes, Centre de Recherche en Cancérologie de Marseille, Marseille, France
| | - Philippe Jonveaux
- Laboratoire de Génétique, Centre Hospitalier Universitaire de Nancy, Nancy, France
| | - Claude Preudhomme
- Service d'Hématologie Biologique, Centre de Biologie Pathologie, Centre Hospitalier Régional Universitaire de Lille, Lille, France
| | - Paquita Nurden
- Laboratoire d'Hématologie, Hôpital La Timone, Marseille, France.,Centre de Référence des Pathologies Plaquettaires (CRPP), Hôpital La Timone, Marseille, France
| | - Thomas Lecompte
- Service d'Hématologie, Hôpitaux Universitaires de Genève, Geneva, Switzerland.,Faculté de Médecine, Université de Genève, Geneva, Switzerland
| | - Remi Favier
- Assistance Publique-Hôpitaux de Paris, Département d'Hématologie, Hôpital Armand Trousseau, Paris, France. .,Inserm U1170, Villejuif, France. .,Centre de Référence des Pathologies Plaquettaires (CRPP), Hôpital La Timone, Marseille, France. .,Service d'Hématologie Biologique, Hôpital d'enfants Armand Trousseau, 26 Avenue du Dr Netter, 75012, Paris, France.
| |
Collapse
|
62
|
Runx1 downregulates stem cell and megakaryocytic transcription programs that support niche interactions. Blood 2016; 127:3369-81. [PMID: 27076172 DOI: 10.1182/blood-2015-09-668129] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 03/31/2016] [Indexed: 12/20/2022] Open
Abstract
Disrupting mutations of the RUNX1 gene are found in 10% of patients with myelodysplasia (MDS) and 30% of patients with acute myeloid leukemia (AML). Previous studies have revealed an increase in hematopoietic stem cells (HSCs) and multipotent progenitor (MPP) cells in conditional Runx1-knockout (KO) mice, but the molecular mechanism is unresolved. We investigated the myeloid progenitor (MP) compartment in KO mice, arguing that disruptions at the HSC/MPP level may be amplified in downstream cells. We demonstrate that the MP compartment is increased by more than fivefold in Runx1 KO mice, with a prominent skewing toward megakaryocyte (Meg) progenitors. Runx1-deficient granulocyte-macrophage progenitors are characterized by increased cloning capacity, impaired development into mature cells, and HSC and Meg transcription signatures. An HSC/MPP subpopulation expressing Meg markers was also increased in Runx1-deficient mice. Rescue experiments coupled with transcriptome analysis and Runx1 DNA-binding assays demonstrated that granulocytic/monocytic (G/M) commitment is marked by Runx1 suppression of genes encoding adherence and motility proteins (Tek, Jam3, Plxnc1, Pcdh7, and Selp) that support HSC-Meg interactions with the BM niche. In vitro assays confirmed that enforced Tek expression in HSCs/MPPs increases Meg output. Interestingly, besides this key repressor function of Runx1 to control lineage decisions and cell numbers in progenitors, our study also revealed a critical activating function in erythroblast differentiation, in addition to its known importance in Meg and G/M maturation. Thus both repressor and activator functions of Runx1 at multiple hematopoietic stages and lineages likely contribute to the tumor suppressor activity in MDS and AML.
Collapse
|
63
|
de Gonzalez AB, Salotti JA, McHugh K, Little MP, Harbron RW, Lee C, Ntowe E, Braganza MZ, Parker L, Rajaraman P, Stiller C, Stewart DR, Craft AW, Pearce MS. Relationship between paediatric CT scans and subsequent risk of leukaemia and brain tumours: assessment of the impact of underlying conditions. Br J Cancer 2016; 114:388-94. [PMID: 26882064 PMCID: PMC4815765 DOI: 10.1038/bjc.2015.415] [Citation(s) in RCA: 178] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 09/10/2015] [Accepted: 11/01/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND We previously reported evidence of a dose-response relationship between ionising-radiation exposure from paediatric computed tomography (CT) scans and the risk of leukaemia and brain tumours in a large UK cohort. Underlying unreported conditions could have introduced bias into these findings. METHODS We collected and reviewed additional clinical information from radiology information systems (RIS) databases, underlying cause of death and pathology reports. We conducted sensitivity analyses excluding participants with cancer-predisposing conditions or previous unreported cancers and compared the dose-response analyses with our original results. RESULTS We obtained information from the RIS and death certificates for about 40% of the cohort (n∼180 000) and found cancer-predisposing conditions in 4 out of 74 leukaemia/myelodysplastic syndrome (MDS) cases and 13 out of 135 brain tumour cases. As these conditions were unrelated to CT exposure, exclusion of these participants did not alter the dose-response relationships. We found evidence of previous unreported cancers in 2 leukaemia/MDS cases, 7 brain tumour cases and 232 in non-cases. These previous cancers were related to increased number of CTs. Exclusion of these cancers reduced the excess relative risk per mGy by 15% from 0.036 to 0.033 for leukaemia/MDS (P-trend=0.02) and by 30% from 0.023 to 0.016 (P-trend<0.0001) for brain tumours. When we included pathology reports we had additional clinical information for 90% of the cases. Additional exclusions from these reports further reduced the risk estimates, but this sensitivity analysis may have underestimated risks as reports were only available for cases. CONCLUSIONS Although there was evidence of some bias in our original risk estimates, re-analysis of the cohort with additional clinical data still showed an increased cancer risk after low-dose radiation exposure from CT scans in young patients.
Collapse
Affiliation(s)
| | - Jane A Salotti
- Institute of Health and Society, Newcastle University, Sir James Spence Institute, Royal Victoria Infirmary, Newcastle upon Tyne NE1 4LP, UK
| | - Kieran McHugh
- Great Ormond Street Hospital for Children NHS Trust, London WC1N 3JH, UK
| | - Mark P Little
- Radiation Epidemiology Unit, Division of Cancer Epidemiology and Genetics, NCI, Bethesda, MD, USA
| | - Richard W Harbron
- Institute of Health and Society, Newcastle University, Sir James Spence Institute, Royal Victoria Infirmary, Newcastle upon Tyne NE1 4LP, UK
| | - Choonsik Lee
- Radiation Epidemiology Unit, Division of Cancer Epidemiology and Genetics, NCI, Bethesda, MD, USA
| | - Estelle Ntowe
- Radiation Epidemiology Unit, Division of Cancer Epidemiology and Genetics, NCI, Bethesda, MD, USA
| | - Melissa Z Braganza
- Radiation Epidemiology Unit, Division of Cancer Epidemiology and Genetics, NCI, Bethesda, MD, USA
| | - Louise Parker
- Departments of Medicine and Paediatrics, Population Cancer Research Program, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Preetha Rajaraman
- Radiation Epidemiology Unit, Division of Cancer Epidemiology and Genetics, NCI, Bethesda, MD, USA
| | | | - Douglas R Stewart
- Radiation Epidemiology Unit, Division of Cancer Epidemiology and Genetics, NCI, Bethesda, MD, USA
| | - Alan W Craft
- Institute of Health and Society, Newcastle University, Sir James Spence Institute, Royal Victoria Infirmary, Newcastle upon Tyne NE1 4LP, UK
| | - Mark S Pearce
- Institute of Health and Society, Newcastle University, Sir James Spence Institute, Royal Victoria Infirmary, Newcastle upon Tyne NE1 4LP, UK
| |
Collapse
|
64
|
Schmiegelow K. Treatment-related toxicities in children with acute lymphoblastic leukaemia predisposition syndromes. Eur J Med Genet 2016; 59:654-660. [PMID: 26876989 DOI: 10.1016/j.ejmg.2016.02.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 02/09/2016] [Accepted: 02/09/2016] [Indexed: 12/11/2022]
Abstract
Although most children with acute lymphoblastic leukaemia (ALL) do not harbor germline mutations that strongly predispose them to development of this malignancy, large syndrome registries and detailed mapping of exomes or whole genomes of familial leukaemia kindreds have revealed that 3-5% of all childhood ALL cases are due to such germline mutations, but the figure may be higher. Most of these syndromes are primarily characterized by their non-malignant phenotype, whereas ALL may be the dominating or even only striking manifestation of the syndrome in some families. Identification of such ALL patients is important in order to adjust therapy and offer genetic counseling and cancer surveillance to mutation carriers in the family. In the coming years large genomic screening projects are expected to reveal further hitherto unrecognised familial ALL syndromes. The treatment of ALL cases harboring cancer predisposing mutations can be challenging for both the physician and the patient due to their preexisting symptoms, their reduced tolerance to radio- and/or chemotherapy with enhanced risk of life-threatening organ toxicities, and the paucity of data from ALL patients with the same or similar syndromes being treated by contemporary protocols. Recent studies clearly indicate that many of these patients stand a good chance of cure, and that they should be offered chemotherapy with the intention to cure. Some of these syndromes are characterized by reduced tolerance to radiotherapy and/or specific anticancer agents, while others are not. This review summarises our current knowledge on the risk of acute toxicities for these ALL patients and provides guidance for treatment adjustments.
Collapse
Affiliation(s)
- Kjeld Schmiegelow
- Department of Pediatrics and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark; Institute of Clinical Medicine, University of Copenhagen, Denmark; Division of Pediatric Hematology/Oncology, New York, USA; Perlmutter Cancer Center, NYU Langone Medical Center, New York, USA.
| |
Collapse
|
65
|
Ok CY, Leventaki V, Wang SA, Dinardo C, Medeiros LJ, Konoplev S. Detection of an Abnormal Myeloid Clone by Flow Cytometry in Familial Platelet Disorder With Propensity to Myeloid Malignancy. Am J Clin Pathol 2016; 145:271-6. [PMID: 26800764 DOI: 10.1093/ajcp/aqv080] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES To report aberrant myeloblasts detected by flow cytometry immunophenotypic studies in an asymptomatic patient with familial platelet disorder with propensity to myeloid malignancy, a rare autosomal dominant disease caused by germline heterozygous mutations in Runt-related transcription factor 1. METHODS Morphologic evaluation, flow cytometry immunophenotypic studies, nanofluidics-based qualitative multiplex reverse transcriptase polymerase chain reaction, Sanger sequencing, and next-generation sequencing-based mutational hotspot analysis of 53 genes were performed on bone marrow biopsy and aspirate samples. RESULTS Flow cytometry immunophenotypic analysis showed 0.6% CD34+ blasts with an abnormal immunophenotype: CD13 increased, CD33+, CD38 decreased, CD117 increased, and CD123 increased. CONCLUSIONS The acquisition of new phenotypic aberrancies in myeloblasts as detected by flow cytometry immunophenotypic studies might be a harbinger of impending myelodysplastic syndrome or acute myeloid leukemia in a patient with familial platelet disorder with propensity to myeloid malignancy.
Collapse
Affiliation(s)
| | - Vasiliki Leventaki
- Department of Pathology, St Jude Children's Research Hospital, Memphis, TN
| | - Sa A Wang
- From the Departments of Hematopathology and
| | - Courtney Dinardo
- Leukemia, University of Texas MD Anderson Cancer Center, Houston
| | | | | |
Collapse
|
66
|
Linkage between the mechanisms of thrombocytopenia and thrombopoiesis. Blood 2016; 127:1234-41. [PMID: 26787737 DOI: 10.1182/blood-2015-07-607903] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 08/19/2015] [Indexed: 12/30/2022] Open
Abstract
Thrombocytopenia is defined as a status in which platelet numbers are reduced. Imbalance between the homeostatic regulation of platelet generation and destruction is 1 potential cause of thrombocytopenia. In adults, platelet generation is a 2-stage process entailing the differentiation of hematopoietic stem cells into mature megakaryocytes (MKs; known as megakaryopoiesis) and release of platelets from MKs (known as thrombopoiesis or platelet biogenesis). Until recently, information about the genetic defects responsible for congenital thrombocytopenia was only available for a few forms of the disease. However, investigations over the past 15 years have identified mutations in genes encoding >20 different proteins that are responsible for these disorders, which has advanced our understanding of megakaryopoiesis and thrombopoiesis. The underlying pathogenic mechanisms can be categorized as (1) defects in MK lineage commitment and differentiation, (2) defects in MK maturation, and (3) defect in platelet release. Using these developmental stage categories, we here update recently described mechanisms underlying megakaryopoiesis and thrombopoiesis and discuss the association between platelet generation systems and thrombocytopenia.
Collapse
|
67
|
Bitan M, van Walraven SM, Worel N, Ball LM, Styczynski J, Torrabadella M, Witt V, Shaw BE, Seber A, Yabe H, Greinix HT, Peters C, Gluckman E, Rocha V, Halter J, Pulsipher MA. Determination of Eligibility in Related Pediatric Hematopoietic Cell Donors: Ethical and Clinical Considerations. Recommendations from a Working Group of the Worldwide Network for Blood and Marrow Transplantation Association. Biol Blood Marrow Transplant 2016; 22:96-103. [DOI: 10.1016/j.bbmt.2015.08.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 08/12/2015] [Indexed: 11/27/2022]
|
68
|
Babushok DV, Bessler M, Olson TS. Genetic predisposition to myelodysplastic syndrome and acute myeloid leukemia in children and young adults. Leuk Lymphoma 2015; 57:520-36. [PMID: 26693794 DOI: 10.3109/10428194.2015.1115041] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Myelodysplastic syndrome (MDS) is a clonal blood disorder characterized by ineffective hematopoiesis, cytopenias, dysplasia and an increased risk of acute myeloid leukemia (AML). With the growing availability of clinical genetic testing, there is an increasing appreciation that a number of genetic predisposition syndromes may underlie apparent de novo presentations of MDS/AML, particularly in children and young adults. Recent findings of clonal hematopoiesis in acquired aplastic anemia add another facet to our understanding of the mechanisms of MDS/AML predisposition. As more predisposition syndromes are recognized, it is becoming increasingly important for hematologists and oncologists to have familiarity with the common as well as emerging syndromes, and to have a systematic approach to diagnosis and screening of at risk patient populations. Here, we provide a practical algorithm for approaching a patient with a suspected MDS/AML predisposition, and provide an in-depth review of the established and emerging familial MDS/AML syndromes caused by mutations in the ANKRD26, CEBPA, DDX41, ETV6, GATA2, RUNX1, SRP72 genes. Finally, we discuss recent data on the role of somatic mutations in malignant transformation in acquired aplastic anemia, and review the practical aspects of MDS/AML management in patients and families with predisposition syndromes.
Collapse
Affiliation(s)
- Daria V Babushok
- a Division of Hematology-Oncology, Department of Medicine , Hospital of the University of Pennsylvania , Philadelphia , PA , USA ;,b Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of Pediatrics , Children's Hospital of Philadelphia , Philadelphia , PA , USA
| | - Monica Bessler
- a Division of Hematology-Oncology, Department of Medicine , Hospital of the University of Pennsylvania , Philadelphia , PA , USA ;,b Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of Pediatrics , Children's Hospital of Philadelphia , Philadelphia , PA , USA
| | - Timothy S Olson
- b Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of Pediatrics , Children's Hospital of Philadelphia , Philadelphia , PA , USA ;,c Blood and Marrow Transplant Program, Division of Oncology, Department of Pediatrics , Children's Hospital of Philadelphia and University of Pennsylvania , Philadelphia , PA , USA
| |
Collapse
|
69
|
Baccini V, Alessi MC. [Diagnosis of inherited thrombocytopenia]. Rev Med Interne 2015; 37:117-26. [PMID: 26617290 DOI: 10.1016/j.revmed.2015.10.346] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 10/19/2015] [Indexed: 12/19/2022]
Abstract
Inherited thrombocytopenias are rare, heterogenous and probably under-diagnosed because often classified as autoimmune thrombocytopenia. About 20 genes were described responsible for these thrombocytopenias. Precise diagnosis is necessary because the prognosis is different and some of them can evolve into hemopathies. First of all, it is important to gather a body of evidence to orientate towards an inherited cause: presence of the thrombocytopenia since childhood and of other family cases is a strong argument. Secondly, it is difficult to target the genetic investigations that settle the precise diagnosis. Genetic variants responsible for inherited thrombocytopenias affect different stage during megakaryocytopoiesis and cause thrombocytopenias with distinct characteristics. Presence of extra-hematological features, platelets' size measurement and evaluation of bone marrow megakaryocyte morphology when it is possible allow a primary orientation. We propose a diagnostic approach considering extra-hematological features, mode of inheritance, morphology, molecular and functional platelets' studies and bone marrow megakaryocyte morphology in order to better target genetic study. Nevertheless, despite this approach, some inherited thrombocytopenias remain still unexplained and could benefit from new methods of new generation sequencing in the future.
Collapse
Affiliation(s)
- V Baccini
- Laboratoire d'hématologie, hôpital Nord, CHU de Marseille, chemin des Bourrelly, 13015 Marseille, France; Centre de référence des pathologies plaquettaires (CRPP), CHU Timone, 264, rue Saint-Pierre, 13385 Marseille cedex 5, France.
| | - M C Alessi
- Laboratoire d'hématologie, hôpital Nord, CHU de Marseille, chemin des Bourrelly, 13015 Marseille, France; Centre de référence des pathologies plaquettaires (CRPP), CHU Timone, 264, rue Saint-Pierre, 13385 Marseille cedex 5, France
| |
Collapse
|
70
|
Wu D, Du X, Jin J, Xiao Z, Shen Z, Shao Z, Li X, Huang X, Liu T, Yu L, Li J, Chen B, He G, Cai Z, Liang H, Li J, Ruan C. Decitabine for Treatment of Myelodysplastic Syndromes in Chinese Patients: An Open-Label, Phase-3b Study. Adv Ther 2015; 32:1140-59. [PMID: 26568466 PMCID: PMC4662721 DOI: 10.1007/s12325-015-0263-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Indexed: 02/05/2023]
Abstract
Introduction The objective of this study was to evaluate the
efficacy and safety of decitabine in Chinese patients with myelodysplastic syndrome (MDS). Methods Patients (≥18 years) who had a de novo or secondary MDS diagnosis according to French–American–British classification and an International Prognostic Scoring System score ≥0.5 were enrolled and randomized (1:1) to one of two decitabine regimens: 3-day treatment (3-h intravenous infusion of 15 mg/m2 given every 8 h for three consecutive days/cycle/6 weeks) or 5-day treatment (1-h intravenous infusion of 20 mg/m2 once daily on days 1–5/cycle/4 weeks). After a minimum of 30 patients were assigned to 3-day schedule, the remaining were assigned to the 5-day schedule. The primary efficacy endpoint was the overall response rate (ORR). Secondary outcome measures included hematologic improvement (HI), cytogenetic response rate, the time to acute myeloid leukemia (AML) progression, and overall survival (OS). Results In total, 132 of 135 enrolled patients (3-day treatment, n = 36; 5-day treatment, n = 99) discontinued treatment (major reasons included patient withdrawal/lack of efficacy, n = 48; adverse events, n = 23; and disease progression, n = 22). During the study, 35 of 132 (26.5%) patients from the intent-to-treat (ITT) group achieved significant (P < 0.001) ORR [3-day group (n = 10, 29.4%), P = 0.003; 5-day group (n = 25, 25.5%), P < 0.001]. The HI rate was similar between the 3-day (47.1%) and 5-day groups (48.0%). Cytogenetic response was achieved in 20 of the 30 (66.7%) patients who had a baseline cytogenetic abnormality. Fifty-three (40.2%) AML transformations or deaths occurred and the median AML-free survival time was 23.8 months for all patients from the ITT set; 24-month OS rate was 48.9%. Adverse events of myelosuppression-related disorders (85.6%) and infections (43.2%) were commonly reported. Conclusion Decitabine treatment was efficacious in Chinese patients with MDS with its safety profile comparable to the global studies of decitabine conducted to date. Funding Xian-Janssen Pharmaceutical Ltd. China (a company of Johnson & Johnson). Trial registration ClinicalTrials.gov identifier, NCT01751867. Electronic supplementary material The online version of this article (doi:10.1007/s12325-015-0263-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Depei Wu
- The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Xin Du
- Guangdong General Hospital, Guangzhou, People's Republic of China
| | - Jie Jin
- The First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Zhijian Xiao
- Institute of Hematology and Blood Diseases Hospital, Tianjin, People's Republic of China
| | - Zhixiang Shen
- Shanghai Jiaotong University Medical College Affiliated Ruijin Hospital, Shanghai, People's Republic of China
| | - Zonghong Shao
- Tianjin Medical University of General Hospital, Tianjin, People's Republic of China
| | - Xiao Li
- Shanghai 6th People's Hospital, Shanghai, People's Republic of China
| | - Xiaojun Huang
- Peking University People's Hospital, Beijing, People's Republic of China
| | - Ting Liu
- West China Hospital of Sichuan University, Chengdu, People's Republic of China
| | - Li Yu
- Chinese People's Liberation Army General Hospital, Beijing, People's Republic of China
| | - Jianyong Li
- Jiangsu Province Hospital/The First Affiliated Hospital With Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | - Baoan Chen
- Zhongda Hospital Affiliated To Southeast University, Nanjing, People's Republic of China
| | - Guangsheng He
- The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Zhen Cai
- The First Affiliated Hospital, Zhejiang University College of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Hongchuang Liang
- Xian-Janssen Pharmaceutical Ltd., Beijing, People's Republic of China
| | - Jigang Li
- Xian-Janssen Pharmaceutical Ltd., Beijing, People's Republic of China
| | - Changgeng Ruan
- The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China.
| |
Collapse
|
71
|
Noris P, Balduini CL. Inherited thrombocytopenias in the era of personalized medicine. Haematologica 2015; 100:145-8. [PMID: 25638803 DOI: 10.3324/haematol.2014.122549] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Patrizia Noris
- Department of Internal Medicine, University of Pavia - IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Carlo L Balduini
- Department of Internal Medicine, University of Pavia - IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| |
Collapse
|
72
|
Duchmann M, Fenaux P, Cluzeau T. [Management of myelodysplastic syndromes]. Bull Cancer 2015; 102:946-57. [PMID: 26410692 DOI: 10.1016/j.bulcan.2015.07.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Revised: 06/25/2015] [Accepted: 07/10/2015] [Indexed: 12/16/2022]
Abstract
Myelodysplastic syndromes are heterogeneous diseases whose molecular characteristics have only been identified in recent years. Better identification of prognostic factors, larger access to allogeneic stem cell transplantation and the advent of new drugs notably hypomethylating agents (azacitidine, decitabine) and lenalidomide have improved patient outcome.
Collapse
Affiliation(s)
| | - Pierre Fenaux
- Université Paris 7, Assistance publique des Hôpitaux de Paris, hôpital Saint-Louis, service d'hématologie séniors, 75010 Paris, France
| | - Thomas Cluzeau
- Assistance publique des Hôpitaux de Paris, hôpital Saint-Louis, service d'hématologie, 75010 Paris, France; Centre méditerranéen de médecine moléculaire, Inserm U1065, 06204 Nice, France.
| |
Collapse
|
73
|
Somatic mutations associated with leukemic progression of familial platelet disorder with predisposition to acute myeloid leukemia. Leukemia 2015; 30:999-1002. [DOI: 10.1038/leu.2015.236] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
74
|
Abstract
OBJECTIVES Acute myeloid leukemia with myelodysplasia-related changes (AML-MRC) is a heterogeneous disorder defined by morphologic, genetic, or clinical features. Genetic abnormalities associated with AML-MRC are often associated with adverse prognostic features, and many cases are preceded by a myelodysplastic syndrome (MDS) or a myelodysplastic/myeloproliferative neoplasm. METHODS Although the criteria of 20% or more blasts in blood or bone marrow and multilineage dysplasia affecting 50% or more of cells in two or more of the myeloid lineages seem straightforward for AML-MRC, identification of morphologic dysplasia among observers is not always consistent, and there is morphologic overlap with other leukemic disorders such as acute erythroleukemia. RESULTS Session 3 of the workshop cases displayed heterogeneity as expected within AML-MRC, yet several cases suggested that recently recognized entities may exist within this category, such as familial MDS/AML predisposition syndromes and rare cases of high-risk AML associated with the cryptic t(5;11)(q35;p15);NUP98-NSD1 that may masquerade as a del(5q). However, most cases of AML-MRC were usually associated with adverse genetic abnormalities, particularly -5/del(5q), -7/del(7q), and/or complex karyotypes. CONCLUSIONS Whole-genome sequencing and array studies may identify genetic abnormalities, such as those affecting TP53, which may provide prognostic information.
Collapse
|
75
|
Topka S, Vijai J, Walsh MF, Jacobs L, Maria A, Villano D, Gaddam P, Wu G, McGee RB, Quinn E, Inaba H, Hartford C, Pui CH, Pappo A, Edmonson M, Zhang MY, Stepensky P, Steinherz P, Schrader K, Lincoln A, Bussel J, Lipkin SM, Goldgur Y, Harit M, Stadler ZK, Mullighan C, Weintraub M, Shimamura A, Zhang J, Downing JR, Nichols KE, Offit K. Germline ETV6 Mutations Confer Susceptibility to Acute Lymphoblastic Leukemia and Thrombocytopenia. PLoS Genet 2015; 11:e1005262. [PMID: 26102509 PMCID: PMC4477877 DOI: 10.1371/journal.pgen.1005262] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 05/05/2015] [Indexed: 12/30/2022] Open
Abstract
Somatic mutations affecting ETV6 often occur in acute lymphoblastic leukemia (ALL), the most common childhood malignancy. The genetic factors that predispose to ALL remain poorly understood. Here we identify a novel germline ETV6 p. L349P mutation in a kindred affected by thrombocytopenia and ALL. A second ETV6 p. N385fs mutation was identified in an unrelated kindred characterized by thrombocytopenia, ALL and secondary myelodysplasia/acute myeloid leukemia. Leukemic cells from the proband in the second kindred showed deletion of wild type ETV6 with retention of the ETV6 p. N385fs. Enforced expression of the ETV6 mutants revealed normal transcript and protein levels, but impaired nuclear localization. Accordingly, these mutants exhibited significantly reduced ability to regulate the transcription of ETV6 target genes. Our findings highlight a novel role for ETV6 in leukemia predisposition.
Collapse
Affiliation(s)
- Sabine Topka
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, United States of America
- Cancer Biology and Genetics Program, Sloan Kettering Institute, New York, New York, United States of America
| | - Joseph Vijai
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, United States of America
- Cancer Biology and Genetics Program, Sloan Kettering Institute, New York, New York, United States of America
| | - Michael F. Walsh
- St Jude Children’s Research Hospital (SJCRH), Memphis, Tennessee, United States of America
| | - Lauren Jacobs
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, United States of America
| | - Ann Maria
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, United States of America
| | - Danylo Villano
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, United States of America
| | - Pragna Gaddam
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, United States of America
| | - Gang Wu
- St Jude Children’s Research Hospital (SJCRH), Memphis, Tennessee, United States of America
| | - Rose B. McGee
- St Jude Children’s Research Hospital (SJCRH), Memphis, Tennessee, United States of America
| | - Emily Quinn
- St Jude Children’s Research Hospital (SJCRH), Memphis, Tennessee, United States of America
| | - Hiroto Inaba
- St Jude Children’s Research Hospital (SJCRH), Memphis, Tennessee, United States of America
| | - Christine Hartford
- St Jude Children’s Research Hospital (SJCRH), Memphis, Tennessee, United States of America
| | - Ching-hon Pui
- St Jude Children’s Research Hospital (SJCRH), Memphis, Tennessee, United States of America
| | - Alberto Pappo
- St Jude Children’s Research Hospital (SJCRH), Memphis, Tennessee, United States of America
| | - Michael Edmonson
- St Jude Children’s Research Hospital (SJCRH), Memphis, Tennessee, United States of America
| | - Michael Y. Zhang
- Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington, United States of America
| | - Polina Stepensky
- Pediatric Hematology/Oncology Department, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Peter Steinherz
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, United States of America
| | | | - Anne Lincoln
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, United States of America
| | - James Bussel
- Weill Cornell Medical College, New York, New York, United States of America
| | - Steve M. Lipkin
- Weill Cornell Medical College, New York, New York, United States of America
| | - Yehuda Goldgur
- Structural Biology Program, Sloan Kettering Institute, New York, New York, United States of America
| | - Mira Harit
- Pediatric Hematology/Oncology Department, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Zsofia K. Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, United States of America
| | - Charles Mullighan
- St Jude Children’s Research Hospital (SJCRH), Memphis, Tennessee, United States of America
| | - Michael Weintraub
- Pediatric Hematology/Oncology Department, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Akiko Shimamura
- Fred Hutchinson Cancer Research Center and University of Washington, Seattle, Washington, United States of America
- Seattle Children’s Hospital, Seattle, Washington, United States of America
| | - Jinghui Zhang
- St Jude Children’s Research Hospital (SJCRH), Memphis, Tennessee, United States of America
| | - James R. Downing
- St Jude Children’s Research Hospital (SJCRH), Memphis, Tennessee, United States of America
| | - Kim E. Nichols
- St Jude Children’s Research Hospital (SJCRH), Memphis, Tennessee, United States of America
| | - Kenneth Offit
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, United States of America
- Cancer Biology and Genetics Program, Sloan Kettering Institute, New York, New York, United States of America
- Weill Cornell Medical College, New York, New York, United States of America
| |
Collapse
|
76
|
Savoia A. Molecular basis of inherited thrombocytopenias. Clin Genet 2015; 89:154-62. [DOI: 10.1111/cge.12607] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Revised: 05/04/2015] [Accepted: 05/05/2015] [Indexed: 02/01/2023]
Affiliation(s)
- A. Savoia
- Department of Medical SciencesUniversity of Trieste Trieste Italy
- Institute for Maternal and Child HealthIRCCS Burlo Garofolo Trieste Italy
| |
Collapse
|
77
|
Songdej N, Rao AK. Hematopoietic transcription factor mutations and inherited platelet dysfunction. F1000PRIME REPORTS 2015; 7:66. [PMID: 26097739 PMCID: PMC4447035 DOI: 10.12703/p7-66] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The molecular and genetic mechanisms in most patients with inherited platelet dysfunction are unknown. There is increasing evidence that mutations in hematopoietic transcription factors are major players in the pathogenesis of defective megakaryopoiesis and platelet dysfunction in patients with inherited platelet disorders. These hematopoietic transcription factors include RUNX1, FLI1, GATA-1, and GFI1B. Mutations involving these transcription factors affect diverse aspects of platelet production and function at the genetic and molecular levels, culminating in clinical manifestations of thrombocytopenia and platelet dysfunction. This review focuses on these hematopoietic transcription factors in the pathobiology of inherited platelet dysfunction.
Collapse
|
78
|
Pecci A. Diagnosis and treatment of inherited thrombocytopenias. Clin Genet 2015; 89:141-53. [PMID: 25920516 DOI: 10.1111/cge.12603] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Revised: 04/23/2015] [Accepted: 04/23/2015] [Indexed: 12/26/2022]
Abstract
Knowledge in the field of inherited thrombocytopenias (ITs) has greatly improved over the last 15 years. Several new genes responsible for thrombocytopenia have been identified leading to the definition of novel nosographic entities and to a much better characterization of the phenotypes of these diseases. To date, ITs encompass 22 disorders caused by mutations in 24 genes and characterized by different degrees of complexity and great variability in prognosis. Making a definite diagnosis is important for setting an appropriate follow-up, choosing the best treatments and providing proper counseling. Despite the abovementioned progress, diagnosis of ITs remains difficult and these disorders are still underdiagnosed. The purpose of this review is to provide an updated guide to the diagnosis of ITs based on simple procedures. Moreover, the currently available therapeutic options for these conditions are recapitulated and discussed.
Collapse
Affiliation(s)
- A Pecci
- Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation and University of Pavia, Pavia, Italy
| |
Collapse
|
79
|
Polprasert C, Schulze I, Sekeres MA, Makishima H, Przychodzen B, Hosono N, Singh J, Padgett RA, Gu X, Phillips JG, Clemente M, Parker Y, Lindner D, Dienes B, Jankowsky E, Saunthararajah Y, Du Y, Oakley K, Nguyen N, Mukherjee S, Pabst C, Godley LA, Churpek JE, Pollyea DA, Krug U, Berdel WE, Klein HU, Dugas M, Shiraishi Y, Chiba K, Tanaka H, Miyano S, Yoshida K, Ogawa S, Müller-Tidow C, Maciejewski JP. Inherited and Somatic Defects in DDX41 in Myeloid Neoplasms. Cancer Cell 2015; 27:658-70. [PMID: 25920683 PMCID: PMC8713504 DOI: 10.1016/j.ccell.2015.03.017] [Citation(s) in RCA: 320] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 02/09/2015] [Accepted: 03/30/2015] [Indexed: 01/25/2023]
Abstract
Most cases of adult myeloid neoplasms are routinely assumed to be sporadic. Here, we describe an adult familial acute myeloid leukemia (AML) syndrome caused by germline mutations in the DEAD/H-box helicase gene DDX41. DDX41 was also found to be affected by somatic mutations in sporadic cases of myeloid neoplasms as well as in a biallelic fashion in 50% of patients with germline DDX41 mutations. Moreover, corresponding deletions on 5q35.3 present in 6% of cases led to haploinsufficient DDX41 expression. DDX41 lesions caused altered pre-mRNA splicing and RNA processing. DDX41 is exemplary of other RNA helicase genes also affected by somatic mutations, suggesting that they constitute a family of tumor suppressor genes.
Collapse
Affiliation(s)
- Chantana Polprasert
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland, OH 44195, USA; Department of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Isabell Schulze
- Department of Hematology and Oncology, University of Halle, Halle 06108, Germany; Department of Hematology and Oncology, University of Muenster, Muenster 48149, Germany
| | - Mikkael A Sekeres
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland, OH 44195, USA; Leukemia Program, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA
| | - Hideki Makishima
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland, OH 44195, USA
| | - Bartlomiej Przychodzen
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland, OH 44195, USA
| | - Naoko Hosono
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland, OH 44195, USA; First Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui 910-8507, Japan
| | - Jarnail Singh
- Department of Molecular Genetics, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Richard A Padgett
- Department of Molecular Genetics, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Xiaorong Gu
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland, OH 44195, USA
| | - James G Phillips
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland, OH 44195, USA
| | - Michael Clemente
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland, OH 44195, USA
| | - Yvonne Parker
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland, OH 44195, USA
| | - Daniel Lindner
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland, OH 44195, USA
| | - Brittney Dienes
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland, OH 44195, USA
| | - Eckhard Jankowsky
- Department of Biochemistry, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Yogen Saunthararajah
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland, OH 44195, USA
| | - Yang Du
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Kevin Oakley
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Nhu Nguyen
- Department of Pediatrics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Sudipto Mukherjee
- Leukemia Program, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA
| | - Caroline Pabst
- Department of Hematology and Oncology, University of Halle, Halle 06108, Germany
| | - Lucy A Godley
- Department of Medicine, Comprehensive Cancer Center and Center for Clinical Cancer Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Jane E Churpek
- Department of Medicine, Comprehensive Cancer Center and Center for Clinical Cancer Genetics, University of Chicago, Chicago, IL 60637, USA
| | - Daniel A Pollyea
- University of Colorado School of Medicine and University of Colorado Cancer Center, Aurora, CO 80045, USA
| | - Utz Krug
- Department of Hematology and Oncology, University of Muenster, Muenster 48149, Germany
| | - Wolfgang E Berdel
- Department of Hematology and Oncology, University of Muenster, Muenster 48149, Germany
| | - Hans-Ulrich Klein
- Institute of Medical Informatics, University of Muenster, Muenster 48149, Germany
| | - Martin Dugas
- Institute of Medical Informatics, University of Muenster, Muenster 48149, Germany
| | - Yuichi Shiraishi
- Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo 113-8654, Japan
| | - Kenichi Chiba
- Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo 113-8654, Japan
| | - Hiroko Tanaka
- Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo 113-8654, Japan
| | - Satoru Miyano
- Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo 113-8654, Japan
| | - Kenichi Yoshida
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto 606-8501, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto 606-8501, Japan
| | - Carsten Müller-Tidow
- Department of Hematology and Oncology, University of Halle, Halle 06108, Germany; Department of Hematology and Oncology, University of Muenster, Muenster 48149, Germany.
| | - Jaroslaw P Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland, OH 44195, USA; Leukemia Program, Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH 44195, USA.
| |
Collapse
|
80
|
Arbogast T, Raveau M, Chevalier C, Nalesso V, Dembele D, Jacobs H, Wendling O, Roux M, Duchon A, Herault Y. Deletion of the App-Runx1 region in mice models human partial monosomy 21. Dis Model Mech 2015; 8:623-34. [PMID: 26035870 PMCID: PMC4457029 DOI: 10.1242/dmm.017814] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Accepted: 04/10/2015] [Indexed: 02/01/2023] Open
Abstract
Partial monosomy 21 (PM21) is a rare chromosomal abnormality that is characterized by the loss of a variable segment along human chromosome 21 (Hsa21). The clinical phenotypes of this loss are heterogeneous and range from mild alterations to lethal consequences, depending on the affected region of Hsa21. The most common features include intellectual disabilities, craniofacial dysmorphology, short stature, and muscular and cardiac defects. As a complement to human genetic approaches, our team has developed new monosomic mouse models that carry deletions on Hsa21 syntenic regions in order to identify the dosage-sensitive genes that are responsible for the symptoms. We focus here on the Ms5Yah mouse model, in which a 7.7-Mb region has been deleted from the App to Runx1 genes. Ms5Yah mice display high postnatal lethality, with a few surviving individuals showing growth retardation, motor coordination deficits, and spatial learning and memory impairments. Further studies confirmed a gene dosage effect in the Ms5Yah hippocampus, and pinpointed disruptions of pathways related to cell adhesion (involving App, Cntnap5b, Lgals3bp, Mag, Mcam, Npnt, Pcdhb2, Pcdhb3, Pcdhb4, Pcdhb6, Pcdhb7, Pcdhb8, Pcdhb16 and Vwf). Our PM21 mouse model is the first to display morphological abnormalities and behavioural phenotypes similar to those found in affected humans, and it therefore demonstrates the major contribution that the App-Runx1 region has in the pathophysiology of PM21. Summary: The Del(16App-Runx1)5Yah mouse model displays morphological abnormalities and behavioural phenotypes similar to those found in humans with partial monosomy 21, and it therefore demonstrates the major contribution of the App-Runx1 region to the pathophysiology of partial monosomy 21.
Collapse
Affiliation(s)
- Thomas Arbogast
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, 1 rue Laurent Fries, Illkirch 67404, France Centre National de la Recherche Scientifique, UMR7104, Illkirch 67404, France Institut National de la Santé et de la Recherche Médicale, U964, Illkirch 67404, France Université de Strasbourg, Illkirch 67404, France
| | - Matthieu Raveau
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, 1 rue Laurent Fries, Illkirch 67404, France Centre National de la Recherche Scientifique, UMR7104, Illkirch 67404, France Institut National de la Santé et de la Recherche Médicale, U964, Illkirch 67404, France Université de Strasbourg, Illkirch 67404, France
| | - Claire Chevalier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, 1 rue Laurent Fries, Illkirch 67404, France Centre National de la Recherche Scientifique, UMR7104, Illkirch 67404, France Institut National de la Santé et de la Recherche Médicale, U964, Illkirch 67404, France Université de Strasbourg, Illkirch 67404, France
| | - Valérie Nalesso
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, 1 rue Laurent Fries, Illkirch 67404, France Centre National de la Recherche Scientifique, UMR7104, Illkirch 67404, France Institut National de la Santé et de la Recherche Médicale, U964, Illkirch 67404, France Université de Strasbourg, Illkirch 67404, France
| | - Doulaye Dembele
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, 1 rue Laurent Fries, Illkirch 67404, France Centre National de la Recherche Scientifique, UMR7104, Illkirch 67404, France Institut National de la Santé et de la Recherche Médicale, U964, Illkirch 67404, France Université de Strasbourg, Illkirch 67404, France
| | - Hugues Jacobs
- Institut Clinique de la Souris, PHENOMIN-ICS, CNRS, INSERM, Université de Strasbourg, 1 rue Laurent Fries, Illkirch 67404, France
| | - Olivia Wendling
- Institut Clinique de la Souris, PHENOMIN-ICS, CNRS, INSERM, Université de Strasbourg, 1 rue Laurent Fries, Illkirch 67404, France
| | - Michel Roux
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, 1 rue Laurent Fries, Illkirch 67404, France Centre National de la Recherche Scientifique, UMR7104, Illkirch 67404, France Institut National de la Santé et de la Recherche Médicale, U964, Illkirch 67404, France Université de Strasbourg, Illkirch 67404, France Institut Clinique de la Souris, PHENOMIN-ICS, CNRS, INSERM, Université de Strasbourg, 1 rue Laurent Fries, Illkirch 67404, France
| | - Arnaud Duchon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, 1 rue Laurent Fries, Illkirch 67404, France Centre National de la Recherche Scientifique, UMR7104, Illkirch 67404, France Institut National de la Santé et de la Recherche Médicale, U964, Illkirch 67404, France Université de Strasbourg, Illkirch 67404, France
| | - Yann Herault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Department of Translational Medicine and Neurogenetics, 1 rue Laurent Fries, Illkirch 67404, France Centre National de la Recherche Scientifique, UMR7104, Illkirch 67404, France Institut National de la Santé et de la Recherche Médicale, U964, Illkirch 67404, France Université de Strasbourg, Illkirch 67404, France Institut Clinique de la Souris, PHENOMIN-ICS, CNRS, INSERM, Université de Strasbourg, 1 rue Laurent Fries, Illkirch 67404, France
| |
Collapse
|
81
|
Schmit JM, Turner DJ, Hromas RA, Wingard JR, Brown RA, Li Y, Li MM, Slayton WB, Cogle CR. Two novel RUNX1 mutations in a patient with congenital thrombocytopenia that evolved into a high grade myelodysplastic syndrome. Leuk Res Rep 2015; 4:24-7. [PMID: 25893166 PMCID: PMC4398854 DOI: 10.1016/j.lrr.2015.03.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 03/23/2015] [Accepted: 03/25/2015] [Indexed: 11/26/2022] Open
Abstract
Here we report two new RUNX1 mutations in one patient with congenital thrombocytopenia that transformed into a high grade myelodysplastic syndrome with myelomonocytic features. The first mutation was a nucleotide base substitution from guanine to adenine within exon 8, resulting in a nonsense mutation in the DNA-binding inhibitory domain of the Runx1 protein. This nonsense mutation is suspected a de novo germline mutation since both parents are negative for the mutation. The second mutation identified was an in-frame six nucleotide base pair insertion in exon 5 of the RUNX1 gene, which is predicted to result in an insertion in the DNA-binding runt homology domain (RHD). This mutation is believed to be a somatic mutation as it was mosaic before allogeneic hematopoietic cell transplantation and disappeared after transplant. As no other genetic mutation was found using genetic screening, it is speculated that the combined effect of these two RUNX1 mutations may have exerted a stronger dominant negative effect than either RUNX1 mutation alone, thus leading to a myeloid malignancy. We report two new RUNX1 mutations in a patient with thrombocytopenia and MDS. We demonstrate that a second hit to RUNX1 results in transformed MDS. Allogeneic transplant was successfully used to treat double RUNX1 mutant MDS.
Collapse
Affiliation(s)
- Jessica M Schmit
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Daniel J Turner
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Robert A Hromas
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - John R Wingard
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Randy A Brown
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Ying Li
- Division of Hematopathology, Department of Pathology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Marilyn M Li
- Cancer Genetics Laboratory, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - William B Slayton
- Division of Hematology and Oncology, Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Christopher R Cogle
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
82
|
McGraw KL, Zhang LM, Rollison DE, Basiorka AA, Fulp W, Rawal B, Jerez A, Billingsley DL, Lin HY, Kurtin SE, Yoder S, Zhang Y, Guinta K, Mallo M, Solé F, Calasanz MJ, Cervera J, Such E, González T, Nevill TJ, Haferlach T, Smith AE, Kulasekararaj A, Mufti G, Karsan A, Maciejewski JP, Sokol L, Epling-Burnette PK, Wei S, List AF. The relationship of TP53 R72P polymorphism to disease outcome and TP53 mutation in myelodysplastic syndromes. Blood Cancer J 2015; 5:e291. [PMID: 25768405 PMCID: PMC4382654 DOI: 10.1038/bcj.2015.11] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 01/13/2015] [Indexed: 01/22/2023] Open
Abstract
Nonsynonymous TP53 exon 4 single-nucleotide polymorphism (SNP), R72P, is linked to cancer and mutagen susceptibility. R72P associations with specific cancer risk, particularly hematological malignancies, have been conflicting. Myelodysplastic syndrome (MDS) with chromosome 5q deletion is characterized by erythroid hypoplasia arising from lineage-specific p53 accumulation resulting from ribosomal insufficiency. We hypothesized that apoptotically diminished R72P C-allele may influence predisposition to del(5q) MDS. Bone marrow and blood DNA was sequenced from 705 MDS cases (333 del(5q), 372 non-del(5q)) and 157 controls. Genotype distribution did not significantly differ between del(5q) cases (12.6% CC, 38.1% CG, 49.2% GG), non-del(5q) cases (9.7% CC, 44.6% CG, 45.7% GG) and controls (7.6% CC, 37.6% CG, 54.8% GG) (P=0.13). Allele frequency did not differ between non-del(5q) and del(5q) cases (P=0.91) but trended towards increased C-allele frequency comparing non-del(5q) (P=0.08) and del(5q) (P=0.10) cases with controls. Median lenalidomide response duration increased proportionate to C-allele dosage in del(5q) patients (2.2 (CC), 1.3 (CG) and 0.89 years (GG)). Furthermore, C-allele homozygosity in del(5q) was associated with prolonged overall and progression-free survival and non-terminal interstitial deletions that excluded 5q34, whereas G-allele homozygozity was associated with inferior outcome and terminal deletions involving 5q34 (P=0.05). These findings comprise the largest MDS R72P SNP analysis.
Collapse
Affiliation(s)
- K L McGraw
- Hematology Department, H Lee Moffitt Cancer Center, Tampa, FL, USA
| | - L M Zhang
- Molecular Genomics Core Lab, H Lee Moffitt Cancer Center, Tampa, FL, USA
| | - D E Rollison
- Cancer Epidemiology, H Lee Moffitt Cancer Center, Tampa, FL, USA
| | - A A Basiorka
- 1] Hematology Department, H Lee Moffitt Cancer Center, Tampa, FL, USA [2] Cancer Biology PhD Program, University of South Florida, Tampa, FL, USA
| | - W Fulp
- Biostatistics and Bioinformatics Department, H Lee Moffitt Cancer Center, Tampa, FL, USA
| | - B Rawal
- Mayo Clinic, Biostatistics-Division of Health Sciences Research, Jacksonville, FL, USA
| | - A Jerez
- Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH, USA
| | | | - H-Y Lin
- Biostatistics and Bioinformatics Department, H Lee Moffitt Cancer Center, Tampa, FL, USA
| | | | - S Yoder
- Molecular Genomics Core Lab, H Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Y Zhang
- Biostatistics and Bioinformatics Department, H Lee Moffitt Cancer Center, Tampa, FL, USA
| | - K Guinta
- Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH, USA
| | - M Mallo
- Institut de Recerca Contra la Leucèmia Josep Carreras (IJC) Badalona, Barcelona, Spain
| | - F Solé
- Institut de Recerca Contra la Leucèmia Josep Carreras (IJC) Badalona, Barcelona, Spain
| | - M J Calasanz
- Hematology Department, Hospital Universitario La Fe, Valencia, Spain
| | - J Cervera
- Hematology Department, Hospital Universitario La Fe, Valencia, Spain
| | - E Such
- Hematology Department, Hospital Universitario La Fe, Valencia, Spain
| | - T González
- Genomics Medicine Public Foundation, Hospital Clinico Universitario, Santiago de Compostela, Spain
| | - T J Nevill
- British Columbia Cancer Agency, Vancouver, BC, Canada
| | | | - A E Smith
- King's College London, King's College Hospital, London, UK
| | | | - G Mufti
- King's College London, King's College Hospital, London, UK
| | - A Karsan
- British Columbia Cancer Agency, Vancouver, BC, Canada
| | - J P Maciejewski
- Cleveland Clinic, Taussig Cancer Institute, Cleveland, OH, USA
| | - L Sokol
- Hematology Department, H Lee Moffitt Cancer Center, Tampa, FL, USA
| | | | - S Wei
- Immunology Department, H Lee Moffitt Cancer Center, Tampa, FL, USA
| | - A F List
- Hematology Department, H Lee Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
83
|
Harada H, Harada Y. Recent advances in myelodysplastic syndromes: Molecular pathogenesis and its implications for targeted therapies. Cancer Sci 2015; 106:329-36. [PMID: 25611784 PMCID: PMC4409874 DOI: 10.1111/cas.12614] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 01/12/2015] [Accepted: 01/13/2015] [Indexed: 02/06/2023] Open
Abstract
Myelodysplastic syndromes (MDS) are defined as stem cell disorders caused by various gene abnormalities. Recent analysis using next-generation sequencing has provided great advances in identifying relationships between gene mutations and clinical phenotypes of MDS. Gene mutations affecting RNA splicing machinery, DNA methylation, histone modifications, transcription factors, signal transduction proteins and components of the cohesion complex participate in the pathogenesis and progression of MDS. Mutations in RNA splicing and DNA methylation occur early and are considered “founding mutations”, whereas others that occur later are regarded as “subclonal mutations”. RUNX1 mutations are more likely to subclonal; however, they apparently play a pivotal role in familial MDS. These genetic findings may lead to future therapies for MDS.
Collapse
Affiliation(s)
- Hironori Harada
- Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan
| | | |
Collapse
|
84
|
Zhang MY, Keel SB, Walsh T, Lee MK, Gulsuner S, Watts AC, Pritchard CC, Salipante SJ, Jeng MR, Hofmann I, Williams DA, Fleming MD, Abkowitz JL, King MC, Shimamura A. Genomic analysis of bone marrow failure and myelodysplastic syndromes reveals phenotypic and diagnostic complexity. Haematologica 2014; 100:42-8. [PMID: 25239263 DOI: 10.3324/haematol.2014.113456] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Accurate and timely diagnosis of inherited bone marrow failure and inherited myelodysplastic syndromes is essential to guide clinical management. Distinguishing inherited from acquired bone marrow failure/myelodysplastic syndrome poses a significant clinical challenge. At present, diagnostic genetic testing for inherited bone marrow failure/myelodysplastic syndrome is performed gene-by-gene, guided by clinical and laboratory evaluation. We hypothesized that standard clinically-directed genetic testing misses patients with cryptic or atypical presentations of inherited bone marrow failure/myelodysplastic syndrome. In order to screen simultaneously for mutations of all classes in bone marrow failure/myelodysplastic syndrome genes, we developed and validated a panel of 85 genes for targeted capture and multiplexed massively parallel sequencing. In patients with clinical diagnoses of Fanconi anemia, genomic analysis resolved subtype assignment, including those of patients with inconclusive complementation test results. Eight out of 71 patients with idiopathic bone marrow failure or myelodysplastic syndrome were found to harbor damaging germline mutations in GATA2, RUNX1, DKC1, or LIG4. All 8 of these patients lacked classical clinical stigmata or laboratory findings of these syndromes and only 4 had a family history suggestive of inherited disease. These results reflect the extensive genetic heterogeneity and phenotypic complexity of bone marrow failure/myelodysplastic syndrome phenotypes. This study supports the integration of broad unbiased genetic screening into the diagnostic workup of children and young adults with bone marrow failure and myelodysplastic syndromes.
Collapse
Affiliation(s)
- Michael Y Zhang
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Siobán B Keel
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA
| | - Tom Walsh
- Department of Medicine and Department of Genome Sciences, University of Washington, Seattle, WA
| | - Ming K Lee
- Department of Medicine and Department of Genome Sciences, University of Washington, Seattle, WA
| | - Suleyman Gulsuner
- Department of Medicine and Department of Genome Sciences, University of Washington, Seattle, WA
| | - Amanda C Watts
- Department of Medicine and Department of Genome Sciences, University of Washington, Seattle, WA
| | - Colin C Pritchard
- Department of Laboratory Medicine, University of Washington, Seattle, WA
| | | | - Michael R Jeng
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA
| | - Inga Hofmann
- Division of Hematology/Oncology, Boston Children's Hospital, Dana Farber Cancer Institute, and Harvard Medical School, Boston, MA
| | - David A Williams
- Division of Hematology/Oncology, Boston Children's Hospital, Dana Farber Cancer Institute, and Harvard Medical School, Boston, MA Harvard Stem Cell Institute, Boston, MA
| | | | - Janis L Abkowitz
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA
| | - Mary-Claire King
- Department of Medicine and Department of Genome Sciences, University of Washington, Seattle, WA
| | - Akiko Shimamura
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA Department of Pediatric Hematology/Oncology, Seattle Children's Hospital, WA Department of Pediatrics, University of Washington, Seattle, WA, USA
| |
Collapse
|
85
|
Yoshimi A, Toya T, Kawazu M, Ueno T, Tsukamoto A, Iizuka H, Nakagawa M, Nannya Y, Arai S, Harada H, Usuki K, Hayashi Y, Ito E, Kirito K, Nakajima H, Ichikawa M, Mano H, Kurokawa M. Recurrent CDC25C mutations drive malignant transformation in FPD/AML. Nat Commun 2014; 5:4770. [PMID: 25159113 DOI: 10.1038/ncomms5770] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 07/21/2014] [Indexed: 11/09/2022] Open
Abstract
Familial platelet disorder (FPD) with predisposition to acute myelogenous leukaemia (AML) is characterized by platelet defects with a propensity for the development of haematological malignancies. Its molecular pathogenesis is poorly understood, except for the role of germline RUNX1 mutations. Here we show that CDC25C mutations are frequently found in FPD/AML patients (53%). Mutated CDC25C disrupts the G2/M checkpoint and promotes cell cycle progression even in the presence of DNA damage, suggesting a critical role for CDC25C in malignant transformation in FPD/AML. The predicted hierarchical architecture shows that CDC25C mutations define a founding pre-leukaemic clone, followed by stepwise acquisition of subclonal mutations that contribute to leukaemia progression. In three of seven individuals with CDC25C mutations, GATA2 is the target of subsequent mutation. Thus, CDC25C is a novel gene target identified in haematological malignancies. CDC25C is also useful as a clinical biomarker that predicts progression of FPD/AML in the early stage.
Collapse
Affiliation(s)
- Akihide Yoshimi
- 1] Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan [2]
| | - Takashi Toya
- 1] Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan [2]
| | - Masahito Kawazu
- Department of Medical Genomics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Toshihide Ueno
- Department of Cellular Signaling, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Ayato Tsukamoto
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Hiromitsu Iizuka
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Masahiro Nakagawa
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Yasuhito Nannya
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Shunya Arai
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Hironori Harada
- Department of Hematology, Juntendo University School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo 113-8431, Japan
| | - Kensuke Usuki
- Department of Hematology, NTT Medical Center Tokyo, 5-9-22 Higashi-Gotanda, Shinagawa-ku, Tokyo 141-8625, Japan
| | - Yasuhide Hayashi
- Department of Hematology/Oncology, Gunma Children's Medical Center, 779 Simohakoda, Kitaakebonocho, Shibukawa-shi, Gunma 377-8577, Japan
| | - Etsuro Ito
- Department of Pediatrics, Graduate School of Medicine, Hirosaki University, 53 Honmachi, Hirosaki-shi, Aomori 036-8563, Japan
| | - Keita Kirito
- Department of Hematology and Oncology, University of Yamanashi, 1110 Simokawakita, Chuou-shi, Yamanashi 409-3898, Japan
| | - Hideaki Nakajima
- Division of Hematology, Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjyuku-ku, Tokyo 160-8582, Japan
| | - Motoshi Ichikawa
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Hiroyuki Mano
- Department of Cellular Signaling, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| | - Mineo Kurokawa
- Department of Hematology and Oncology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
| |
Collapse
|
86
|
Obata M, Tsutsumi S, Makino S, Takahashi K, Watanabe N, Yoshida T, Tamiya G, Kurachi H. Whole-exome sequencing confirmation of a novel heterozygous mutation in RUNX1 in a pregnant woman with platelet disorder. Platelets 2014; 26:364-9. [PMID: 24853048 DOI: 10.3109/09537104.2014.912750] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We describe a successful pregnancy and delivery in a patient with platelet disorder. Prophylactic platelet transfusions ensured that there were no bleeding complications during and after cesarean section. Following delivery, we performed whole exome sequencing, using next generation sequencing, to analyze the DNA samples of the patient and her family, and to identify the disease-causing mutation or variant. To identify de-novo mutations systematically, we also analyzed DNA isolated from the parents of the patient and the neonate. We successfully identified a causative novel mutation c.419 G > A (p.S140N) in RUNX1 in the patient and the neonate. Mutations of RUNX1 have been reported to be associated with familial platelet disorder and with a predisposition for myelodysplasia and/or acute myeloid leukemia. The patient and the neonate require careful long-term hematological follow-up. Identification of mutations by a through whole-exome analysis using next-generation sequencing may be useful in the determination of a long-term follow-up schedule for the patient.
Collapse
Affiliation(s)
- Miyuki Obata
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine , Yamagata , Japan and
| | | | | | | | | | | | | | | |
Collapse
|
87
|
Pecci A, Balduini CL. Lessons in platelet production from inherited thrombocytopenias. Br J Haematol 2014; 165:179-92. [PMID: 24480030 DOI: 10.1111/bjh.12752] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Our knowledge of the cellular and molecular mechanisms of platelet production has greatly expanded in recent years due to the opportunity to culture in vitro megakaryocytes and to create transgenic animals with specific genetic defects that interfere with platelet biogenesis. However, in vitro models do not reproduce the complexity of the bone marrow microenvironment where megakaryopoiesis takes place, and experience shows that what is seen in animals does not always happen in humans. So, these experimental models tell us what might happen in humans, but does not assure us that these events really occur. In contrast, inherited thrombocytopenias offer the unique opportunity to verify in humans the actual effects of abnormalities in specific molecules on platelet production. There are currently 20 genes whose defects are known to result in thrombocytopenia and, on this basis, this review tries to outline a model of megakaryopoiesis based on firm evidence. Inherited thrombocytopenias have not yet yielded all the information they can provide, because nearly half of patients have forms that do not fit with any known disorder. So, further investigation of inherited thrombocytopenias will advance not only the knowledge of human illnesses, but also our understanding of human platelet production.
Collapse
Affiliation(s)
- Alessandro Pecci
- Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation - University of Pavia, Pavia, Italy
| | | |
Collapse
|
88
|
West AH, Godley LA, Churpek JE. Familial myelodysplastic syndrome/acute leukemia syndromes: a review and utility for translational investigations. Ann N Y Acad Sci 2014; 1310:111-8. [PMID: 24467820 DOI: 10.1111/nyas.12346] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The familial myelodysplastic (MDS)/acute leukemia (AL) predisposition syndromes are inherited disorders that lead to significantly increased lifetime risks of MDS and AL development. At present, four recognized syndromes have Clinical Laboratory Improvement Amendments--certified testing for their respective germ-line mutations: telomere biology disorders due to mutation of TERC or TERT, familial acute myeloid leukemia (AML) with mutated CEBPA, familial MDS/AML with mutated GATA2, and familial platelet disorder with propensity to myeloid malignancy. These disorders are heterogeneous with regard to their causative genetic mutations, clinical presentation, and progression to MDS/AL. However, as a group, they all share the unique requirement for a high index of clinical suspicion to allow appropriate genetic counseling, genetic testing, and mutation-specific clinical management. In addition, translational investigations of individuals and families with these syndromes provide a rare opportunity to understand key pathways underlying susceptibility and progression to MDS/AL and allow the possibility of novel strategies for the prevention and treatment of both familial and sporadic forms of MDS/AL.
Collapse
Affiliation(s)
- Allison H West
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | | | | |
Collapse
|
89
|
Abstract
One of the most common hematologic malignancies in adults, myelodysplastic syndrome (MDS) is a heterogenous group of clonal disorders characterized by peripheral cytopenia(s) and normal or hypercellular bone marrow with dysplasia in ≥1 blood cell lineages. MDS frequently evolves to secondary acute myeloid leukemia with poor prognosis. Although uncommon among pediatric hematologic malignancies, both de novo and secondary MDS occur in children and may be the first presentation of an inherited bone marrow failure syndrome. Unlike its adult counterpart, pediatric MDS is more frequently associated with hypocellular bone marrow and monosomy 7. Refractory cytopenia is more typical than refractory anemia, as seen in the elderly. Its recognition and management can be quite challenging and requires the expertise of an experienced hematopathologist. In this review, we describe the epidemiology, genetics, and clinical spectrum of pediatric MDS along with its diagnostic and therapeutic challenges. We also compare and contrast pediatric and adult MDS.
Collapse
|
90
|
Kumar R, Kahr WHA. Congenital thrombocytopenia: clinical manifestations, laboratory abnormalities, and molecular defects of a heterogeneous group of conditions. Hematol Oncol Clin North Am 2013; 27:465-94. [PMID: 23714308 DOI: 10.1016/j.hoc.2013.02.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Once considered exceptionally rare, congenital thrombocytopenias are increasingly recognized as a heterogeneous group of disorders characterized by a reduction in platelet number and a bleeding tendency that may range from very mild to life threatening. Although some of these disorders affect only megakaryocytes and platelets, others involve different cell types and may result in characteristic phenotypic abnormalities. This review elaborates the clinical presentation and laboratory manifestations of common congenital thrombocytopenias in addition to exploring our understanding of the molecular basis of these disorders and therapeutic interventions available.
Collapse
Affiliation(s)
- Riten Kumar
- Division of Haematology/Oncology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
91
|
Abstract
Myelodysplasia is a diagnostic feature of myelodysplastic syndromes (MDSs) but is also found in other myeloid neoplasms. Its molecular basis has been recently elucidated by means of massive parallel sequencing studies. About 90% of MDS patients carry ≥1 oncogenic mutations, and two thirds of them are found in individuals with a normal karyotype. Driver mutant genes include those of RNA splicing (SF3B1, SRSF2, U2AF1, and ZRSR2), DNA methylation (TET2, DNMT3A, and IDH1/2), chromatin modification (ASXL1 and EZH2), transcription regulation (RUNX1), DNA repair (TP53), signal transduction (CBL, NRAS, and KRAS), and cohesin complex (STAG2). Only 4 to 6 genes are consistently mutated in ≥10% MDS patients, whereas a long tail of ∼50 genes are mutated less frequently. At presentation, most patients typically have 2 or 3 driver oncogenic mutations and hundreds of background mutations. MDS driver genes are also frequently mutated in other myeloid neoplasms. Reliable genotype/phenotype relationships include the association of the SF3B1 mutation with refractory anemia with ring sideroblasts, TET2/SRSF2 comutation with chronic myelomonocytic leukemia, and activating CSF3R mutation with chronic neutrophilic leukemia. Although both founding and subclonal driver mutations have been shown to have prognostic significance, prospective clinical trials that include the molecular characterization of the patient's genome are now needed.
Collapse
|
92
|
|
93
|
Abstract
The diagnosis of inherited thrombocytopenias is difficult, for many reasons. First, as they are all rare diseases, they are little known by clinicians, who therefore tend to suspect the most common forms. Second, making a definite diagnosis often requires complex laboratory techniques that are available in only a few centers. Finally, half of the patients have forms that have not yet been described. As a consequence, many patients with inherited thrombocytopenias are misdiagnosed with immune thrombocytopenia, and are at risk of receiving futile treatments. Misdiagnosis is particularly frequent in patients whose low platelet count is discovered in adult life, because, in these cases, even the inherited origin of thrombocytopenia may be missed. Making the correct diagnosis promptly is important, as we recently learned that some forms of inherited thrombocytopenia predispose to other illnesses, such as leukemia or kidney failure, and affected subjects therefore require close surveillance and, if necessary, prompt treatments. Moreover, medical treatment can increase platelet counts in specific disorders, and affected subjects can therefore receive drugs instead of platelet transfusions when selective surgery is required. In this review, we will discuss how to suspect, diagnose and manage inherited thrombocytopenias, with particular attention to the forms that frequently present in adults. Moreover, we describe four recently identified disorders that belong to this group of disorders that are often diagnosed in adults: MYH9-related disease, monoallelic Bernard-Soulier syndrome, ANKRD26-related thrombocytopenia, and familial platelet disorder with predisposition to acute leukemia.
Collapse
Affiliation(s)
- C L Balduini
- Department of Internal Medicine, University of Pavia-IRCCS Policlinico San Matteo Foundation, Pavia, Italy.
| | | | | |
Collapse
|
94
|
Pecci A. Pathogenesis and management of inherited thrombocytopenias: rationale for the use of thrombopoietin-receptor agonists. Int J Hematol 2013; 98:34-47. [PMID: 23636669 DOI: 10.1007/s12185-013-1351-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2013] [Revised: 04/18/2013] [Accepted: 04/19/2013] [Indexed: 12/30/2022]
Abstract
Knowledge in the field of inherited thrombocytopenias (ITs) has considerably improved over the recent years. In the last 5 years, nine new genes whose mutations are responsible for thrombocytopenia have been identified, and this also led to the recognition of several novel nosographic entities, such as thrombocytopenias deriving from mutations in CYCS, TUBB1, FLNA, ITGA2B/ITGB3, ANKRD26 and ACTN1. The identification of novel molecular alterations causing thrombocytopenia together with improvement of methodologies to study megakaryopoiesis led to considerable advances in understanding pathophysiology of ITs, thus providing the background for proposing new treatments. Thrombopoietin-receptor agonists (TPO-RAs) represent an appealing therapeutic hypothesis for ITs and have been tested in a limited number of patients. In this review, we provide an updated description of pathogenetic mechanisms of thrombocytopenia in the different forms of ITs and recapitulate the current management of these disorders. Moreover, we report the available clinical and preclinical data about the role of TPO-RAs in ITs and discuss the rationale for the use of these molecules in view of pathogenesis of the different forms of thrombocytopenia of genetic origin.
Collapse
Affiliation(s)
- Alessandro Pecci
- Department of Internal Medicine, IRCCS Policlinico San Matteo Foundation, University of Pavia, Piazzale Golgi, 27100 Pavia, Italy.
| |
Collapse
|
95
|
RUNX1/AML1 mutant collaborates with BMI1 overexpression in the development of human and murine myelodysplastic syndromes. Blood 2013; 121:3434-46. [DOI: 10.1182/blood-2012-06-434423] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Key Points
BMI1 overexpression is one of the second hit partner genes of RUNX1 mutations that contribute to the development of MDSs.
Collapse
|
96
|
Identifying familial myelodysplastic/acute leukemia predisposition syndromes through hematopoietic stem cell transplantation donors with thrombocytopenia. Blood 2013; 120:5247-9. [PMID: 23258901 DOI: 10.1182/blood-2012-09-457945] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
97
|
|
98
|
Chromosome anomalies in bone marrow as primary cause of aplastic or hypoplastic conditions and peripheral cytopenia: disorders due to secondary impairment of RUNX1 and MPL genes. Mol Cytogenet 2012; 5:39. [PMID: 23025896 PMCID: PMC3542585 DOI: 10.1186/1755-8166-5-39] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 09/06/2012] [Indexed: 11/18/2022] Open
Abstract
Background Chromosome changes in the bone marrow (BM) of patients with persistent cytopenia are often considered diagnostic for a myelodysplastic syndrome (MDS). Comprehensive cytogenetic evaluations may give evidence of the real pathogenetic role of these changes in cases with cytopenia without morphological signs of MDS. Results Chromosome anomalies were found in the BM of three patients, without any morphological evidence of MDS: 1) an acquired complex rearrangement of chromosome 21 in a boy with severe aplastic anaemia (SAA); the rearrangement caused the loss of exons 2–8 of the RUNX1 gene with subsequent hypoexpression. 2) a constitutional complex rearrangement of chromosome 21 in a girl with congenital thrombocytopenia; the rearrangement led to RUNX1 disruption and hypoexpression. 3) an acquired paracentric inversion of chromosome 1, in which two regions at the breakpoints were shown to be lost, in a boy with aplastic anaemia; the MPL gene, localized in chromosome 1 short arms was not mutated neither disrupted, but its expression was severely reduced: we postulate that the aplastic anaemia was due to position effects acting both in cis and in trans, and causing Congenital Amegakaryocytic Thrombocytopenia (CAMT). Conclusions A clonal anomaly in BM does not imply per se a diagnosis of MDS: a subgroup of BM hypoplastic disorders is directly due to chromosome structural anomalies with effects on specific genes, as was the case of RUNX1 and MPL in the patients here reported with diagnosis of SAA, thrombocytopenia, and CAMT. The anomaly may be either acquired or constitutional, and it may act by deletion/disruption of the gene, or by position effects. Full cytogenetic investigations, including a-CGH, should always be part of the diagnostic evaluation of patients with BM aplasia/hypoplasia and peripheral cytopenias.
Collapse
|
99
|
Balduini CL, Pecci A, Noris P. Inherited thrombocytopenias: the evolving spectrum. Hamostaseologie 2012; 32:259-70. [PMID: 22972471 DOI: 10.5482/ha12050001] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Accepted: 08/28/2012] [Indexed: 12/23/2022] Open
Abstract
The chapter of inherited thrombocytopenias has expanded greatly over the last decade and many "new" forms deriving from mutations in "new" genes have been identified. Nevertheless, nearly half of patients remain without a definite diagnosis because their illnesses have not yet been described. The diagnostic approach to these diseases can still take advantage of the algorithm proposed by the Italian Platelet Study Group in 2003, although an update is required to include the recently described disorders. So far, transfusions of platelet concentrates have represented the main tool for preventing or treating bleedings, while haematopoietic stem cell transplantation has been reserved for patients with very severe forms. However, recent disclosure that an oral thrombopoietin mimetic is effective in increasing platelet count in patients with MYH9-related thrombocytopenia opened new therapeutic perspectives. This review summarizes the general aspects of inherited thrombocytopenias and describes in more detail MYH9-related diseases (encompassing four thrombocytopenias previously recognized as separate diseases) and the recently described ANKRD26-related thrombocytopenia, which are among the most frequent forms of inherited thrombocytopenia.
Collapse
Affiliation(s)
- C L Balduini
- Department of Internal Medicine, University of Pavia – IRCCS Policlinico San Matteo Foundation, Pavia, Italy.
| | | | | |
Collapse
|
100
|
Genetics of familial forms of thrombocytopenia. Hum Genet 2012; 131:1821-32. [DOI: 10.1007/s00439-012-1215-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 08/01/2012] [Indexed: 12/21/2022]
|