1
|
Dey A, Butcher M, Gegonne A, Singer DS, Zhu J, Ozato K. BRD4 expression in microglia supports recruitment of T cells into the CNS and exacerbates EAE. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.13.612948. [PMID: 39372778 PMCID: PMC11451676 DOI: 10.1101/2024.09.13.612948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
In EAE, a mouse model of multiple sclerosis, immunization with MOG autoantigen results in the generation of Th1/Th17 T cells in the periphery. MOG-specific T cells then invade into the central nervous system (CNS), resulting in neuronal demyelination. Microglia, innate immune cells in the CNS are known to regulate various neuronal diseases. However, the role of microglia in EAE has remained elusive. BRD4 is a BET protein expressed in microglia, whether BRD4 in microglia contributes to EAE has not been determined. We show that EAE pathology was markedly reduced with microglia-specific Brd4 conditional knockout (cKO). In these mice, microglia- T cell interactions were greatly reduced, leading to the lack of T cell reactivation. Microglia specific transcriptome data showed downregulation of genes required for interaction with and reactivation of T cells in Brd4 cKO samples. In summary, BRD4 plays a critical role in regulating microglia function in normal and EAE CNS. Summary This study demonstrates that in a EAE model, microglia-specific Brd4 conditional knockout mice were defective in expressing genes required for microglia- T cells interaction and those involved in neuroinflammation, and demyelination resulting in fewer CNS T cell invasion and display marked reduction in EAE pathology.
Collapse
|
2
|
Wu J, Fan C, Kabir AU, Krchma K, Kim M, Kwon Y, Xing X, Wang T, Choi K. Baf155 controls hematopoietic differentiation and regeneration through chromatin priming. Cell Rep 2024; 43:114558. [PMID: 39088321 PMCID: PMC11465209 DOI: 10.1016/j.celrep.2024.114558] [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: 10/12/2023] [Revised: 05/06/2024] [Accepted: 07/14/2024] [Indexed: 08/03/2024] Open
Abstract
Chromatin priming promotes cell-type-specific gene expression, lineage differentiation, and development. The mechanism of chromatin priming has not been fully understood. Here, we report that mouse hematopoietic stem and progenitor cells (HSPCs) lacking the Baf155 subunit of the BAF (BRG1/BRM-associated factor) chromatin remodeling complex produce a significantly reduced number of mature blood cells, leading to a failure of hematopoietic regeneration upon transplantation and 5-fluorouracil (5-FU) injury. Baf155-deficient HSPCs generate particularly fewer neutrophils, B cells, and CD8+ T cells at homeostasis, supporting a more immune-suppressive tumor microenvironment and enhanced tumor growth. Single-nucleus multiomics analysis reveals that Baf155-deficient HSPCs fail to establish accessible chromatin in selected regions that are enriched for putative enhancers and binding motifs of hematopoietic lineage transcription factors. Our study provides a fundamental mechanistic understanding of the role of Baf155 in hematopoietic lineage chromatin priming and the functional consequences of Baf155 deficiency in regeneration and tumor immunity.
Collapse
Affiliation(s)
- Jun Wu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Changxu Fan
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA; Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ashraf Ul Kabir
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Karen Krchma
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Minseo Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yoojung Kwon
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xiaoyun Xing
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA; Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ting Wang
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA; Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kyunghee Choi
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
3
|
Miyauchi J. The hematopoietic microenvironment of the fetal liver and transient abnormal myelopoiesis associated with Down syndrome: A review. Crit Rev Oncol Hematol 2024; 199:104382. [PMID: 38723838 DOI: 10.1016/j.critrevonc.2024.104382] [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: 11/02/2023] [Revised: 04/21/2024] [Accepted: 05/02/2024] [Indexed: 05/23/2024] Open
Abstract
Transient abnormal myelopoiesis (TAM) in neonates with Down syndrome is a distinct form of leukemia or preleukemia that mirrors the hematological features of acute megakaryoblastic leukemia. However, it typically resolves spontaneously in the early stages. TAM originates from fetal liver (FL) hematopoietic precursor cells and emerges due to somatic mutations in GATA1 in utero. In TAM, progenitor cells proliferate and differentiate into mature megakaryocytes and granulocytes. This process occurs both in vitro, aided by hematopoietic growth factors (HGFs) produced in the FL, and in vivo, particularly in specific anatomical sites like the FL and blood vessels. The FL's hematopoietic microenvironment plays a crucial role in TAM's pathogenesis and may contribute to its spontaneous regression. This review presents an overview of current knowledge regarding the unique features of TAM in relation to the FL hematopoietic microenvironment, focusing on the functions of HGFs and the pathological features of TAM.
Collapse
Affiliation(s)
- Jun Miyauchi
- Department of Diagnostic Pathology, Saitama City Hospital, Saitama, Saitama-ken, Japan.
| |
Collapse
|
4
|
Baruchel A, Bourquin JP, Crispino J, Cuartero S, Hasle H, Hitzler J, Klusmann JH, Izraeli S, Lane AA, Malinge S, Rabin KR, Roberts I, Ryeom S, Tasian SK, Wagenblast E. Down syndrome and leukemia: from basic mechanisms to clinical advances. Haematologica 2023; 108:2570-2581. [PMID: 37439336 PMCID: PMC10542835 DOI: 10.3324/haematol.2023.283225] [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: 05/02/2023] [Accepted: 06/29/2023] [Indexed: 07/14/2023] Open
Abstract
Children with Down syndrome (DS, trisomy 21) are at a significantly higher risk of developing acute leukemia compared to the overall population. Many studies investigating the link between trisomy 21 and leukemia initiation and progression have been conducted over the last two decades. Despite improved treatment regimens and significant progress in iden - tifying genes on chromosome 21 and the mechanisms by which they drive leukemogenesis, there is still much that is unknown. A focused group of scientists and clinicians with expertise in leukemia and DS met in October 2022 at the Jérôme Lejeune Foundation in Paris, France for the 1st International Symposium on Down Syndrome and Leukemia. This meeting was held to discuss the most recent advances in treatment regimens and the biology underlying the initiation, progression, and relapse of acute lymphoblastic leukemia and acute myeloid leukemia in children with DS. This review provides a summary of what is known in the field, challenges in the management of DS patients with leukemia, and key questions in the field.
Collapse
Affiliation(s)
- André Baruchel
- Hôpital Universitaire Robert Debré (APHP and Université Paris Cité), Paris, France
| | | | - John Crispino
- St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Sergi Cuartero
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Henrik Hasle
- Department of Pediatrics and Adolescent Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Johann Hitzler
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | - Shai Izraeli
- Schneider Children's Medical Center of Israel, Petah Tikva, Israel
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Aviv University, Aviv, Israel
| | | | - Sébastien Malinge
- Telethon Kids Institute - Cancer Centre, Perth, Western Australia, Australia
| | - Karen R. Rabin
- Baylor College of Medicine, Texas Children's Cancer Center, Houston, TX, USA
| | | | - Sandra Ryeom
- Department of Surgery, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Sarah K. Tasian
- Children’s Hospital of Philadelphia and University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | |
Collapse
|
5
|
Ling T, Zhang K, Yang J, Gurbuxani S, Crispino JD. Gata1s mutant mice display persistent defects in the erythroid lineage. Blood Adv 2023; 7:3253-3264. [PMID: 36350717 PMCID: PMC10336263 DOI: 10.1182/bloodadvances.2022008124] [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: 05/18/2022] [Revised: 09/23/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022] Open
Abstract
GATA1 mutations that result in loss of the N-terminal 83 amino acids are a feature of myeloid leukemia in children with Down syndrome, rare familial cases of dyserythropoietic anemia, and a subset of cases of Diamond-Blackfan anemia. The Gata1s mouse model, which expresses only the short GATA1 isoform that begins at methionine 84, has been shown to have a defect in hematopoiesis, especially impaired erythropoiesis with expanded megakaryopoiesis, during gestation. However, these mice reportedly did not show any postnatal phenotype. Here, we demonstrate that Gata1s mutant mice display macrocytic anemia and features of aberrant megakaryopoiesis throughout life, culminating in profound splenomegaly and bone marrow fibrosis. These data support the use of this animal model for studies of GATA1 deficiencies.
Collapse
Affiliation(s)
- Te Ling
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Kevin Zhang
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Jiayue Yang
- Department of Microbiology, School of Molecular and Cellular Biology, University of Illinois Urbana-Champaign, Champaign, IL
| | | | - John D. Crispino
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN
| |
Collapse
|
6
|
Mendoza-Castrejon J, Magee JA. Layered immunity and layered leukemogenicity: Developmentally restricted mechanisms of pediatric leukemia initiation. Immunol Rev 2023; 315:197-215. [PMID: 36588481 PMCID: PMC10301262 DOI: 10.1111/imr.13180] [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: 01/03/2023]
Abstract
Hematopoietic stem cells (HSCs) and multipotent progenitor cells (MPPs) arise in successive waves during ontogeny, and their properties change significantly throughout life. Ontological changes in HSCs/MPPs underlie corresponding changes in mechanisms of pediatric leukemia initiation. As HSCs and MPPs progress from fetal to neonatal, juvenile and adult stages of life, they undergo transcriptional and epigenetic reprogramming that modifies immune output to meet age-specific pathogenic challenges. Some immune cells arise exclusively from fetal HSCs/MPPs. We propose that this layered immunity instructs cell fates that underlie a parallel layered leukemogenicity. Indeed, some pediatric leukemias, such as juvenile myelomonocytic leukemia, myeloid leukemia of Down syndrome, and infant pre-B-cell acute lymphoblastic leukemia, are age-restricted. They only present during infancy or early childhood. These leukemias likely arise from fetal progenitors that lose competence for transformation as they age. Other childhood leukemias, such as non-infant pre-B-cell acute lymphoblastic leukemia and acute myeloid leukemia, have mutation profiles that are common in childhood but rare in morphologically similar adult leukemias. These differences could reflect temporal changes in mechanisms of mutagenesis or changes in how progenitors respond to a given mutation at different ages. Interactions between leukemogenic mutations and normal developmental switches offer potential targets for therapy.
Collapse
Affiliation(s)
- Jonny Mendoza-Castrejon
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110
| | - Jeffrey A. Magee
- Department of Pediatrics, Division of Hematology and Oncology, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110
- Department of Genetics, Washington University School of Medicine, 660 S. Euclid Ave, St. Louis, MO 63110
| |
Collapse
|
7
|
Gialesaki S, Bräuer-Hartmann D, Issa H, Bhayadia R, Alejo-Valle O, Verboon L, Schmell AL, Laszig S, Regényi E, Schuschel K, Labuhn M, Ng M, Winkler R, Ihling C, Sinz A, Glaß M, Hüttelmaier S, Matzk S, Schmid L, Strüwe FJ, Kadel SK, Reinhardt D, Yaspo ML, Heckl D, Klusmann JH. RUNX1 isoform disequilibrium promotes the development of trisomy 21-associated myeloid leukemia. Blood 2023; 141:1105-1118. [PMID: 36493345 PMCID: PMC10023736 DOI: 10.1182/blood.2022017619] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 11/08/2022] [Accepted: 11/22/2022] [Indexed: 12/14/2022] Open
Abstract
Gain of chromosome 21 (Hsa21) is among the most frequent aneuploidies in leukemia. However, it remains unclear how partial or complete amplifications of Hsa21 promote leukemogenesis and why children with Down syndrome (DS) (ie, trisomy 21) are particularly at risk of leukemia development. Here, we propose that RUNX1 isoform disequilibrium with RUNX1A bias is key to DS-associated myeloid leukemia (ML-DS). Starting with Hsa21-focused CRISPR-CRISPR-associated protein 9 screens, we uncovered a strong and specific RUNX1 dependency in ML-DS cells. Expression of the RUNX1A isoform is elevated in patients with ML-DS, and mechanistic studies using murine ML-DS models and patient-derived xenografts revealed that excess RUNX1A synergizes with the pathognomonic Gata1s mutation during leukemogenesis by displacing RUNX1C from its endogenous binding sites and inducing oncogenic programs in complex with the MYC cofactor MAX. These effects were reversed by restoring the RUNX1A:RUNX1C equilibrium in patient-derived xenografts in vitro and in vivo. Moreover, pharmacological interference with MYC:MAX dimerization using MYCi361 exerted strong antileukemic effects. Thus, our study highlights the importance of alternative splicing in leukemogenesis, even on a background of aneuploidy, and paves the way for the development of specific and targeted therapies for ML-DS, as well as for other leukemias with Hsa21 aneuploidy or RUNX1 isoform disequilibrium.
Collapse
Affiliation(s)
- Sofia Gialesaki
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Daniela Bräuer-Hartmann
- Pediatric Hematology and Oncology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Hasan Issa
- Department of Pediatrics, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Raj Bhayadia
- Department of Pediatrics, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Oriol Alejo-Valle
- Pediatric Hematology and Oncology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Lonneke Verboon
- Department of Pediatrics, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Anna-Lena Schmell
- Department of Pediatrics, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Stephanie Laszig
- Department of Pediatrics, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Enikő Regényi
- Pediatric Hematology and Oncology, Martin Luther University Halle-Wittenberg, Halle, Germany
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Konstantin Schuschel
- Department of Pediatrics, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Maurice Labuhn
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Michelle Ng
- Pediatric Hematology and Oncology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Robert Winkler
- Department of Pediatrics, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Christian Ihling
- Department of Pharmaceutical Chemistry and Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Andrea Sinz
- Department of Pharmaceutical Chemistry and Bioanalytics, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Markus Glaß
- Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Stefan Hüttelmaier
- Institute of Molecular Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Sören Matzk
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Lena Schmid
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | | | - Sofie-Katrin Kadel
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Dirk Reinhardt
- Pediatric Hematology and Oncology, Pediatrics III, University Hospital Essen, Essen, Germany
| | | | - Dirk Heckl
- Pediatric Hematology and Oncology, Martin Luther University Halle-Wittenberg, Halle, Germany
- Dirk Heckl, Pediatric Hematology and Oncology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Str. 40, 06120 Halle, Germany;
| | - Jan-Henning Klusmann
- Department of Pediatrics, Goethe University Frankfurt, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Correspondence: Jan-Henning Klusmann, Department of Pediatrics, Goethe University Frankfurt, Theodor Stern Kai 7, 60590 Frankfurt, Germany;
| |
Collapse
|
8
|
Roberts I. Leukemogenesis in infants and young children with trisomy 21. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2022; 2022:1-8. [PMID: 36485097 PMCID: PMC9820574 DOI: 10.1182/hematology.2022000395] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Children with Down syndrome (DS) have a greater than 100-fold increased risk of developing acute myeloid leukemia (ML) and an approximately 30-fold increased risk of acute lymphoblastic leukemia (ALL) before their fifth birthday. ML-DS originates in utero and typically presents with a self-limiting, neonatal leukemic syndrome known as transient abnormal myelopoiesis (TAM) that is caused by cooperation between trisomy 21-associated abnormalities of fetal hematopoiesis and somatic N-terminal mutations in the transcription factor GATA1. Around 10% of neonates with DS have clinical signs of TAM, although the frequency of hematologically silent GATA1 mutations in DS neonates is much higher (~25%). While most cases of TAM/silent TAM resolve without treatment within 3 to 4 months, in 10% to 20% of cases transformation to full-blown leukemia occurs within the first 4 years of life when cells harboring GATA1 mutations persist and acquire secondary mutations, most often in cohesin genes. By contrast, DS-ALL, which is almost always B-lineage, presents after the first few months of life and is characterized by a high frequency of rearrangement of the CRLF2 gene (60%), often co-occurring with activating mutations in JAK2 or RAS genes. While treatment of ML-DS achieves long-term survival in approximately 90% of children, the outcome of DS-ALL is inferior to ALL in children without DS. Ongoing studies in primary cells and model systems indicate that the role of trisomy 21 in DS leukemogenesis is complex and cell context dependent but show promise in improving management and the treatment of relapse, in which the outcome of both ML-DS and DS-ALL remains poor.
Collapse
Affiliation(s)
- Irene Roberts
- Correspondence Irene Roberts, Department of Paediatrics, MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Headington, Oxford OX3 9DS, United Kingdom; e-mail: ,
| |
Collapse
|
9
|
Kanezaki R, Toki T, Terui K, Sato T, Kobayashi A, Kudo K, Kamio T, Sasaki S, Kawaguchi K, Watanabe K, Ito E. Mechanism of KIT gene regulation by GATA1 lacking the N-terminal domain in Down syndrome-related myeloid disorders. Sci Rep 2022; 12:20587. [PMID: 36447001 PMCID: PMC9708825 DOI: 10.1038/s41598-022-25046-z] [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: 05/16/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022] Open
Abstract
Children with Down syndrome (DS) are at high risk of transient abnormal myelopoiesis (TAM) and myeloid leukemia of DS (ML-DS). GATA1 mutations are detected in almost all TAM and ML-DS samples, with exclusive expression of short GATA1 protein (GATA1s) lacking the N-terminal domain (NTD). However, it remains to be clarified how GATA1s is involved with both disorders. Here, we established the K562 GATA1s (K562-G1s) clones expressing only GATA1s by CRISPR/Cas9 genome editing. The K562-G1s clones expressed KIT at significantly higher levels compared to the wild type of K562 (K562-WT). Chromatin immunoprecipitation studies identified the GATA1-bound regulatory sites upstream of KIT in K562-WT, K562-G1s clones and two ML-DS cell lines; KPAM1 and CMK11-5. Sonication-based chromosome conformation capture (3C) assay demonstrated that in K562-WT, the - 87 kb enhancer region of KIT was proximal to the - 115 kb, - 109 kb and + 1 kb region, while in a K562-G1s clone, CMK11-5 and primary TAM cells, the - 87 kb region was more proximal to the KIT transcriptional start site. These results suggest that the NTD of GATA1 is essential for proper genomic conformation and regulation of KIT gene expression, and that perturbation of this function might be involved in the pathogenesis of TAM and ML-DS.
Collapse
Affiliation(s)
- Rika Kanezaki
- grid.257016.70000 0001 0673 6172Department of Pediatrics, Hirosaki University Graduate School of Medicine, 5 Zaifucho, Hirosaki, Aomori 036-8562 Japan
| | - Tsutomu Toki
- grid.257016.70000 0001 0673 6172Department of Pediatrics, Hirosaki University Graduate School of Medicine, 5 Zaifucho, Hirosaki, Aomori 036-8562 Japan
| | - Kiminori Terui
- grid.257016.70000 0001 0673 6172Department of Pediatrics, Hirosaki University Graduate School of Medicine, 5 Zaifucho, Hirosaki, Aomori 036-8562 Japan
| | - Tomohiko Sato
- grid.257016.70000 0001 0673 6172Department of Pediatrics, Hirosaki University Graduate School of Medicine, 5 Zaifucho, Hirosaki, Aomori 036-8562 Japan
| | - Akie Kobayashi
- grid.257016.70000 0001 0673 6172Department of Pediatrics, Hirosaki University Graduate School of Medicine, 5 Zaifucho, Hirosaki, Aomori 036-8562 Japan
| | - Ko Kudo
- grid.257016.70000 0001 0673 6172Department of Pediatrics, Hirosaki University Graduate School of Medicine, 5 Zaifucho, Hirosaki, Aomori 036-8562 Japan
| | - Takuya Kamio
- grid.257016.70000 0001 0673 6172Department of Pediatrics, Hirosaki University Graduate School of Medicine, 5 Zaifucho, Hirosaki, Aomori 036-8562 Japan
| | - Shinya Sasaki
- grid.257016.70000 0001 0673 6172Department of Pediatrics, Hirosaki University Graduate School of Medicine, 5 Zaifucho, Hirosaki, Aomori 036-8562 Japan
| | - Koji Kawaguchi
- grid.415798.60000 0004 0378 1551Department of Hematology and Oncology, Shizuoka Children’s Hospital, Shizuoka, Japan
| | - Kenichiro Watanabe
- grid.415798.60000 0004 0378 1551Department of Hematology and Oncology, Shizuoka Children’s Hospital, Shizuoka, Japan
| | - Etsuro Ito
- grid.257016.70000 0001 0673 6172Department of Pediatrics, Hirosaki University Graduate School of Medicine, 5 Zaifucho, Hirosaki, Aomori 036-8562 Japan ,grid.257016.70000 0001 0673 6172Department of Community Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| |
Collapse
|
10
|
Chen T, Zhang Q, Shang X, Zou S, Qin J, Li K, Lin B, Tao Z, Long X, Xu X. Diamond-Blackfan anaemia caused by a de novo initiation codon mutation resulting in a shorter isoform of GATA1. Clin Genet 2022; 102:548-554. [PMID: 36029112 DOI: 10.1111/cge.14218] [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: 07/17/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 11/30/2022]
Abstract
Diamond-Blackfan Anaemia (DBA) is an inherited marrow failure disorder characterised by selective erythroid aplasia. Herein, we reported a case of DBA caused by a novel GATA1 gene mutation. The proband manifested normocytic normochromic anaemia, while the parents were asymptomatic. Next-generation sequencing identified a novel de novo mutation at GATA1 initiation codon (GATA1:c.3G>A) in the proband. The mutation led to a shortened GATA1 protein (GATA1s), which caused a reduction in full-length functional GATA1 protein (GATA1fl). This is the first report of GATA1-related DBA patient in the East Asian population, which expanded the mutational spectrum of DBA furthering understanding of its pathogenesis.
Collapse
Affiliation(s)
- Tongtong Chen
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Qianqian Zhang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Xuan Shang
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Shaomin Zou
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiaofeng Qin
- Department of Pediatrics, Liuzhou People's Hospital, Liuzhou, Guangxi, China
| | - Kui Li
- Guangzhou Huayin Medical Laboratory Center Co., Ltd., Guangzhou, Guangdong, China.,Guangzhou Jiexu Gene Technology Co., Ltd., Guangzhou, Guangdong, China
| | - Bin Lin
- Guangzhou Huayin Medical Laboratory Center Co., Ltd., Guangzhou, Guangdong, China.,Guangzhou Jiexu Gene Technology Co., Ltd., Guangzhou, Guangdong, China
| | - Zhenzhong Tao
- Guangzhou Huayin Medical Laboratory Center Co., Ltd., Guangzhou, Guangdong, China.,Guangzhou Jiexu Gene Technology Co., Ltd., Guangzhou, Guangdong, China
| | - Xingjiang Long
- Department of Pediatrics, Liuzhou People's Hospital, Liuzhou, Guangxi, China
| | - Xiangmin Xu
- Department of Medical Genetics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China.,Innovative Research Center for Diagnosis and Therapy of Thalassemias, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
11
|
Arkoun B, Robert E, Boudia F, Mazzi S, Dufour V, Siret A, Mammasse Y, Aid Z, Vieira M, Imanci A, Aglave M, Cambot M, Petermann R, Souquere S, Rameau P, Catelain C, Diot R, Tachdjian G, Hermine O, Droin N, Debili N, Plo I, Malinge S, Soler E, Raslova H, Mercher T, Vainchenker W. Stepwise GATA1 and SMC3 mutations alter megakaryocyte differentiation in a Down syndrome leukemia model. J Clin Invest 2022; 132:156290. [PMID: 35587378 PMCID: PMC9282925 DOI: 10.1172/jci156290] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 05/13/2022] [Indexed: 11/22/2022] Open
Abstract
Acute megakaryoblastic leukemia of Down syndrome (DS-AMKL) is a model of clonal evolution from a preleukemic transient myeloproliferative disorder requiring both a trisomy 21 (T21) and a GATA1s mutation to a leukemia driven by additional driver mutations. We modeled the megakaryocyte differentiation defect through stepwise gene editing of GATA1s, SMC3+/–, and MPLW515K, providing 20 different T21 or disomy 21 (D21) induced pluripotent stem cell (iPSC) clones. GATA1s profoundly reshaped iPSC-derived hematopoietic architecture with gradual myeloid-to-megakaryocyte shift and megakaryocyte differentiation alteration upon addition of SMC3 and MPL mutations. Transcriptional, chromatin accessibility, and GATA1-binding data showed alteration of essential megakaryocyte differentiation genes, including NFE2 downregulation that was associated with loss of GATA1s binding and functionally involved in megakaryocyte differentiation blockage. T21 enhanced the proliferative phenotype, reproducing the cellular and molecular abnormalities of DS-AMKL. Our study provides an array of human cell–based models revealing individual contributions of different mutations to DS-AMKL differentiation blockage, a major determinant of leukemic progression.
Collapse
Affiliation(s)
- Brahim Arkoun
- INSERM, UMR1287, Institut Gustave Roussy, Villejuif, France
| | - Elie Robert
- INSERM, UMR1170, Institut Gustave Roussy, Villejuif, France
| | - Fabien Boudia
- INSERM, UMR1170, Institut Gustave Roussy, Villejuif, France
| | - Stefania Mazzi
- INSERM, UMR1287, Institut Gustave Roussy, Villejuif, France
| | - Virginie Dufour
- INSERM, UMR1287, Institut National de la Transfusion Sanguine, Villejuif, France
| | - Aurelie Siret
- INSERM, UMR1170, Institut Gustave Roussy, Villejuif, France
| | - Yasmine Mammasse
- Département d'Immunologie Plaquettaire, Institut National de la Transfusion Sanguine, Paris, France
| | - Zakia Aid
- INSERM, UMR1170, Institut Gustave Roussy, Villejuif, France
| | - Mathieu Vieira
- INSERM, UMR1287, Institut Gustave Roussy, Villejuif, France
| | - Aygun Imanci
- INSERM, UMR1287, Institut Gustave Roussy, Villejuif, France
| | - Marine Aglave
- Plateforme de Bioinformatique, Institut Gustave Roussy, Villejuif, France
| | - Marie Cambot
- Département d'Immunologie Plaquettaire, Institut National de la Transfusion Sanguine, Paris, France
| | - Rachel Petermann
- Département d'Immunologie Plaquettaire, Institut National de Transfusion Sanguine, Paris, France
| | - Sylvie Souquere
- Centre National de la Recherche Scientifique, UMR8122, Institut Gustave Roussy, Villejuif, France
| | - Philippe Rameau
- UMS AMMICA, INSERM US23, Institut Gustave Roussy, Villejuif, France
| | - Cyril Catelain
- UMS AMMICA, INSERM US23, Institut Gustave Roussy, Villejuif, France
| | - Romain Diot
- Service d'Histologie, Embryologie et Cytogénétique, Hôpital Antoine Béclère, Clamart, France
| | - Gerard Tachdjian
- Service d'Histologie, Embryologie et Cytogénétique, Hôpital Antoine Béclère, Clamart, France
| | | | - Nathalie Droin
- INSERM, UMR1170, Institut Gustave Roussy, Villejuif, France
| | - Najet Debili
- INSERM, UMR1287, Institut Gustave Roussy, Villejuif, France
| | - Isabelle Plo
- INSERM, UMR1287, Institut Gustave Roussy, Villejuif, France
| | - Sebastien Malinge
- Telethon Kids Cancer Centre, Telethon Kids Institute, University of Western Australia, Perth, Australia
| | - Eric Soler
- IGMM, University of Montpellier, Montpellier, France
| | - Hana Raslova
- INSERM, UMR1287, Institut Gustave Roussy, Villejuif, France
| | - Thomas Mercher
- INSERM, UMR1170, Institut Gustave Roussy, Villejuif, France
| | | |
Collapse
|
12
|
Kwon B, Fansler MM, Patel ND, Lee J, Ma W, Mayr C. Enhancers regulate 3' end processing activity to control expression of alternative 3'UTR isoforms. Nat Commun 2022; 13:2709. [PMID: 35581194 PMCID: PMC9114392 DOI: 10.1038/s41467-022-30525-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 05/02/2022] [Indexed: 12/12/2022] Open
Abstract
Multi-UTR genes are widely transcribed and express their alternative 3'UTR isoforms in a cell type-specific manner. As transcriptional enhancers regulate mRNA expression, we investigated if they also regulate 3'UTR isoform expression. Endogenous enhancer deletion of the multi-UTR gene PTEN did not impair transcript production but prevented 3'UTR isoform switching which was recapitulated by silencing of an enhancer-bound transcription factor. In reporter assays, enhancers increase transcript production when paired with single-UTR gene promoters. However, when combined with multi-UTR gene promoters, they change 3'UTR isoform expression by increasing 3' end processing activity of polyadenylation sites. Processing activity of polyadenylation sites is affected by transcription factors, including NF-κB and MYC, transcription elongation factors, chromatin remodelers, and histone acetyltransferases. As endogenous cell type-specific enhancers are associated with genes that increase their short 3'UTRs in a cell type-specific manner, our data suggest that transcriptional enhancers integrate cellular signals to regulate cell type-and condition-specific 3'UTR isoform expression.
Collapse
Affiliation(s)
- Buki Kwon
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Mervin M Fansler
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
- Tri-Institutional Training Program in Computational Biology and Medicine, Weill Cornell Graduate College, New York, NY, 10021, USA
| | - Neil D Patel
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Jihye Lee
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Weirui Ma
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Christine Mayr
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
- Tri-Institutional Training Program in Computational Biology and Medicine, Weill Cornell Graduate College, New York, NY, 10021, USA.
| |
Collapse
|
13
|
Wells M, Steiner L. Epigenetic and Transcriptional Control of Erythropoiesis. Front Genet 2022; 13:805265. [PMID: 35330735 PMCID: PMC8940284 DOI: 10.3389/fgene.2022.805265] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 02/16/2022] [Indexed: 12/21/2022] Open
Abstract
Erythropoiesis is a process of enormous magnitude, with the average person generating two to three million red cells every second. Erythroid progenitors start as large cells with large nuclei, and over the course of three to four cell divisions they undergo a dramatic decrease in cell size accompanied by profound nuclear condensation, which culminates in enucleation. As maturing erythroblasts are undergoing these dramatic phenotypic changes, they accumulate hemoglobin and express high levels of other erythroid-specific genes, while silencing much of the non-erythroid transcriptome. These phenotypic and gene expression changes are associated with distinct changes in the chromatin landscape, and require close coordination between transcription factors and epigenetic regulators, as well as precise regulation of RNA polymerase II activity. Disruption of these processes are associated with inherited anemias and myelodysplastic syndromes. Here, we review the epigenetic mechanisms that govern terminal erythroid maturation, and their role in human disease.
Collapse
Affiliation(s)
- Maeve Wells
- Department of Pediatrics, University of Rochester, Rochester, NY, United States
| | - Laurie Steiner
- Department of Pediatrics, University of Rochester, Rochester, NY, United States
| |
Collapse
|
14
|
Alejo-Valle O, Weigert K, Bhayadia R, Ng M, Issa H, Beyer C, Emmrich S, Schuschel K, Ihling C, Sinz A, Zimmermann M, Wickenhauser C, Flasinski M, Regenyi E, Labuhn M, Reinhardt D, Yaspo ML, Heckl D, Klusmann JH. The megakaryocytic transcription factor ARID3A suppresses leukemia pathogenesis. Blood 2022; 139:651-665. [PMID: 34570885 PMCID: PMC9632760 DOI: 10.1182/blood.2021012231] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 09/03/2021] [Indexed: 11/22/2022] Open
Abstract
Given the plasticity of hematopoietic stem and progenitor cells, multiple routes of differentiation must be blocked in the the pathogenesis of acute myeloid leukemia, the molecular basis of which is incompletely understood. We report that posttranscriptional repression of the transcription factor ARID3A by miR-125b is a key event in the pathogenesis of acute megakaryoblastic leukemia (AMKL). AMKL is frequently associated with trisomy 21 and GATA1 mutations (GATA1s), and children with Down syndrome are at a high risk of developing the disease. The results of our study showed that chromosome 21-encoded miR-125b synergizes with Gata1s to drive leukemogenesis in this context. Leveraging forward and reverse genetics, we uncovered Arid3a as the main miR-125b target behind this synergy. We demonstrated that, during normal hematopoiesis, this transcription factor promotes megakaryocytic differentiation in concert with GATA1 and mediates TGFβ-induced apoptosis and cell cycle arrest in complex with SMAD2/3. Although Gata1s mutations perturb erythroid differentiation and induce hyperproliferation of megakaryocytic progenitors, intact ARID3A expression assures their megakaryocytic differentiation and growth restriction. Upon knockdown, these tumor suppressive functions are revoked, causing a blockade of dual megakaryocytic/erythroid differentiation and subsequently of AMKL. Inversely, restoring ARID3A expression relieves the arrest of megakaryocytic differentiation in AMKL patient-derived xenografts. This work illustrates how mutations in lineage-determining transcription factors and perturbation of posttranscriptional gene regulation can interact to block multiple routes of hematopoietic differentiation and cause leukemia. In AMKL, surmounting this differentiation blockade through restoration of the tumor suppressor ARID3A represents a promising strategy for treating this lethal pediatric disease.
Collapse
Affiliation(s)
- Oriol Alejo-Valle
- Pediatric Hematology and Oncology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Karoline Weigert
- Pediatric Hematology and Oncology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Raj Bhayadia
- Pediatric Hematology and Oncology, Department of Pediatrics, Goethe University Frankfurt, Frankfurt (Main), Germany
| | - Michelle Ng
- Pediatric Hematology and Oncology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Hasan Issa
- Pediatric Hematology and Oncology, Department of Pediatrics, Goethe University Frankfurt, Frankfurt (Main), Germany
| | - Christoph Beyer
- Pediatric Hematology and Oncology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Stephan Emmrich
- Department of Biology, University of Rochester, Rochester NY
| | - Konstantin Schuschel
- Pediatric Hematology and Oncology, Department of Pediatrics, Goethe University Frankfurt, Frankfurt (Main), Germany
| | - Christian Ihling
- Department of Pharmaceutical Chemistry and Bioanalytics, Institute of Pharmacy, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Andrea Sinz
- Department of Pharmaceutical Chemistry and Bioanalytics, Institute of Pharmacy, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Martin Zimmermann
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | | | - Marius Flasinski
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Hospital Tauberbischofsheim, Tauberbischofsheim, Germany
| | - Eniko Regenyi
- Pediatric Hematology and Oncology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Maurice Labuhn
- Institute for Experimental Virology, Twincore, Center for Experimental and Clinical Infection Research, Hannover, Germany; and
| | - Dirk Reinhardt
- Pediatric Hematology and Oncology, Pediatrics III, University Hospital Essen, Essen, Germany
| | | | - Dirk Heckl
- Pediatric Hematology and Oncology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Jan-Henning Klusmann
- Pediatric Hematology and Oncology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
- Pediatric Hematology and Oncology, Department of Pediatrics, Goethe University Frankfurt, Frankfurt (Main), Germany
| |
Collapse
|
15
|
OUP accepted manuscript. Brief Funct Genomics 2022; 21:159-176. [DOI: 10.1093/bfgp/elac002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 11/14/2022] Open
|
16
|
Ren Y, Zhu J, Han Y, Li P, Wu J, Qu H, Zhang Z, Fang X. Regulatory association of long noncoding RNAs and chromatin accessibility facilitates erythroid differentiation. Blood Adv 2021; 5:5396-5409. [PMID: 34644394 PMCID: PMC9153002 DOI: 10.1182/bloodadvances.2021005167] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/13/2021] [Indexed: 01/17/2023] Open
Abstract
Erythroid differentiation is a dynamic process regulated by multiple factors, whereas the interaction between long noncoding RNAs (lncRNAs) and chromatin accessibility and its influence on erythroid differentiation remains unclear. To elucidate this interaction, we used hematopoietic stem cells, multipotent progenitor cells, common myeloid progenitor cells, megakaryocyte-erythroid progenitor cells, and erythroblasts from human cord blood as an erythroid differentiation model to explore the coordinated regulatory functions of lncRNAs and chromatin accessibility by integrating RNA-seq and ATAC-seq data. We revealed that the integrated network of chromatin accessibility and lncRNAs exhibits stage-specific changes throughout the erythroid differentiation process and that the changes at the erythroblast stage of maturation are dramatic. We identified a subset of stage-specific lncRNAs and transcription factors (TFs) that associate with chromatin accessibility during erythroid differentiation, in which lncRNAs are key regulators of terminal erythroid differentiation via an lncRNA-TF-gene network. LncRNA PCED1B-AS1 was revealed to regulate terminal erythroid differentiation by coordinating GATA1 dynamically binding to the chromatin and interacting with the cytoskeleton network during erythroid differentiation. DANCR, another lncRNA that is highly expressed at the megakaryocyte-erythroid progenitor cell stage, was verified to promote erythroid differentiation by compromising megakaryocyte differentiation and coordinating with chromatin accessibility and TFs, such as RUNX1. Overall, our results identify the associated network of lncRNAs and chromatin accessibility in erythropoiesis and provide novel insights into erythroid differentiation and abundant resources for further study.
Collapse
Affiliation(s)
| | - Junwei Zhu
- CAS Key Laboratory of Genome Science & Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Key Laboratory of Genome and Precision Medicine Technologies, Beijing, China
| | - Yuanyuan Han
- Guizhou University, Medical College, Guiyang, China, China; and
| | - Pin Li
- CAS Key Laboratory of Genome Science & Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, China
| | - Jing Wu
- CAS Key Laboratory of Genome Science & Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China
| | - Hongzhu Qu
- CAS Key Laboratory of Genome Science & Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Key Laboratory of Genome and Precision Medicine Technologies, Beijing, China
| | | | - Xiangdong Fang
- CAS Key Laboratory of Genome Science & Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Key Laboratory of Genome and Precision Medicine Technologies, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
17
|
Wu YY, Zhang HL, Lu X, Du H, Li YC, Zhang PA, Xu GY. Targeting GATA1 and p2x7r Locus Binding in Spinal Astrocytes Suppresses Chronic Visceral Pain by Promoting DNA Demethylation. Neurosci Bull 2021; 38:359-372. [PMID: 34890016 PMCID: PMC9068853 DOI: 10.1007/s12264-021-00799-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 09/04/2021] [Indexed: 11/26/2022] Open
Abstract
Irritable bowel syndrome is a gastrointestinal disorder of unknown etiology characterized by widespread, chronic abdominal pain associated with altered bowel movements. Increasing amounts of evidence indicate that injury and inflammation during the neonatal period have long-term effects on tissue structure and function in the adult that may predispose to gastrointestinal diseases. In this study we aimed to investigate how the epigenetic regulation of DNA demethylation of the p2x7r locus guided by the transcription factor GATA binding protein 1 (GATA1) in spinal astrocytes affects chronic visceral pain in adult rats with neonatal colonic inflammation (NCI). The spinal GATA1 targeting to DNA demethylation of p2x7r locus in these rats was assessed by assessing GATA1 function with luciferase assay, chromatin immunoprecipitation, patch clamp, and interference in vitro and in vivo. In addition, a decoy oligodeoxynucleotide was designed and applied to determine the influence of GATA1 on the DNA methylation of a p2x7r CpG island. We showed that NCI caused the induction of GATA1, Ten-eleven translocation 3 (TET3), and purinergic receptors (P2X7Rs) in astrocytes of the spinal dorsal horn, and demonstrated that inhibiting these molecules markedly increased the pain threshold, inhibited the activation of astrocytes, and decreased the spinal sEPSC frequency. NCI also markedly demethylated the p2x7r locus in a manner dependent on the enhancement of both a GATA1–TET3 physical interaction and GATA1 binding at the p2x7r promoter. Importantly, we showed that demethylation of the p2x7r locus (and the attendant increase in P2X7R expression) was reversed upon knockdown of GATA1 or TET3 expression, and demonstrated that a decoy oligodeoxynucleotide that selectively blocked the GATA1 binding site increased the methylation of a CpG island in the p2x7r promoter. These results demonstrate that chronic visceral pain is mediated synergistically by GATA1 and TET3 via a DNA-demethylation mechanism that controls p2x7r transcription in spinal dorsal horn astrocytes, and provide a potential therapeutic strategy by targeting GATA1 and p2x7r locus binding.
Collapse
Affiliation(s)
- Yan-Yan Wu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Hai-Long Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Xiaomin Lu
- Department of Oncology, The Affiliated Haian Hospital of Nantong University, Nantong, 226600, China
| | - Han Du
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Yong-Chang Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou, 215123, China
| | - Ping-An Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, 215600, China.
| | - Guang-Yin Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou, 215600, China.
| |
Collapse
|
18
|
When Pol II sees red. Blood 2021; 138:1648-1649. [PMID: 34735000 DOI: 10.1182/blood.2021012453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 05/14/2021] [Indexed: 11/20/2022] Open
|
19
|
Murphy ZC, Murphy K, Myers J, Getman M, Couch T, Schulz VP, Lezon-Geyda K, Palumbo C, Yan H, Mohandas N, Gallagher PG, Steiner LA. Regulation of RNA polymerase II activity is essential for terminal erythroid maturation. Blood 2021; 138:1740-1756. [PMID: 34075391 PMCID: PMC8569412 DOI: 10.1182/blood.2020009903] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 04/30/2021] [Indexed: 11/20/2022] Open
Abstract
The terminal maturation of human erythroblasts requires significant changes in gene expression in the context of dramatic nuclear condensation. Defects in this process are associated with inherited anemias and myelodysplastic syndromes. The progressively dense appearance of the condensing nucleus in maturing erythroblasts led to the assumption that heterochromatin accumulation underlies this process, but despite extensive study, the precise mechanisms underlying this essential biologic process remain elusive. To delineate the epigenetic changes associated with the terminal maturation of human erythroblasts, we performed mass spectrometry of histone posttranslational modifications combined with chromatin immunoprecipitation coupled with high-throughput sequencing, Assay for Transposase Accessible Chromatin, and RNA sequencing. Our studies revealed that the terminal maturation of human erythroblasts is associated with a dramatic decline in histone marks associated with active transcription elongation, without accumulation of heterochromatin. Chromatin structure and gene expression were instead correlated with dynamic changes in occupancy of elongation competent RNA polymerase II, suggesting that terminal erythroid maturation is controlled largely at the level of transcription. We further demonstrate that RNA polymerase II "pausing" is highly correlated with transcriptional repression, with elongation competent RNA polymerase II becoming a scare resource in late-stage erythroblasts, allocated to erythroid-specific genes. Functional studies confirmed an essential role for maturation stage-specific regulation of RNA polymerase II activity during erythroid maturation and demonstrate a critical role for HEXIM1 in the regulation of gene expression and RNA polymerase II activity in maturing erythroblasts. Taken together, our findings reveal important insights into the mechanisms that regulate terminal erythroid maturation and provide a novel paradigm for understanding normal and perturbed erythropoiesis.
Collapse
Affiliation(s)
| | | | - Jacquelyn Myers
- Department of Pediatrics and
- Genomics Resource Center, University of Rochester, Rochester, NY
| | | | | | | | | | - Cal Palumbo
- Genomics Resource Center, University of Rochester, Rochester, NY
| | | | | | | | | |
Collapse
|
20
|
de Castro CPM, Cadefau M, Cuartero S. The Mutational Landscape of Myeloid Leukaemia in Down Syndrome. Cancers (Basel) 2021; 13:4144. [PMID: 34439298 PMCID: PMC8394284 DOI: 10.3390/cancers13164144] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 07/30/2021] [Accepted: 08/11/2021] [Indexed: 12/12/2022] Open
Abstract
Children with Down syndrome (DS) are particularly prone to haematopoietic disorders. Paediatric myeloid malignancies in DS occur at an unusually high frequency and generally follow a well-defined stepwise clinical evolution. First, the acquisition of mutations in the GATA1 transcription factor gives rise to a transient myeloproliferative disorder (TMD) in DS newborns. While this condition spontaneously resolves in most cases, some clones can acquire additional mutations, which trigger myeloid leukaemia of Down syndrome (ML-DS). These secondary mutations are predominantly found in chromatin and epigenetic regulators-such as cohesin, CTCF or EZH2-and in signalling mediators of the JAK/STAT and RAS pathways. Most of them are also found in non-DS myeloid malignancies, albeit at extremely different frequencies. Intriguingly, mutations in proteins involved in the three-dimensional organization of the genome are found in nearly 50% of cases. How the resulting mutant proteins cooperate with trisomy 21 and mutant GATA1 to promote ML-DS is not fully understood. In this review, we summarize and discuss current knowledge about the sequential acquisition of genomic alterations in ML-DS.
Collapse
Affiliation(s)
| | - Maria Cadefau
- Josep Carreras Leukaemia Research Institute (IJC), Campus Can Ruti, 08916 Badalona, Spain; (C.P.M.d.C); (M.C.)
- Germans Trias i Pujol Research Institute (IGTP), Campus Can Ruti, 08916 Badalona, Spain
| | - Sergi Cuartero
- Josep Carreras Leukaemia Research Institute (IJC), Campus Can Ruti, 08916 Badalona, Spain; (C.P.M.d.C); (M.C.)
- Germans Trias i Pujol Research Institute (IGTP), Campus Can Ruti, 08916 Badalona, Spain
| |
Collapse
|
21
|
Wagenblast E, Araújo J, Gan OI, Cutting SK, Murison A, Krivdova G, Azkanaz M, McLeod JL, Smith SA, Gratton BA, Marhon SA, Gabra M, Medeiros JJF, Manteghi S, Chen J, Chan-Seng-Yue M, Garcia-Prat L, Salmena L, De Carvalho DD, Abelson S, Abdelhaleem M, Chong K, Roifman M, Shannon P, Wang JCY, Hitzler JK, Chitayat D, Dick JE, Lechman ER. Mapping the cellular origin and early evolution of leukemia in Down syndrome. Science 2021; 373:eabf6202. [PMID: 34244384 DOI: 10.1126/science.abf6202] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 03/09/2021] [Accepted: 05/21/2021] [Indexed: 12/14/2022]
Abstract
Children with Down syndrome have a 150-fold increased risk of developing myeloid leukemia, but the mechanism of predisposition is unclear. Because Down syndrome leukemogenesis initiates during fetal development, we characterized the cellular and developmental context of preleukemic initiation and leukemic progression using gene editing in human disomic and trisomic fetal hematopoietic cells and xenotransplantation. GATA binding protein 1 (GATA1) mutations caused transient preleukemia when introduced into trisomy 21 long-term hematopoietic stem cells, where a subset of chromosome 21 microRNAs affected predisposition to preleukemia. By contrast, progression to leukemia was independent of trisomy 21 and originated in various stem and progenitor cells through additional mutations in cohesin genes. CD117+/KIT proto-oncogene (KIT) cells mediated the propagation of preleukemia and leukemia, and KIT inhibition targeted preleukemic stem cells.
Collapse
MESH Headings
- Animals
- Antigens, CD34/analysis
- Cell Cycle Proteins/genetics
- Cell Cycle Proteins/metabolism
- Cell Lineage
- Cell Proliferation
- Cell Transformation, Neoplastic
- Chromosomal Proteins, Non-Histone/genetics
- Chromosomes, Human, Pair 21/genetics
- Chromosomes, Human, Pair 21/metabolism
- Disease Models, Animal
- Disease Progression
- Down Syndrome/complications
- Down Syndrome/genetics
- Female
- GATA1 Transcription Factor/genetics
- GATA1 Transcription Factor/metabolism
- Hematopoiesis
- Hematopoietic Stem Cell Transplantation
- Hematopoietic Stem Cells/physiology
- Heterografts
- Humans
- Leukemia, Myeloid/genetics
- Leukemia, Myeloid/metabolism
- Leukemia, Myeloid/pathology
- Liver/embryology
- Male
- Megakaryocytes/physiology
- Mice
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Mutation
- Preleukemia/genetics
- Preleukemia/metabolism
- Preleukemia/pathology
- Protein Kinase Inhibitors/pharmacology
- Proto-Oncogene Mas
- Proto-Oncogene Proteins c-kit/analysis
- Proto-Oncogene Proteins c-kit/antagonists & inhibitors
- Cohesins
Collapse
Affiliation(s)
- Elvin Wagenblast
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada.
| | - Joana Araújo
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
- Department of Hematology, Centro Hospitalar Universitário de São João, Porto, 4200-319, Portugal
- Faculty of Medicine, University of Porto, Porto, 4200-319, Portugal
- Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, 4200-135, Portugal
- Instituto Nacional de Investigação Biomédica, University of Porto, Porto, 4200-135, Portugal
| | - Olga I Gan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Sarah K Cutting
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Alex Murison
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Gabriela Krivdova
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Maria Azkanaz
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Jessica L McLeod
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Sabrina A Smith
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Blaise A Gratton
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Sajid A Marhon
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Martino Gabra
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jessie J F Medeiros
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | - Sanaz Manteghi
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 1X8, Canada
| | - Jian Chen
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 1X8, Canada
| | - Michelle Chan-Seng-Yue
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
- Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | - Laura Garcia-Prat
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Leonardo Salmena
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Daniel D De Carvalho
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sagi Abelson
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada
| | - Mohamed Abdelhaleem
- Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Karen Chong
- The Prenatal Diagnosis and Medical Genetics Program, Department of Obstetrics and Gynecology, Mount Sinai Hospital, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Clinical and Metabolic Genetics, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Maian Roifman
- The Prenatal Diagnosis and Medical Genetics Program, Department of Obstetrics and Gynecology, Mount Sinai Hospital, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Clinical and Metabolic Genetics, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Patrick Shannon
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jean C Y Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Division of Medical Oncology and Hematology, University Health Network, Toronto, Ontario M5G 2M9, Canada
| | - Johann K Hitzler
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children Research Institute, Toronto, ON M5G 1X8, Canada
- Department of Pediatrics, University of Toronto, Toronto, ON M5G 1X8, Canada
- Division of Hematology and Oncology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - David Chitayat
- The Prenatal Diagnosis and Medical Genetics Program, Department of Obstetrics and Gynecology, Mount Sinai Hospital, University of Toronto, Toronto, ON M5S 1A8, Canada
- Division of Clinical and Metabolic Genetics, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Eric R Lechman
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada.
| |
Collapse
|
22
|
Sun L, Xu N, Shen M, Wang R, Sun Y, Zhuang J, Zhao Y, Zeng X, Zhang X. GATA2 mutation with recurrent haemophagocytic lymphohistiocytosis and panniculitis: a case report. Rheumatology (Oxford) 2021; 60:e229-e231. [PMID: 33410496 DOI: 10.1093/rheumatology/keaa913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 11/09/2020] [Accepted: 12/11/2020] [Indexed: 11/14/2022] Open
Affiliation(s)
- Luxi Sun
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education.,Department of Internal Medicine
| | - Na Xu
- Department of General Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College
| | - Min Shen
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education
| | - Rongrong Wang
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College
| | - Yang Sun
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College
| | - Junling Zhuang
- Hematological Department, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, China
| | - Yan Zhao
- Department of Rheumatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education
| | - Xuejun Zeng
- Department of General Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College
| | - Xue Zhang
- McKusick-Zhang Center for Genetic Medicine, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College
| |
Collapse
|
23
|
Fu YK, Tan Y, Wu B, Dai YT, Xu XG, Pan MM, Chen ZW, Qiao N, Wu J, Jiang L, Lu J, Chen B, Rein A, Izraeli S, Sun XJ, Huang JY, Huang QH, Chen Z, Chen SJ. Gata2-L359V impairs primitive and definitive hematopoiesis and blocks cell differentiation in murine chronic myelogenous leukemia model. Cell Death Dis 2021; 12:568. [PMID: 34078881 PMCID: PMC8173010 DOI: 10.1038/s41419-021-03826-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 05/01/2021] [Accepted: 05/11/2021] [Indexed: 02/05/2023]
Abstract
GATA2, a key transcription factor in hematopoiesis, is frequently mutated in hematopoietic malignancies. How the GATA2 mutants contribute to hematopoiesis and malignant transformation remains largely unexplored. Here, we report that Gata2-L359V mutation impeded hematopoietic differentiation in murine embryonic and adult hematopoiesis and blocked murine chronic myeloid leukemia (CML) cell differentiation. We established a Gata2-L359V knockin mouse model in which the homozygous Gata2-L359V mutation caused major defects in primitive erythropoiesis with an accumulation of erythroid precursors and severe anemia, leading to embryonic lethality around E11.5. During adult life, the Gata2-L359V heterozygous mice exhibited a notable decrease in bone marrow (BM) recovery under stress induction with cytotoxic drug 5-fluorouracil. Using RNA sequencing, it was revealed that homozygous Gata2-L359V suppressed genes related to embryonic hematopoiesis in yolk sac, while heterozygous Gata2-L359V dysregulated genes related to cell cycle and proliferation in BM Lin-Sca1+c-kit+ cells. Furthermore, through chromatin immunoprecipitation sequencing and transactivation experiments, we found that this mutation enhanced the DNA-binding capacity and transcriptional activities of Gata2, which was likely associated with the altered expression of some essential genes during embryonic and adult hematopoiesis. In mice model harboring BCR/ABL, single-cell RNA-sequencing demonstrated that Gata2-L359V induced additional gene expression profile abnormalities and partially affected cell differentiation at the early stage of myelomonocytic lineage, evidenced by the increase of granulocyte-monocyte progenitors and monocytosis. Taken together, our study unveiled that Gata2-L359V mutation induces defective hematopoietic development and blocks the differentiation of CML cells.
Collapse
Affiliation(s)
- Ya-Kai Fu
- grid.412277.50000 0004 1760 6738Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China ,grid.415869.7Present Address: Department of Rheumatology, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yun Tan
- grid.412277.50000 0004 1760 6738Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Bo Wu
- grid.412277.50000 0004 1760 6738Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China ,grid.16821.3c0000 0004 0368 8293Institute of Health Sciences, Shanghai Institutes for Biological Sciences and Graduate School, Chinese Academy of Sciences and SJTU School of Medicine, Shanghai, China
| | - Yu-Ting Dai
- grid.412277.50000 0004 1760 6738Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Xiao-Guang Xu
- grid.412277.50000 0004 1760 6738Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Meng-Meng Pan
- grid.412277.50000 0004 1760 6738Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Zhi-Wei Chen
- grid.412277.50000 0004 1760 6738Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Niu Qiao
- grid.412277.50000 0004 1760 6738Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Jing Wu
- grid.412277.50000 0004 1760 6738Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Lu Jiang
- grid.412277.50000 0004 1760 6738Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Jing Lu
- grid.412277.50000 0004 1760 6738Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Bing Chen
- grid.412277.50000 0004 1760 6738Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Avigail Rein
- grid.12136.370000 0004 1937 0546Cancer Research Center, Sheba Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shai Izraeli
- grid.12136.370000 0004 1937 0546Division of Pediatric Hemato-Oncology, Schneider Children’s Medical Center of Israel, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Xiao-Jian Sun
- grid.412277.50000 0004 1760 6738Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Jin-Yan Huang
- grid.412277.50000 0004 1760 6738Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Qiu-Hua Huang
- grid.412277.50000 0004 1760 6738Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Zhu Chen
- grid.412277.50000 0004 1760 6738Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| | - Sai-Juan Chen
- grid.412277.50000 0004 1760 6738Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University (SJTU) School of Medicine, Shanghai, China
| |
Collapse
|
24
|
Grimm J, Heckl D, Klusmann JH. Molecular Mechanisms of the Genetic Predisposition to Acute Megakaryoblastic Leukemia in Infants With Down Syndrome. Front Oncol 2021; 11:636633. [PMID: 33777792 PMCID: PMC7992977 DOI: 10.3389/fonc.2021.636633] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/12/2021] [Indexed: 01/28/2023] Open
Abstract
Individuals with Down syndrome are genetically predisposed to developing acute megakaryoblastic leukemia. This myeloid leukemia associated with Down syndrome (ML–DS) demonstrates a model of step-wise leukemogenesis with perturbed hematopoiesis already presenting in utero, facilitating the acquisition of additional driver mutations such as truncating GATA1 variants, which are pathognomonic to the disease. Consequently, the affected individuals suffer from a transient abnormal myelopoiesis (TAM)—a pre-leukemic state preceding the progression to ML–DS. In our review, we focus on the molecular mechanisms of the different steps of clonal evolution in Down syndrome leukemogenesis, and aim to provide a comprehensive view on the complex interplay between gene dosage imbalances, GATA1 mutations and somatic mutations affecting JAK-STAT signaling, the cohesin complex and epigenetic regulators.
Collapse
Affiliation(s)
- Juliane Grimm
- Pediatric Hematology and Oncology, Martin Luther University Halle-Wittenberg, Halle, Germany.,Department of Internal Medicine IV, Oncology/Hematology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Dirk Heckl
- Pediatric Hematology and Oncology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Jan-Henning Klusmann
- Pediatric Hematology and Oncology, Martin Luther University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
25
|
Barwe SP, Sidhu I, Kolb EA, Gopalakrishnapillai A. Modeling Transient Abnormal Myelopoiesis Using Induced Pluripotent Stem Cells and CRISPR/Cas9 Technology. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 19:201-209. [PMID: 33102613 PMCID: PMC7558799 DOI: 10.1016/j.omtm.2020.09.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 09/13/2020] [Indexed: 01/18/2023]
Abstract
Approximately 1%–2% of children with Down syndrome (DS) develop acute myeloid leukemia (AML) prior to age 5 years. AML in DS children (ML-DS) is characterized by the pathognomonic mutation in the gene encoding the essential hematopoietic transcription factor GATA1, resulting in N-terminally truncated short form of GATA1 (GATA1s). Trisomy 21 and GATA1s together are sufficient to induce transient abnormal myelopoiesis (TAM) exhibiting pre-leukemic characteristics. Approximately 30% of these cases progress into ML-DS by acquisition of additional somatic mutations. We employed disease modeling in vitro by the use of customizable induced pluripotent stem cells (iPSCs) to generate a TAM model. Isogenic iPSC lines derived from the fibroblasts of DS individuals with trisomy 21 and with disomy 21 were used. The CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats)/Cas9 system was used to introduce GATA1 mutation in disomic and trisomic iPSC lines. The hematopoietic stem and progenitor cells (HSPCs) derived from GATA1 mutant iPSC lines expressed GATA1s. The expression of GATA1s concomitant with loss of full-length GATA1 reduced the erythroid population, whereas it augmented megakaryoid and myeloid populations, characteristic of TAM. In conclusion, we have developed a model system representing TAM, which can be used for modeling ML-DS by stepwise introduction of additional mutations.
Collapse
Affiliation(s)
- Sonali P Barwe
- Nemours Center for Childhood Cancer Research, A.I. DuPont Hospital for Children, Wilmington, DE 19803, USA.,University of Delaware, Newark, DE 19711, USA
| | - Ishnoor Sidhu
- Nemours Center for Childhood Cancer Research, A.I. DuPont Hospital for Children, Wilmington, DE 19803, USA.,University of Delaware, Newark, DE 19711, USA
| | - E Anders Kolb
- Nemours Center for Childhood Cancer Research, A.I. DuPont Hospital for Children, Wilmington, DE 19803, USA
| | - Anilkumar Gopalakrishnapillai
- Nemours Center for Childhood Cancer Research, A.I. DuPont Hospital for Children, Wilmington, DE 19803, USA.,University of Delaware, Newark, DE 19711, USA
| |
Collapse
|
26
|
|
27
|
Schmidt L, Heyes E, Grebien F. Gain-of-Function Effects of N-Terminal CEBPA Mutations in Acute Myeloid Leukemia. Bioessays 2019; 42:e1900178. [PMID: 31867767 PMCID: PMC7115832 DOI: 10.1002/bies.201900178] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/20/2019] [Indexed: 12/12/2022]
Abstract
Mutations in the CEBPA gene are present in 10–15% of acute myeloid leukemia (AML) patients. The most frequent type of mutations leads to the expression of an N-terminally truncated variant of the transcription factor CCAAT/enhancer-binding protein alpha (C/EBPα), termed p30. While initial reports proposed that p30 represents a dominant-negative version of the wild-type C/EBPα protein, other studies show that p30 retains the capacity to actively regulate gene expression. Recent global transcriptomic and epigenomic analyses have advanced the understanding of the distinct roles of the p30 isoform in leukemogenesis. This review outlines direct and indirect effects of the C/EBPα p30 variant on oncogenic transformation of hematopoietic progenitor cells and discusses how studies of N-terminal CEBPA mutations in AML can be extrapolated to identify novel gain-of-function features in oncoproteins that arise from recurrent truncating mutations in transcription factors.
Collapse
Affiliation(s)
- Luisa Schmidt
- Institute for Medical Biochemistry, University of Veterinary Medicine, Vienna, 1210, Austria
| | - Elizabeth Heyes
- Institute for Medical Biochemistry, University of Veterinary Medicine, Vienna, 1210, Austria
| | - Florian Grebien
- Institute for Medical Biochemistry, University of Veterinary Medicine, Vienna, 1210, Austria
| |
Collapse
|
28
|
Garnett C, Cruz Hernandez D, Vyas P. GATA1 and cooperating mutations in myeloid leukaemia of Down syndrome. IUBMB Life 2019; 72:119-130. [PMID: 31769932 DOI: 10.1002/iub.2197] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 10/24/2019] [Indexed: 12/22/2022]
Abstract
Myeloid leukaemia of Down syndrome (ML-DS) is an acute megakaryoblastic/erythroid leukaemia uniquely found in children with Down syndrome (constitutive trisomy 21). It has a unique clinical course, being preceded by a pre-leukaemic condition known as transient abnormal myelopoiesis (TAM), and provides an excellent model to study multistep leukaemogenesis. Both TAM and ML-DS blasts carry acquired N-terminal truncating mutations in the erythro-megakaryocytic transcription factor GATA1. These result in exclusive production of a shorter isoform (GATA1s). The majority of TAM cases resolve spontaneously without the need for treatment; however, around 10% acquire additional cooperating mutations and transform to leukaemia, with differentiation block and clinically significant cytopenias. Transformation is driven by the acquisition of additional mutation(s), which cooperate with GATA1s to perturb normal haematopoiesis.
Collapse
Affiliation(s)
- Catherine Garnett
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, United Kingdom of Great Britain and Northern Ireland
| | - David Cruz Hernandez
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, United Kingdom of Great Britain and Northern Ireland
| | - Paresh Vyas
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
29
|
Gutiérrez L, Caballero N, Fernández-Calleja L, Karkoulia E, Strouboulis J. Regulation of GATA1 levels in erythropoiesis. IUBMB Life 2019; 72:89-105. [PMID: 31769197 DOI: 10.1002/iub.2192] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022]
Abstract
GATA1 is considered as the "master" transcription factor in erythropoiesis. It regulates at the transcriptional level all aspects of erythroid maturation and function, as revealed by gene knockout studies in mice and by genome-wide occupancies in erythroid cells. The GATA1 protein contains two zinc finger domains and an N-terminal transactivation domain. GATA1 translation results in the production of the full-length protein and of a shorter variant (GATA1s) lacking the N-terminal transactivation domain, which is functionally deficient in supporting erythropoiesis. GATA1 protein abundance is highly regulated in erythroid cells at different levels, including transcription, mRNA translation, posttranslational modifications, and protein degradation, in a differentiation-stage-specific manner. Maintaining high GATA1 protein levels is essential in the early stages of erythroid maturation, whereas downregulating GATA1 protein levels is a necessary step in terminal erythroid differentiation. The importance of maintaining proper GATA1 protein homeostasis in erythropoiesis is demonstrated by the fact that both GATA1 loss and its overexpression result in lethal anemia. Importantly, alterations in any of those GATA1 regulatory checkpoints have been recognized as an important cause of hematological disorders such as dyserythropoiesis (with or without thrombocytopenia), β-thalassemia, Diamond-Blackfan anemia, myelodysplasia, or leukemia. In this review, we provide an overview of the multilevel regulation of GATA1 protein homeostasis in erythropoiesis and of its deregulation in hematological disease.
Collapse
Affiliation(s)
- Laura Gutiérrez
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain.,Department of Medicine, Universidad de Oviedo, Oviedo, Spain
| | - Noemí Caballero
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Luis Fernández-Calleja
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Elena Karkoulia
- Institute of Molecular Biology and Biotechnology, Foundation of Research & Technology Hellas, Heraklion, Crete, Greece
| | - John Strouboulis
- Cancer Comprehensive Center, School of Cancer and Pharmaceutical Sciences, King's College London, London, United Kingdom
| |
Collapse
|
30
|
The long and the short of it. Blood 2019; 134:1565-1566. [PMID: 31698422 DOI: 10.1182/blood.2019002983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
31
|
Ling T, Crispino JD. GATA1 mutations in red cell disorders. IUBMB Life 2019; 72:106-118. [PMID: 31652397 DOI: 10.1002/iub.2177] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 09/18/2019] [Indexed: 01/01/2023]
Abstract
GATA1 is an essential regulator of erythroid cell gene expression and maturation. In its absence, erythroid progenitors are arrested in differentiation and undergo apoptosis. Much has been learned about GATA1 function through animal models, which include genetic knockouts as well as ones with decreased levels of expression. However, even greater insights have come from the finding that a number of rare red cell disorders, including Diamond-Blackfan anemia, are associated with GATA1 mutations. These mutations affect the amino-terminal zinc finger (N-ZF) and the amino-terminus of the protein, and in both cases can alter the DNA-binding activity, which is primarily conferred by the third functional domain, the carboxyl-terminal zinc finger (C-ZF). Here we discuss the role of GATA1 in erythropoiesis with an emphasis on the mutations found in human patients with red cell disorders.
Collapse
Affiliation(s)
- Te Ling
- Division of Hematology/Oncology, Northwestern University, Chicago, Illinois
| | - John D Crispino
- Division of Hematology/Oncology, Northwestern University, Chicago, Illinois
| |
Collapse
|