1
|
McConville M, Thomas T, Beckner R, Kroger B, Valadez C, Chook YM, Chung S, Liszczak G. A germline ETV6 mutation disrupts hematopoiesis via de novo creation of a nuclear export signal. SCIENCE ADVANCES 2025; 11:eadu4058. [PMID: 40238870 PMCID: PMC12002123 DOI: 10.1126/sciadv.adu4058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 03/10/2025] [Indexed: 04/18/2025]
Abstract
Germline mutations in the transcriptional regulator ETV6 are a root cause of familial inherited thrombocytopenia and predispose carriers to myelodysplastic syndromes and acute leukemias. Here, we report that the ETV6 P214L mutation creates an XPO1-dependent nuclear export signal to cause protein mislocalization. Strategies to disrupt XPO1 nuclear export activity fully restore ETV6 P214L protein nuclear localization and transcription regulation activity, establishing XPO1-dependent mislocalization as a critical mechanism underscoring ETV6 P214L dysfunction. Mechanistic insight inspired the design of "humanized" ETV6 mice in which the germline P214L mutation is sufficient to elicit severe defects in thrombopoiesis and hematopoietic stem cell maintenance and survival in animals. These studies define a unique mechanism by which the ETV6 P214L mutation exerts a dominant negative effect on protein function and reveal critical mutation-dependent disruptions to hematopoiesis that underlie disease phenotypes.
Collapse
Affiliation(s)
- Michael McConville
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Dallas, TX, USA
| | - Toby Thomas
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ryan Beckner
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Dallas, TX, USA
| | - Benjamin Kroger
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Catherine Valadez
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Dallas, TX, USA
| | - Yuh Min Chook
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Stephen Chung
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Glen Liszczak
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Dallas, TX, USA
| |
Collapse
|
2
|
Ammeti D, De Simone T, Zampieri S, Bon C, Zanchetta ME, Bottega R, Faleschini M, Savoia A. Functional Testing of ETV6 Variants: Is the Evaluation of Their Intracellular Localization Sufficient in Assessing Pathogenicity? Genes (Basel) 2025; 16:112. [PMID: 40004441 PMCID: PMC11854649 DOI: 10.3390/genes16020112] [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: 12/06/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES ETV6-related thrombocytopenia (ETV6-RT) is a rare autosomal dominant disorder characterized by mild thrombocytopenia since birth and an increased predisposition to hematologic malignancies. ETV6 functions as a transcriptional repressor, and its pathogenic variants, predominantly within the ETS domain, disrupt nuclear localization and transcriptional activity. In individuals with congenital thrombocytopenia, we identified two missense variants: c.1110A>G, p.Ile370Met, a novel variant, and c.1133G>A, p.Arg378Gln, a known variant with conflicting pathogenicity interpretations, for which functional characterization is necessary to provide an accurate molecular diagnosis. METHODS In silico bioinformatic tools and structural modeling were used to predict the impact of the variants. Functional assays included a dual-luciferase reporter assay to measure transcriptional activity and immunofluorescence/immunoblotting to assess intracellular localization in transfected HEK293T and HeLa cells. RESULTS Bioinformatic predictions and structural analyses suggested that the two variants might play a role in altering the folding or function of the ETS domain. Functional analysis revealed that p.Ile370Met abolished the ETV6 transcriptional repression activity, confirming its pathogenicity. p.Arg378Gln had no effect on the reporter gene levels, and, as expected, it localized in the nucleus. Interestingly, unlike the known mutations, which fail to enter the nucleus, p.Ile370Met retained partial nuclear localization. CONCLUSIONS Since we described the first ETV6 mutation localized in the ETS domain that causes a loss of transcriptional activity, although it maintains the ability to enter the nucleus, we suggest that both transcriptional activity and intracellular localization assays are important for the accurate classification of ETV6 variants by functional studies.
Collapse
Affiliation(s)
- Daniele Ammeti
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy; (D.A.); (S.Z.); (C.B.); (M.E.Z.); (R.B.)
| | - Tonia De Simone
- Department of Engineering for Innovation Medicine, University of Verona, 37134 Verona, Italy;
| | - Stefania Zampieri
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy; (D.A.); (S.Z.); (C.B.); (M.E.Z.); (R.B.)
| | - Cristina Bon
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy; (D.A.); (S.Z.); (C.B.); (M.E.Z.); (R.B.)
| | - Melania Eva Zanchetta
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy; (D.A.); (S.Z.); (C.B.); (M.E.Z.); (R.B.)
| | - Roberta Bottega
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy; (D.A.); (S.Z.); (C.B.); (M.E.Z.); (R.B.)
| | - Michela Faleschini
- Institute for Maternal and Child Health, IRCCS Burlo Garofolo, 34137 Trieste, Italy; (D.A.); (S.Z.); (C.B.); (M.E.Z.); (R.B.)
| | - Anna Savoia
- Department of Engineering for Innovation Medicine, University of Verona, 37134 Verona, Italy;
| |
Collapse
|
3
|
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
|
4
|
Zhang S, Gao H, You G, Cao H, Wang Y, Gao L, Zheng SJ. A novel role of ETV6 as a pro-viral factor in host response by inhibiting TBK1 phosphorylation. Int J Biol Macromol 2024; 279:135525. [PMID: 39260650 DOI: 10.1016/j.ijbiomac.2024.135525] [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: 07/14/2024] [Revised: 08/23/2024] [Accepted: 09/08/2024] [Indexed: 09/13/2024]
Abstract
E26-transforming specific (ETS) variant 6 (ETV6) is a transcription factor regulating the expression of interferon stimulating genes (ISGs) and involved in the embryonic development and hematopoietic regulation, but the role of ETV6 in host response to virus infection is not clear. In this study, we show that ETV6 was upregulated in DF-1 cells with poly(I:C) stimulation or IBDV, AIV and ARV infection via engagement of dsRNA by MDA5. Overexpression of ETV6 in DF-1 cells markedly inhibited IBDV-induced type I interferon (IFN-I) and ISGs expressions. In contrast, knockdown, or knockout of ETV6 remarkably inhibited IBDV replication via promoting IFN-I response. Furthermore, our data show that ETV6 negatively regulated host antiviral response to IBDV infection by interaction with TANK binding kinase 1 (TBK1) and subsequently inhibited its phosphorylation. These results uncovered a novel role of ETV6 as a pro-viral factor in host response by inhibiting TBK1 phosphorylation, furthering our understandings of RNA virus immunosuppression and providing a valuable clue to the development of antiviral reagents for the control of avian RNA virus infection.
Collapse
Affiliation(s)
- Shujun Zhang
- National Key Laboratory of Veterinary Public Health Security, China; Animal Epidemiology of the Ministry of Agriculture, China; College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Hui Gao
- National Key Laboratory of Veterinary Public Health Security, China; Animal Epidemiology of the Ministry of Agriculture, China; College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Guangju You
- Laboratory of Animal Virology, Institute of Animal Husbandry and Veterinary Medicine, Fujian Academy of Agriculture Sciences, Fuzhou, China
| | - Hong Cao
- National Key Laboratory of Veterinary Public Health Security, China; Animal Epidemiology of the Ministry of Agriculture, China; College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yongqiang Wang
- National Key Laboratory of Veterinary Public Health Security, China; Animal Epidemiology of the Ministry of Agriculture, China; College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Li Gao
- National Key Laboratory of Veterinary Public Health Security, China; Animal Epidemiology of the Ministry of Agriculture, China; College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| | - Shijun J Zheng
- National Key Laboratory of Veterinary Public Health Security, China; Animal Epidemiology of the Ministry of Agriculture, China; College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
5
|
McConville BM, Thomas T, Beckner R, Valadez C, Chook Y, Chung S, Liszczak G. Enigmatic missense mutations can cause disease via creation of de novo nuclear export signals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.590854. [PMID: 38712034 PMCID: PMC11071533 DOI: 10.1101/2024.04.24.590854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Disease-causing missense mutations that occur within structurally and functionally unannotated protein regions can guide researchers to new mechanisms of protein regulation and dysfunction. Here, we report that the thrombocytopenia-, myelodysplastic syndromes-, and leukemia-associated P214L mutation in the transcriptional regulator ETV6 creates an XPO1-dependent nuclear export signal to cause protein mislocalization. Strategies to disrupt XPO1 activity fully restore ETV6 P214L protein nuclear localization and transcription regulation activity. Mechanistic insight inspired the design of a 'humanized' ETV6 mice, which we employ to demonstrate that the germline P214L mutation is sufficient to elicit severe defects in thrombopoiesis and hematopoietic stem cell maintenance. Beyond ETV6, we employed computational methods to uncover rare disease-associated missense mutations in unrelated proteins that create a nuclear export signal to disrupt protein function. Thus, missense mutations that operate through this mechanism should be predictable and may suggest rational therapeutic strategies for associated diseases.
Collapse
|
6
|
Singh M, Sharma P, Bhatia P, Trehan A, Thakur R, Sreedharanunni S. Integrated analysis of transcriptome and genome variations in pediatric T cell acute lymphoblastic leukemia: data from north Indian tertiary care center. BMC Cancer 2024; 24:325. [PMID: 38459434 PMCID: PMC10924344 DOI: 10.1186/s12885-024-12063-6] [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: 11/23/2023] [Accepted: 02/26/2024] [Indexed: 03/10/2024] Open
Abstract
INTRODUCTION T-cell acute lymphoblastic leukemia (T-ALL) is a genetically heterogeneous disease with poor prognosis and inferior outcome. Although multiple studies have been perform on genomics of T-ALL, data from Indian sub-continent is scarce. METHODS In the current study we aimed to identify the genetic variability of T-ALL in an Indian cohort of pediatric (age ≤ 12 years) T-ALL patients (n = 25) by whole transcriptome sequencing along with whole exome sequencing and correlated the findings with clinical characteristics and disease outcome. RESULTS The median age was 7 years (range 3 -12 years). RNA sequencing revealed a definitive fusion event in 14 cases (56%) (including a novel fusions) with STIL::TAL1 in 4 (16%), followed by NUP21::ABL1, TCF7::SPI1, ETV6::HDAC8, LMO1::RIC3, DIAPH1::JAK2, SETD2::CCDC12 and RCBTB2::LPAR6 in 1 (4%) case each. Significant aberrant expression was noted in RAG1 (64%), RAG2 (80%), MYCN (52%), NKX3-1 (52%), NKX3-2 (32%), TLX3 (28%), LMO1 (20%) and MYB (16%) genes. WES data showed frequent mutations in NOTCH1 (35%) followed by WT1 (23%), FBXW7 (12%), KRAS (12%), PHF6 (12%) and JAK3 (12%). Nearly 88.2% of cases showed a deletion of CDKN2A/CDKN2B/MTAP genes. Clinically significant association of a better EFS and OS (p=0.01) was noted with RAG2 over-expression at a median follow up of 22 months, while a poor EFS (p=0.041) and high relapse rate (p=0.045) was observed with MYB over-expression. CONCLUSION Overall, the present study demonstrates the frequencies of transcriptomic and genetic alterations from Indian cohort of pediatric T-ALL and is a salient addition to current genomics data sets available in T-ALL.
Collapse
Affiliation(s)
- Minu Singh
- Haematology-Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Sector -12, 160012, Chandigarh, India.
| | - Pankaj Sharma
- Haematology-Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Sector -12, 160012, Chandigarh, India
| | - Prateek Bhatia
- Haematology-Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Sector -12, 160012, Chandigarh, India
| | - Amita Trehan
- Haematology-Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Sector -12, 160012, Chandigarh, India
| | - Rozy Thakur
- Haematology-Oncology Unit, Department of Paediatrics, Postgraduate Institute of Medical Education and Research, Sector -12, 160012, Chandigarh, India
| | - Sreejesh Sreedharanunni
- Department of Haematology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
7
|
He F, Laranjeira AB, Kong T, Lin S, Ashworth KJ, Liu A, Lasky NM, Fisher DA, Cox MJ, Fulbright MC, Antunes-Heck L, Yu L, Brakhane M, Gao B, Sykes SM, D’Alessandro A, Di Paola J, Oh ST. Multiomic profiling reveals metabolic alterations mediating aberrant platelet activity and inflammation in myeloproliferative neoplasms. J Clin Invest 2024; 134:e172256. [PMID: 38060311 PMCID: PMC10836808 DOI: 10.1172/jci172256] [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: 05/12/2023] [Accepted: 12/06/2023] [Indexed: 02/02/2024] Open
Abstract
Platelets from patients with myeloproliferative neoplasms (MPNs) exhibit a hyperreactive phenotype. Here, we found elevated P-selectin exposure and platelet-leukocyte aggregates indicating activation of platelets from essential thrombocythemia (ET) patients. Single-cell RNA-seq analysis of primary samples revealed significant enrichment of transcripts related to platelet activation, mTOR, and oxidative phosphorylation in ET patient platelets. These observations were validated via proteomic profiling. Platelet metabolomics revealed distinct metabolic phenotypes consisting of elevated ATP generation accompanied by increases in the levels of multiple intermediates of the tricarboxylic acid cycle, but lower α-ketoglutarate (α-KG) in MPN patients. Inhibition of PI3K/AKT/mTOR signaling significantly reduced metabolic responses and hyperreactivity in MPN patient platelets, while α-KG supplementation markedly reduced oxygen consumption and ATP generation. Ex vivo incubation of platelets from both MPN patients and Jak2 V617F-knockin mice with α-KG supplementation significantly reduced platelet activation responses. Oral α-KG supplementation of Jak2 V617F mice decreased splenomegaly and reduced hematocrit, monocyte, and platelet counts. Finally, α-KG treatment significantly decreased proinflammatory cytokine secretion from MPN CD14+ monocytes. Our results reveal a previously unrecognized metabolic disorder in conjunction with aberrant PI3K/AKT/mTOR signaling that contributes to platelet hyperreactivity in MPN patients.
Collapse
Affiliation(s)
- Fan He
- Division of Hematology, Department of Medicine, and
| | | | - Tim Kong
- Division of Hematology, Department of Medicine, and
| | - Shuyang Lin
- Division of Hematology, Department of Medicine, and
| | - Katrina J. Ashworth
- Division of Hematology & Oncology, Department of Pediatrics, School of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Alice Liu
- Division of Hematology & Oncology, Department of Pediatrics, School of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nina M. Lasky
- Division of Hematology & Oncology, Department of Pediatrics, School of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | | | - Lilian Antunes-Heck
- Division of Hematology & Oncology, Department of Pediatrics, School of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - LaYow Yu
- Division of Hematology, Department of Medicine, and
| | | | - Bei Gao
- Division of Hematology & Oncology, Department of Pediatrics, School of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Stephen M. Sykes
- Division of Hematology & Oncology, Department of Pediatrics, School of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jorge Di Paola
- Division of Hematology & Oncology, Department of Pediatrics, School of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Stephen T. Oh
- Division of Hematology, Department of Medicine, and
- Immunomonitoring Laboratory, Center for Human Immunology and Immunotherapy Programs, and
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
8
|
Martinez TC, McNerney ME. Haploinsufficient Transcription Factors in Myeloid Neoplasms. ANNUAL REVIEW OF PATHOLOGY 2024; 19:571-598. [PMID: 37906947 DOI: 10.1146/annurev-pathmechdis-051222-013421] [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: 11/02/2023]
Abstract
Many transcription factors (TFs) function as tumor suppressor genes with heterozygous phenotypes, yet haploinsufficiency generally has an underappreciated role in neoplasia. This is no less true in myeloid cells, which are normally regulated by a delicately balanced and interconnected transcriptional network. Detailed understanding of TF dose in this circuitry sheds light on the leukemic transcriptome. In this review, we discuss the emerging features of haploinsufficient transcription factors (HITFs). We posit that: (a) monoallelic and biallelic losses can have distinct cellular outcomes; (b) the activity of a TF exists in a greater range than the traditional Mendelian genetic doses; and (c) how a TF is deleted or mutated impacts the cellular phenotype. The net effect of a HITF is a myeloid differentiation block and increased intercellular heterogeneity in the course of myeloid neoplasia.
Collapse
Affiliation(s)
- Tanner C Martinez
- Department of Pathology, Department of Pediatrics, Section of Hematology/Oncology, The University of Chicago Medicine Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois, USA;
- Medical Scientist Training Program, The University of Chicago, Chicago, Illinois, USA
| | - Megan E McNerney
- Department of Pathology, Department of Pediatrics, Section of Hematology/Oncology, The University of Chicago Medicine Comprehensive Cancer Center, The University of Chicago, Chicago, Illinois, USA;
| |
Collapse
|
9
|
Monovich AC, Gurumurthy A, Ryan RJH. The Diverse Roles of ETV6 Alterations in B-Lymphoblastic Leukemia and Other Hematopoietic Cancers. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1459:291-320. [PMID: 39017849 DOI: 10.1007/978-3-031-62731-6_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Genetic alterations of the repressive ETS family transcription factor gene ETV6 are recurrent in several categories of hematopoietic malignancy, including subsets of B-cell and T-cell acute lymphoblastic leukemias (B-ALL and T-ALL), myeloid neoplasms, and mature B-cell lymphomas. ETV6 is essential for adult hematopoietic stem cells (HSCs), contributes to specific functions of some mature immune cells, and plays a key role in thrombopoiesis as demonstrated by familial ETV6 mutations associated with thrombocytopenia and predisposition to hematopoietic cancers, particularly B-ALL. ETV6 appears to have a tumor suppressor role in several hematopoietic lineages, as demonstrated by recurrent somatic loss-of-function (LoF) and putative dominant-negative alterations in leukemias and lymphomas. ETV6 rearrangements contribute to recurrent fusion oncogenes such as the B-ALL-associated transcription factor (TF) fusions ETV6::RUNX1 and PAX5::ETV6, rare drivers such as ETV6::NCOA6, and a spectrum of tyrosine kinase gene fusions encoding hyperactive signaling proteins that self-associate via the ETV6 N-terminal pointed domain. Another subset of recurrent rearrangements involving the ETV6 gene locus appear to function primarily to drive overexpression of the partner gene. This review surveys what is known about the biochemical and genome regulatory properties of ETV6 as well as our current understanding of how alterations in these functions contribute to hematopoietic and nonhematopoietic cancers.
Collapse
Affiliation(s)
- Alexander C Monovich
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Aishwarya Gurumurthy
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Russell J H Ryan
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
10
|
Bloom M, Oak N, Baskin-Doerfler R, Feng R, Iacobucci I, Baviskar P, Zhao X, Stroh AN, Li C, Ozark P, Tillman HS, Li Y, Verbist KC, Albeituni S, Scott DC, King MT, McKinney-Freeman SL, Weiss MJ, Yang JJ, Nichols KE. ETV6 represses inflammatory response genes and regulates HSPC function during stress hematopoiesis in mice. Blood Adv 2023; 7:5608-5623. [PMID: 37522715 PMCID: PMC10514086 DOI: 10.1182/bloodadvances.2022009313] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 07/14/2023] [Accepted: 07/19/2023] [Indexed: 08/01/2023] Open
Abstract
ETS variant 6 (ETV6) encodes a transcriptional repressor expressed in hematopoietic stem and progenitor cells (HSPCs), where it is required for adult hematopoiesis. Heterozygous pathogenic germline ETV6 variants are associated with thrombocytopenia 5 (T5), a poorly understood genetic condition resulting in thrombocytopenia and predisposition to hematologic malignancies. To elucidate how germline ETV6 variants affect HSPCs and contribute to disease, we generated a mouse model harboring an Etv6R355X loss-of-function variant, equivalent to the T5-associated variant ETV6R359X. Under homeostatic conditions, all HSPC subpopulations are present in the bone marrow (BM) of Etv6R355X/+ mice; however, these animals display shifts in the proportions and/or numbers of progenitor subtypes. To examine whether the Etv6R355X/+ mutation affects HSPC function, we performed serial competitive transplantation and observed that Etv6R355X/+ lineage-sca1+cKit+ (LSK) cells exhibit impaired reconstitution, with near complete failure to repopulate irradiated recipients by the tertiary transplant. Mechanistic studies incorporating cleavage under target and release under nuclease assay, assay for transposase accessible chromatin sequencing, and high-throughput chromosome conformation capture identify ETV6 binding at inflammatory gene loci, including multiple genes within the tumor necrosis factor (TNF) signaling pathway in ETV6-sufficient mouse and human HSPCs. Furthermore, single-cell RNA sequencing of BM cells isolated after transplantation reveals upregulation of inflammatory genes in Etv6R355X/+ progenitors when compared to Etv6+/+ counterparts. Corroborating these findings, Etv6R355X/+ HSPCs produce significantly more TNF than Etv6+/+ cells post-transplantation. We conclude that ETV6 is required to repress inflammatory gene expression in HSPCs under conditions of hematopoietic stress, and this mechanism may be critical to sustain HSPC function.
Collapse
Affiliation(s)
- Mackenzie Bloom
- St. Jude Graduate School of Biomedical Sciences, Memphis, TN
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Ninad Oak
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | | | - Ruopeng Feng
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Ilaria Iacobucci
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Pradyumna Baviskar
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Xujie Zhao
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN
| | - Alexa N. Stroh
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Chunliang Li
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Patrick Ozark
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Heather S. Tillman
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Yichao Li
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | | | - Sabrin Albeituni
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Danny C. Scott
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Moeko T. King
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN
| | | | - Mitchell J. Weiss
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Jun J. Yang
- Department of Pharmaceutical Sciences, St. Jude Children’s Research Hospital, Memphis, TN
| | - Kim E. Nichols
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
11
|
Camacho V. Translate or differentiate? Molecular mechanisms of ETV6-related thrombocytopenia. J Thromb Haemost 2023; 21:2367-2369. [PMID: 37597896 DOI: 10.1016/j.jtha.2023.04.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 04/27/2023] [Accepted: 04/29/2023] [Indexed: 08/21/2023]
Affiliation(s)
- Virginia Camacho
- Vascular Biology Program, Boston Children's Hospital, Boston, Massachusetts, USA; Department of Surgery, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
12
|
Zerella JR, Homan CC, Arts P, Brown AL, Scott HS, Hahn CN. Transcription factor genetics and biology in predisposition to bone marrow failure and hematological malignancy. Front Oncol 2023; 13:1183318. [PMID: 37377909 PMCID: PMC10291195 DOI: 10.3389/fonc.2023.1183318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
Transcription factors (TFs) play a critical role as key mediators of a multitude of developmental pathways, with highly regulated and tightly organized networks crucial for determining both the timing and pattern of tissue development. TFs can act as master regulators of both primitive and definitive hematopoiesis, tightly controlling the behavior of hematopoietic stem and progenitor cells (HSPCs). These networks control the functional regulation of HSPCs including self-renewal, proliferation, and differentiation dynamics, which are essential to normal hematopoiesis. Defining the key players and dynamics of these hematopoietic transcriptional networks is essential to understanding both normal hematopoiesis and how genetic aberrations in TFs and their networks can predispose to hematopoietic disease including bone marrow failure (BMF) and hematological malignancy (HM). Despite their multifaceted and complex involvement in hematological development, advances in genetic screening along with elegant multi-omics and model system studies are shedding light on how hematopoietic TFs interact and network to achieve normal cell fates and their role in disease etiology. This review focuses on TFs which predispose to BMF and HM, identifies potential novel candidate predisposing TF genes, and examines putative biological mechanisms leading to these phenotypes. A better understanding of the genetics and molecular biology of hematopoietic TFs, as well as identifying novel genes and genetic variants predisposing to BMF and HM, will accelerate the development of preventative strategies, improve clinical management and counseling, and help define targeted treatments for these diseases.
Collapse
Affiliation(s)
- Jiarna R. Zerella
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
| | - Claire C. Homan
- 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
| | - Peer Arts
- 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
| | - Anna L. Brown
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- 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
| | - Hamish S. Scott
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- 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
| | - Christopher N. Hahn
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
- 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
| |
Collapse
|
13
|
Wang Y, Huang Z, Sun M, Huang W, Xia L. ETS transcription factors: Multifaceted players from cancer progression to tumor immunity. Biochim Biophys Acta Rev Cancer 2023; 1878:188872. [PMID: 36841365 DOI: 10.1016/j.bbcan.2023.188872] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/18/2023] [Accepted: 01/28/2023] [Indexed: 02/26/2023]
Abstract
The E26 transformation specific (ETS) family comprises 28 transcription factors, the majority of which are involved in tumor initiation and development. Serving as a group of functionally heterogeneous gene regulators, ETS factors possess a structurally conserved DNA-binding domain. As one of the most prominent families of transcription factors that control diverse cellular functions, ETS activation is modulated by multiple intracellular signaling pathways and post-translational modifications. Disturbances in ETS activity often lead to abnormal changes in oncogenicity, including cancer cell survival, growth, proliferation, metastasis, genetic instability, cell metabolism, and tumor immunity. This review systematically addresses the basics and advances in studying ETS factors, from their tumor relevance to clinical translational utility, with a particular focus on elucidating the role of ETS family in tumor immunity, aiming to decipher the vital role and clinical potential of regulation of ETS factors in the cancer field.
Collapse
Affiliation(s)
- Yufei Wang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Zhao Huang
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei 430030, China
| | - Mengyu Sun
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Wenjie Huang
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei 430030, China.
| | - Limin Xia
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China.
| |
Collapse
|
14
|
Lu DY, Ellegast JM, Ross KN, Malone CF, Lin S, Mabe NW, Dharia NV, Meyer A, Conway A, Su AH, Selich-Anderson J, Taslim C, Byrum AK, Seong BKA, Adane B, Gray NS, Rivera MN, Lessnick SL, Stegmaier K. The ETS transcription factor ETV6 constrains the transcriptional activity of EWS-FLI to promote Ewing sarcoma. Nat Cell Biol 2023; 25:285-297. [PMID: 36658220 PMCID: PMC9928584 DOI: 10.1038/s41556-022-01059-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 11/24/2022] [Indexed: 01/21/2023]
Abstract
Transcription factors (TFs) are frequently mutated in cancer. Paediatric cancers exhibit few mutations genome-wide but frequently harbour sentinel mutations that affect TFs, which provides a context to precisely study the transcriptional circuits that support mutant TF-driven oncogenesis. A broadly relevant mechanism that has garnered intense focus involves the ability of mutant TFs to hijack wild-type lineage-specific TFs in self-reinforcing transcriptional circuits. However, it is not known whether this specific type of circuitry is equally crucial in all mutant TF-driven cancers. Here we describe an alternative yet central transcriptional mechanism that promotes Ewing sarcoma, wherein constraint, rather than reinforcement, of the activity of the fusion TF EWS-FLI supports cancer growth. We discover that ETV6 is a crucial TF dependency that is specific to this disease because it, counter-intuitively, represses the transcriptional output of EWS-FLI. This work discovers a previously undescribed transcriptional mechanism that promotes cancer.
Collapse
Affiliation(s)
- Diana Y Lu
- Harvard/MIT MD-PhD Program, Harvard Medical School, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jana M Ellegast
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kenneth N Ross
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Clare F Malone
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shan Lin
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nathaniel W Mabe
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Neekesh V Dharia
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ashleigh Meyer
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Amy Conway
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Angela H Su
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Julia Selich-Anderson
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Cenny Taslim
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Andrea K Byrum
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Bo Kyung A Seong
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Biniam Adane
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Miguel N Rivera
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Stephen L Lessnick
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Division of Pediatric Hematology, Oncology and BMT, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA.
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
15
|
Kaczmarska A, Derebas J, Pinkosz M, Niedźwiecki M, Lejman M. The Landscape of Secondary Genetic Rearrangements in Pediatric Patients with B-Cell Acute Lymphoblastic Leukemia with t(12;21). Cells 2023; 12:cells12030357. [PMID: 36766699 PMCID: PMC9913634 DOI: 10.3390/cells12030357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
The most frequent chromosomal rearrangement in childhood B-cell acute lymphoblastic leukemia (B-ALL) is translocation t(12;21)(p13;q22). It results in the fusion of the ETV6::RUNX1 gene, which is active in the regulation of multiple crucial cellular pathways. Recent studies hypothesize that many translocations are influenced by RAG-initiated deletions, as well as defects in the RAS and NRAS pathways. According to a "two-hit" model for the molecular pathogenesis of pediatric ETV6::RUNX1-positive B-ALL, the t(12;21) translocation requires leukemia-causing secondary mutations. Patients with ETV6::RUNX1 express up to 60 different aberrations, which highlights the heterogeneity of this B-ALL subtype and is reflected in differences in patient response to treatment and chances of relapse. Most studies of secondary genetic changes have concentrated on deletions of the normal, non-rearranged ETV6 allele. Other predominant structural changes included deletions of chromosomes 6q and 9p, loss of entire chromosomes X, 8, and 13, duplications of chromosome 4q, or trisomy of chromosomes 21 and 16, but the impact of these changes on overall survival remains unclarified. An equally genetically diverse group is the recently identified new B-ALL subtype ETV6::RUNX1-like ALL. In our review, we provide a comprehensive description of recurrent secondary mutations in pediatric B-ALL with t(12;21) to emphasize the value of investigating detailed molecular mechanisms in ETV6::RUNX1-positive B-ALL, both for our understanding of the etiology of the disease and for future clinical advances in patient treatment and management.
Collapse
Affiliation(s)
- Agnieszka Kaczmarska
- Student Scientific Society of Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, A. Gębali 6, 20-093 Lublin, Poland
| | - Justyna Derebas
- Student Scientific Society of Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, A. Gębali 6, 20-093 Lublin, Poland
| | - Michalina Pinkosz
- Student Scientific Society of Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, A. Gębali 6, 20-093 Lublin, Poland
| | - Maciej Niedźwiecki
- Department of Pediatrics, Hematology and Oncology Medical University of Gdansk, Debinki 7, 80-211 Gdansk, Poland
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, A. Gębali 6, 20-093 Lublin, Poland
- Correspondence:
| |
Collapse
|
16
|
Villar J, Cros A, De Juan A, Alaoui L, Bonte PE, Lau CM, Tiniakou I, Reizis B, Segura E. ETV3 and ETV6 enable monocyte differentiation into dendritic cells by repressing macrophage fate commitment. Nat Immunol 2023; 24:84-95. [PMID: 36543959 PMCID: PMC9810530 DOI: 10.1038/s41590-022-01374-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 10/31/2022] [Indexed: 12/24/2022]
Abstract
In inflamed tissues, monocytes differentiate into macrophages (mo-Macs) or dendritic cells (mo-DCs). In chronic nonresolving inflammation, mo-DCs are major drivers of pathogenic events. Manipulating monocyte differentiation would therefore be an attractive therapeutic strategy. However, how the balance of mo-DC versus mo-Mac fate commitment is regulated is not clear. In the present study, we show that the transcriptional repressors ETV3 and ETV6 control human monocyte differentiation into mo-DCs. ETV3 and ETV6 inhibit interferon (IFN)-stimulated genes; however, their action on monocyte differentiation is independent of IFN signaling. Instead, we find that ETV3 and ETV6 directly repress mo-Mac development by controlling MAFB expression. Mice deficient for Etv6 in monocytes have spontaneous expression of IFN-stimulated genes, confirming that Etv6 regulates IFN responses in vivo. Furthermore, these mice have impaired mo-DC differentiation during inflammation and reduced pathology in an experimental autoimmune encephalomyelitis model. These findings provide information about the molecular control of monocyte fate decision and identify ETV6 as a therapeutic target to redirect monocyte differentiation in inflammatory disorders.
Collapse
Affiliation(s)
- Javiera Villar
- Institut Curie, PSL Research University, INSERM, U932,, Paris, France
| | - Adeline Cros
- Institut Curie, PSL Research University, INSERM, U932,, Paris, France
| | - Alba De Juan
- Institut Curie, PSL Research University, INSERM, U932,, Paris, France
| | - Lamine Alaoui
- Institut Curie, PSL Research University, INSERM, U932,, Paris, France
| | | | - Colleen M Lau
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Ioanna Tiniakou
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Boris Reizis
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Elodie Segura
- Institut Curie, PSL Research University, INSERM, U932,, Paris, France.
| |
Collapse
|
17
|
Zhou C, Uluisik R, Rowley JW, David C, Jones CL, Scharer CD, Noetzli L, Fisher MH, Kirkpatrick GD, Bark K, Boss JM, Henry CJ, Pietras EM, Di Paola J, Porter CC. Germline ETV6 mutation promotes inflammation and disrupts lymphoid development of early hematopoietic progenitors. Exp Hematol 2022; 112-113:24-34. [PMID: 35803545 PMCID: PMC9885892 DOI: 10.1016/j.exphem.2022.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 06/10/2022] [Accepted: 06/26/2022] [Indexed: 02/01/2023]
Abstract
Germline mutations in ETV6 are associated with a syndrome of thrombocytopenia and leukemia predisposition, and ETV6 is among the most commonly mutated genes in leukemias, especially childhood B-cell acute lymphoblastic leukemia. However, the mechanisms underlying disease caused by ETV6 dysfunction are poorly understood. To address these gaps in knowledge, using CRISPR/Cas9, we developed a mouse model of the most common recurrent, disease-causing germline mutation in ETV6. We found defects in hematopoiesis related primarily to abnormalities of the multipotent progenitor population 4 (MPP4) subset of hematopoietic progenitor cells and evidence of sterile inflammation. Expression of ETV6 in Ba/F3 cells altered the expression of several cytokines, some of which were also detected at higher levels in the bone marrow of the mice with Etv6 mutation. Among these, interleukin-18 and interleukin-13 abrogated B-cell development of sorted MPP4 cells, but not common lymphoid progenitors, suggesting that inflammation contributes to abnormal hematopoiesis by impairing lymphoid development. These data, along with those from humans, support a model in which ETV6 dysfunction promotes inflammation, which adversely affects thrombopoiesis and promotes leukemogenesis.
Collapse
Affiliation(s)
- Chengjing Zhou
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Rizvan Uluisik
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Jesse W Rowley
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT
| | - Camille David
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | | | - Christopher D Scharer
- Department of Microbiology & Immunology, Emory University School of Medicine, Atlanta, GA
| | | | - Marlie H Fisher
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, CO
| | | | - Katrina Bark
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO
| | - Jeremy M Boss
- Department of Microbiology & Immunology, Emory University School of Medicine, Atlanta, GA
| | - Curtis J Henry
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Eric M Pietras
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO
| | - Jorge Di Paola
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Christopher C Porter
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA; Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA.
| |
Collapse
|
18
|
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
|
19
|
Liu Y, Wang Y, Zhang C, Feng Q, Hou M, Peng J, Hu X, Wang S. HDAC3 single-nucleotide polymorphism rs2530223 is associated with increased susceptibility and severity of primary immune thrombocytopenia. Int J Lab Hematol 2022; 44:875-882. [PMID: 35484920 DOI: 10.1111/ijlh.13857] [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: 12/03/2021] [Revised: 03/03/2022] [Accepted: 04/03/2022] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Primary immune thrombocytopenia (ITP) is an autoimmune hemorrhagic disorder characterized by a low platelet count and increased risk of bleeding. We previously reported that low-dose chidamide, a histone deacetylase (HDAC) inhibitor, restores immune tolerance in patients with ITP. This study aimed to evaluate the association of a single-nucleotide polymorphism (SNP) rs2530223 in the HDAC3 gene with susceptibility to ITP and its clinical features. METHODS Patients with ITP and age-matched healthy participants were recruited for this case-control study. Genotyping of the HDAC3 rs2530223 polymorphism was performed using MassARRAY platform. RESULTS Individuals with T allele of HDAC3 rs2530223 exhibited a 1.472-fold increased risk of ITP susceptibility (OR 1.472; 95% CI 1.100-1.969; p = .009), while ones with the TT genotype under the codominant and recessive models, and the TC/TT genotypes under the dominant model all revealed increased risk of ITP susceptibility (dominant odds ratio[OR] 1.965; 95% CI: 1.046-3.656; p = .036; codominant OR 2.264; 95% CI 1.175-4.360; p = .015; and recessive OR 1.512; 95% CI 1.028-2.224; p = .036, respectively). Regarding platelet counts in ITP patients, we observed that the TC/TT genotypes exhibited a 3.932-fold increased risk for platelet (PLT) <30 × 109 /L (OR 3.932; 95% CI 1.426-10.842; p = .008). CONCLUSION This study indicates that HDAC3 rs2530223 may be an important genetic factor related to ITP susceptibility and platelet count in ITP patients, providing new perspectives on disease progression, new therapeutic targets, and severity prediction.
Collapse
Affiliation(s)
- Yan Liu
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yin Wang
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Cheng Zhang
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qi Feng
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ming Hou
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jun Peng
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiang Hu
- Shandong Provincial Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shuwen Wang
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
20
|
Montenont E, Bhatlekar S, Jacob S, Kosaka Y, Manne BK, Lee O, Parra-Izquierdo I, Tugolukova E, Tolley ND, Rondina MT, Bray PF, Rowley JW. CRISPR-edited megakaryocytes for rapid screening of platelet gene functions. Blood Adv 2021; 5:2362-2374. [PMID: 33944898 PMCID: PMC8114553 DOI: 10.1182/bloodadvances.2020004112] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/09/2021] [Indexed: 01/07/2023] Open
Abstract
Human anucleate platelets cannot be directly modified using traditional genetic approaches. Instead, studies of platelet gene function depend on alternative models. Megakaryocytes (the nucleated precursor to platelets) are the nearest cell to platelets in origin, structure, and function. However, achieving consistent genetic modifications in primary megakaryocytes has been challenging, and the functional effects of induced gene deletions on human megakaryocytes for even well-characterized platelet genes (eg, ITGA2B) are unknown. Here we present a rapid and systematic approach to screen genes for platelet functions in CD34+ cell-derived megakaryocytes called CRIMSON (CRISPR-edited megakaryocytes for rapid screening of platelet gene functions). By using CRISPR/Cas9, we achieved efficient nonviral gene editing of a panel of platelet genes in megakaryocytes without compromising megakaryopoiesis. Gene editing induced loss of protein in up to 95% of cells for platelet function genes GP6, RASGRP2, and ITGA2B; for the immune receptor component B2M; and for COMMD7, which was previously associated with cardiovascular disease and platelet function. Gene deletions affected several select responses to platelet agonists in megakaryocytes in a manner largely consistent with those expected for platelets. Deletion of B2M did not significantly affect platelet-like responses, whereas deletion of ITGA2B abolished agonist-induced integrin activation and spreading on fibrinogen without affecting the translocation of P-selectin. Deletion of GP6 abrogated responses to collagen receptor agonists but not thrombin. Deletion of RASGRP2 impaired functional responses to adenosine 5'-diphosphate (ADP), thrombin, and collagen receptor agonists. Deletion of COMMD7 significantly impaired multiple responses to platelet agonists. Together, our data recommend CRIMSON for rapid evaluation of platelet gene phenotype associations.
Collapse
Affiliation(s)
- Emilie Montenont
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Seema Bhatlekar
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Shancy Jacob
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Yasuhiro Kosaka
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Bhanu K Manne
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Olivia Lee
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | | | - Emilia Tugolukova
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Neal D Tolley
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
| | - Matthew T Rondina
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
- Department of Internal Medicine
- George E. Wahlen Department of Veterans Affairs Medical Center
- Department of Internal Medicine and Geriatric Research and Education Clinical Center, and
- Department of Pathology, The University of Utah, Salt Lake City, UT
| | - Paul F Bray
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
- Department of Internal Medicine
| | - Jesse W Rowley
- Molecular Medicine Program, The University of Utah, Salt Lake City, UT
- Department of Internal Medicine
| |
Collapse
|