1
|
Ramanan R, Verstraete A, Van Laer C, Freson K. Implementation and clinical utility of multigene panels for bleeding, platelet, and thrombotic disorders. J Thromb Haemost 2025:S1538-7836(25)00277-6. [PMID: 40345666 DOI: 10.1016/j.jtha.2025.04.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/21/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025]
Abstract
High-throughput sequencing, with its capacity to simultaneously sequence large volumes of genomic data, has evolved from a research-focused technology to a clinical tool. This review outlines key steps in the development of a clinical hemostasis and thrombosis genetics service leveraging a multigene panel. We discuss its value across inherited bleeding, platelet, and thrombotic disorders (BPTDs) in the context of published studies utilizing multigene panels for these conditions. Benefits of sequencing include establishing a diagnosis through the simultaneous assessment of multiple candidate genes, exclusion of genocopies, and predictions of phenotype to deliver targeted therapy. The presence of concomitant variants may modify phenotype; however, predictions on disease course from oligogenic modifiers are not yet used to guide patient care or counseling. Limitations in the widespread roll-out of multigene panels for clinical diagnosis of BPTDs exist. These challenges relate to detection of structural variants, variable diagnostic hit rates, and management of incidental findings. Variants of uncertain significance also frustrate diagnostic yield. Family segregation studies, in vitro characterization, and protein modeling aid interpretation of variant pathogenicity. Although multigene panels offer substantial opportunities to improve BPTD diagnostics, their implementation should be guided by appropriate expertise and in conjunction with clinical research to ensure safe and ethical care.
Collapse
Affiliation(s)
- Radha Ramanan
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria, Australia; Department of Human Molecular Pathology, Alfred Hospital, Melbourne, Victoria, Australia.
| | - Andreas Verstraete
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Department of Cardiovascular Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Christine Van Laer
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium; Clinical Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Kathleen Freson
- Centre for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
2
|
Zheng L, Wu Z, Yada N, Liu S, Lin C, Bignotti A, Zhao X, Zheng XL. Modeling ANKRD26 5'-UTR mutation-related thrombocytopenia. Dis Model Mech 2025; 18:dmm052222. [PMID: 40170493 PMCID: PMC12067082 DOI: 10.1242/dmm.052222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/26/2025] [Indexed: 04/03/2025] Open
Abstract
Mutations in the 5'-untranslated region (5'-UTR) of ankyrin repeat domain-containing protein 26 (ANKRD26) are associated with hereditary thrombocytopenia 2 (THC2). However, the causative role of these mutations and the mechanisms underlying THC2 are not fully established. Here, we report, for the first time, that zebrafish carrying a deletion of two nucleotides (Δ2) in the 5'-UTR of ankrd26 recapitulate some of the key laboratory features of THC2. ankrd26ku6 (homozygous for the Δ2 deletion in the 5'-UTR) fish larvae exhibited significantly increased expression of ankrd26 mRNA and protein. Adult ankrd26ku6 fish exhibited spontaneous thrombocytopenia. Furthermore, the thrombocytes from ankrd26ku6 fish showed enhanced ability to adhere and aggregate on a collagen surface under flow. Proteomic profiling demonstrated marked upregulation of Ninjurin 1 in young thrombocytes from ankrd26ku6 fish compared with those from wild-type controls. The ankrd26ku6 fish with a homozygous nacre allele developed myelodysplastic syndrome at old age. ANKRD26 protein levels were also significantly increased in platelets and plasma from patients with immune thrombotic thrombocytopenic purpura compared with those from unaffected controls. We conclude that ANKRD26 overexpression, resulting from either hereditary or acquired mechanisms, contributes to thrombocytopenia, thrombosis and hematologic malignancies.
Collapse
Affiliation(s)
- Liang Zheng
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66126, USA
- Institute of Reproductive and Developmental Sciences, The University of Kansas Medical Center, Kansas City, KS 66126, USA
| | - Zhijian Wu
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66126, USA
- Department of Pulmonary and Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha 410010, China
| | - Noritaka Yada
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66126, USA
| | - Szumam Liu
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66126, USA
| | - Cindy Lin
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66126, USA
| | - Antonia Bignotti
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66126, USA
| | - Xinyang Zhao
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66126, USA
- Institute of Reproductive and Developmental Sciences, The University of Kansas Medical Center, Kansas City, KS 66126, USA
| | - X. Long Zheng
- Department of Pathology and Laboratory Medicine, The University of Kansas Medical Center, Kansas City, KS 66126, USA
- Institute of Reproductive and Developmental Sciences, The University of Kansas Medical Center, Kansas City, KS 66126, USA
| |
Collapse
|
3
|
Liu YC, Eldomery MK, Maciaszek JL, Klco JM. Inherited Predispositions to Myeloid Neoplasms: Pathogenesis and Clinical Implications. ANNUAL REVIEW OF PATHOLOGY 2025; 20:87-114. [PMID: 39357070 PMCID: PMC12048009 DOI: 10.1146/annurev-pathmechdis-111523-023420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Myeloid neoplasms with and without preexisting platelet disorders frequently develop in association with an underlying germline predisposition. Germline alterations affecting ANKRD26, CEBPA, DDX41, ETV6, and RUNX1 are associated with nonsyndromic predisposition to the development of myeloid neoplasms including acute myeloid leukemia and myelodysplastic syndrome. However, germline predisposition to myeloid neoplasms is also associated with a wide range of other syndromes, including SAMD9/9L associated predisposition, GATA2 deficiency, RASopathies, ribosomopathies, telomere biology disorders, Fanconi anemia, severe congenital neutropenia, Down syndrome, and others. In the fifth edition of the World Health Organization (WHO) series on the classification of tumors of hematopoietic and lymphoid tissues, myeloid neoplasms associated with germline predisposition have been recognized as a separate entity. Here, we review several disorders from this WHO entity as well as other related conditions with an emphasis on the molecular pathogenesis of disease and accompanying somatic alterations. Finally, we provide an overview of establishing the molecular diagnosis of these germline genetic conditions and general recommendations for screening and management of the associated hematologic conditions.
Collapse
Affiliation(s)
- Yen-Chun Liu
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA;
| | - Mohammad K Eldomery
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA;
| | - Jamie L Maciaszek
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA;
| | - Jeffery M Klco
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee, USA;
| |
Collapse
|
4
|
Capaci V, Zanchetta ME, Fontana G, Ammeti D, Bottega R, Faleschini M, Savoia A. Inherited Thrombocytopenia Related Genes: GPS2 Mediates the Interplay Between ANKRD26 and ETV6. Cells 2024; 14:23. [PMID: 39791724 PMCID: PMC11720448 DOI: 10.3390/cells14010023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/17/2024] [Accepted: 12/24/2024] [Indexed: 01/30/2025] Open
Abstract
Mutations in the genes ANKRD26, RUNX1, and ETV6 cause three clinically overlapping thrombocytopenias characterized by a predisposition to hematological neoplasms. The ANKRD26 gene, which encodes a protein involved in protein-protein interactions, is downregulated by RUNX1 during megakaryopoiesis. Mutations in 5'UTR of ANKRD26, leading to ANKRD26-RT, disrupt this regulation, resulting in the persistent expression of ANKRD26, which leads to impaired platelet biogenesis and an increased risk of leukemia. Although ANKRD26 and ETV6 exhibit inverse expression during megakaryopoiesis, ETV6 does not regulate the ANKRD26 expression. Hypothesizing an interplay between ETV6 and ANKRD26 through in vitro studies, we explored the interactions between the two proteins. In this study, we found that ANKRD26 interacts with ETV6 and retains it in the cytoplasm, phenocopying ETV6-RT-related mutants. We found that GPS2, a component of the NCoR complex, binds both ANKRD26 and ETV6, mediating this interaction. Furthermore, ANKRD26 overexpression deregulates ETV6 transcriptional repression, supporting a common pathogenic mechanism underlying ANKRD26-RT, FPD/AML, and ETV6-RT. Our results unveil a novel ANKRD26-ETV6-GPS2 axis, providing new insights to investigate the molecular mechanism underlying thrombocytopenias with a predisposition to myeloid neoplasms that need to be further characterized.
Collapse
Affiliation(s)
- Valeria Capaci
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy; (M.E.Z.); (G.F.); (D.A.); (R.B.); (M.F.)
| | - Melania Eva Zanchetta
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy; (M.E.Z.); (G.F.); (D.A.); (R.B.); (M.F.)
| | - Giorgia Fontana
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy; (M.E.Z.); (G.F.); (D.A.); (R.B.); (M.F.)
| | - Daniele Ammeti
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy; (M.E.Z.); (G.F.); (D.A.); (R.B.); (M.F.)
| | - Roberta Bottega
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy; (M.E.Z.); (G.F.); (D.A.); (R.B.); (M.F.)
| | - Michela Faleschini
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy; (M.E.Z.); (G.F.); (D.A.); (R.B.); (M.F.)
| | - Anna Savoia
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy
| |
Collapse
|
5
|
Trizuljak J, Likavcová P, Staňo Kozubík K, Vrzalová Z, Hynšt J, Deissová T, Štika J, Radová L, Prudková M, Vaculová J, Blaháková I, Smejkal P, Kamelander J, Pospíšilová Š, Doubek M. Impact of thrombocytopenia-associated c.-118C>T and c.-140C>G ANKRD26 5'UTR variants in three-generational pedigree. Platelets 2024; 35:2388103. [PMID: 39212265 DOI: 10.1080/09537104.2024.2388103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/02/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024]
Abstract
Inherited thrombocytopenias (ITs) encompass a group of rare disorders characterized by diminished platelet count. Recent advancements have unveiled various forms of IT, with inherited thrombocytopenia 2 (THC2) emerging as a prevalent subtype associated with germline variants in the critical 5' untranslated region of the ANKRD26 gene. This region is crucial in regulating the gene expression of ANKRD26, particularly in megakaryocytes. THC2 is an autosomal dominant disorder presenting as mild-to-moderate thrombocytopenia with minimal symptoms, with an increased risk of myeloproliferative malignancies. In our study of a family with suspected IT, three affected individuals harbored the c.-118C>T ANKRD26 variant, while four healthy members carried the c.-140C>G ANKRD26 variant. We performed a functional analysis by studying platelet-specific ANKRD26 gene expression levels using quantitative real-time polymerase-chain reaction. Functional analysis of the c.-118C>T variant showed a significant increase in ANKRD26 expression in affected individuals, supporting its pathogenicity. On the contrary, carriers of the c.-140C>G variant exhibited normal platelet counts and no significant elevation in the ANKRD26 expression, indicating the likely benign nature of this variant. Our findings provide evidence confirming the pathogenicity of the c.-118C>T ANKRD26 variant in THC2 and suggest the likely benign nature of the c.-140C>G variant.
Collapse
Affiliation(s)
- Jakub Trizuljak
- Institute of Medical Genetics and Genomics, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine - Haematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University and ERN EuroBloodNet Centre, Brno, Czech Republic
- Centre of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Paulína Likavcová
- Institute of Medical Genetics and Genomics, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Kateřina Staňo Kozubík
- Institute of Medical Genetics and Genomics, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine - Haematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University and ERN EuroBloodNet Centre, Brno, Czech Republic
- Centre of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Zuzana Vrzalová
- Department of Internal Medicine - Haematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University and ERN EuroBloodNet Centre, Brno, Czech Republic
- Centre of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Jakub Hynšt
- Centre of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Tereza Deissová
- Centre of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Jiří Štika
- Institute of Medical Genetics and Genomics, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Centre of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Lenka Radová
- Centre of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Marie Prudková
- Department of Internal Medicine - Haematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University and ERN EuroBloodNet Centre, Brno, Czech Republic
- Department of Clinical Haematology, University Hospital Brno, Masaryk University, Brno, Czech Republic
- Department of Laboratory Methods, Faculty of Medicine, Masaryk University, Brno, Czech Republicand
| | - Jana Vaculová
- Department of Clinical Haematology and Haematooncology, Hospital Havířov, Havířov, Czech Republic
| | - Ivona Blaháková
- Department of Internal Medicine - Haematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University and ERN EuroBloodNet Centre, Brno, Czech Republic
- Centre of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Petr Smejkal
- Department of Clinical Haematology, University Hospital Brno, Masaryk University, Brno, Czech Republic
- Department of Laboratory Methods, Faculty of Medicine, Masaryk University, Brno, Czech Republicand
| | - Jan Kamelander
- Department of Clinical Haematology, University Hospital Brno, Masaryk University, Brno, Czech Republic
- Department of Laboratory Methods, Faculty of Medicine, Masaryk University, Brno, Czech Republicand
| | - Šárka Pospíšilová
- Institute of Medical Genetics and Genomics, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine - Haematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University and ERN EuroBloodNet Centre, Brno, Czech Republic
- Centre of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Michael Doubek
- Institute of Medical Genetics and Genomics, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine - Haematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University and ERN EuroBloodNet Centre, Brno, Czech Republic
- Centre of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| |
Collapse
|
6
|
Sapozhnikov DM, Szyf M. Genetic confounds of transgenerational epigenetic inheritance in mice. Epigenetics 2024; 19:2318519. [PMID: 38369744 PMCID: PMC10878023 DOI: 10.1080/15592294.2024.2318519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Accepted: 02/07/2024] [Indexed: 02/20/2024] Open
Abstract
Transgenerational epigenetic inheritance in mammals remains a controversial phenomenon. A recent study by Takahashi et al. provides evidence for this mode of inheritance in mice by using a CRISPR/Cas9-based epigenetic editing technique to modify DNA methylation levels at specific promoters and then demonstrating the inheritance of the gain in methylation in offspring. In this technical commentary, we argue that the method used in the original study inherently amplifies the likelihood of genetic changes that thereafter lead to the heritability of epigenetic changes. We provide evidence that genetic changes from multiple sources do indeed occur in these experiments and explore several avenues by which these changes could be causal to the apparent inheritance of epigenetic changes. We conclude a genetic basis of inheritance cannot be ruled out and thus transgenerational epigenetic inheritance has not been adequately established by the original study.
Collapse
Affiliation(s)
- Daniel M. Sapozhnikov
- Department of Pharmacology and Therapeutics, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Moshe Szyf
- Department of Pharmacology and Therapeutics, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
7
|
Sato D, Kirikae H, Nakano T, Katayama S, Yaoita H, Takayama J, Tamiya G, Kure S, Kikuchi A, Sasahara Y. Comprehensive genetic analysis for identification of monogenic disorders and selection of appropriate treatments in pediatric patients with persistent thrombocytopenia. Pediatr Hematol Oncol 2024; 41:541-556. [PMID: 39318204 DOI: 10.1080/08880018.2024.2395358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/27/2024] [Accepted: 08/18/2024] [Indexed: 09/26/2024]
Abstract
Persistent thrombocytopenia is caused by various diseases, including immune thrombocytopenia, inherited thrombocytopenia, and inherited bone marrow failure syndromes. Considering the large number of genes responsible for inherited disorders, comprehensive genetic analysis is required to diagnose monogenic disorders. In this study, we enrolled 53 pediatric patients with persistent thrombocytopenia exhibiting visually small or normal-sized platelets. We performed whole-exome sequencing, including 56 genes responsible for inherited thrombocytopenia, and evaluated clinical parameters according to disease type. Among 53 patients, 12 patients (22.6%) were diagnosed with monogenic disorders. Nine patients had a family history of thrombocytopenia. Pathogenic or novel variants of genes responsible for inherited thrombocytopenia were identified in three and six patients, respectively. The variants in genes for inherited thrombocytopenia with large or giant platelets were unexpectedly identified in six patients. Pathogenic variants in genes for inherited bone marrow failure syndromes with systemic features were identified in three patients with atypical symptoms. Since the definitive diagnostic methods for immune thrombocytopenia are limited, and a substantial number of patients with inherited thrombocytopenia are at a high risk of developing malignancies, comprehensive genetic analysis is indispensable for selecting appropriate therapies, avoidance of unnecessary treatments for immune thrombocytopenia, and long-term follow-up of patients with inherited thrombocytopenia.
Collapse
Affiliation(s)
- Daichi Sato
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Hinako Kirikae
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Tomohiro Nakano
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Saori Katayama
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Hisao Yaoita
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Jun Takayama
- Department of Rare Disease Genomics, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Department of AI and Innovative Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Statistical Genetics Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
| | - Gen Tamiya
- Department of Rare Disease Genomics, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Department of AI and Innovative Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Statistical Genetics Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
| | - Shigeo Kure
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Department of Rare Disease Genomics, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Miyagi Children's Hospital, Miyagi, Japan
| | - Atsuo Kikuchi
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Department of Rare Disease Genomics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Yoji Sasahara
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| |
Collapse
|
8
|
Zerella JR, Homan CC, Arts P, Lin X, Spinelli SJ, Venugopal P, Babic M, Brautigan PJ, Truong L, Arriola-Martinez L, Moore S, Hollins R, Parker WT, Nguyen H, Kassahn KS, Branford S, Feurstein S, Larcher L, Sicre de Fontbrune F, Demirdas S, de Munnik S, Antoine-Poirel H, Brichard B, Mansour S, Gordon K, Wlodarski MW, Koppayi A, Dobbins S, Mutsaers PGNJ, Nichols KE, Oak N, DeMille D, Mao R, Crawford A, McCarrier J, Basel D, Flores-Daboub J, Drazer MW, Phillips K, Poplawski NK, Birdsey GM, Pirri D, Ostergaard P, Simons A, Godley LA, Ross DM, Hiwase DK, Soulier J, Brown AL, Carmichael CL, Scott HS, Hahn CN. Germ line ERG haploinsufficiency defines a new syndrome with cytopenia and hematological malignancy predisposition. Blood 2024; 144:1765-1780. [PMID: 38991192 PMCID: PMC11530364 DOI: 10.1182/blood.2024024607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/21/2024] [Accepted: 06/21/2024] [Indexed: 07/13/2024] Open
Abstract
ABSTRACT The genomics era has facilitated the discovery of new genes that predispose individuals to bone marrow failure (BMF) and hematological malignancy (HM). We report the discovery of ETS-related gene (ERG), a novel, autosomal dominant BMF/HM predisposition gene. ERG is a highly constrained transcription factor that is critical for definitive hematopoiesis, stem cell function, and platelet maintenance. ERG colocalizes with other transcription factors, including RUNX family transcription factor 1 (RUNX1) and GATA binding protein 2 (GATA2), on promoters or enhancers of genes that orchestrate hematopoiesis. We identified a rare heterozygous ERG missense variant in 3 individuals with thrombocytopenia from 1 family and 14 additional ERG variants in unrelated individuals with BMF/HM, including 2 de novo cases and 3 truncating variants. Phenotypes associated with pathogenic germ line ERG variants included cytopenias (thrombocytopenia, neutropenia, and pancytopenia) and HMs (acute myeloid leukemia, myelodysplastic syndrome, and acute lymphoblastic leukemia) with onset before 40 years. Twenty ERG variants (19 missense and 1 truncating), including 3 missense population variants, were functionally characterized. Thirteen potentially pathogenic erythroblast transformation specific (ETS) domain missense variants displayed loss-of-function (LOF) characteristics, thereby disrupting transcriptional transactivation, DNA binding, and/or nuclear localization. Selected variants overexpressed in mouse fetal liver cells failed to drive myeloid differentiation and cytokine-independent growth in culture and to promote acute erythroleukemia when transplanted into mice, concordant with these being LOF variants. Four individuals displayed somatic genetic rescue by copy neutral loss of heterozygosity. Identification of predisposing germ line ERG variants has clinical implications for patient and family diagnoses, counseling, surveillance, and treatment strategies, including selection of bone marrow donors and cell or gene therapy.
Collapse
Affiliation(s)
- Jiarna R. Zerella
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Claire C. Homan
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Peer Arts
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Xuzhu Lin
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Sam J. Spinelli
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Parvathy Venugopal
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Milena Babic
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Peter J. Brautigan
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Lynda Truong
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Luis Arriola-Martinez
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Sarah Moore
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Rachel Hollins
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Wendy T. Parker
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Hung Nguyen
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Karin S. Kassahn
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Susan Branford
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Simone Feurstein
- Department of Internal Medicine, Section of Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Lise Larcher
- Université Paris Cité, INSERM and Hôpital Saint-Louis, Assistance Publique–Hôpitaux de Paris, Paris, France
| | | | - Serwet Demirdas
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sonja de Munnik
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Benedicte Brichard
- Department of Pediatric Hematology and Oncology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Sahar Mansour
- Cardiovascular and Genomics Research Institute, St. George's University of London, London, United Kingdom
- South West Thames Regional Centre for Genomics, St. George's Universities National Health Service Foundation Trust, London, United Kingdom
| | - Kristiana Gordon
- Cardiovascular and Genomics Research Institute, St. George's University of London, London, United Kingdom
- Dermatology and Lymphovascular Medicine, St. George's Universities National Health Service Foundation Trust, London, United Kingdom
| | - Marcin W. Wlodarski
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN
| | - Ashwin Koppayi
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | - Sara Dobbins
- Cardiovascular and Genomics Research Institute, St. George's University of London, London, United Kingdom
| | - Pim G. N. J. Mutsaers
- Department of Hematology, Erasmus Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Kim E. Nichols
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Ninad Oak
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Desiree DeMille
- Associated Regional and University Pathologists Institute for Clinical and Experimental Pathology, Associated Regional and University Pathologists Laboratories, Salt Lake City, UT
| | - Rong Mao
- Associated Regional and University Pathologists Institute for Clinical and Experimental Pathology, Associated Regional and University Pathologists Laboratories, Salt Lake City, UT
- Department of Pathology, University of Utah, Salt Lake City, UT
| | | | - Julie McCarrier
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| | - Donald Basel
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| | | | - Michael W. Drazer
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL
| | - Kerry Phillips
- Adult Genetics Unit, Royal Adelaide Hospital, Adelaide, SA, Australia
| | | | - Graeme M. Birdsey
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Daniela Pirri
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Pia Ostergaard
- Cardiovascular and Genomics Research Institute, St. George's University of London, London, United Kingdom
| | - Annet Simons
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lucy A. Godley
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | - David M. Ross
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA, Australia
| | - Devendra K. Hiwase
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA, Australia
| | - Jean Soulier
- Université Paris Cité, INSERM and Hôpital Saint-Louis, Assistance Publique–Hôpitaux de Paris, Paris, France
| | - Anna L. Brown
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | - Catherine L. Carmichael
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Hamish S. Scott
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
- Australian Cancer Research Foundation Genomics Facility, Centre for Cancer Biology, Adelaide, SA, Australia
| | - Christopher N. Hahn
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| |
Collapse
|
9
|
Nagata Y. Molecular pathophysiology of germline mutations in acute myeloid leukemia. Int J Hematol 2024; 120:417-426. [PMID: 39150677 DOI: 10.1007/s12185-024-03824-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 06/30/2024] [Accepted: 07/17/2024] [Indexed: 08/17/2024]
Abstract
Germline (GL) predisposition to acute myeloid leukemia (AML) has been established as an independent disease entity in the latest World Health Organization classification. Following the American College of Medical Genetics and Genomics guidelines, GL variants were interpreted as causal if they were classified as "pathogenic." GL predisposition can be divided into three groups with different phenotypes, and play an important role in the pathogenesis of adult-onset AML. The clinical course and age of onset of myeloid neoplasms varied considerably for each gene. For example, patients with GATA2 GL variants develop AML before the age of 30 along with bone marrow failure, whereas those with DDX41 GL variants tend to develop AML after the age of 50 without any preceding hematological abnormalities or organ dysfunction. A comprehensive analysis of adult-onset myelodysplastic syndromes in transplant donors showed a 7% frequency of pathogenic GL variants, with DDX41 being the most frequent gene mutation at approximately 3.8%. Future research on GL predisposition at any age of myeloid neoplasm onset will assist in early and accurate diagnosis, development of effective treatment strategies, and selection of suitable donors for stem cell transplantation.
Collapse
Affiliation(s)
- Yasunobu Nagata
- Department of Hematology, Nippon Medical School, Sendagi 1-1-5, Bunkyo-ku, Tokyo, 113-8603, Japan.
| |
Collapse
|
10
|
Dunstan-Harrison C, Morison IM, Ledgerwood EC. Inherited thrombocytopenia associated with a variant in the FLI1 binding site in the 5' UTR of ANKRD26. Clin Genet 2024; 106:315-320. [PMID: 38757516 DOI: 10.1111/cge.14547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 05/07/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024]
Abstract
Variants in the 5' UTR of ANKRD26 are a common cause of inherited thrombocytopenia (ANKRD26-RT), and are associated with sustained ANKRD26 expression, which inhibits megakaryocyte maturation and proplatelet formation. ANKRD26 expression is controlled by the binding of a RUNX1/FLI1 complex to the 5' UTR. To date, all reported ANKRD26-RD associated variants have been within the RUNX1 binding site and a 22 base pair flanking region. Here, we report a novel variant in the 5' UTR of ANKRD26, c.-107C>T. This variant is in the FLI1 binding site, and is predicted to disrupt FLI1 binding due to loss of a hydrogen bond with FLI1. Differentiated PBMCs from affected family members showed impaired megakaryocyte maturation and proplatelet formation and sustained expression of ANKRD26, and platelets from affected family members had higher ANKRD26 expression than control platelets. The variant increased activity of the ANKRD26 promotor in a reporter assay. We also provide evidence that the previously reported c.-140C>G ANKRD26 5' UTR variant is benign and not associated with thrombocytopenia. Identification of the c.-107C>T variant extends the range of the regulatory region in the 5' UTR of ANKRD26 that is associated with ANKRD26-RT.
Collapse
Affiliation(s)
- Caitlin Dunstan-Harrison
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Ian M Morison
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Elizabeth C Ledgerwood
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
11
|
Chen CP, Chen CY, Wu FT, Pan YT, Wu PS, Wang W. Prenatal diagnosis of a de novo 10p12.1p11.23 microdeletion encompassing the WAC gene in a fetus associated with bilateral hydronephrosis and right clubfoot on prenatal ultrasound. Taiwan J Obstet Gynecol 2024; 63:545-548. [PMID: 39004484 DOI: 10.1016/j.tjog.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2024] [Indexed: 07/16/2024] Open
Abstract
OBJECTIVE We present prenatal diagnosis of de novo 10p12.1p11.23 microdeletion encompassing the WAC gene in a fetus associated with bilateral hydronephrosis on prenatal ultrasound. CASE REPORT A 40-year-old, gravida 2, para 1, woman underwent amniocentesis at 17 weeks of gestation because of advanced maternal age. Amniocentesis revealed a karyotype of 46,XY. Level II ultrasound at 22 weeks of gestation revealed bilateral hydronephrosis and right clubfoot. At 23 weeks of gestation, repeat amniocentesis revealed the result of arr [GRCh37] 10p12.1p11.23 (26,182,512-29,826,276) × 1 dn with a 3.6-Mb microdeletion of 10p12.1p11.23 encompassing the genes of MYO3A, GAD2, APBB1IP, PDSS1, ABI1, ANKRD26, YME1L1, MASTL, ACBD5, PTCHD3, RAB18, MKX, ODAD2, MPP7, WAC and BAMBI. The pregnancy was subsequently terminated, and a malformed fetus was delivered with facial dysmorphism of low-set ears, broad forehead and flat nasal bridge. Array comparative genomic hybridization (aCGH) analysis of umbilical cord confirmed a 3.6-Mb 10p12.1p11.23 microdeletion encompassing WAC. CONCLUSION Application of aCGH is useful in the pregnancy with a normal fetal karyotype and abnormal fetal ultrasound.
Collapse
Affiliation(s)
- Chih-Ping Chen
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan; Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan; School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan; Institute of Clinical and Community Health Nursing, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Obstetrics and Gynecology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Medical Laboratory Science and Biotechnology, College of Medical & Health Science, Asia University, Taichung, Taiwan.
| | - Chen-Yu Chen
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City, Taiwan; MacKay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
| | - Fang-Tzu Wu
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Yen-Ting Pan
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan
| | | | - Wayseen Wang
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| |
Collapse
|
12
|
Banaszak LG, Cabral PL, Smith-Simmer K, Hassan A, Brunner M, Fallon M, Shoger K, Lovrien L, Golner D, Zurbriggen L, Mattison R, Gahvari Z, Hall A, Nadiminti K, Reinig E, Churpek JE. Implementation of and Systems-Level Barriers to Guideline-Driven Germline Genetic Evaluation in the Care of Patients With Myelodysplastic Syndrome and Acute Myeloid Leukemia. JCO Precis Oncol 2024; 8:e2300518. [PMID: 38848520 PMCID: PMC11234342 DOI: 10.1200/po.23.00518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 02/19/2024] [Accepted: 04/02/2024] [Indexed: 06/09/2024] Open
Abstract
PURPOSE Knowledge of an inherited predisposition to myelodysplastic syndrome (MDS) and AML has important clinical implications for treatment decisions, surveillance, and care of at-risk relatives. National Comprehensive Cancer Network (NCCN) guidelines recently incorporated recommendations for germline genetic evaluation of patients with MDS/AML on the basis of personal and family history features, but the practicality of implementing these recommendations has not been studied. METHODS A hereditary hematology quality improvement (QI) committee was formed to implement these guidelines in a prospective cohort of patients diagnosed with MDS/AML. Referral for germline genetic testing was recommended for patients meeting NCCN guideline criteria. Referral patterns and genetic evaluation outcomes were compared with a historical cohort of patients with MDS/AML. Barriers to evaluation were identified. RESULTS Of the 90 patients with MDS/AML evaluated by the QI committee, 59 (66%) met criteria for germline evaluation. Implementation of the QI committee led to more referrals for germline evaluation in accordance with NCCN guidelines (31% v 14%, P = .03). However, the majority of those meeting criteria were never referred due to high medical acuity or being deceased or in hospice at the time of QI committee recommendations. Despite this, two (17%) of the 12 patients undergoing genetic testing were diagnosed with a hereditary myeloid malignancy syndrome. CONCLUSION Current NCCN guidelines resulted in two thirds of patients with MDS/AML meeting criteria for germline evaluation. A hereditary hematology-focused QI committee aided initial implementation and modestly improved NCCN guideline adherence. However, the high morbidity and mortality and prolonged inpatient stays associated with MDS/AML challenged traditional outpatient genetic counseling models. Further improvements in guideline adherence require innovating new models of genetic counseling and testing for this patient population.
Collapse
Affiliation(s)
- Lauren G. Banaszak
- Department of Medicine; University of Wisconsin-Madison; Madison, Wisconsin, USA
| | - Paloma L. Cabral
- Department of Medicine; University of Wisconsin-Madison; Madison, Wisconsin, USA
| | - Kelcy Smith-Simmer
- Oncology Genetics; University of Wisconsin Carbone Cancer Center; UWHealth; Madison, Wisconsin, USA
| | - Ayesha Hassan
- Department of Medicine; University of Wisconsin-Madison; Madison, Wisconsin, USA
| | - Matthew Brunner
- Department of Medicine; University of Wisconsin-Madison; Madison, Wisconsin, USA
| | - Michael Fallon
- Department of Medicine; University of Wisconsin-Madison; Madison, Wisconsin, USA
| | - Kyle Shoger
- Department of Medicine; University of Wisconsin-Madison; Madison, Wisconsin, USA
| | - Lauren Lovrien
- Department of Medicine; University of Wisconsin-Madison; Madison, Wisconsin, USA
| | - Danielle Golner
- Department of Medicine; University of Wisconsin-Madison; Madison, Wisconsin, USA
| | - Luke Zurbriggen
- Department of Medicine; University of Wisconsin-Madison; Madison, Wisconsin, USA
| | - Ryan Mattison
- Department of Medicine; University of Wisconsin-Madison; Madison, Wisconsin, USA
| | - Zhubin Gahvari
- Department of Medicine; University of Wisconsin-Madison; Madison, Wisconsin, USA
| | - Aric Hall
- Department of Medicine; University of Wisconsin-Madison; Madison, Wisconsin, USA
| | - Kalyan Nadiminti
- Department of Medicine; University of Wisconsin-Madison; Madison, Wisconsin, USA
| | - Erica Reinig
- Department of Pathology and Laboratory Medicine; University of Wisconsin-Madison; Madison, Wisconsin, USA
| | - Jane E. Churpek
- Department of Medicine; University of Wisconsin-Madison; Madison, Wisconsin, USA
| |
Collapse
|
13
|
Jiang YZ, Hu LY, Chen MS, Wang XJ, Tan CN, Xue PP, Yu T, He XY, Xiang LX, Xiao YN, Li XL, Ran Q, Li ZJ, Chen L. GATA binding protein 2 mediated ankyrin repeat domain containing 26 high expression in myeloid-derived cell lines. World J Stem Cells 2024; 16:538-550. [PMID: 38817334 PMCID: PMC11135246 DOI: 10.4252/wjsc.v16.i5.538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/12/2024] [Accepted: 04/12/2024] [Indexed: 05/24/2024] Open
Abstract
BACKGROUND Thrombocytopenia 2, an autosomal dominant inherited disease characterized by moderate thrombocytopenia, predisposition to myeloid malignancies and normal platelet size and function, can be caused by 5'-untranslated region (UTR) point mutations in ankyrin repeat domain containing 26 (ANKRD26). Runt related transcription factor 1 (RUNX1) and friend leukemia integration 1 (FLI1) have been identified as negative regulators of ANKRD26. However, the positive regulators of ANKRD26 are still unknown. AIM To prove the positive regulatory effect of GATA binding protein 2 (GATA2) on ANKRD26 transcription. METHODS Human induced pluripotent stem cells derived from bone marrow (hiPSC-BM) and urothelium (hiPSC-U) were used to examine the ANKRD26 expression pattern in the early stage of differentiation. Then, transcriptome sequencing of these iPSCs and three public transcription factor (TF) databases (Cistrome DB, animal TFDB and ENCODE) were used to identify potential TF candidates for ANKRD26. Furthermore, overexpression and dual-luciferase reporter experiments were used to verify the regulatory effect of the candidate TFs on ANKRD26. Moreover, using the GENT2 platform, we analyzed the relationship between ANKRD26 expression and overall survival in cancer patients. RESULTS In hiPSC-BMs and hiPSC-Us, we found that the transcription levels of ANKRD26 varied in the absence of RUNX1 and FLI1. We sequenced hiPSC-BM and hiPSC-U and identified 68 candidate TFs for ANKRD26. Together with three public TF databases, we found that GATA2 was the only candidate gene that could positively regulate ANKRD26. Using dual-luciferase reporter experiments, we showed that GATA2 directly binds to the 5'-UTR of ANKRD26 and promotes its transcription. There are two identified binding sites of GATA2 that are located 2 kb upstream of the TSS of ANKRD26. In addition, we discovered that high ANKRD26 expression is always related to a more favorable prognosis in breast and lung cancer patients. CONCLUSION We first discovered that the transcription factor GATA2 plays a positive role in ANKRD26 transcription and identified its precise binding sites at the promoter region, and we revealed the importance of ANKRD26 in many tissue-derived cancers.
Collapse
Affiliation(s)
- Yang-Zhou Jiang
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Third Military Medical University, Chongqing 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing 400037, China
| | - Lan-Yue Hu
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Third Military Medical University, Chongqing 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing 400037, China
| | - Mao-Shan Chen
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Third Military Medical University, Chongqing 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing 400037, China
| | - Xiao-Jie Wang
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Third Military Medical University, Chongqing 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing 400037, China
| | - Cheng-Ning Tan
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Third Military Medical University, Chongqing 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing 400037, China
| | - Pei-Pei Xue
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Third Military Medical University, Chongqing 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing 400037, China
| | - Teng Yu
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Third Military Medical University, Chongqing 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing 400037, China
| | - Xiao-Yan He
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Third Military Medical University, Chongqing 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing 400037, China
| | - Li-Xin Xiang
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Third Military Medical University, Chongqing 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing 400037, China
| | - Yan-Ni Xiao
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Third Military Medical University, Chongqing 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing 400037, China
| | - Xiao-Liang Li
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Third Military Medical University, Chongqing 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing 400037, China
| | - Qian Ran
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Third Military Medical University, Chongqing 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing 400037, China
| | - Zhong-Jun Li
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Third Military Medical University, Chongqing 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing 400037, China
| | - Li Chen
- Laboratory of Radiation Biology, Department of Blood Transfusion, Laboratory Medicine Center, The Second Affiliated Hospital, Third Military Medical University, Chongqing 400037, China
- Hematopoietic Acute Radiation Syndrome Medical and Pharmaceutical Basic Research Innovation Center, Ministry of Education of the People's Republic of China, Chongqing 400037, China.
| |
Collapse
|
14
|
Shiozawa Y, Fujita S, Nannya Y, Ogawa S, Nomura N, Kiguchi T, Sezaki N, Kudo H, Toyama T. First report of familial mixed phenotype acute leukemia: shared clinical characteristics, Philadelphia translocation, and germline variants. Int J Hematol 2024; 119:465-471. [PMID: 38424413 DOI: 10.1007/s12185-024-03724-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 01/17/2024] [Accepted: 01/25/2024] [Indexed: 03/02/2024]
Abstract
While our understanding of the molecular basis of mixed phenotype acute leukemia (MPAL) has progressed over the decades, our knowledge is limited and the prognosis remains poor. Investigating cases of familial leukemia can provide insights into the role of genetic and environmental factors in leukemogenesis. Although familial cases and associated mutations have been identified in some leukemias, familial occurrence of MPAL has never been reported. Here, we report the first cases of MPAL in a family. A 68-year-old woman was diagnosed with MPAL and received haploidentical stem cell transplantation from her 44-year-old son. In four years, the son himself developed MPAL. Both cases exhibited similar characteristics such as biphenotypic leukemia with B/myeloid cell antigens, Philadelphia translocation (BCR-ABL1 mutation), and response to acute lymphoblastic leukemia-type chemotherapy. These similarities suggest the presence of hereditary factors contributing to the development of MPAL. Targeted sequencing identified shared germline variants in these cases; however, in silico analyses did not strongly support their pathogenicity. Intriguingly, when the son developed MPAL, the mother did not develop donor-derived leukemia and remained in remission. Our cases provide valuable insights to guide future research on familial MPAL.
Collapse
Affiliation(s)
- Yuka Shiozawa
- Department of Hematology, Federation of National Public Service Personnel Mutual Aid Associations Tachikawa Hospital, 4-2-22 Nishiki-Cho, Tachikawa-Shi, Tokyo, 190-8531, Japan
| | - Shinya Fujita
- Department of Hematology, Federation of National Public Service Personnel Mutual Aid Associations Tachikawa Hospital, 4-2-22 Nishiki-Cho, Tachikawa-Shi, Tokyo, 190-8531, Japan.
| | - Yasuhito Nannya
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
- Division of Hematopoietic Disease Control, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
| | - Naho Nomura
- Department of Hematology, Chugoku Central Hospital of Japan Mutual Aid Association of Public School Teachers, Hiroshima, Japan
| | - Toru Kiguchi
- Saitama Medical Center, Department of Diabetes, Endocrinology and Hematology, Dokkyo Medical University, Saitama, Japan
| | - Nobuo Sezaki
- Department of Hematology, Chugoku Central Hospital of Japan Mutual Aid Association of Public School Teachers, Hiroshima, Japan
| | - Himari Kudo
- Department of Hematology, Federation of National Public Service Personnel Mutual Aid Associations Tachikawa Hospital, 4-2-22 Nishiki-Cho, Tachikawa-Shi, Tokyo, 190-8531, Japan
| | - Takaaki Toyama
- Department of Hematology, Federation of National Public Service Personnel Mutual Aid Associations Tachikawa Hospital, 4-2-22 Nishiki-Cho, Tachikawa-Shi, Tokyo, 190-8531, Japan
| |
Collapse
|
15
|
Cheng YHH, Bohaczuk SC, Stergachis AB. Functional categorization of gene regulatory variants that cause Mendelian conditions. Hum Genet 2024; 143:559-605. [PMID: 38436667 PMCID: PMC11078748 DOI: 10.1007/s00439-023-02639-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 12/30/2023] [Indexed: 03/05/2024]
Abstract
Much of our current understanding of rare human diseases is driven by coding genetic variants. However, non-coding genetic variants play a pivotal role in numerous rare human diseases, resulting in diverse functional impacts ranging from altered gene regulation, splicing, and/or transcript stability. With the increasing use of genome sequencing in clinical practice, it is paramount to have a clear framework for understanding how non-coding genetic variants cause disease. To this end, we have synthesized the literature on hundreds of non-coding genetic variants that cause rare Mendelian conditions via the disruption of gene regulatory patterns and propose a functional classification system. Specifically, we have adapted the functional classification framework used for coding variants (i.e., loss-of-function, gain-of-function, and dominant-negative) to account for features unique to non-coding gene regulatory variants. We identify that non-coding gene regulatory variants can be split into three distinct categories by functional impact: (1) non-modular loss-of-expression (LOE) variants; (2) modular loss-of-expression (mLOE) variants; and (3) gain-of-ectopic-expression (GOE) variants. Whereas LOE variants have a direct corollary with coding loss-of-function variants, mLOE and GOE variants represent disease mechanisms that are largely unique to non-coding variants. These functional classifications aim to provide a unified terminology for categorizing the functional impact of non-coding variants that disrupt gene regulatory patterns in Mendelian conditions.
Collapse
Affiliation(s)
- Y H Hank Cheng
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Stephanie C Bohaczuk
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Andrew B Stergachis
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, USA.
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.
| |
Collapse
|
16
|
Englisch AS, Hofbrucker-MacKenzie SA, Izadi-Seitz M, Kessels MM, Qualmann B. Ankrd26 is a retinoic acid-responsive plasma membrane-binding and -shaping protein critical for proper cell differentiation. Cell Rep 2024; 43:113939. [PMID: 38493476 DOI: 10.1016/j.celrep.2024.113939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 01/17/2024] [Accepted: 02/23/2024] [Indexed: 03/19/2024] Open
Abstract
Morphogens are important triggers for differentiation processes. Yet, downstream effectors that organize cell shape changes in response to morphogenic cues, such as retinoic acid, largely remain elusive. Additionally, derailed plasma membrane-derived signaling often is associated with cancer. We identify Ankrd26 as a critical player in cellular differentiation and as plasma membrane-localized protein able to self-associate and form clusters at the plasma membrane in response to retinoic acid. We show that Ankrd26 uses an N-terminal amphipathic structure for membrane binding and bending. Importantly, in an acute myeloid leukemia-associated Ankrd26 mutant, this critical structure was absent, and Ankrd26's membrane association and shaping abilities were impaired. In line with this, the mutation rendered Ankrd26 inactive in both gain-of-function and loss-of-function/rescue studies addressing retinoic acid/brain-derived neurotrophic factor (BDNF)-induced neuroblastoma differentiation. Our results highlight the importance and molecular details of Ankrd26-mediated organizational platforms for cellular differentiation at the plasma membrane and how impairment of these platforms leads to cancer-associated pathomechanisms involving these Ankrd26 properties.
Collapse
Affiliation(s)
- Anna Sofie Englisch
- Institute of Biochemistry I, Jena University Hospital - Friedrich Schiller University Jena, Nonnenplan 2-4, 07743 Jena, Germany
| | - Sarah Ann Hofbrucker-MacKenzie
- Institute of Biochemistry I, Jena University Hospital - Friedrich Schiller University Jena, Nonnenplan 2-4, 07743 Jena, Germany
| | - Maryam Izadi-Seitz
- Institute of Biochemistry I, Jena University Hospital - Friedrich Schiller University Jena, Nonnenplan 2-4, 07743 Jena, Germany
| | - Michael Manfred Kessels
- Institute of Biochemistry I, Jena University Hospital - Friedrich Schiller University Jena, Nonnenplan 2-4, 07743 Jena, Germany.
| | - Britta Qualmann
- Institute of Biochemistry I, Jena University Hospital - Friedrich Schiller University Jena, Nonnenplan 2-4, 07743 Jena, Germany.
| |
Collapse
|
17
|
Arai H, Matsui H, Chi S, Utsu Y, Masuda S, Aotsuka N, Minami Y. Germline Variants and Characteristic Features of Hereditary Hematological Malignancy Syndrome. Int J Mol Sci 2024; 25:652. [PMID: 38203823 PMCID: PMC10779750 DOI: 10.3390/ijms25010652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/25/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Due to the proliferation of genetic testing, pathogenic germline variants predisposing to hereditary hematological malignancy syndrome (HHMS) have been identified in an increasing number of genes. Consequently, the field of HHMS is gaining recognition among clinicians and scientists worldwide. Patients with germline genetic abnormalities often have poor outcomes and are candidates for allogeneic hematopoietic stem cell transplantation (HSCT). However, HSCT using blood from a related donor should be carefully considered because of the risk that the patient may inherit a pathogenic variant. At present, we now face the challenge of incorporating these advances into clinical practice for patients with myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML) and optimizing the management and surveillance of patients and asymptomatic carriers, with the limitation that evidence-based guidelines are often inadequate. The 2016 revision of the WHO classification added a new section on myeloid malignant neoplasms, including MDS and AML with germline predisposition. The main syndromes can be classified into three groups. Those without pre-existing disease or organ dysfunction; DDX41, TP53, CEBPA, those with pre-existing platelet disorders; ANKRD26, ETV6, RUNX1, and those with other organ dysfunctions; SAMD9/SAMD9L, GATA2, and inherited bone marrow failure syndromes. In this review, we will outline the role of the genes involved in HHMS in order to clarify our understanding of HHMS.
Collapse
Affiliation(s)
- Hironori Arai
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan; (H.A.); (S.C.)
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Iidacho, Narita 286-0041, Japan; (Y.U.); (S.M.); (N.A.)
| | - Hirotaka Matsui
- Department of Laboratory Medicine, National Cancer Center Hospital, Tsukiji, Chuoku 104-0045, Japan;
- Department of Medical Oncology and Translational Research, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8665, Japan
| | - SungGi Chi
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan; (H.A.); (S.C.)
| | - Yoshikazu Utsu
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Iidacho, Narita 286-0041, Japan; (Y.U.); (S.M.); (N.A.)
| | - Shinichi Masuda
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Iidacho, Narita 286-0041, Japan; (Y.U.); (S.M.); (N.A.)
| | - Nobuyuki Aotsuka
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Iidacho, Narita 286-0041, Japan; (Y.U.); (S.M.); (N.A.)
| | - Yosuke Minami
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan; (H.A.); (S.C.)
| |
Collapse
|
18
|
Manohar S, Gofin Y, Streff H, Vossaert L, Camacho P, Murali CN. A familial deletion of 10p12.1 associated with thrombocytopenia. Am J Med Genet A 2024; 194:77-81. [PMID: 37746810 DOI: 10.1002/ajmg.a.63403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 08/23/2023] [Accepted: 09/01/2023] [Indexed: 09/26/2023]
Abstract
Thrombocytopenia can be inherited or acquired from a variety of causes. While hereditary causes of thrombocytopenia are rare, several genes have been associated with the condition. In this report, we describe an 18-year-old man and his mother, both of whom have congenital thrombocytopenia. Exome sequencing in the man revealed a 1006 kb maternally inherited deletion in the 10p12.1 region (arr[GRCh37] 10p12.1(27378928_28384564)x1) of uncertain clinical significance. This deletion in the THC2 locus includes genes ANKRD26, known to be involved in normal megakaryocyte differentiation, and MASTL, which some studies suggest is linked to autosomal dominant thrombocytopenia. In the family presented here, the deletion segregated with the congenital thrombocytopenia phenotype, suggesting that haploinsufficiency of one or both genes may be the cause. To our knowledge, this is the first report of a deletion of the THC2 locus associated with thrombocytopenia. Future functional studies of deletions of the THC2 locus may elucidate the mechanism for this phenotype observed clinically.
Collapse
Affiliation(s)
- Sujal Manohar
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Yoel Gofin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Haley Streff
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Liesbeth Vossaert
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Pamela Camacho
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children's Cancer and Hematology Centers, Houston, Texas, USA
| | - Chaya N Murali
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
19
|
Pang C, Wu X, Nikuze L, Wei H. Analysis of clinical characteristics and treatment efficacy in two pediatric cases of ANKRD26-related thrombocytopenia. Platelets 2023; 34:2262607. [PMID: 37852929 DOI: 10.1080/09537104.2023.2262607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/19/2023] [Indexed: 10/20/2023]
Abstract
ANKRD26-related thrombocytopenia (ANKRD26-RT or THC2, MIM 188 000), an autosomal dominant thrombocytopenia, is unresponsive to immunosuppressive therapy and susceptible to hematological malignancies. A large number of pediatric patients are diagnosed with immune thrombocytopenia (ITP) every year; however, thrombocytopenia of genetic origin is often missed. Extensive characterization of ANKRD26-RT will help prevent missed diagnosis and misdiagnosis. Furthermore, identification of ANKRD26-RT will help in the formulation of an accurate diagnosis and a treatment plan. In our study, we report cases of two Chinese pediatric patients with ANKRD26-RT and analyze their clinical characteristics, gene mutations, and treatment modalities. Both patients were 1-year-old and presented with mild bleeding (World Health Organization(WHO) score grade 1), different degrees of platelet reduction, normal mean platelet volume, and megakaryocyte maturation impairment not obvious. Genetic tests revealed that both patients had ANKRD26 gene mutations.Patient 1 had a mutation c.-140C>G of the 5' untranslated region (UTR), and patient 2 had a mutation of c.-127A>T of 5'UTR. Both patients were treated with eltrombopag, and the treatment was no response, with no adverse reactions.
Collapse
Affiliation(s)
- Congfei Pang
- Department of Pediatrics, The Sixth Affiliated Hospital of Guangxi Medical University: The First People's Hospital of Yulin, Yulin, Guangxi, P.R. China
| | - Xiaomei Wu
- Department of Pediatrics, Red Cross Hospital of Yulin city, Yulin, Guangxi, P.R. China
| | | | - Hongying Wei
- Department of Pediatrics, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, P.R. China
| |
Collapse
|
20
|
Obiorah IE, Upadhyaya KD, Calvo KR. Germline Predisposition to Myeloid Neoplasms: Diagnostic Concepts and Classifications. Clin Lab Med 2023; 43:615-638. [PMID: 37865507 DOI: 10.1016/j.cll.2023.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2023]
Abstract
Molecular and sequencing advances have led to substantial breakthroughs in the discovery of new genes and inherited mutations associated with increased risk of developing myeloid malignancies. Many of the same germline mutated genes are also drivers of malignancy in sporadic cancer. Recognition of myeloid malignancy associated with germline mutations is essential for proper therapy, disease surveillance, informing related donor selection for hematopoietic stem cell transplantation, and genetic counseling of the patient and affected family members. Some germline mutations are associated with syndromic features that precede the development of malignancy; however, penetrance may be highly variable leading to masking of the syndromic phenotype and/or inherited etiology.
Collapse
Affiliation(s)
- Ifeyinwa E Obiorah
- Department of Pathology, Division of Hematopathology, University of Virginia Health, Charlottesville, VA, USA
| | - Kalpana D Upadhyaya
- Hematology Section, Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Katherine R Calvo
- Hematology Section, Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, USA; Myeloid Malignancies Program, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
21
|
Bonjoch L, Castellví-Bel S, Ruiz-Ponte C. Reply. Gastroenterology 2023; 165:1577-1578. [PMID: 37741422 DOI: 10.1053/j.gastro.2023.09.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 09/19/2023] [Indexed: 09/25/2023]
Affiliation(s)
- Laia Bonjoch
- Gastroenterology Department, Hospital Clínic, Institut d'Investigacions Biomediques August Pi i Sunyer, Centro de Investigacion Biomedica en Red de Enfermedades Hepaticas y Digestivas, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Sergi Castellví-Bel
- Gastroenterology Department, Hospital Clínic, Institut d'Investigacions Biomediques August Pi i Sunyer, Centro de Investigacion Biomedica en Red de Enfermedades Hepaticas y Digestivas, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Clara Ruiz-Ponte
- Fundación Pública Galega de Medicina Xenómica, Instituto de Investigacion Sanitaria de Santiago, Grupo de Medicina Xenomica-Universidad de Santiago de Compostela, Santiago de Compostela, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras, Madrid, Spain
| |
Collapse
|
22
|
Kanagal-Shamanna R, Schafernak KT, Calvo KR. Diagnostic work-up of hematological malignancies with underlying germline predisposition disorders (GPD). Semin Diagn Pathol 2023; 40:443-456. [PMID: 37977953 DOI: 10.1053/j.semdp.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/07/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023]
Abstract
Hematological malignancies with underlying germline predisposition disorders have been recognized by the World Health Organization 5th edition and International Consensus Classification (ICC) classification systems. The list of genes and the associated phenotypes are expanding and involve both pediatric and adult populations. While the clinical presentation and underlying molecular pathogenesis are relatively well described, the knowledge regarding the bone marrow morphologic features, the landscape of somatic aberrations associated with progression to hematological malignancies is limited. These pose challenges in the diagnosis of low-grade myelodysplastic syndrome (MDS) to hematopathologists which carries direct implication for various aspects of clinical management of the patient, donor selection for transplantation, and family members. Here in, we provide a focused review on the diagnostic work-up of hematological malignancies with underlying germline predisposition disorders with emphasis on the spectrum of hematological malignancies associated with each entity, and characteristic bone marrow morphologic, somatic cytogenetic and molecular alterations at the time of diagnosis of hematological malignancies. We also review the key clinical, morphologic, and molecular features, that should initiate screening for these entities.
Collapse
Affiliation(s)
- Rashmi Kanagal-Shamanna
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Kristian T Schafernak
- Division of Pathology and Laboratory Medicine, Phoenix Children's Hospital, Phoenix, AZ, United States
| | - Katherine R Calvo
- Hematology Section, Department of Laboratory Medicine, National Institutes of Health Clinical Center, Bethesda, MD, United States.
| |
Collapse
|
23
|
Kemmler CL, Smolikova J, Moran HR, Mannion BJ, Knapp D, Lim F, Czarkwiani A, Hermosilla Aguayo V, Rapp V, Fitch OE, Bötschi S, Selleri L, Farley E, Braasch I, Yun M, Visel A, Osterwalder M, Mosimann C, Kozmik Z, Burger A. Conserved enhancers control notochord expression of vertebrate Brachyury. Nat Commun 2023; 14:6594. [PMID: 37852970 PMCID: PMC10584899 DOI: 10.1038/s41467-023-42151-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/29/2023] [Indexed: 10/20/2023] Open
Abstract
The cell type-specific expression of key transcription factors is central to development and disease. Brachyury/T/TBXT is a major transcription factor for gastrulation, tailbud patterning, and notochord formation; however, how its expression is controlled in the mammalian notochord has remained elusive. Here, we identify the complement of notochord-specific enhancers in the mammalian Brachyury/T/TBXT gene. Using transgenic assays in zebrafish, axolotl, and mouse, we discover three conserved Brachyury-controlling notochord enhancers, T3, C, and I, in human, mouse, and marsupial genomes. Acting as Brachyury-responsive, auto-regulatory shadow enhancers, in cis deletion of all three enhancers in mouse abolishes Brachyury/T/Tbxt expression selectively in the notochord, causing specific trunk and neural tube defects without gastrulation or tailbud defects. The three Brachyury-driving notochord enhancers are conserved beyond mammals in the brachyury/tbxtb loci of fishes, dating their origin to the last common ancestor of jawed vertebrates. Our data define the vertebrate enhancers for Brachyury/T/TBXTB notochord expression through an auto-regulatory mechanism that conveys robustness and adaptability as ancient basis for axis development.
Collapse
Affiliation(s)
- Cassie L Kemmler
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jana Smolikova
- Institute of Molecular Genetics of the ASCR, v. v. i., Prague, Czech Republic
| | - Hannah R Moran
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Brandon J Mannion
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Comparative Biochemistry Program, University of California, Berkeley, CA, 94720, USA
| | - Dunja Knapp
- Technische Universität Dresden, CRTD Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Fabian Lim
- Department of Medicine, Health Sciences, University of California San Diego, La Jolla, CA, USA
- Department of Molecular Biology, Biological Sciences, University of California San Diego, La Jolla, CA, USA
- Biological Sciences Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Anna Czarkwiani
- Technische Universität Dresden, CRTD Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Viviana Hermosilla Aguayo
- Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA
| | - Vincent Rapp
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Olivia E Fitch
- Department of Integrative Biology and Ecology, Evolution and Behavior Program, Michigan State University, East Lansing, MI, USA
| | - Seraina Bötschi
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Licia Selleri
- Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA
| | - Emma Farley
- Department of Medicine, Health Sciences, University of California San Diego, La Jolla, CA, USA
- Department of Molecular Biology, Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Ingo Braasch
- Department of Integrative Biology and Ecology, Evolution and Behavior Program, Michigan State University, East Lansing, MI, USA
| | - Maximina Yun
- Technische Universität Dresden, CRTD Center for Regenerative Therapies Dresden, Dresden, Germany
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden, Germany
| | - Axel Visel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- US Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- School of Natural Sciences, University of California Merced, Merced, CA, USA
| | - Marco Osterwalder
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Cardiology, Bern University Hospital, Bern, Switzerland
| | - Christian Mosimann
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Zbynek Kozmik
- Institute of Molecular Genetics of the ASCR, v. v. i., Prague, Czech Republic.
| | - Alexa Burger
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
24
|
Li Y, Wang S, Xiao H, Lu F, Zhang B, Zhou T. Evaluation and validation of the prognostic value of platelet indices in patients with leukemia. Clin Exp Med 2023; 23:1835-1844. [PMID: 36622510 DOI: 10.1007/s10238-022-00985-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 12/29/2022] [Indexed: 01/10/2023]
Abstract
Platelets (PLTs) are believed to play a role in the process by which tumors can accelerate their growth rate, as well as offer the physical and mechanical support necessary to evade the immunological system and metastasis. There is, however, no literature available if PLTs have a role in leukemia. It is significant for PLTs to play a part in hematological malignancies from a therapeutic standpoint and to have the capacity to serve as a prognostic marker in the evolution of leukemia. This is because PLTs play a crucial role in the development of cancer and tumors. In this study, it will be shown that PLT count can be used to predict long-term prognosis after chemotherapy especially in the case of acute myeloid leukemia patients. Furthermore, low PLT-to-lymphocyte ratio and mean PLT volume, as well as high PLT distribution width, are associated with poor prognosis and may represent a novel independent prognostic factor.
Collapse
Affiliation(s)
- Yuyan Li
- Department of Experimental Diagnostic, Jilin Kingmed for Clinical Laboratory Co., Ltd., Changchun, 130000, China
| | - Shuangge Wang
- Department of Experimental Diagnostic, Jilin Kingmed for Clinical Laboratory Co., Ltd., Changchun, 130000, China
| | - Han Xiao
- Department of Experimental Diagnostic, Jilin Kingmed for Clinical Laboratory Co., Ltd., Changchun, 130000, China
| | - Fang Lu
- Department of Experimental Diagnostic, Jilin Kingmed for Clinical Laboratory Co., Ltd., Changchun, 130000, China
| | - Bin Zhang
- Department of Experimental Diagnostic, Jilin Kingmed for Clinical Laboratory Co., Ltd., Changchun, 130000, China
| | - Tingting Zhou
- Department of Experimental Diagnostic, Jilin Kingmed for Clinical Laboratory Co., Ltd., Changchun, 130000, China.
| |
Collapse
|
25
|
Bohn E, Lau TTY, Wagih O, Masud T, Merico D. A curated census of pathogenic and likely pathogenic UTR variants and evaluation of deep learning models for variant effect prediction. Front Mol Biosci 2023; 10:1257550. [PMID: 37745687 PMCID: PMC10517338 DOI: 10.3389/fmolb.2023.1257550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/28/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction: Variants in 5' and 3' untranslated regions (UTR) contribute to rare disease. While predictive algorithms to assist in classifying pathogenicity can potentially be highly valuable, the utility of these tools is often unclear, as it depends on carefully selected training and validation conditions. To address this, we developed a high confidence set of pathogenic (P) and likely pathogenic (LP) variants and assessed deep learning (DL) models for predicting their molecular effects. Methods: 3' and 5' UTR variants documented as P or LP (P/LP) were obtained from ClinVar and refined by reviewing the annotated variant effect and reassessing evidence of pathogenicity following published guidelines. Prediction scores from sequence-based DL models were compared between three groups: P/LP variants acting though the mechanism for which the model was designed (model-matched), those operating through other mechanisms (model-mismatched), and putative benign variants. PhyloP was used to compare conservation scores between P/LP and putative benign variants. Results: 295 3' and 188 5' UTR variants were obtained from ClinVar, of which 26 3' and 68 5' UTR variants were classified as P/LP. Predictions by DL models achieved statistically significant differences when comparing modelmatched P/LP variants to both putative benign variants and modelmismatched P/LP variants, as well as when comparing all P/LP variants to putative benign variants. PhyloP conservation scores were significantly higher among P/LP compared to putative benign variants for both the 3' and 5' UTR. Discussion: In conclusion, we present a high-confidence set of P/LP 3' and 5' UTR variants spanning a range of mechanisms and supported by detailed pathogenicity and molecular mechanism evidence curation. Predictions from DL models further substantiate these classifications. These datasets will support further development and validation of DL algorithms designed to predict the functional impact of variants that may be implicated in rare disease.
Collapse
Affiliation(s)
- Emma Bohn
- Deep Genomics Inc., Toronto, ON, Canada
| | | | | | | | - Daniele Merico
- Deep Genomics Inc., Toronto, ON, Canada
- The Centre for Applied Genomics, Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
26
|
Reinig EF, Rubinstein JD, Patil AT, Schussman AL, Horner VL, Kanagal-Shamanna R, Churpek JE, Matson DR. Needle in a haystack or elephant in the room? Identifying germline predisposition syndromes in the setting of a new myeloid malignancy diagnosis. Leukemia 2023; 37:1589-1599. [PMID: 37393344 PMCID: PMC10529926 DOI: 10.1038/s41375-023-01955-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 06/03/2023] [Accepted: 06/22/2023] [Indexed: 07/03/2023]
Abstract
Myeloid malignancies associated with germline predisposition syndromes account for up to 10% of myeloid neoplasms. They are classified into three categories by the proposed 5th Edition of the World Health Organization Classification of Hematolymphoid Tumors: (1) neoplasms with germline predisposition without a pre-existing platelet disorder or organ dysfunction, (2) neoplasms with germline predisposition and pre-existing platelet disorder, or (3) neoplasms with germline predisposition and potential organ dysfunction. Recognizing these entities is critical because patients and affected family members benefit from interfacing with hematologists who specialize in these disorders and can facilitate tailored treatment strategies. However, identification of these syndromes in routine pathology practice is often challenging, as characteristic findings associated with these diagnoses at baseline are frequently absent, nonspecific, or impossible to evaluate in the setting of a myeloid malignancy. Here we review the formally classified germline predisposition syndromes associated with myeloid malignancies and summarize practical recommendations for pathologists evaluating a new myeloid malignancy diagnosis. Our intent is to empower clinicians to better screen for germline disorders in this common clinical setting. Recognizing when to suspect a germline predisposition syndrome, pursue additional ancillary testing, and ultimately recommend referral to a cancer predisposition clinic or hematology specialist, will ensure optimal patient care and expedite research to improve outcomes for these individuals.
Collapse
Affiliation(s)
- Erica F Reinig
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Jeremy D Rubinstein
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Apoorva T Patil
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Amanda L Schussman
- Department of Surgery, University of Wisconsin-Madison, Madison, WI, USA
- Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Vanessa L Horner
- Wisconsin State Laboratory of Hygiene, University of Wisconsin-Madison, Madison, WI, USA
| | - Rashmi Kanagal-Shamanna
- Department of Hematopathology and Molecular Diagnostics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jane E Churpek
- Division of Hematology, Medical Oncology, and Palliative Care, Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Blood Cancer Research Institute, Madison, WI, USA
| | - Daniel R Matson
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA.
- Wisconsin Blood Cancer Research Institute, Madison, WI, USA.
| |
Collapse
|
27
|
Mohan S, Mayers M, Weaver M, Baudet H, De Biase I, Goldstein J, Mao R, McGlaughon J, Moser A, Pujol A, Suchy S, Yuzyuk T, Braverman NE. Evaluating the strength of evidence for genes implicated in peroxisomal disorders using the ClinGen clinical validity framework and providing updates to the peroxisomal disease nomenclature. Mol Genet Metab 2023; 139:107604. [PMID: 37236006 PMCID: PMC10484331 DOI: 10.1016/j.ymgme.2023.107604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 03/09/2023] [Accepted: 05/01/2023] [Indexed: 05/28/2023]
Abstract
Peroxisomal disorders are heterogeneous in nature, with phenotypic overlap that is indistinguishable without molecular testing. Newborn screening and gene sequencing for a panel of genes implicated in peroxisomal diseases are critical tools for the early and accurate detection of these disorders. It is therefore essential to evaluate the clinical validity of the genes included in sequencing panels for peroxisomal disorders. The Peroxisomal Gene Curation Expert Panel (GCEP) assessed genes frequently included on clinical peroxisomal testing panels using the Clinical Genome Resource (ClinGen) gene-disease validity curation framework and classified gene-disease relationships as Definitive, Strong, Moderate, Limited, Disputed, Refuted, or No Known Disease Relationship. Subsequent to gene curation, the GCEP made recommendations to update the disease nomenclature and ontology in the Monarch Disease Ontology (Mondo) database. Thirty-six genes were assessed for the strength of evidence supporting their role in peroxisomal disease, leading to 36 gene-disease relationships, after two genes were removed for their lack of a role in peroxisomal disease and two genes were curated for two different disease entities each. Of these, 23 were classified as Definitive (64%), one as Strong (3%), eight as Moderate (23%), two as Limited (5%), and two as No known disease relationship (5%). No contradictory evidence was found to classify any relationships as Disputed or Refuted. The gene-disease relationship curations are publicly available on the ClinGen website (https://clinicalgenome.org/affiliation/40049/). The changes to peroxisomal disease nomenclature are displayed on the Mondo website (http://purl.obolibrary.org/obo/MONDO_0019053). The Peroxisomal GCEP-curated gene-disease relationships will inform clinical and laboratory diagnostics and enhance molecular testing and reporting. As new data will emerge, the gene-disease classifications asserted by the Peroxisomal GCEP will be re-evaluated periodically.
Collapse
Affiliation(s)
- Shruthi Mohan
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, NC, USA
| | - Megan Mayers
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, NC, USA
| | - Meredith Weaver
- American College of Medical Genetics and Genomics, Bethesda, MD, USA
| | - Heather Baudet
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, NC, USA
| | | | - Jennifer Goldstein
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, NC, USA
| | - Rong Mao
- ARUP Laboratories, Salt Lake City, UT, USA
| | | | - Ann Moser
- Kennedy Krieger Institute, Baltimore, MD, USA
| | - Aurora Pujol
- Bellvitge Biomedical Research Institute (IDIBELL Instituto de Investigación Biomédica de Bellvitge), Barcelona, Spain
| | | | | | - Nancy E Braverman
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada.
| |
Collapse
|
28
|
Gener-Ricos G, Gerstein YS, Hammond D, DiNardo CD. Germline Predisposition to Myelodysplastic Syndromes. Cancer J 2023; 29:143-151. [PMID: 37195770 DOI: 10.1097/ppo.0000000000000660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
ABSTRACT While germline predisposition to myelodysplastic syndromes is well-established, knowledge has advanced rapidly resulting in more cases of inherited hematologic malignancies being identified. Understanding the biological features and main clinical manifestations of hereditary hematologic malignancies is essential to recognizing and referring patients with myelodysplastic syndrome, who may underlie inherited predisposition, for appropriate genetic evaluation. Importance lies in individualized genetic counseling along with informed treatment decisions, especially with regard to hematopoietic stem cell transplant-related donor selection. Future studies will improve comprehension of these disorders, enabling better management of affected patients and their families.
Collapse
Affiliation(s)
| | - Yoheved S Gerstein
- Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | |
Collapse
|
29
|
Kemmler CL, Smolikova J, Moran HR, Mannion BJ, Knapp D, Lim F, Czarkwiani A, Hermosilla Aguayo V, Rapp V, Fitch OE, Bötschi S, Selleri L, Farley E, Braasch I, Yun M, Visel A, Osterwalder M, Mosimann C, Kozmik Z, Burger A. Conserved enhancer logic controls the notochord expression of vertebrate Brachyury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.20.536761. [PMID: 37131681 PMCID: PMC10153258 DOI: 10.1101/2023.04.20.536761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The cell type-specific expression of key transcription factors is central to development. Brachyury/T/TBXT is a major transcription factor for gastrulation, tailbud patterning, and notochord formation; however, how its expression is controlled in the mammalian notochord has remained elusive. Here, we identify the complement of notochord-specific enhancers in the mammalian Brachyury/T/TBXT gene. Using transgenic assays in zebrafish, axolotl, and mouse, we discover three Brachyury-controlling notochord enhancers T3, C, and I in human, mouse, and marsupial genomes. Acting as Brachyury-responsive, auto-regulatory shadow enhancers, deletion of all three enhancers in mouse abolishes Brachyury/T expression selectively in the notochord, causing specific trunk and neural tube defects without gastrulation or tailbud defects. Sequence and functional conservation of Brachyury-driving notochord enhancers with the brachyury/tbxtb loci from diverse lineages of fishes dates their origin to the last common ancestor of jawed vertebrates. Our data define the enhancers for Brachyury/T/TBXTB notochord expression as ancient mechanism in axis development.
Collapse
Affiliation(s)
- Cassie L. Kemmler
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jana Smolikova
- Institute of Molecular Genetics of the ASCR, v. v. i., Prague, Czech Republic
| | - Hannah R. Moran
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Brandon J. Mannion
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Comparative Biochemistry Program, University of California, Berkeley, CA 94720, USA
| | - Dunja Knapp
- Technische Universität Dresden, CRTD/Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Fabian Lim
- Department of Medicine, Health Sciences, University of California San Diego, La Jolla, CA, USA
- Department of Molecular Biology, Biological Sciences, University of California San Diego, La Jolla, CA USA
- Biological Sciences Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Anna Czarkwiani
- Technische Universität Dresden, CRTD/Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Viviana Hermosilla Aguayo
- Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA
| | - Vincent Rapp
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Olivia E. Fitch
- Department of Integrative Biology and Ecology, Evolution and Behavior Program, Michigan State University, East Lansing, MI, USA
| | - Seraina Bötschi
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Licia Selleri
- Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA
| | - Emma Farley
- Department of Medicine, Health Sciences, University of California San Diego, La Jolla, CA, USA
- Department of Molecular Biology, Biological Sciences, University of California San Diego, La Jolla, CA USA
| | - Ingo Braasch
- Department of Integrative Biology and Ecology, Evolution and Behavior Program, Michigan State University, East Lansing, MI, USA
| | - Maximina Yun
- Technische Universität Dresden, CRTD/Center for Regenerative Therapies Dresden, Dresden, Germany
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden, Germany
| | - Axel Visel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- US Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- School of Natural Sciences, University of California Merced, Merced, CA, USA
| | - Marco Osterwalder
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Cardiology, Berne University Hospital, Berne, Switzerland
| | - Christian Mosimann
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Zbynek Kozmik
- Institute of Molecular Genetics of the ASCR, v. v. i., Prague, Czech Republic
| | - Alexa Burger
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
30
|
Förster A, Davenport C, Duployez N, Erlacher M, Ferster A, Fitzgibbon J, Göhring G, Hasle H, Jongmans MC, Kolenova A, Kronnie G, Lammens T, Mecucci C, Mlynarski W, Niemeyer CM, Sole F, Szczepanski T, Waanders E, Biondi A, Wlodarski M, Schlegelberger B, Ripperger T. European standard clinical practice - Key issues for the medical care of individuals with familial leukemia. Eur J Med Genet 2023; 66:104727. [PMID: 36775010 DOI: 10.1016/j.ejmg.2023.104727] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 02/02/2023] [Accepted: 02/10/2023] [Indexed: 02/12/2023]
Abstract
Although hematologic malignancies (HM) are no longer considered exclusively sporadic, additional awareness of familial cases has yet to be created. Individuals carrying a (likely) pathogenic germline variant (e.g., in ETV6, GATA2, SAMD9, SAMD9L, or RUNX1) are at an increased risk for developing HM. Given the clinical and psychological impact associated with the diagnosis of a genetic predisposition to HM, it is of utmost importance to provide high-quality, standardized patient care. To address these issues and harmonize care across Europe, the Familial Leukemia Subnetwork within the ERN PaedCan has been assigned to draft an European Standard Clinical Practice (ESCP) document reflecting current best practices for pediatric patients and (healthy) relatives with (suspected) familial leukemia. The group was supported by members of the German network for rare diseases MyPred, of the Host Genome Working Group of SIOPE, and of the COST action LEGEND. The ESCP on familial leukemia is proposed by an interdisciplinary team of experts including hematologists, oncologists, and human geneticists. It is intended to provide general recommendations in areas where disease-specific recommendations do not yet exist. Here, we describe key issues for the medical care of familial leukemia that shall pave the way for a future consensus guideline: (i) identification of individuals with or suggestive of familial leukemia, (ii) genetic analysis and variant interpretation, (iii) genetic counseling and patient education, and (iv) surveillance and (psychological) support. To address the question on how to proceed with individuals suggestive of or at risk of familial leukemia, we developed an algorithm covering four different, partially linked clinical scenarios, and additionally a decision tree to guide clinicians in their considerations regarding familial leukemia in minors with HM. Our recommendations cover, not only patients but also relatives that both should have access to adequate medical care. We illustrate the importance of natural history studies and the need for respective registries for future evidence-based recommendations that shall be updated as new evidence-based standards are established.
Collapse
Affiliation(s)
- Alisa Förster
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Claudia Davenport
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Nicolas Duployez
- Department of Hematology, CHU Lille, INSERM, University Lille, Lille, France
| | - Miriam Erlacher
- Division of Pediatric Hematology-Oncology, Department of Pediatric and Adolescent Medicine, University of Freiburg, Freiburg, Germany
| | - Alina Ferster
- Department of Pediatric Rheumatology, Hôpital Universitaire des Enfants Reine Fabiola, Brussels, Belgium
| | - Jude Fitzgibbon
- Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Gudrun Göhring
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Henrik Hasle
- Department of Paediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Marjolijn C Jongmans
- Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Alexandra Kolenova
- Department of Pediatric Hematology and Oncology, Comenius University Medical School and University Children's Hospital, Bratislava, Slovakia
| | | | - Tim Lammens
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Cristina Mecucci
- Institute of Hematology and Center for Hemato-Oncology Research, University and Hospital of Perugia, Perugia, Italy
| | - Wojciech Mlynarski
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Lodz, Poland
| | - Charlotte M Niemeyer
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Francesc Sole
- Josep Carreras Leukemia Research Institute (IJC), Campus ICO-Hospital Germans Trias i Pujol, Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Tomasz Szczepanski
- Polish Pediatric Leukemia/Lymphoma Study Group, Zabrze, Poland; Medical University of Silesia, Katowice, Poland
| | - Esmé Waanders
- Department of Genetics, University Medical Center Utrecht, Utrecht, the Netherlands; Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Andrea Biondi
- Clinica Pediatrica and Centro Ricerca Tettamanti, Università di Milano-Bicocca, Monza, Italy
| | - Marcin Wlodarski
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Tim Ripperger
- Department of Human Genetics, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
31
|
Homan CC, Scott HS, Brown AL. Hereditary platelet disorders associated with germ line variants in RUNX1, ETV6, and ANKRD26. Blood 2023; 141:1533-1543. [PMID: 36626254 PMCID: PMC10651873 DOI: 10.1182/blood.2022017735] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/22/2022] [Accepted: 12/24/2022] [Indexed: 01/11/2023] Open
Abstract
Hereditary platelet disorders (HPDs) are a group of blood disorders with variable severity and clinical impact. Although phenotypically there is much overlap, known genetic causes are many, prompting the curation of multigene panels for clinical use, which are being deployed in increasingly large-scale populations to uncover missing heritability more efficiently. For some of these disorders, in particular RUNX1, ETV6, and ANKRD26, pathogenic germ line variants in these genes also come with a risk of developing hematological malignancy (HM). Although they may initially present as similarly mild-moderate thrombocytopenia, each of these 3 disorders have distinct penetrance of HM and a different range of somatic alterations associated with malignancy development. As our ability to diagnose HPDs has improved, we are now faced with the challenges of integrating these advances into routine clinical practice for patients and how to optimize management and surveillance of patients and carriers who have not developed malignancy. The volume of genetic information now being generated has created new challenges in how to accurately assess and report identified variants. The answers to all these questions involve international initiatives on rare diseases to better understand the biology of these disorders and design appropriate models and therapies for preclinical testing and clinical trials. Partnered with this are continued technological developments, including the rapid sharing of genetic variant information and automated integration with variant classification relevant data, such as high-throughput functional data. Collective progress in this area will drive timely diagnosis and, in time, leukemia preventive therapeutic interventions.
Collapse
Affiliation(s)
- Claire C. Homan
- Department of Genetics and Molecular Pathology, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
| | - Hamish S. Scott
- Department of Genetics and Molecular Pathology, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
- Australian Cancer Research Foundation (ACRF) Genomics Facility, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Anna L. Brown
- Department of Genetics and Molecular Pathology, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- UniSA Clinical and Health Sciences, University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
32
|
McCormick BJ, Chirila RM. ANKRD26 Gene Variant of Uncertain Significance in a Patient With Acute Myeloid Leukemia. Cureus 2023; 15:e36152. [PMID: 37065357 PMCID: PMC10101738 DOI: 10.7759/cureus.36152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2023] [Indexed: 03/16/2023] Open
Abstract
ANKRD26-related thrombocytopenia is a rare inherited disorder associated with an increased risk of malignancy. While the genetic mutations underlying this condition are well understood, there is limited knowledge regarding its contribution to myeloid neoplasms, such as acute myeloid leukemia (AML). We present a case of ANKRD26-related thrombocytopenia with a variant of uncertain significance in a patient with AML and review the pathogenesis and implications of hereditary germline mutations in disease management.
Collapse
|
33
|
Santiago M, Liquori A, Such E, Zúñiga Á, Cervera J. The Clinical Spectrum, Diagnosis, and Management of GATA2 Deficiency. Cancers (Basel) 2023; 15:cancers15051590. [PMID: 36900380 PMCID: PMC10000430 DOI: 10.3390/cancers15051590] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/02/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Hereditary myeloid malignancy syndromes (HMMSs) are rare but are becoming increasingly significant in clinical practice. One of the most well-known syndromes within this group is GATA2 deficiency. The GATA2 gene encodes a zinc finger transcription factor essential for normal hematopoiesis. Insufficient expression and function of this gene as a result of germinal mutations underlie distinct clinical presentations, including childhood myelodysplastic syndrome and acute myeloid leukemia, in which the acquisition of additional molecular somatic abnormalities can lead to variable outcomes. The only curative treatment for this syndrome is allogeneic hematopoietic stem cell transplantation, which should be performed before irreversible organ damage happens. In this review, we will examine the structural characteristics of the GATA2 gene, its physiological and pathological functions, how GATA2 genetic mutations contribute to myeloid neoplasms, and other potential clinical manifestations. Finally, we will provide an overview of current therapeutic options, including recent transplantation strategies.
Collapse
Affiliation(s)
- Marta Santiago
- Hematology Department, Hospital La Fe, 46026 Valencia, Spain; (M.S.); (E.S.); (J.C.)
- Hematology Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
| | - Alessandro Liquori
- Hematology Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Correspondence:
| | - Esperanza Such
- Hematology Department, Hospital La Fe, 46026 Valencia, Spain; (M.S.); (E.S.); (J.C.)
- Hematology Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Ángel Zúñiga
- Genetics Unit, Hospital La Fe, 46026 Valencia, Spain;
| | - José Cervera
- Hematology Department, Hospital La Fe, 46026 Valencia, Spain; (M.S.); (E.S.); (J.C.)
- Hematology Research Group, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Genetics Unit, Hospital La Fe, 46026 Valencia, Spain;
| |
Collapse
|
34
|
Speight B, Hanson H, Turnbull C, Hardy S, Drummond J, Khorashad J, Wragg C, Page P, Parkin NW, Rio-Machin A, Fitzgibbon J, Kulasekararaj AG, Hamblin A, Talley P, McVeigh TP, Snape K. Germline predisposition to haematological malignancies: Best practice consensus guidelines from the UK Cancer Genetics Group (UKCGG), CanGene-CanVar and the NHS England Haematological Oncology Working Group. Br J Haematol 2023; 201:25-34. [PMID: 36744544 DOI: 10.1111/bjh.18675] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/10/2023] [Accepted: 01/17/2023] [Indexed: 02/07/2023]
Abstract
The implementation of whole genome sequencing and large somatic gene panels in haematological malignancies is identifying an increasing number of individuals with either potential or confirmed germline predisposition to haematological malignancy. There are currently no national or international best practice guidelines with respect to management of carriers of such variants or of their at-risk relatives. To address this gap, the UK Cancer Genetics Group (UKCGG), CanGene-CanVar and the NHS England Haematological Oncology Working Group held a workshop over two days on 28-29th April 2022, with the aim of establishing consensus guidelines on relevant clinical and laboratory pathways. The workshop focussed on the management of disease-causing germline variation in the following genes: DDX41, CEBPA, RUNX1, ANKRD26, ETV6, GATA2. Using a pre-workshop survey followed by structured discussion and in-meeting polling, we achieved consensus for UK best practice in several areas. In particular, high consensus was achieved on issues regarding standardised reporting, variant classification, multidisciplinary team working and patient support. The best practice recommendations from this meeting may be applicable to an expanding number of other genes in this setting.
Collapse
Affiliation(s)
- Beverley Speight
- East Anglian Medical Genetics Service, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, UK
| | - Helen Hanson
- South West Thames Regional Genetics Service, St George's University Hospitals NHS Foundation Trust, London, UK
- Institute of Cancer Research, Sutton, London, UK
| | - Clare Turnbull
- Institute of Cancer Research, Sutton, London, UK
- Cancer Genetics Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - Steven Hardy
- National Disease Registration Service, NHS Digital, London, UK
| | - James Drummond
- East Anglian Medical Genetics Service, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, UK
| | | | - Christopher Wragg
- South West Genomics Laboratory Hub, Bristol Genetics Laboratory, North Bristol NHS Trust, Pathology Building, Southmead Hospital, Bristol, UK
| | - Paula Page
- West Midlands Regional Genetics Laboratory, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Nicholas W Parkin
- Molecular Pathology Laboratory, Synnovis Analytics, King's College Hospital, London, UK
| | - Ana Rio-Machin
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Jude Fitzgibbon
- Centre for Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Austin Gladston Kulasekararaj
- King's College Hospital NHS Foundation Trust, London, UK
- National Institute for Health and Care Research and Wellcome King's Research Facility, London, UK
- King's College London, London, UK
| | - Angela Hamblin
- Oxford University Hospitals NHS Foundation Trust and Central and South Genomic Laboratory Hub, Oxford, UK
| | - Polly Talley
- Genomics Unit, NHS UK and NHS Improvement, Leeds, UK
- North East and Yorkshire Genomic Laboratory Hub, Leeds, UK
| | - Terri P McVeigh
- Institute of Cancer Research, Sutton, London, UK
- Cancer Genetics Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - Katie Snape
- South West Thames Regional Genetics Service, St George's University Hospitals NHS Foundation Trust, London, UK
- King's College Hospital NHS Foundation Trust, London, UK
| | | |
Collapse
|
35
|
Zaninetti C, Leinøe E, Lozano ML, Rossing M, Bastida JM, Zetterberg E, Rivera J, Greinacher A. Validation of immunofluorescence analysis of blood smears in patients with inherited platelet disorders. JOURNAL OF THROMBOSIS AND HAEMOSTASIS : JTH 2023; 21:1010-1019. [PMID: 36732160 DOI: 10.1016/j.jtha.2022.12.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 12/08/2022] [Accepted: 12/27/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND Inherited platelet disorders (IPDs) are rare diseases characterized by reduced blood platelet counts and/or impaired platelet function. Recognizing IPDs is advisable but often challenging. The diagnostic tools include clinical evaluation, platelet function tests, and molecular analyses. Demonstration of a pathogenic genetic variant confirms IPDs. We established a method to assess the platelet phenotype on blood smears using immunofluorescence microscopy as a diagnostic tool for IPDs. OBJECTIVES The aim of the present study was to validate immunofluorescence microscopy as a screening tool for IPDs in comparison with genetic screening. METHODS We performed a blinded comparison between the diagnosis made using immunofluorescence microscopy on blood smears and genetic findings in a cohort of 43 families affected with 20 different genetically confirmed IPDs. In total, 76% of the cases had inherited thrombocytopenia. RESULTS Immunofluorescence correctly predicted the underlying IPD in the vast majority of patients with 1 of 9 IPDs for which the typical morphologic pattern is known. Thirty of the 43 enrolled families (70%) were affected by 1 of these 9 IPDs. For the other 11 forms of IPD, we describe alterations of platelet structure in 9 disorders and normal findings in 2 disorders. CONCLUSION Immunofluorescence microscopy on blood smears is an effective screening tool for 9 forms of IPD, which include the most frequent forms of inherited thrombocytopenia. Using this approach, typical changes in the phenotype may also be identified for other rare IPDs.
Collapse
Affiliation(s)
- Carlo Zaninetti
- Institut für Transfusionsmedizin, Universitätsmedizin Greifswald, Greifswald, Germany. https://twitter.com/ZaninettiCarlo
| | - Eva Leinøe
- Department of Haematology, Rigshospitalet University Hospital, Copenhagen, Denmark; Department of Genomic Medicine, Rigshospitalet University Hospital, Copenhagen, Denmark
| | - María Luisa Lozano
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Pascual Parrilla, CIBERER-U765, Murcia, Spain
| | - Maria Rossing
- Centre for Genomic Medicine, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jose Maria Bastida
- Department of Hematology, Complejo Asistencial Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca, Universidad de Salamanca, Salamanca, Spain; Grupo Español de Alteraciones Plaquetarias Congénitas, Spanish Society of Thrombosis and Haemostasis, Madrid, Spain
| | - Eva Zetterberg
- Clinical Coagulation Research Unit, Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Jose Rivera
- Servicio de Hematología y Oncología Médica, Hospital Universitario Morales Meseguer, Centro Regional de Hemodonación, Universidad de Murcia, IMIB-Pascual Parrilla, CIBERER-U765, Murcia, Spain; Grupo Español de Alteraciones Plaquetarias Congénitas, Spanish Society of Thrombosis and Haemostasis, Madrid, Spain
| | - Andreas Greinacher
- Institut für Transfusionsmedizin, Universitätsmedizin Greifswald, Greifswald, Germany.
| |
Collapse
|
36
|
The International Consensus Classification (ICC) of hematologic neoplasms with germline predisposition, pediatric myelodysplastic syndrome, and juvenile myelomonocytic leukemia. Virchows Arch 2023; 482:113-130. [PMID: 36445482 DOI: 10.1007/s00428-022-03447-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/23/2022] [Accepted: 10/27/2022] [Indexed: 11/30/2022]
Abstract
Updating the classification of hematologic neoplasia with germline predisposition, pediatric myelodysplastic syndrome (MDS), and juvenile myelomonocytic leukemia (JMML) is critical for diagnosis, therapy, research, and clinical trials. Advances in next-generation sequencing technology have led to the identification of an expanding group of genes that predispose to the development of hematolymphoid neoplasia when mutated in germline configuration and inherited. This review encompasses recent advances in the classification of myeloid and lymphoblastic neoplasia with germline predisposition summarizing important genetic and phenotypic information, relevant laboratory testing, and pathologic bone marrow features. Genes are organized into three major categories including (1) those that are not associated with constitutional disorder and include CEBPA, DDX41, and TP53; (2) those associated with thrombocytopenia or platelet dysfunction including RUNX1, ANKRD26, and ETV6; and (3) those associated with constitutional disorders affecting multiple organ systems including GATA2, SAMD9, and SAMD9L, inherited genetic mutations associated with classic bone marrow failure syndromes and JMML, and Down syndrome. A provisional category of germline predisposition genes is created to recognize genes with growing evidence that may be formally included in future revised classifications as substantial supporting data emerges. We also detail advances in the classification of pediatric myelodysplastic syndrome (MDS), expanding the definition of refractory cytopenia of childhood (RCC) to include early manifestation of MDS in patients with germline predisposition. Finally, updates in the classification of juvenile myelomonocytic leukemia are presented which genetically define JMML as a myeloproliferative/myelodysplastic disease harboring canonical RAS pathway mutations. Diseases with features overlapping with JMML that do not carry RAS pathway mutations are classified as JMML-like. The review is based on the International Consensus Classification (ICC) of Myeloid and Lymphoid Neoplasms as reported by Arber et al. (Blood 140(11):1200-1228, 2022).
Collapse
|
37
|
Whole exome and genome sequencing in mendelian disorders: a diagnostic and health economic analysis. Eur J Hum Genet 2022; 30:1121-1131. [PMID: 35970915 PMCID: PMC9553973 DOI: 10.1038/s41431-022-01162-2] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 06/01/2022] [Accepted: 07/20/2022] [Indexed: 12/15/2022] Open
Abstract
Whole genome sequencing (WGS) improves Mendelian disorder diagnosis over whole exome sequencing (WES); however, additional diagnostic yields and costs remain undefined. We investigated differences between diagnostic and cost outcomes of WGS and WES in a cohort with suspected Mendelian disorders. WGS was performed in 38 WES-negative families derived from a 64 family Mendelian cohort that previously underwent WES. For new WGS diagnoses, contemporary WES reanalysis determined whether variants were diagnosable by original WES or unique to WGS. Diagnostic rates were estimated for WES and WGS to simulate outcomes if both had been applied to the 64 families. Diagnostic costs were calculated for various genomic testing scenarios. WGS diagnosed 34% (13/38) of WES-negative families. However, contemporary WES reanalysis on average 2 years later would have diagnosed 18% (7/38 families) resulting in a WGS-specific diagnostic yield of 19% (6/31 remaining families). In WES-negative families, the incremental cost per additional diagnosis using WGS following WES reanalysis was AU$36,710 (£19,407;US$23,727) and WGS alone was AU$41,916 (£22,159;US$27,093) compared to WES-reanalysis. When we simulated the use of WGS alone as an initial genomic test, the incremental cost for each additional diagnosis was AU$29,708 (£15,705;US$19,201) whereas contemporary WES followed by WGS was AU$36,710 (£19,407;US$23,727) compared to contemporary WES. Our findings confirm that WGS is the optimal genomic test choice for maximal diagnosis in Mendelian disorders. However, accepting a small reduction in diagnostic yield, WES with subsequent reanalysis confers the lowest costs. Whether WES or WGS is utilised will depend on clinical scenario and local resourcing and availability.
Collapse
|
38
|
Gilad O, Dgany O, Noy-Lotan S, Krasnov T, Yacobovich J, Rabinowicz R, Goldberg T, Kuperman AA, Abu-Quider A, Miskin H, Kapelushnik N, Mandel-Shorer N, Shimony S, Harlev D, Ben-Ami T, Adam E, Levin C, Aviner S, Elhasid R, Berger-Achituv S, Chaitman-Yerushalmi L, Kodman Y, Oniashvilli N, Hameiri-Grosman M, Izraeli S, Tamary H, Steinberg-Shemer O. Syndromes predisposing to leukemia are a major cause of inherited cytopenias in children. Haematologica 2022; 107:2081-2095. [PMID: 35295078 PMCID: PMC9425329 DOI: 10.3324/haematol.2021.280116] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 03/10/2022] [Indexed: 11/09/2022] Open
Abstract
Prolonged cytopenias are a non-specific sign with a wide differential diagnosis. Among inherited disorders, cytopenias predisposing to leukemia require a timely and accurate diagnosis to ensure appropriate medical management, including adequate monitoring and stem cell transplantation prior to the development of leukemia. We aimed to define the types and prevalences of the genetic causes leading to persistent cytopenias in children. The study comprises children with persistent cytopenias, myelodysplastic syndrome, aplastic anemia, or suspected inherited bone marrow failure syndromes, who were referred for genetic evaluation from all pediatric hematology centers in Israel during 2016-2019. For variant detection, we used Sanger sequencing of commonly mutated genes and a custom-made targeted next-generation sequencing panel covering 226 genes known to be mutated in inherited cytopenias; the minority subsequently underwent whole exome sequencing. In total, 189 children with persistent cytopenias underwent a genetic evaluation. Pathogenic and likely pathogenic variants were identified in 59 patients (31.2%), including 47 with leukemia predisposing syndromes. Most of the latter (32, 68.1%) had inherited bone marrow failure syndromes, nine (19.1%) had inherited thrombocytopenia predisposing to leukemia, and three each (6.4%) had predisposition to myelodysplastic syndrome or congenital neutropenia. Twelve patients had cytopenias with no known leukemia predisposition, including nine children with inherited thrombocytopenia and three with congenital neutropenia. In summary, almost one third of 189 children referred with persistent cytopenias had an underlying inherited disorder; 79.7% of whom had a germline predisposition to leukemia. Precise diagnosis of children with cytopenias should direct follow-up and management programs and may positively impact disease outcome.
Collapse
Affiliation(s)
- Oded Gilad
- Department of Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel; Sackler Faculty of Medicine, Aviv University, Aviv
| | - Orly Dgany
- Pediatric Hematology Laboratory, Felsenstein Medical Research Center, Petach Tikva
| | - Sharon Noy-Lotan
- Pediatric Hematology Laboratory, Felsenstein Medical Research Center, Petach Tikva
| | - Tanya Krasnov
- Pediatric Hematology Laboratory, Felsenstein Medical Research Center, Petach Tikva
| | - Joanne Yacobovich
- Department of Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel; Sackler Faculty of Medicine, Aviv University, Aviv
| | - Ron Rabinowicz
- Department of Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel; Sackler Faculty of Medicine, Aviv University, Aviv
| | - Tracie Goldberg
- Department of Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel; Sackler Faculty of Medicine, Aviv University, Aviv
| | - Amir A Kuperman
- Blood Coagulation Service and Pediatric Hematology Clinic, Galilee Medical Center, Nahariya, Israel; Azrieli Faculty of Medicine, Bar-Ilan University, Safed
| | - Abed Abu-Quider
- Pediatric Hematology, Soroka University Medical Center, Ben-Gurion University, Beer Sheva
| | - Hagit Miskin
- Pediatric Hematology Unit, Shaare Zedek Medical Center, Jerusalem, Israel; Faculty of Medicine, Hebrew University, Jerusalem
| | - Noa Kapelushnik
- Sackler Faculty of Medicine, Aviv University, Aviv, Israel; Goldschleger Eye Institute, Sheba Medical Center, Hashomer
| | - Noa Mandel-Shorer
- Department of Pediatric Hematology-Oncology, Ruth Rappaport Children's Hospital, Rambam Healthcare Campus; Rappaport Faculty of Medicine, Technion-Institute of Technology, Haifa
| | - Shai Shimony
- Sackler Faculty of Medicine, Aviv University, Aviv, Israel; Rabin Medical Center, Institute of Hematology, Davidoff Cancer Centre, Beilinson Hospital, Petach-Tikva, Israel; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Dan Harlev
- Pediatric Hematology-Oncology Department, Hadassah University Medical Center, Jerusalem
| | - Tal Ben-Ami
- Pediatric Hematology Unit, Kaplan Medical Center, Rehovot, Israel; Faculty of Medicine, Hebrew University of Jerusalem
| | - Etai Adam
- Pediatric Hematology-Oncology Department, Sheba Medical Center, Hashomer
| | - Carina Levin
- Rappaport Faculty of Medicine, Technion-Institute of Technology, Haifa, Israel; Pediatric Hematology Unit and Research Laboratory, Emek Medical Center, Afula
| | - Shraga Aviner
- Department of Pediatrics, Barzilai University Medical Center, Ashkelon, affiliated to Ben Gurion University, Beer-Sheva
| | - Ronit Elhasid
- Sackler Faculty of Medicine, Aviv University, Aviv, Israel; Department of Pediatric Hemato-Oncology, Aviv Medical Center
| | - Sivan Berger-Achituv
- Sackler Faculty of Medicine, Aviv University, Aviv, Israel; Department of Pediatric Hemato-Oncology, Aviv Medical Center
| | | | - Yona Kodman
- Department of Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva
| | - Nino Oniashvilli
- Department of Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva
| | - Michal Hameiri-Grosman
- Department of Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva
| | - Shai Izraeli
- Department of Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel; Sackler Faculty of Medicine, Aviv University, Aviv
| | - Hannah Tamary
- Department of Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel; Sackler Faculty of Medicine, Aviv University, Aviv, Israel; Pediatric Hematology Laboratory, Felsenstein Medical Research Center, Petach Tikva.
| | - Orna Steinberg-Shemer
- Department of Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel; Sackler Faculty of Medicine, Aviv University, Aviv, Israel; Pediatric Hematology Laboratory, Felsenstein Medical Research Center, Petach Tikva
| |
Collapse
|
39
|
Sullivan MJ, Palmer EL, Botero JP. ANKRD26-Related Thrombocytopenia and Predisposition to Myeloid Neoplasms. Curr Hematol Malig Rep 2022; 17:105-112. [PMID: 35751752 DOI: 10.1007/s11899-022-00666-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW This review describes ANKRD26-related thrombocytopenia (RT) from a molecular, clinical, and laboratory perspective, with a focus on the clinical decision-making that takes place in the diagnosis and management of families with ANKRD26-RT. RECENT FINDINGS ANKRD26-related thrombocytopenia (ANKRD26-RT) is a non-syndromic autosomal dominant thrombocytopenia with predisposition to hematologic neoplasm. The clinical presentation is variable with moderate thrombocytopenia with normal platelet size and absent to mild bleeding being the hallmark which makes it difficult to distinguish from other inherited thrombocytopenias. The pathophysiology involves overexpression of ANKRD26 through loss of inhibitory control by transcription factors RUNX1 and FLI1. The great majority of disease-causing variants are in the 5' untranslated region. Acute myeloid leukemia, myelodysplastic syndrome, and chronic myelomonocytic leukemia have been reported to occur in the context of germline variants in ANKRD26, with the development of somatic driver mutations in hematopoietic regulators playing an important role in malignant transformation. In the absence of clear risk estimates of development of malignancy, optimal surveillance strategies and interventions to reduce risk of evolution to a myeloid disorder, multidisciplinary evaluation, with a strong genetic counseling framework is essential in the approach to these patients and their families. Gene-specific expertise and a multidisciplinary approach are important in the diagnosis and treatment of patients and families with ANKRD26-RT. These strategies help overcome the challenges faced by clinicians in the evaluation of individuals with a rare, non-syndromic, inherited disorder with predisposition to hematologic malignancy for which large data to guide decision-making is not available.
Collapse
Affiliation(s)
- Mia J Sullivan
- Diagnostic Laboratories, Versiti, 638 N 18th St, Milwaukee, WI, 53233, USA
| | - Elizabeth L Palmer
- Diagnostic Laboratories, Versiti, 638 N 18th St, Milwaukee, WI, 53233, USA
| | - Juliana Perez Botero
- Diagnostic Laboratories, Versiti, 638 N 18th St, Milwaukee, WI, 53233, USA. .,Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
40
|
Warren JT, Di Paola J. Genetics of inherited thrombocytopenias. Blood 2022; 139:3264-3277. [PMID: 35167650 PMCID: PMC9164741 DOI: 10.1182/blood.2020009300] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 02/04/2022] [Indexed: 01/19/2023] Open
Abstract
The inherited thrombocytopenia syndromes are a group of disorders characterized primarily by quantitative defects in platelet number, though with a variety demonstrating qualitative defects and/or extrahematopoietic findings. Through collaborative international efforts applying next-generation sequencing approaches, the list of genetic syndromes that cause thrombocytopenia has expanded significantly in recent years, now with over 40 genes implicated. In this review, we focus on what is known about the genetic etiology of inherited thrombocytopenia syndromes and how the field has worked to validate new genetic discoveries. We highlight the important role for the clinician in identifying a germline genetic diagnosis and strategies for identifying novel causes through research-based endeavors.
Collapse
Affiliation(s)
- Julia T Warren
- Division of Hematology-Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Jorge Di Paola
- Division of Hematology-Oncology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
41
|
Vyas H, Alcheikh A, Lowe G, Stevenson WS, Morgan NV, Rabbolini DJ. Prevalence and natural history of variants in the ANKRD26 gene: a short review and update of reported cases. Platelets 2022; 33:1107-1112. [PMID: 35587581 PMCID: PMC9555274 DOI: 10.1080/09537104.2022.2071853] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
ANKRD26 is a highly conserved gene located on chromosome 10p12.1 which has shown to play a role in normal megakaryocyte differentiation. ANKRD26-related thrombocytopenia, or thrombocytopenia 2, is an inherited thrombocytopenia with mild bleeding diathesis resulting from point mutations the 5ʹUTR of the ANKRD26 gene. Point mutations in the 5ʹUTR region have been shown to prevent transcription factor-mediated downregulation of ANKRD26 in normal megakaryocyte differentiation. Patients with ANKRD26-related thrombocytopenia have a predisposition to developing hematological malignancies, with acute myeloid leukemia and myelodysplastic syndrome most commonly described in the literature. We review the clinical features and biological mechanisms of ANKRD26-related thrombocytopenia and summarize known cases in the literature.
Collapse
Affiliation(s)
- Hrushikesh Vyas
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Ahmad Alcheikh
- Northern Blood Research Centre, Kolling Institute, University of Sydney, Sydney, Australia
| | - Gillian Lowe
- Comprehensive Care Haemophilia Centre, University Hospital Birmingham NHS Foundation Trust, Birmingham, UK
| | - William S Stevenson
- Northern Blood Research Centre, Kolling Institute, University of Sydney, Sydney, Australia.,Department of Haematology and Transfusion Medicine, Royal North Shore Hospital, Sydney, Australia
| | - Neil V Morgan
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - David J Rabbolini
- Northern Blood Research Centre, Kolling Institute, University of Sydney, Sydney, Australia
| |
Collapse
|
42
|
Feurstein S, Hahn CN, Mehta N, Godley LA. A practical guide to interpreting germline variants that drive hematopoietic malignancies, bone marrow failure, and chronic cytopenias. Genet Med 2022; 24:931-954. [PMID: 35063349 DOI: 10.1016/j.gim.2021.12.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 12/14/2021] [Indexed: 10/19/2022] Open
Abstract
PURPOSE The American College of Medical Genetics and Genomics and the Association for Molecular Pathology guidelines for germline variant interpretation are implemented as a broad framework by standardizing variant interpretation. These rules were designed to be specified, but this process has not been performed for most of the 200 genes associated with inherited hematopoietic malignancies, bone marrow failure, and cytopenias. Because guidelines on how to perform these gene specifications are lacking, variant interpretation is less reliable and reproducible. METHODS We have used a variety of methods such as calculations of minor allele frequencies, quasi-case-control studies to establish thresholds, proband counting, and plotting of receiver operating characteristic curves to compare different in silico prediction tools to design recommendations for variant interpretation. RESULTS We herein provide practical recommendations for the creation of thresholds for minor allele frequencies, in silico predictions, counting of probands, identification of functional domains with minimal benign variation, use of constraint Z-scores and functional evidence, prediction of nonsense-mediated decay, and assessment of phenotype specificity. CONCLUSION These guidelines can be used by anyone interpreting variants associated with inherited hematopoietic malignancies, bone marrow failure, and cytopenias to develop criteria for reliable, accurate, and reproducible germline variant interpretation.
Collapse
Affiliation(s)
- Simone Feurstein
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL; Section of Hematology, Oncology and Rheumatology, Department of Internal Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Christopher N Hahn
- Molecular Pathology Research Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia; Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| | - Nikita Mehta
- Diagnostic Molecular Genetics Laboratory, Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Lucy A Godley
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL; Department of Human Genetics, The University of Chicago, Chicago, IL.
| |
Collapse
|
43
|
Bourguignon A, Tasneem S, Hayward CP. Screening and diagnosis of inherited platelet disorders. Crit Rev Clin Lab Sci 2022; 59:405-444. [PMID: 35341454 DOI: 10.1080/10408363.2022.2049199] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Inherited platelet disorders are important conditions that often manifest with bleeding. These disorders have heterogeneous underlying pathologies. Some are syndromic disorders with non-blood phenotypic features, and others are associated with an increased predisposition to developing myelodysplasia and leukemia. Platelet disorders can present with thrombocytopenia, defects in platelet function, or both. As the underlying pathogenesis of inherited thrombocytopenias and platelet function disorders are quite diverse, their evaluation requires a thorough clinical assessment and specialized diagnostic tests, that often challenge diagnostic laboratories. At present, many of the commonly encountered, non-syndromic platelet disorders do not have a defined molecular cause. Nonetheless, significant progress has been made over the past few decades to improve the diagnostic evaluation of inherited platelet disorders, from the assessment of the bleeding history to improved standardization of light transmission aggregometry, which remains a "gold standard" test of platelet function. Some platelet disorder test findings are highly predictive of a bleeding disorder and some show association to symptoms of prolonged bleeding, surgical bleeding, and wound healing problems. Multiple assays can be required to diagnose common and rare platelet disorders, each requiring control of preanalytical, analytical, and post-analytical variables. The laboratory investigations of platelet disorders include evaluations of platelet counts, size, and morphology by light microscopy; assessments for aggregation defects; tests for dense granule deficiency; analyses of granule constituents and their release; platelet protein analysis by immunofluorescent staining or flow cytometry; tests of platelet procoagulant function; evaluations of platelet ultrastructure; high-throughput sequencing and other molecular diagnostic tests. The focus of this article is to review current methods for the diagnostic assessment of platelet function, with a focus on contemporary, best diagnostic laboratory practices, and relationships between clinical and laboratory findings.
Collapse
Affiliation(s)
- Alex Bourguignon
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - Subia Tasneem
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - Catherine P Hayward
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada.,Department of Medicine, McMaster University, Hamilton, Canada
| |
Collapse
|
44
|
Lassandro G, Palladino V, Faleschini M, Barone A, Boscarol G, Cesaro S, Chiocca E, Farruggia P, Giona F, Gorio C, Maggio A, Marinoni M, Marzollo A, Palumbo G, Russo G, Saracco P, Spinelli M, Verzegnassi F, Morga F, Savoia A, Giordano P. "CHildren with Inherited Platelet disorders Surveillance" (CHIPS) retrospective and prospective observational cohort study by Italian Association of Pediatric Hematology and Oncology (AIEOP). Front Pediatr 2022; 10:967417. [PMID: 36507135 PMCID: PMC9728612 DOI: 10.3389/fped.2022.967417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 10/18/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Inherited thrombocytopenias (ITs) are rare congenital bleeding disorders characterized by different clinical expression and variable prognosis. ITs are poorly known by clinicians and often misdiagnosed with most common forms of thrombocytopenia. MATERIAL AND METHODS "CHildren with Inherited Platelet disorders Surveillance" study (CHIPS) is a retrospective - prospective observational cohort study conducted between January 2003 and January 2022 in 17 centers affiliated to the Italian Association of Pediatric Hematology and Oncology (AIEOP). The primary objective of this study was to collect clinical and laboratory data on Italian pediatric patients with inherited thrombocytopenias. Secondary objectives were to calculate prevalence of ITs in Italian pediatric population and to assess frequency and genotype-phenotype correlation of different types of mutations in our study cohort. RESULTS A total of 139 children, with ITs (82 male - 57 female) were enrolled. ITs prevalence in Italy ranged from 0.7 per 100,000 children during 2010 to 2 per 100,000 children during 2022. The median time between the onset of thrombocytopenia and the diagnosis of ITs was 1 years (range 0 - 18 years). A family history of thrombocytopenia has been reported in 90 patients (65%). Among 139 children with ITs, in 73 (53%) children almost one defective gene has been identified. In 61 patients a pathogenic mutation has been identified. Among them, 2 patients also carry a variant of uncertain significance (VUS), and 4 others harbour 2 VUS variants. VUS variants were identified in further 8 patients (6%), 4 of which carry more than one variant VUS. Three patients (2%) had a likely pathogenic variant while in 1 patient (1%) a variant was identified that was initially given an uncertain significance but was later classified as benign. In addition, in 17 patients the genetic diagnosis is not available, but their family history and clinical/laboratory features strongly suggest the presence of a specific genetic cause. In 49 children (35%) no genetic defect were identified. In ninetyseven patients (70%), thrombocytopenia was not associated with other clinically apparent disorders. However, 42 children (30%) had one or more additional clinical alterations. CONCLUSION Our study provides a descriptive collection of ITs in the pediatric Italian population.
Collapse
Affiliation(s)
- Giuseppe Lassandro
- Interdisciplinary Department of Medicine, Pediatric Section, University of Bari "Aldo Moro", Bari, Italy
| | - Valentina Palladino
- Interdisciplinary Department of Medicine, Pediatric Section, University of Bari "Aldo Moro", Bari, Italy
| | - Michela Faleschini
- Department of Medical Genetics, Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
| | - Angelica Barone
- Pediatric Hematology Oncology, Dipartimento Materno-Infantile, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Gianluca Boscarol
- Department of Pediatrics, Central Teaching Hospital of Bolzano/Bozen, Bolzano, Italy
| | - Simone Cesaro
- Pediatric Hematology Oncology, Department of Mother and Child, Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Elena Chiocca
- Pediatric Hematology Oncology, Department of Pediatric Hematology/Oncology and HSCT, Meyer Children's University Hospital, Florence, Italy
| | - Piero Farruggia
- Pediatric Hematology and Oncology Unit, ARNAS (Azienda di Rilievo Nazionale ad Alta Specializzazione) Ospedale Civico, Palermo, Italy
| | - Fiorina Giona
- Department of Translational and Precision Medicine, Sapienza University of Rome, AOU Policlinico Umberto I, Rome, Italy
| | - Chiara Gorio
- Hematology Oncology Unit, Children's Hospital, ASST Spedali Civili, Brescia, Italy
| | - Angela Maggio
- UOC Oncoematologia Pediatrica-IRCCS Ospedale Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
| | - Maddalena Marinoni
- Pediatric Hematology Oncology, Department of Mother and Child, Azienda Socio Sanitaria Settelaghi, Varese, Italy
| | - Antonio Marzollo
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Padua, Italy
| | - Giuseppe Palumbo
- Department of Pediatric Hematology and Oncology Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Giovanna Russo
- Pediatric Hematology Oncology, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Paola Saracco
- Pediatric Hematology, Department of Pediatrics, University Hospital Città Della Salute e Della Scienza, Turin, Italy
| | - Marco Spinelli
- Pediatric Hematology Oncology, Department of Pediatrics, MBBM Foundation, Monza, Italy
| | - Federico Verzegnassi
- Department of Medical Genetics, Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy
| | - Francesca Morga
- Interdisciplinary Department of Medicine, Pediatric Section, University of Bari "Aldo Moro", Bari, Italy
| | - Anna Savoia
- Department of Medical Genetics, Institute for Maternal and Child Health-IRCCS Burlo Garofolo, Trieste, Italy.,Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Paola Giordano
- Interdisciplinary Department of Medicine, Pediatric Section, University of Bari "Aldo Moro", Bari, Italy
| |
Collapse
|
45
|
Boeckelmann D, Glonnegger H, Sandrock-Lang K, Zieger B. Pathogenic Aspects of Inherited Platelet Disorders. Hamostaseologie 2021; 41:460-468. [PMID: 34942659 DOI: 10.1055/a-1665-6249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Inherited platelet disorders (IPDs) constitute a large heterogeneous group of rare bleeding disorders. These are classified into: (1) quantitative defects, (2) qualitative disorders, or (3) altered platelet production rate disorders or increased platelet turnover. Classically, IPD diagnostic is based on clinical phenotype characterization, comprehensive laboratory analyses (platelet function analysis), and, in former times, candidate gene sequencing. Today, molecular genetic analysis is performed using next-generation sequencing, mostly by targeting enrichment of a gene panel or by whole-exome sequencing. Still, the biochemical and molecular genetic characterization of patients with congenital thrombocytopathias/thrombocytopenia is essential, since postoperative or posttraumatic bleeding often occurs due to undiagnosed platelet defects. Depending upon the kind of surgery or trauma, this bleeding may be life-threatening, e.g., after tonsillectomy or in brain surgery. Undiagnosed platelet defects may lead to additional surgery, hysterectomy, pulmonary bleeding, and even resuscitation. In addition, these increased bleeding symptoms can lead to wound healing problems. Only specialized laboratories can perform the special platelet function analyses (aggregometry, flow cytometry, or immunofluorescent microscopy of the platelets); therefore, many IPDs are still undetected.
Collapse
Affiliation(s)
- Doris Boeckelmann
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| | - Hannah Glonnegger
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| | - Kirstin Sandrock-Lang
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| | - Barbara Zieger
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| |
Collapse
|
46
|
Tawana K, Brown AL, Churpek JE. Integrating germline variant assessment into routine clinical practice for myelodysplastic syndrome and acute myeloid leukaemia: current strategies and challenges. Br J Haematol 2021; 196:1293-1310. [PMID: 34658019 DOI: 10.1111/bjh.17855] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 08/24/2021] [Accepted: 09/12/2021] [Indexed: 12/28/2022]
Abstract
Over the last decade, the field of hereditary haematological malignancy syndromes (HHMSs) has gained increasing recognition among clinicians and scientists worldwide. Germline mutations now account for almost 10% of adult and paediatric myelodysplasia/acute myeloid leukaemia (MDS/AML). As our ability to diagnose HHMSs has improved, we are now faced with the challenges of integrating these advances into routine clinical practice for patients with MDS/AML and how to optimise management and surveillance of patients and asymptomatic carriers. Discoveries of novel syndromes combined with clinical, genetic and epigenetic profiling of tumour samples, have highlighted unique patterns of disease evolution across HHMSs. Despite these advances, causative lesions are detected in less than half of familial cases and evidence-based guidelines are often lacking, suggesting there is much still to learn. Future research efforts are needed to sustain current momentum within the field, led not only by advancing genetic technology but essential collaboration between clinical and academic communities.
Collapse
Affiliation(s)
- Kiran Tawana
- Department of Haematology, Addenbrooke's Hospital, Cambridge, UK
| | - Anna L Brown
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia.,Centre for Cancer Biology, SA Pathology, University of South Australia, Adelaide, SA, Australia.,Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Jane E Churpek
- Division of Hematology, Medical Oncology, and Palliative Care, Department of Medicine, School of Medicine and Public Health, The University of Wisconsin, Madison, WI, USA
| |
Collapse
|
47
|
Andrés‐Zayas C, Suárez‐González J, Rodríguez‐Macías G, Dorado N, Osorio S, Font P, Carbonell D, Chicano M, Muñiz P, Bastos M, Kwon M, Díez‐Martín JL, Buño I, Martínez‐Laperche C. Clinical utility of targeted next-generation sequencing for the diagnosis of myeloid neoplasms with germline predisposition. Mol Oncol 2021; 15:2273-2284. [PMID: 33533142 PMCID: PMC8410541 DOI: 10.1002/1878-0261.12921] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 12/30/2020] [Accepted: 01/11/2021] [Indexed: 12/30/2022] Open
Abstract
Myeloid neoplasms (MN) with germline predisposition (MNGP) are likely to be more common than currently appreciated. Many of the genes involved in MNGP are also recurrently mutated in sporadic MN. Therefore, routine analysis of gene panels by next-generation sequencing provides an effective approach to detect germline variants with clinical significance in patients with hematological malignancies. Gene panel sequencing was performed in 88 consecutive and five nonconsecutive patients with MN diagnosis. Disease-causing germline mutations in CEBPα, ASXL1, TP53, MPL, GATA2, DDX41, and ETV6 genes were identified in nine patients. Six out of the nine patients with germline variants had a strong family history. These patients presented great heterogeneity in the age of diagnosis and phenotypic characteristics. In our study, there were families in which all the affected members presented the same subtype of disease, whereas members of other families presented various disease phenotypes. This intrafamiliar heterogeneity suggests that the acquisition of particular somatic variants may drive the evolution of the disease. This approach enabled high-throughput detection of MNGP in patients with MN diagnosis, which is of great relevance for both the patients themselves and the asymptomatic mutation carriers within the family. It is crucial to make a proper diagnosis of these patients to provide them with the most suitable treatment, follow-up, and genetic counseling.
Collapse
Affiliation(s)
- Cristina Andrés‐Zayas
- Genomics UnitGregorio Marañón General University HospitalGregorio Marañón Health Research Institute (IiSGM)MadridSpain
- Gregorio Marañón Health Research Institute (IiSGM)MadridSpain
| | - Julia Suárez‐González
- Genomics UnitGregorio Marañón General University HospitalGregorio Marañón Health Research Institute (IiSGM)MadridSpain
- Gregorio Marañón Health Research Institute (IiSGM)MadridSpain
| | | | - Nieves Dorado
- Gregorio Marañón Health Research Institute (IiSGM)MadridSpain
- Department of HematologyGregorio Marañón General University HospitalMadridSpain
| | - Santiago Osorio
- Gregorio Marañón Health Research Institute (IiSGM)MadridSpain
- Department of HematologyGregorio Marañón General University HospitalMadridSpain
| | - Patricia Font
- Gregorio Marañón Health Research Institute (IiSGM)MadridSpain
- Department of HematologyGregorio Marañón General University HospitalMadridSpain
| | - Diego Carbonell
- Gregorio Marañón Health Research Institute (IiSGM)MadridSpain
- Department of HematologyGregorio Marañón General University HospitalMadridSpain
| | - María Chicano
- Gregorio Marañón Health Research Institute (IiSGM)MadridSpain
- Department of HematologyGregorio Marañón General University HospitalMadridSpain
| | - Paula Muñiz
- Gregorio Marañón Health Research Institute (IiSGM)MadridSpain
- Department of HematologyGregorio Marañón General University HospitalMadridSpain
| | - Mariana Bastos
- Gregorio Marañón Health Research Institute (IiSGM)MadridSpain
- Department of HematologyGregorio Marañón General University HospitalMadridSpain
| | - Mi Kwon
- Gregorio Marañón Health Research Institute (IiSGM)MadridSpain
- Department of HematologyGregorio Marañón General University HospitalMadridSpain
| | - José Luis Díez‐Martín
- Gregorio Marañón Health Research Institute (IiSGM)MadridSpain
- Department of HematologyGregorio Marañón General University HospitalMadridSpain
- Department of MedicineSchool of MedicineComplutense University of MadridSpain
| | - Ismael Buño
- Genomics UnitGregorio Marañón General University HospitalGregorio Marañón Health Research Institute (IiSGM)MadridSpain
- Gregorio Marañón Health Research Institute (IiSGM)MadridSpain
- Department of HematologyGregorio Marañón General University HospitalMadridSpain
- Department of Cell BiologySchool of MedicineComplutense University of MadridSpain
| | - Carolina Martínez‐Laperche
- Gregorio Marañón Health Research Institute (IiSGM)MadridSpain
- Department of HematologyGregorio Marañón General University HospitalMadridSpain
| |
Collapse
|
48
|
Tsai FD, Battinelli EM. Inherited Platelet Disorders. Hematol Oncol Clin North Am 2021; 35:1069-1084. [PMID: 34391603 DOI: 10.1016/j.hoc.2021.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Bleeding disorders due to platelet dysfunction are a common hematologic complication affecting patients, and typically present with mucocutaneous bleeding or hemorrhage. An inherited platelet disorder should be suspected in individuals with a suggestive family history and no identified secondary causes of bleeding. Genetic defects have been described at all levels of platelet activation, including receptor binding, signaling, granule release, cytoskeletal remodeling, and platelet hematopoiesis. Management of these disorders is typically supportive, with an emphasis on awareness, patient education, and anticipatory guidance to prevent future episodes of bleeding.
Collapse
Affiliation(s)
- Frederick D Tsai
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115, USA; Division of Hematologic Neoplasia, Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA; Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA
| | - Elisabeth M Battinelli
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, 75 Francis St, Boston, MA 02115, USA; Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA.
| |
Collapse
|
49
|
Zarubina KI, Parovichnikova EN, Surin VL, Pshenichnikova OS, Gavrilina OA, Isinova GA, Troitskaya VV, Sokolov AN, Galtseva IV, Kapranov NM, Davydova JO, Obukhova TN, Nikulina EE, Sudarikov AB, Savchenko VG. Li–Fraumeni syndrome in adult patients with acute lymphoblastic leukemia. TERAPEVT ARKH 2021; 93:763-769. [DOI: 10.26442/00403660.2021.07.200913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 07/20/2021] [Indexed: 11/22/2022]
Abstract
Background. LiFraumeni syndrome (LFS) is a rare, autosomal dominant, hereditary disorder that is characterized by an increased risk for certain types of cancer, acute lymphoblastic leukemia (ALL), particularly. Germline TP53 mutations are associated with LFS. Genetic counseling and follow-up is essential for patients with LFS and their relatives. Special therapeutic approaches are needed for treatment of oncological disease in these patients. The article presents a series of clinical cases of patients with ALL and SLF, considers general issues of diagnosis and treatment of adult patients with this hereditary genetic syndrome.
Aim. Describe clinical observations of patients with acute lymphoblastic leukemia (ALL) and LFS and consider general issues of diagnosis and treatment of adult patients with LFS and ALL.
Materials and methods. TP53 gene mutations were screened using Sanger sequencing in 180 de novo patients with Ph-negative (B- and T-cell) and Ph-positive ALL treated by Russian multicenter protocols (ALL-2009, ALL-2012, ALL-2016) at the National Research Center for Hematology, Moscow, Russia, and at the hematology departments of regional clinics of Russia (multicenter study participants).
Results. TP53 gene mutations were found in 7.8% (n=14) of de novo ALL patients. In patients, whose biological material was available TP53 gene mutational status was determined in non-tumor cells (bone marrow and peripheral blood during remission, bone marrow samples after allogeneic hematopoietic stem cells transplantation and in tissue of non-hematopoietic origin) for discriminating germline mutations. The analysis included 5 patients (out of 14 with TP53 mutations), whose non-tumor biological material was available for research. Germline status was confirmed in 4 out of 5 B-cell ALL (n=3), T-cell ALL (n=1) investigated patients.
Conclusion. Practical value of the research is the observation that the greater part of TP53 gene mutations in patients with Ph-negative B-cell ALL are germinal and associated with LFS.
Collapse
|
50
|
Abstract
The genetic basis for pediatric acute myeloid leukemia (AML) is highly heterogeneous, often involving the cooperative action of characteristic chromosomal rearrangements and somatic mutations in progrowth and antidifferentiation pathways that drive oncogenesis. Although some driver mutations are shared with adult AML, many genetic lesions are unique to pediatric patients, and their appropriate identification is essential for patient care. The increased understanding of these malignancies through broad genomic studies has begun to risk-stratify patients based on their combinations of genomic alterations, a trend that will enable precision medicine in this population.
Collapse
Affiliation(s)
- Bryan Krock
- Caris Life Sciences, 4610 South 44th Place, Phoenix, AZ, USA
| | | |
Collapse
|