1
|
Xu H, Hu K, Wang Y, Cai S, Wu F, Bao J, Hu Q, Guan Y, Tao Y, Lu J. Single-cell transcriptome sequencing reveals the mechanism of Realgar improvement on erythropoiesis in mice with myelodysplastic syndrome. Cancer Cell Int 2025; 25:135. [PMID: 40200265 PMCID: PMC11978147 DOI: 10.1186/s12935-025-03768-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 03/28/2025] [Indexed: 04/10/2025] Open
Abstract
Myelodysplastic syndrome (MDS) is a malignant hematologic disorder with limited curative options, primarily reliant on hematopoietic stem cell transplantation. Anemia, a prevalent symptom of MDS, has few effective treatment strategies. Realgar, though known for its therapeutic effects on MDS, remains poorly understood in terms of its mechanism of action. In this study, both in vivo and in vitro experiments were conducted using Realgar and its primary active component, As2S2, to examine their impact on mouse erythroblasts at the single-cell level. Realgar treatment significantly altered the transcriptional profiles and cellular composition of bone marrow in mice, both in vivo and in vitro. Differentially expressed genes in erythroblasts regulated by Realgar were identified, unveiling potential regulatory functions and signaling pathways, such as heme biosynthesis, hemoglobin production, oxygen binding, IL-17 signaling, and MAPK pathways. These findings suggest that Realgar enhances the differentiation of erythroblasts in mouse bone marrow and improves overall blood cell counts. This work offers preliminary insights into Realgar's mechanisms, expands the understanding of this mineral medicine, and may inform strategies to optimize its therapeutic potential in hematologic diseases.
Collapse
Affiliation(s)
- Hao Xu
- Clinical Hematology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Hematology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Kexin Hu
- Clinical Hematology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Hematology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yanlu Wang
- Clinical Hematology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Hematology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shuyang Cai
- Clinical Hematology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Hematology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fan Wu
- Clinical Hematology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Hematology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jizhang Bao
- Clinical Hematology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Hematology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qi Hu
- Clinical Hematology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Hematology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Guan
- Clinical Hematology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Institute of Hematology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yuchen Tao
- Clinical Hematology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Institute of Hematology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Jiahui Lu
- Clinical Hematology Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
- Institute of Hematology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
2
|
Patel SB, Moskop DR, Jordan CT, Pietras EM. Understanding MDS stem cells: Advances and limitations. Semin Hematol 2024; 61:409-419. [PMID: 39472255 DOI: 10.1053/j.seminhematol.2024.09.007] [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: 06/25/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 11/10/2024]
Abstract
In work spanning several decades, extensive studies have focused on the properties of malignant stem cells that drive the pathogenesis of acute myeloid leukemia (AML). However, relatively little attention has been devoted to several serious myeloid malignancies that occur prior to the onset of frank leukemia, including myelodysplastic syndrome (MDS). Like leukemia, MDS is hypothesized to arise from a pool of immature malignant stem and progenitor cells (MDS-SCs) that serve as a reservoir for disease evolution and progression1. While multiple studies have sought to identify and characterize the biology and vulnerabilities of MDS-SCs, yet translation of scientific concepts to therapeutically impactful regimens has been limited. Here, we evaluate the currently known properties of MDS-SCs as well as the post-transcriptional mechanisms that drive MDS pathogenesis at a stem and progenitor level. We highlight limits and gaps in our characterization and understanding of MDS-SCs and address the extent to which the properties of MDS-SC are (and can be) inferred from the characterization of LSCs.
Collapse
Affiliation(s)
- Sweta B Patel
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO
| | - Daniel R Moskop
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO
| | - Craig T Jordan
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO.
| | - Eric M Pietras
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO.
| |
Collapse
|
3
|
Eroz I, Kakkar PK, Lazar RA, El-Jawhari J. Mesenchymal Stem Cells in Myelodysplastic Syndromes and Leukaemia. Biomedicines 2024; 12:1677. [PMID: 39200142 PMCID: PMC11351218 DOI: 10.3390/biomedicines12081677] [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: 06/26/2024] [Revised: 07/17/2024] [Accepted: 07/24/2024] [Indexed: 09/01/2024] Open
Abstract
Mesenchymal stem cells (MSCs) are one of the main residents in the bone marrow (BM) and have an essential role in the regulation of haematopoietic stem cell (HSC) differentiation and proliferation. Myelodysplastic syndromes (MDSs) are a group of myeloid disorders impacting haematopoietic stem and progenitor cells (HSCPs) that are characterised by BM failure, ineffective haematopoiesis, cytopenia, and a high risk of transformation through the expansion of MDS clones together with additional genetic defects. It has been indicated that MSCs play anti-tumorigenic roles such as in cell cycle arrest and pro-tumorigenic roles including the induction of metastasis in MDS and leukaemia. Growing evidence has shown that MSCs have impaired functions in MDS, such as decreased proliferation capacity, differentiation ability, haematopoiesis support, and immunomodulation function and increased inflammatory alterations within the BM through some intracellular pathways such as Notch and Wnt and extracellular modulators abnormally secreted by MSCs, including increased expression of inflammatory factors and decreased expression of haematopoietic factors, contributing to the development and progression of MDSs. Therefore, MSCs can be targeted for the treatment of MDSs and leukaemia. However, it remains unclear what drives MSCs to behave abnormally. In this review, dysregulations in MSCs and their contributions to myeloid haematological malignancies will be discussed.
Collapse
Affiliation(s)
- Ilayda Eroz
- Biosciences Department, School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK (P.K.K.); (R.A.L.)
| | - Prabneet Kaur Kakkar
- Biosciences Department, School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK (P.K.K.); (R.A.L.)
| | - Renal Antoinette Lazar
- Biosciences Department, School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK (P.K.K.); (R.A.L.)
| | - Jehan El-Jawhari
- Biosciences Department, School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK (P.K.K.); (R.A.L.)
- Clinical Pathology Department, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
4
|
Mina A, Pavletic S, Aplan PD. The evolution of preclinical models for myelodysplastic neoplasms. Leukemia 2024; 38:683-691. [PMID: 38396286 PMCID: PMC10997513 DOI: 10.1038/s41375-024-02181-2] [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: 09/20/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024]
Abstract
Myelodysplastic Neoplasms (MDS) are a group of clonal disorders characterized by ineffective hematopoiesis and morphologic dysplasia. Clinical manifestations of MDS vary widely and are dictated in large part by a range of genetic aberrations. The lack of robust in vitro models for MDS has limited the ability to conduct high throughput drug screens, which in turn has hampered the development of novel therapies for MDS. There are very few well-characterized MDS cell lines, and the available cell lines expand poorly in vitro. Conventional xenograft mouse models can provide an in vivo vessel to provide growth of cancer cells, but human MDS cells engraft poorly. Three-dimensional (3D) scaffold models that form human "ossicles" represent a promising new approach and can reproduce the intricate communication between hematopoietic stem and progenitor cells and their environment. Genetically engineered mice utilize specific mutations and may not represent the entire array of human MDS; however, genetically engineered mice provided in vivo proof of principle for novel agents such as luspatercept, demonstrating the clinical utility of this approach. This review offers an overview of available preclinical MDS models and potential approaches to accelerate accurate clinical translation.
Collapse
Affiliation(s)
- Alain Mina
- Myeloid Malignancies Program, Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Steven Pavletic
- Myeloid Malignancies Program, Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Peter D Aplan
- Myeloid Malignancies Program, Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
5
|
Chen GW, Chen MN, Liu L, Zheng YY, Wang JP, Gong SS, Huang RF, Fan CM, Chen YZ. A research review of experimental animal models with myelodysplastic syndrome. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:105-113. [PMID: 36068448 DOI: 10.1007/s12094-022-02931-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/22/2022] [Indexed: 01/07/2023]
Abstract
Myelodysplastic syndrome (MDS) consists of a group of hematologic tumors that are derived from the clonal proliferation of hematopoietic stem cells, featuring abnormal hematopoietic cell development and ineffective hematopoiesis. Animal models are an important scientific research platform that has been widely applied in the research of human diseases, especially tumors. Animal models with MDS can simulate characteristic human genetic variations and tumor phenotypes. They also provide a reliable platform for the exploration of the pathogenesis and diagnostic markers of MDS as well as for a drug efficacy evaluation. This paper reviews the research status of three animal models and a new spontaneous mouse model with MDS.
Collapse
Affiliation(s)
- Gen-Wang Chen
- Clinical Lab and Medical Diagnostics Laboratory, Donghai Hospital District, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Mei-Na Chen
- Clinical Lab, Quanzhou Hospital of Traditional Chinese Medicine, Quanzhou, 362000, China
| | - Lei Liu
- Clinical Lab and Medical Diagnostics Laboratory, Donghai Hospital District, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Yu-Yu Zheng
- Clinical Lab and Medical Diagnostics Laboratory, Donghai Hospital District, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Jin-Peng Wang
- Clinical Lab and Medical Diagnostics Laboratory, Donghai Hospital District, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Si-Si Gong
- Clinical Lab and Medical Diagnostics Laboratory, Donghai Hospital District, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Rong-Fu Huang
- Clinical Lab and Medical Diagnostics Laboratory, Donghai Hospital District, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China
| | - Chun-Mei Fan
- Clinical Lab and Medical Diagnostics Laboratory, Donghai Hospital District, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, 362000, China.
| | - Yue-Zu Chen
- Clinical Lab, Quanzhou Hospital of Traditional Chinese Medicine, Quanzhou, 362000, China
| |
Collapse
|
6
|
Mei Y, Ren K, Liu Y, Ma A, Xia Z, Han X, Li E, Tariq H, Bao H, Xie X, Zou C, Zhang D, Li Z, Dong L, Verma A, Lu X, Abaza Y, Altman JK, Sukhanova M, Yang J, Ji P. Bone marrow confined IL-6 signaling mediates the progression of myelodysplastic syndromes to acute myeloid leukemia. J Clin Invest 2022; 132:152673. [PMID: 35900794 PMCID: PMC9435651 DOI: 10.1172/jci152673] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 07/14/2022] [Indexed: 11/17/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are age-related myeloid neoplasms with increased risk of progression to acute myeloid leukemia (AML). The mechanisms of transformation of MDS to AML are poorly understood, especially in relation to the aging microenvironment. We previously established an mDia1/miR-146a double knockout (DKO) mouse model phenocopying MDS. These mice develop age-related pancytopenia with oversecretion of proinflammatory cytokines. Here, we found that most of the DKO mice underwent leukemic transformation at 12–14 months of age. These mice showed myeloblast replacement of fibrotic bone marrow and widespread leukemic infiltration. Strikingly, depletion of IL-6 in these mice largely rescued the leukemic transformation and markedly extended survival. Single-cell RNA sequencing analyses revealed that DKO leukemic mice had increased monocytic blasts that were reduced with IL-6 knockout. We further revealed that the levels of surface and soluble IL-6 receptor (IL-6R) in the bone marrow were significantly increased in high-risk MDS patients. Similarly, IL-6R was also highly expressed in older DKO mice. Blocking of IL-6 signaling significantly ameliorated AML progression in the DKO model and clonogenicity of CD34-positive cells from MDS patients. Our study establishes a mouse model of progression of age-related MDS to AML and indicates the clinical significance of targeting IL-6 signaling in treating high-risk MDS.
Collapse
Affiliation(s)
- Yang Mei
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, United States of America
| | - Kehan Ren
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, United States of America
| | - Yijie Liu
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, United States of America
| | - Annabel Ma
- Department of Pathology, Northwestern University, Chicago, United States of America
| | - Zongjun Xia
- Department of Pathology, Northwestern University, Chicago, United States of America
| | - Xu Han
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, United States of America
| | - Ermin Li
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, United States of America
| | - Hamza Tariq
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, United States of America
| | - Haiyan Bao
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, United States of America
| | - Xinshu Xie
- School of Biomedical Sciences, Hunan University, Changsha, China
| | - Cheng Zou
- Biology, Hunan University, Changsha, China
| | | | | | - Lili Dong
- Biology, Hunan University, Changsha, China
| | - Amit Verma
- Department of Medicine, Albert Einstein College of Medicine, New York, United States of America
| | - Xinyan Lu
- Pathology, Northwestern University, Chicago, United States of America
| | - Yasmin Abaza
- Medicine, Northwestern University, Chicago, United States of America
| | - Jessica K Altman
- Medicine, Feinberg School of Medicine Northwestern University, Chicago, United States of America
| | - Madina Sukhanova
- Pathology, Feinberg School of Medicine Northwestern University, Chicago, United States of America
| | - Jing Yang
- Pathology, Northwestern University, Chicago, United States of America
| | - Peng Ji
- Pathology, Northwestern University, Chicago, United States of America
| |
Collapse
|
7
|
Liu W, Teodorescu P, Halene S, Ghiaur G. The Coming of Age of Preclinical Models of MDS. Front Oncol 2022; 12:815037. [PMID: 35372085 PMCID: PMC8966105 DOI: 10.3389/fonc.2022.815037] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous group of clonal bone-marrow diseases with ineffective hematopoiesis resulting in cytopenias and morphologic dysplasia of hematopoietic cells. MDS carry a wide spectrum of genetic abnormalities, ranging from chromosomal abnormalities such as deletions/additions, to recurrent mutations affecting the spliceosome, epigenetic modifiers, or transcription factors. As opposed to AML, research in MDS has been hindered by the lack of preclinical models that faithfully replicate the complexity of the disease and capture the heterogeneity. The complex molecular landscape of the disease poses a unique challenge when creating transgenic mouse-models. In addition, primary MDS cells are difficult to manipulate ex vivo limiting in vitro studies and resulting in a paucity of cell lines and patient derived xenograft models. In recent years, progress has been made in the development of both transgenic and xenograft murine models advancing our understanding of individual contributors to MDS pathology as well as the complex primary interplay of genetic and microenvironment aberrations. We here present a comprehensive review of these transgenic and xenograft models for MDS and future directions.
Collapse
Affiliation(s)
- Wei Liu
- Section of Hematology, Yale Cancer Center and Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| | - Patric Teodorescu
- Department of Oncology, The Johns Hopkins Hospital, Johns Hopkins Medicine, Baltimore, MD, United States
| | - Stephanie Halene
- Section of Hematology, Yale Cancer Center and Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| | - Gabriel Ghiaur
- Department of Oncology, The Johns Hopkins Hospital, Johns Hopkins Medicine, Baltimore, MD, United States
| |
Collapse
|
8
|
Li W, Cao L, Li M, Yang X, Zhang W, Song Z, Wang X, Zhang L, Morahan G, Qin C, Gao R. Novel spontaneous myelodysplastic syndrome mouse model. Animal Model Exp Med 2021; 4:169-180. [PMID: 34179724 PMCID: PMC8212821 DOI: 10.1002/ame2.12168] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 04/18/2021] [Indexed: 12/14/2022] Open
Abstract
Background Myelodysplastic syndrome (MDS) is a group of disorders involving hemopoietic dysfunction leading to leukemia. Although recently progress has been made in identifying underlying genetic mutations, many questions still remain. Animal models of MDS have been produced by introduction of specific mutations. However, there is no spontaneous mouse model of MDS, and an animal model to simulate natural MDS pathogenesis is urgently needed. Methods In characterizing the genetically diverse mouse strains of the Collaborative Cross (CC) we observed that one, designated JUN, had abnormal hematological traits. This strain was thus further analyzed for phenotypic and pathological identification, comparing the changes in each cell population in peripheral blood and in bone marrow. Results In a specific-pathogen free environment, mice of the JUN strain are relatively thin, with healthy appearance. However, in a conventional environment, they become lethargic, develop wrinkled yellow hair, have loose and light stools, and are prone to infections. We found that the mice were cytopenic, which was due to abnormal differentiation of multipotent bone marrow progenitor cells. These are common characteristics of MDS. Conclusions A mouse strain, JUN, was found displaying spontaneous myelodysplastic syndrome. This strain has the advantage over existing models in that it develops MDS spontaneously and is more similar to human MDS than genetically modified mouse models. JUN mice will be an important tool for pathogenesis research of MDS and for evaluation of new drugs and treatments.
Collapse
Affiliation(s)
- Weisha Li
- NHC Key Laboratory of Human Disease Comparative MedicineBeijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesInstitute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingChina
| | - Lin Cao
- NHC Key Laboratory of Human Disease Comparative MedicineBeijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesInstitute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingChina
| | - Mengyuan Li
- NHC Key Laboratory of Human Disease Comparative MedicineBeijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesInstitute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingChina
| | - Xingjiu Yang
- NHC Key Laboratory of Human Disease Comparative MedicineBeijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesInstitute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingChina
| | - Wenlong Zhang
- NHC Key Laboratory of Human Disease Comparative MedicineBeijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesInstitute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingChina
| | - Zhiqi Song
- NHC Key Laboratory of Human Disease Comparative MedicineBeijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesInstitute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingChina
| | - Xinpei Wang
- NHC Key Laboratory of Human Disease Comparative MedicineBeijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesInstitute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingChina
| | - Lingyan Zhang
- NHC Key Laboratory of Human Disease Comparative MedicineBeijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesInstitute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingChina
| | - Grant Morahan
- NHC Key Laboratory of Human Disease Comparative MedicineBeijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesInstitute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingChina
| | - Chuan Qin
- NHC Key Laboratory of Human Disease Comparative MedicineBeijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesInstitute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingChina
| | - Ran Gao
- NHC Key Laboratory of Human Disease Comparative MedicineBeijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesInstitute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingChina
| |
Collapse
|
9
|
Current State and Challenges in Development of Targeted Therapies in Myelodysplastic Syndromes (MDS). HEMATO 2021. [DOI: 10.3390/hemato2020013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Myelodysplastic syndromes (MDS) encompass a variety of myeloid neoplasms characterized by ineffective hematopoiesis. The interaction of abnormal clonal hematopoiesis and changes in the bone marrow microenvironment propagate abnormal clones. Advances in next generation sequencing has identified over 100 somatic mutations, but despite deepened understanding of the genetics of MDS, therapeutic discoveries have remained limited. To date, only five drugs have been approved for MDS: Azacitidine, Decitabine, Lenalidomide, Luspatercept, and oral Decitabine with Cedazuridine. Current strategies for low-risk MDS continue to focus on symptomatic management and correction of cytopenias, while treatment for high-risk MDS focuses on delaying progression of disease and improving survival. In this review we discuss some of the challenges in developing pre-clinical models of MDS in which to test therapeutics, the advances that have been made, and promising novel therapeutics in the pipeline.
Collapse
|
10
|
Li W, Li M, Yang X, Zhang W, Cao L, Gao R. Summary of animal models of myelodysplastic syndrome. Animal Model Exp Med 2021; 4:71-76. [PMID: 33738439 PMCID: PMC7954832 DOI: 10.1002/ame2.12144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 12/01/2020] [Indexed: 01/26/2023] Open
Abstract
Myelodysplastic syndrome (MDS) is a malignant tumor of the hematological system characterized by long-term, progressive refractory hemocytopenia. In addition, the risk of leukemia is high, and once it develops, the course of acute leukemia is short with poor curative effect. Animal models are powerful tools for studying human diseases and are highly effective preclinical platforms. Animal models of MDS can accurately show genetic aberrations and hematopoietic clone phenotypes with similar cellular features (such as impaired differentiation and increased apoptosis), and symptoms can be used to assess existing treatments. Animal models are also helpful for understanding the pathogenesis of MDS and its relationship with acute leukemia, which helps with the identification of candidate genes related to the MDS phenotype. This review summarizes the current status of animal models used to research myelodysplastic syndrome (MDS).
Collapse
Affiliation(s)
- Weisha Li
- NHC Key Laboratory of Human Disease Comparative MedicineBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS)BeijingChina
- Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingChina
| | - Mengyuan Li
- NHC Key Laboratory of Human Disease Comparative MedicineBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS)BeijingChina
- Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingChina
| | - Xingjiu Yang
- NHC Key Laboratory of Human Disease Comparative MedicineBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS)BeijingChina
- Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingChina
| | - Wenlong Zhang
- NHC Key Laboratory of Human Disease Comparative MedicineBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS)BeijingChina
- Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingChina
| | - Lin Cao
- Beijing Tongren Hospital Affiliated to Capital Medical UniversityBeijingChina
| | - Ran Gao
- NHC Key Laboratory of Human Disease Comparative MedicineBeijingChina
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesBeijingChina
- Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS)BeijingChina
- Comparative Medicine CenterPeking Union Medical College (PUMC)BeijingChina
| |
Collapse
|
11
|
Sen T, Jain M, Gram M, Mattebo A, Soneji S, Walkley CR, Singbrant S. Enhancing mitochondrial function in vivo rescues MDS-like anemia induced by pRb deficiency. Exp Hematol 2020; 88:28-41. [PMID: 32629063 DOI: 10.1016/j.exphem.2020.06.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 06/25/2020] [Accepted: 06/30/2020] [Indexed: 12/14/2022]
Abstract
Erythropoiesis is intimately coupled to cell division, and deletion of the cell cycle regulator retinoblastoma protein (pRb) causes anemia in mice. Erythroid-specific deletion of pRb has been found to result in inefficient erythropoiesis because of deregulated coordination of cell cycle exit and mitochondrial biogenesis. However, the pathophysiology remains to be fully described, and further characterization of the link between cell cycle regulation and mitochondrial function is needed. To this end we further assessed conditional erythroid-specific deletion of pRb. This resulted in macrocytic anemia, despite elevated levels of erythropoietin (Epo), and an accumulation of erythroid progenitors in the bone marrow, a phenotype strongly resembling refractory anemia associated with myelodysplastic syndromes (MDS). Using high-fractionation fluorescence-activated cell sorting analysis for improved phenotypic characterization, we illustrate that erythroid differentiation was disrupted at the orthochromatic stage. Transcriptional profiling of sequential purified populations revealed failure to upregulate genes critical for mitochondrial function such as Pgc1β, Alas2, and Abcb7 specifically at the block, together with disturbed heme production and iron transport. Notably, deregulated ABCB7 causes ring sideroblastic anemia in MDS patients, and the mitochondrial co-activator PGC1β is heterozygously lost in del5q MDS. Importantly, the anemia could be rescued through enhanced PPAR signaling in vivo via either overexpression of Pgc1β or bezafibrate administration. In conclusion, lack of pRb results in MDS-like anemia with disrupted differentiation and impaired mitochondrial function at the orthochromatic erythroblast stage. Our findings reveal for the first time a role for pRb in heme and iron regulation, and indicate that pRb-induced anemia can be rescued in vivo through therapeutic enhancement of PPAR signaling.
Collapse
Affiliation(s)
- Taha Sen
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Mayur Jain
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Magnus Gram
- Department of Clinical Sciences Lund, Pediatrics, Lund University, Skane University Hospital Lund, Lund, Sweden
| | - Alexander Mattebo
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Shamit Soneji
- Division of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Carl R Walkley
- St. Vincent's Institute of Medical Research and Department of Medicine, University of Melbourne, Fitzroy, VIC, Australia
| | - Sofie Singbrant
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
12
|
Côme C, Balhuizen A, Bonnet D, Porse BT. Myelodysplastic syndrome patient-derived xenografts: from no options to many. Haematologica 2020; 105:864-869. [PMID: 32193253 PMCID: PMC7109759 DOI: 10.3324/haematol.2019.233320] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 11/27/2019] [Indexed: 12/19/2022] Open
Affiliation(s)
- Christophe Côme
- The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Denmark.,Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark.,Danish Stem Cell Center (DanStem), Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Alexander Balhuizen
- The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Denmark.,Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark.,Danish Stem Cell Center (DanStem), Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Dominique Bonnet
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, UK
| | - Bo T Porse
- The Finsen Laboratory, Rigshospitalet, Faculty of Health Sciences, University of Copenhagen, Denmark .,Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark.,Danish Stem Cell Center (DanStem), Faculty of Health Sciences, University of Copenhagen, Denmark
| |
Collapse
|
13
|
Bereshchenko O, Lo Re O, Nikulenkov F, Flamini S, Kotaskova J, Mazza T, Le Pannérer MM, Buschbeck M, Giallongo C, Palumbo G, Li Volti G, Pazienza V, Cervinek L, Riccardi C, Krejci L, Pospisilova S, Stewart AF, Vinciguerra M. Deficiency and haploinsufficiency of histone macroH2A1.1 in mice recapitulate hematopoietic defects of human myelodysplastic syndrome. Clin Epigenetics 2019; 11:121. [PMID: 31439048 PMCID: PMC6704528 DOI: 10.1186/s13148-019-0724-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 08/12/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Epigenetic regulation is important in hematopoiesis, but the involvement of histone variants is poorly understood. Myelodysplastic syndromes (MDS) are heterogeneous clonal hematopoietic stem cell (HSC) disorders characterized by ineffective hematopoiesis. MacroH2A1.1 is a histone H2A variant that negatively correlates with the self-renewal capacity of embryonic, adult, and cancer stem cells. MacroH2A1.1 is a target of the frequent U2AF1 S34F mutation in MDS. The role of macroH2A1.1 in hematopoiesis is unclear. RESULTS MacroH2A1.1 mRNA levels are significantly decreased in patients with low-risk MDS presenting with chromosomal 5q deletion and myeloid cytopenias and tend to be decreased in MDS patients carrying the U2AF1 S34F mutation. Using an innovative mouse allele lacking the macroH2A1.1 alternatively spliced exon, we investigated whether macroH2A1.1 regulates HSC homeostasis and differentiation. The lack of macroH2A1.1 decreased while macroH2A1.1 haploinsufficiency increased HSC frequency upon irradiation. Moreover, bone marrow transplantation experiments showed that both deficiency and haploinsufficiency of macroH2A1.1 resulted in enhanced HSC differentiation along the myeloid lineage. Finally, RNA-sequencing analysis implicated macroH2A1.1-mediated regulation of ribosomal gene expression in HSC homeostasis. CONCLUSIONS Together, our findings suggest a new epigenetic process contributing to hematopoiesis regulation. By combining clinical data with a discrete mutant mouse model and in vitro studies of human and mouse cells, we identify macroH2A1.1 as a key player in the cellular and molecular features of MDS. These data justify the exploration of macroH2A1.1 and associated proteins as therapeutic targets in hematological malignancies.
Collapse
Affiliation(s)
- Oxana Bereshchenko
- Department of Medicine, Department of Philosophy, Social Sciences and Education, University of Perugia, Perugia, Italy.
| | - Oriana Lo Re
- International Clinical Research Center, St'Anne University Hospital, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Fedor Nikulenkov
- International Clinical Research Center, St'Anne University Hospital, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Sara Flamini
- Department of Medicine, Department of Philosophy, Social Sciences and Education, University of Perugia, Perugia, Italy
| | - Jana Kotaskova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine - Hematology and Oncology, Faculty of Medicine, University Hospital Brno and Masaryk University, Brno, Czech Republic
| | - Tommaso Mazza
- IRCCS Casa Sollievo della Sofferenza, Bioinformatics unit, San Giovanni Rotondo, Italy
| | - Marguerite-Marie Le Pannérer
- Josep Carreras Leukemia Research Institute (IJC), Universitat Autònoma de Barcelona, Campus ICO-Germans Trias I Pujol, Badalona, Spain
- Programme of Predictive and Personalized Medicine of Cancer, Germans Trias i Pujol Research Institute (PMPPC-IGTP), Badalona, Spain
| | - Marcus Buschbeck
- Josep Carreras Leukemia Research Institute (IJC), Universitat Autònoma de Barcelona, Campus ICO-Germans Trias I Pujol, Badalona, Spain
- Programme of Predictive and Personalized Medicine of Cancer, Germans Trias i Pujol Research Institute (PMPPC-IGTP), Badalona, Spain
| | - Cesarina Giallongo
- Division of Hematology, A.O.U. Policlinico-OVE, University of Catania, Catania, Italy
| | - Giuseppe Palumbo
- Department of Medical and Surgical Sciences and Advanced Technologies "GF Ingrassia", University of Catania, Catania, Italy
| | - Giovanni Li Volti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Valerio Pazienza
- Gastroenterology unit, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Libor Cervinek
- Department of Internal Medicine - Hematology and Oncology, Faculty of Medicine, University Hospital Brno and Masaryk University, Brno, Czech Republic
| | - Carlo Riccardi
- Department of Medicine, Department of Philosophy, Social Sciences and Education, University of Perugia, Perugia, Italy
| | - Lumir Krejci
- International Clinical Research Center, St'Anne University Hospital, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Sarka Pospisilova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Internal Medicine - Hematology and Oncology, Faculty of Medicine, University Hospital Brno and Masaryk University, Brno, Czech Republic
| | - A Francis Stewart
- Genomics, Biotechnology Center, Center for Molecular and Cellular Bioengineering, Technische Universität Dresden, Dresden, Germany
| | - Manlio Vinciguerra
- International Clinical Research Center, St'Anne University Hospital, Brno, Czech Republic.
| |
Collapse
|
14
|
Ichim CV, Dervovic DD, Chan LSA, Robertson CJ, Chesney A, Reis MD, Wells RA. The orphan nuclear receptor EAR-2 (NR2F6) inhibits hematopoietic cell differentiation and induces myeloid dysplasia in vivo. Biomark Res 2018; 6:36. [PMID: 30555701 PMCID: PMC6286615 DOI: 10.1186/s40364-018-0149-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/13/2018] [Indexed: 01/21/2023] Open
Abstract
Background In patients with myelodysplastic syndrome (MDS), bone marrow cells have an increased predisposition to apoptosis, yet MDS cells outcompete normal bone marrow (BM)-- suggesting that factors regulating growth potential may be important in MDS. We previously identified v-Erb A related-2 (EAR-2, NR2F6) as a gene involved in control of growth ability. Methods Bone marrow obtained from C57BL/6 mice was transfected with a retrovirus containing EAR-2-IRES-GFP. Ex vivo transduced cells were flow sorted. In some experiments cells were cultured in vitro, in other experiments cells were injected into lethally irradiated recipients, along with non-transduced bone marrow cells. Short-hairpin RNA silencing EAR-2 was also introduced into bone marrow cells cultured ex vivo. Results Here, we show that EAR-2 inhibits maturation of normal BM in vitro and in vivo and that EAR-2 transplant chimeras demonstrate key features of MDS. Competitive repopulation of lethally irradiated murine hosts with EAR-2-transduced BM cells resulted in increased engraftment and increased colony formation in serial replating experiments. Recipients of EAR-2-transduced grafts had hypercellular BM, erythroid dysplasia, abnormal localization of immature precursors and increased blasts; secondary transplantation resulted in acute leukemia. Animals were cytopenic, having reduced numbers of erythrocytes, monocytes and granulocytes. Suspension culture confirmed that EAR-2 inhibits granulocytic and monocytic differentiation, while knockdown induced granulocytic differentiation. We observed a reduction in the number of BFU-E and CFU-GM colonies and the size of erythroid and myeloid colonies. Serial replating of transduced hematopoietic colonies revealed extended replating potential in EAR-2-overexpressing BM, while knockdown reduced re-plating ability. EAR-2 functions by recruitment of histone deacetylases, and inhibition of differentiation in 32D cells is dependent on the DNA binding domain. Conclusions This data suggest that NR2F6 inhibits maturation of normal BM in vitro and in vivo and that the NR2F6 transplant chimera system demonstrates key features of MDS, and could provide a mouse model for MDS.
Collapse
Affiliation(s)
- Christine V Ichim
- Nuclear Exploration Inc., Palo Alto, California 94301 USA.,3Department of Medical Biophysics, University of Toronto, Sunnybrook Research Institute, Toronto, ON M4N 3M5 Canada.,4Biological Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5 Canada
| | - Dzana D Dervovic
- 4Biological Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5 Canada.,5Department of Immunology, University of Toronto, Toronto, ON M5S 1A8 Canada
| | - Lap Shu Alan Chan
- 3Department of Medical Biophysics, University of Toronto, Sunnybrook Research Institute, Toronto, ON M4N 3M5 Canada.,4Biological Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5 Canada
| | - Claire J Robertson
- 1Materials Engineering Division, Lawrence Livermore National Lab, 7000 East Ave, Livermore, CA USA
| | - Alden Chesney
- 6VCU Medical Centre, Department of Pathology, Richmond, VA 23298 USA
| | - Marciano D Reis
- 9Department of Laboratory Hematology, University Health Network, Toronto, ON M5G 2C4 Canada
| | - Richard A Wells
- 3Department of Medical Biophysics, University of Toronto, Sunnybrook Research Institute, Toronto, ON M4N 3M5 Canada.,4Biological Sciences, Sunnybrook Research Institute, Toronto, ON M4N 3M5 Canada.,6VCU Medical Centre, Department of Pathology, Richmond, VA 23298 USA.,7Department of Medicine, University of Toronto, Toronto, ON M5G 2C4 Canada.,8Department of Medical Oncology, Myelodysplastic Syndromes Program, Toronto Sunnybrook Regional Cancer Centre, Toronto, ON M4N 3M5 Canada
| |
Collapse
|
15
|
Bhagat TD, Chen S, Bartenstein M, Barlowe AT, Von Ahrens D, Choudhary GS, Tivnan P, Amin E, Marcondes AM, Sanders MA, Hoogenboezem RM, Kambhampati S, Ramachandra N, Mantzaris I, Sukrithan V, Laurence R, Lopez R, Bhagat P, Giricz O, Sohal D, Wickrema A, Yeung C, Gritsman K, Aplan P, Hochedlinger K, Yu Y, Pradhan K, Zhang J, Greally JM, Mukherjee S, Pellagatti A, Boultwood J, Will B, Steidl U, Raaijmakers MHGP, Deeg HJ, Kharas MG, Verma A. Epigenetically Aberrant Stroma in MDS Propagates Disease via Wnt/β-Catenin Activation. Cancer Res 2017; 77:4846-4857. [PMID: 28684528 DOI: 10.1158/0008-5472.can-17-0282] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 06/06/2017] [Accepted: 06/30/2017] [Indexed: 11/16/2022]
Abstract
The bone marrow microenvironment influences malignant hematopoiesis, but how it promotes leukemogenesis has not been elucidated. In addition, the role of the bone marrow stroma in regulating clinical responses to DNA methyltransferase inhibitors (DNMTi) is also poorly understood. In this study, we conducted a DNA methylome analysis of bone marrow-derived stromal cells from myelodysplastic syndrome (MDS) patients and observed widespread aberrant cytosine hypermethylation occurring preferentially outside CpG islands. Stroma derived from 5-azacytidine-treated patients lacked aberrant methylation and DNMTi treatment of primary MDS stroma enhanced its ability to support erythroid differentiation. An integrative expression analysis revealed that the WNT pathway antagonist FRZB was aberrantly hypermethylated and underexpressed in MDS stroma. This result was confirmed in an independent set of sorted, primary MDS-derived mesenchymal cells. We documented a WNT/β-catenin activation signature in CD34+ cells from advanced cases of MDS, where it associated with adverse prognosis. Constitutive activation of β-catenin in hematopoietic cells yielded lethal myeloid disease in a NUP98-HOXD13 mouse model of MDS, confirming its role in disease progression. Our results define novel epigenetic changes in the bone marrow microenvironment, which lead to β-catenin activation and disease progression of MDS. Cancer Res; 77(18); 4846-57. ©2017 AACR.
Collapse
Affiliation(s)
- Tushar D Bhagat
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Si Chen
- Erasmus Medical Center Cancer Institute, Department of Hematology and Erasmus Stem Cell Institute, Rotterdam, the Netherlands
| | | | | | - Dagny Von Ahrens
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Gaurav S Choudhary
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Patrick Tivnan
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elianna Amin
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - A Mario Marcondes
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.,Division of Oncology, University of Washington, Seattle, Washington
| | - Mathijs A Sanders
- Erasmus Medical Center Cancer Institute, Department of Hematology and Erasmus Stem Cell Institute, Rotterdam, the Netherlands
| | - Remco M Hoogenboezem
- Erasmus Medical Center Cancer Institute, Department of Hematology and Erasmus Stem Cell Institute, Rotterdam, the Netherlands
| | | | - Nandini Ramachandra
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Iaonnis Mantzaris
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Vineeth Sukrithan
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Remi Laurence
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Robert Lopez
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Prafullla Bhagat
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Orsi Giricz
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Davendra Sohal
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | | | - Cecilia Yeung
- Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kira Gritsman
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Peter Aplan
- National Institutes of Health, Bethesda, Maryland
| | | | - Yiting Yu
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Kith Pradhan
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Jinghang Zhang
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - John M Greally
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | | | - Andrea Pellagatti
- Bloodwise Molecular Haematology Unit, NDCLS, NIHR Biomedical Research Centre, Oxford University Hospitals, Oxford, United Kingdom
| | - Jacqueline Boultwood
- Bloodwise Molecular Haematology Unit, NDCLS, NIHR Biomedical Research Centre, Oxford University Hospitals, Oxford, United Kingdom
| | - Britta Will
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Ulrich Steidl
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Marc H G P Raaijmakers
- Erasmus Medical Center Cancer Institute, Department of Hematology and Erasmus Stem Cell Institute, Rotterdam, the Netherlands.
| | - H Joachim Deeg
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. .,Division of Oncology, University of Washington, Seattle, Washington
| | | | - Amit Verma
- Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York.
| |
Collapse
|
16
|
Daw S, Chatterjee R, Law A, Law S. Analysis of hematopathology and alteration of JAK1/STAT3/STAT5 signaling axis in experimental myelodysplastic syndrome. Chem Biol Interact 2016; 260:176-185. [PMID: 27725143 DOI: 10.1016/j.cbi.2016.10.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 10/04/2016] [Accepted: 10/06/2016] [Indexed: 11/27/2022]
Abstract
Hematological disorders like myelodysplastic syndrome (MDS) may arise due to cumulative dysregulation of various signalling pathways controlling proliferation, differentiation, maturation and apoptosis of bone marrow cells. This devastating bone marrow condition can be due to consequential abnormalities in haematopoiesis as well as its supportive microenvironment. Although mutations related to JAK/STAT pathway are common in myeloproliferative neoplasms, further studies are required to fully explore the myelodysplastic scenario regarding the concerned pathway. In this study, we have investigated the JAK-STAT signalling pathway which inevitably plays a crucial role in haematopoiesis. MDS was mimicked in a mouse model with an induction of ENU in adult mice. The bone marrow of the control and MDS groups of animals were subjected to a variety of tests, including cell morphology study in peripheral blood and bone marrow, cytochemistry and histochemistry of bone marrow smears, karyotyping and flowcytometric expression analysis of the phosphorylated forms of proteins like JAK1, STAT3 and STAT5 (denoted as pJAK1, pSTAT3 and pSTAT5) and the phenotypic expression of proteins like CD45 and CD71. The results revealed that the morphology of the blood and bone marrow cells were dysplastic compared to the affected blast populations of different lineages. The expression of common leucocyte antigen CD45 was less in comparison to the expression of transferrin receptor CD71 which was increased in the ENU induced MDS mouse model. Moreover, we have observed an upregulated expression of JAK1 followed by STAT5. Therefore, we can conclude that downregulation of CD45 may have helped in the upregulation of JAK-STAT signaling and CD71 expression. This aberrant signaling may be among one of the activated signaling axes that lead to affected hematopoietic lineages in Myelodysplastic syndrome.
Collapse
Affiliation(s)
- Suchismita Daw
- Stem Cell Research and Application Unit, Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical Medicine, 108, C.R Avenue, Kolkata 700073, West Bengal, India
| | - Ritam Chatterjee
- Stem Cell Research and Application Unit, Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical Medicine, 108, C.R Avenue, Kolkata 700073, West Bengal, India
| | - Aditya Law
- Stem Cell Research and Application Unit, Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical Medicine, 108, C.R Avenue, Kolkata 700073, West Bengal, India
| | - Sujata Law
- Stem Cell Research and Application Unit, Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical Medicine, 108, C.R Avenue, Kolkata 700073, West Bengal, India.
| |
Collapse
|
17
|
Obeng EA, Chappell RJ, Seiler M, Chen MC, Campagna DR, Schmidt PJ, Schneider RK, Lord AM, Wang L, Gambe RG, McConkey ME, Ali AM, Raza A, Yu L, Buonamici S, Smith PG, Mullally A, Wu CJ, Fleming MD, Ebert BL. Physiologic Expression of Sf3b1(K700E) Causes Impaired Erythropoiesis, Aberrant Splicing, and Sensitivity to Therapeutic Spliceosome Modulation. Cancer Cell 2016; 30:404-417. [PMID: 27622333 PMCID: PMC5023069 DOI: 10.1016/j.ccell.2016.08.006] [Citation(s) in RCA: 300] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 04/29/2016] [Accepted: 08/16/2016] [Indexed: 12/20/2022]
Abstract
More than 80% of patients with the refractory anemia with ring sideroblasts subtype of myelodysplastic syndrome (MDS) have mutations in Splicing Factor 3B, Subunit 1 (SF3B1). We generated a conditional knockin mouse model of the most common SF3B1 mutation, Sf3b1(K700E). Sf3b1(K700E) mice develop macrocytic anemia due to a terminal erythroid maturation defect, erythroid dysplasia, and long-term hematopoietic stem cell (LT-HSC) expansion. Sf3b1(K700E) myeloid progenitors and SF3B1-mutant MDS patient samples demonstrate aberrant 3' splice-site selection associated with increased nonsense-mediated decay. Tet2 loss cooperates with Sf3b1(K700E) to cause a more severe erythroid and LT-HSC phenotype. Furthermore, the spliceosome modulator, E7017, selectively kills SF3B1(K700E)-expressing cells. Thus, SF3B1(K700E) expression reflects the phenotype of the mutation in MDS and may be a therapeutic target in MDS.
Collapse
Affiliation(s)
- Esther A Obeng
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Division of Hematology/Oncology, Department of Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ryan J Chappell
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | - Michelle C Chen
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Dean R Campagna
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Paul J Schmidt
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Rebekka K Schneider
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Allegra M Lord
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lili Wang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Rutendo G Gambe
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Marie E McConkey
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Abdullah M Ali
- Division of Hematology/Oncology, Columbia University Medical Center, New York, NY 10027, USA
| | - Azra Raza
- Division of Hematology/Oncology, Columbia University Medical Center, New York, NY 10027, USA
| | - Lihua Yu
- H3 Biomedicine, Inc., Cambridge, MA 03129, USA
| | | | | | - Ann Mullally
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Mark D Fleming
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Benjamin L Ebert
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
18
|
Jonas BA, Johnson C, Gratzinger D, Majeti R. Alkylator-Induced and Patient-Derived Xenograft Mouse Models of Therapy-Related Myeloid Neoplasms Model Clinical Disease and Suggest the Presence of Multiple Cell Subpopulations with Leukemia Stem Cell Activity. PLoS One 2016; 11:e0159189. [PMID: 27428079 PMCID: PMC4948781 DOI: 10.1371/journal.pone.0159189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 06/03/2016] [Indexed: 11/19/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous group of aggressive bone marrow cancers arising from transformed hematopoietic stem and progenitor cells (HSPC). Therapy-related AML and MDS (t-AML/MDS) comprise a subset of AML cases occurring after exposure to alkylating chemotherapy and/or radiation and are associated with a very poor prognosis. Less is known about the pathogenesis and disease-initiating/leukemia stem cell (LSC) subpopulations of t-AML/MDS compared to their de novo counterparts. Here, we report the development of mouse models of t-AML/MDS. First, we modeled alkylator-induced t-AML/MDS by exposing wild type adult mice to N-ethyl-N-nitrosurea (ENU), resulting in several models of AML and MDS that have clinical and pathologic characteristics consistent with human t-AML/MDS including cytopenia, myelodysplasia, and shortened overall survival. These models were limited by their inability to transplant clinically aggressive disease. Second, we established three patient-derived xenograft models of human t-AML. These models led to rapidly fatal disease in recipient immunodeficient xenografted mice. LSC activity was identified in multiple HSPC subpopulations suggesting there is no canonical LSC immunophenotype in human t-AML. Overall, we report several new t-AML/MDS mouse models that could potentially be used to further define disease pathogenesis and test novel therapeutics.
Collapse
Affiliation(s)
- Brian A. Jonas
- Department of Internal Medicine, Division of Hematology and Oncology, University of California Davis Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, CA, United States of America
| | - Carl Johnson
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Dita Gratzinger
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Ravindra Majeti
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States of America
- * E-mail:
| |
Collapse
|
19
|
Yao H, Goldman DC, Fan G, Mandel G, Fleming WH. The Corepressor Rcor1 Is Essential for Normal Myeloerythroid Lineage Differentiation. Stem Cells 2015; 33:3304-14. [PMID: 26119982 DOI: 10.1002/stem.2086] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 06/05/2015] [Accepted: 06/11/2015] [Indexed: 12/15/2022]
Abstract
Based on its physical interactions with histone-modifying enzymes, the transcriptional corepressor Rcor1 has been implicated in the epigenetic regulation blood cell development. Previously, we have demonstrated that Rcor1 is essential for the maturation of definitive erythroid cells and fetal survival. To determine the functional role of Rcor1 in steady-state hematopoiesis in the adult, we used a conditional knockout approach. Here, we show that the loss of Rcor1 expression results in the rapid onset of severe anemia due to a complete, cell autonomous block in the maturation of committed erythroid progenitors. By contrast, both the frequency of megakaryocyte progenitors and their capacity to produce platelets were normal. Although the frequency of common lymphoid progenitors and T cells was not altered, B cells were significantly reduced and showed increased apoptosis. However, Rcor1-deficient bone marrow sustained normal levels of B-cells following transplantation, indicating a non-cell autonomous requirement for Rcor1 in B-cell survival. Evaluation of the myelomonocytic lineage revealed an absence of mature neutrophils and a significant increase in the absolute number of monocytic cells. Rcor1-deficient monocytes were less apoptotic and showed ∼100-fold more colony-forming activity than their normal counterparts, but did not give rise to leukemia. Moreover, Rcor1(-/-) monocytes exhibited extensive, cytokine-dependent self-renewal and overexpressed genes associated with hematopoietic stem/progenitor cell expansion including Gata2, Meis1, and Hoxa9. Taken together, these data demonstrate that Rcor1 is essential for the normal differentiation of myeloerythroid progenitors and for appropriately regulating self-renewal activity in the monocyte lineage.
Collapse
Affiliation(s)
- Huilan Yao
- Vollum Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Devorah C Goldman
- Department of Pediatrics, Oregon Health & Science University, Portland, Oregon, USA.,Oregon Stem Cell Center, Oregon Health & Science University, Portland, Oregon, USA.,Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Guang Fan
- Department of Pathology, Oregon Health & Science University, Portland, Oregon, USA
| | - Gail Mandel
- Vollum Institute, Oregon Health & Science University, Portland, Oregon, USA.,Howard Hughes Medical Institute, Portland, Oregon, USA
| | - William H Fleming
- Department of Pediatrics, Oregon Health & Science University, Portland, Oregon, USA.,Oregon Stem Cell Center, Oregon Health & Science University, Portland, Oregon, USA.,Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
20
|
Revisiting the case for genetically engineered mouse models in human myelodysplastic syndrome research. Blood 2015; 126:1057-68. [PMID: 26077396 DOI: 10.1182/blood-2015-01-624239] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 06/01/2015] [Indexed: 01/11/2023] Open
Abstract
Much-needed attention has been given of late to diseases specifically associated with an expanding elderly population. Myelodysplastic syndrome (MDS), a hematopoietic stem cell-based blood disease, is one of these. The lack of clear understanding of the molecular mechanisms underlying the pathogenesis of this disease has hampered the development of efficacious therapies, especially in the presence of comorbidities. Mouse models could potentially provide new insights into this disease, although primary human MDS cells grow poorly in xenografted mice. This makes genetically engineered murine models a more attractive proposition, although this approach is not without complications. In particular, it is unclear if or how myelodysplasia (abnormal blood cell morphology), a key MDS feature in humans, presents in murine cells. Here, we evaluate the histopathologic features of wild-type mice and 23 mouse models with verified myelodysplasia. We find that certain features indicative of myelodysplasia in humans, such as Howell-Jolly bodies and low neutrophilic granularity, are commonplace in healthy mice, whereas other features are similarly abnormal in humans and mice. Quantitative hematopoietic parameters, such as blood cell counts, are required to distinguish between MDS and related diseases. We provide data that mouse models of MDS can be genetically engineered and faithfully recapitulate human disease.
Collapse
|
21
|
Humeniuk R, Koller R, Bies J, Aplan P, Wolff L. Brief report: Loss of p15Ink4b accelerates development of myeloid neoplasms in Nup98-HoxD13 transgenic mice. Stem Cells 2014; 32:1361-6. [PMID: 24449168 DOI: 10.1002/stem.1635] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Accepted: 11/13/2013] [Indexed: 12/26/2022]
Abstract
Homeostasis of hematopoietic stem and progenitor cells is a tightly regulated process. The disturbance of the balance in the hematopoietic progenitor pool can result in favorable conditions for development of diseases such as myelodysplastic syndromes and leukemia. It has been shown recently that mice lacking p15Ink4b have skewed differentiation of common myeloid progenitors toward the myeloid lineage at the expense of erythroid progenitors. The lack of p15INK4B expression in human leukemic blasts has been linked to poor prognosis and increased risk of myelodysplastic syndromes transformation to acute myeloid leukemia. However, the role of p15Ink4b in disease development is just beginning to be elucidated. This study examines the collaboration of the loss of p15Ink4b with Nup98-HoxD13 translocation in the development of hematological malignancies in a mouse model. Here, we report that loss of p15Ink4b collaborates with Nup98-HoxD13 transgene in the development of predominantly myeloid neoplasms, namely acute myeloid leukemia, myeloproliferative disease, and myelodysplastic syndromes. This mouse model could be a very valuable tool for studying p15Ink4b function in tumorigenesis as well as preclinical drug testing.
Collapse
Affiliation(s)
- Rita Humeniuk
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | |
Collapse
|
22
|
Visconte V, Tabarroki A, Zhang L, Parker Y, Hasrouni E, Mahfouz R, Isono K, Koseki H, Sekeres MA, Saunthararajah Y, Barnard J, Lindner D, Rogers HJ, Tiu RV. Splicing factor 3b subunit 1 (Sf3b1) haploinsufficient mice display features of low risk Myelodysplastic syndromes with ring sideroblasts. J Hematol Oncol 2014; 7:89. [PMID: 25481243 PMCID: PMC4266210 DOI: 10.1186/s13045-014-0089-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 11/15/2014] [Indexed: 12/18/2022] Open
Abstract
Background The presence of somatic mutations in splicing factor 3b subunit 1 (SF3B1) in patients with Myelodysplastic syndromes with ring sideroblasts (MDS-RS) highlights the importance of the RNA-splicing machinery in MDS. We previously reported the presence of bone marrow (BM) RS in Sf3b1 heterozygous (Sf3b1+/−) mice which are rarely found in mouse models of MDS. Sf3b1+/− mice were originally engineered to study the interaction between polycomb genes and other proteins. Methods We used routine blood tests and histopathologic analysis of BM, spleen, and liver to evaluate the hematologic and morphologic characteristics of Sf3b1+/− mice in the context of MDS by comparing the long term follow-up (15 months) of Sf3b1+/− and Sf3b1+/+ mice. We then performed a comprehensive RNA-sequencing analysis to evaluate the transcriptome of BM cells from Sf3b1+/− and Sf3b1+/+ mice. Results Sf3b1+/− exhibited macrocytic anemia (MCV: 49.5 ± 1.6 vs 47.2 ± 1.4; Hgb: 5.5 ± 1.7 vs 7.2 ± 1.0) and thrombocytosis (PLTs: 911.4 ± 212.1 vs 878.4 ± 240.9) compared to Sf3b1+/+ mice. BM analysis showed dyserythropoiesis and occasional RS in Sf3b1+/− mice. The splenic architecture showed increased megakaryocytes with hyperchromatic nuclei, and evidence of extramedullary hematopoiesis. RNA-sequencing showed higher expression of a gene set containing Jak2 in Sf3b1+/− compared to Sf3b1+/+. Conclusions Our study indicates that Sf3b1+/− mice manifest features of low risk MDS-RS and may be relevant for preclinical therapeutic studies. Electronic supplementary material The online version of this article (doi:10.1186/s13045-014-0089-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Valeria Visconte
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, 9500 Euclid Avenue R40, Cleveland, OH, USA, 44195.
| | - Ali Tabarroki
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, 9500 Euclid Avenue R40, Cleveland, OH, USA, 44195.
| | - Li Zhang
- Department of Medicine, University of California, School of Medicine, San Francisco, CA, USA.
| | - Yvonne Parker
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, 9500 Euclid Avenue R40, Cleveland, OH, USA, 44195.
| | - Edy Hasrouni
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, 9500 Euclid Avenue R40, Cleveland, OH, USA, 44195.
| | - Reda Mahfouz
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, 9500 Euclid Avenue R40, Cleveland, OH, USA, 44195.
| | - Kyoichi Isono
- Center for Integrative Medical Sciences (IMS), RIKEN, Yokohama Institute, Yokohama, Japan.
| | - Haruhiko Koseki
- Center for Integrative Medical Sciences (IMS), RIKEN, Yokohama Institute, Yokohama, Japan.
| | - Mikkael A Sekeres
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, 9500 Euclid Avenue R40, Cleveland, OH, USA, 44195. .,Leukemia Program, Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA.
| | - Yogen Saunthararajah
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, 9500 Euclid Avenue R40, Cleveland, OH, USA, 44195. .,Leukemia Program, Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA.
| | - John Barnard
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA.
| | - Daniel Lindner
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, 9500 Euclid Avenue R40, Cleveland, OH, USA, 44195.
| | - Heesun J Rogers
- Department of Laboratory Medicine, Cleveland Clinic, Cleveland, OH, USA.
| | - Ramon V Tiu
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, 9500 Euclid Avenue R40, Cleveland, OH, USA, 44195. .,Leukemia Program, Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
23
|
Jhanwar SC. Genetic and epigenetic pathways in myelodysplastic syndromes: A brief overview. Adv Biol Regul 2014; 58:28-37. [PMID: 25499150 DOI: 10.1016/j.jbior.2014.11.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 11/11/2014] [Accepted: 11/12/2014] [Indexed: 12/24/2022]
Abstract
Myelodysplastic syndromes (MDS) are a highly heterogenous group of hematopoietic tumors, mainly due to variable clinical features and diverse set of cytogenetic, molecular genetic and epigenetic lesions. The major clinical features of MDS are ineffective hematopoiesis, peripheral cytopenias, and an increased risk of transformation to acute myeloid leukemias, which in turn is most likely determined by specific genetic abnormalities and other presenting hematologic features. The risk of developing MDS is relatively higher in some genetic syndromes such as Fanconi anemia and receipt of chemotherapy and radiation treatment. In recent years a significant progress has occurred and a vast literatures has become available including the spectrum of cytogenetic abnormalities, gene mutations relating to RNA splicing machinery, epigenetic regulation of gene expression and signaling pathways associated with MDS pathogenesis, which have provided opportunities to understand the molecular mechanisms as well as employ targeted therapeutic approaches to treat MDS. The cytogenetic abnormalities detected in MDS varies from a single abnormality to complex karyotype not easily amenable to conventional cytogenetic analysis. In such cases, array based high resolution genomic analysis detected abnormalities, which are diagnostic as well as prognostic. The most common driver gene mutations detected in patients with MDS include RNA splicing (SF3B1,SRSF2,U2F1,ZRSR2), DNA methylation (TET2,DNMT3A,IDH1/IDH2), chromatin modification (ASXL1,EZH2), transcription regulation (RUNX1,BCOR) and DNA repair control p53. A small subset of MDS arise due to deregulation of RAS pathway, mainly due to NRAS/KRAS/NF1 mutations. Identification of these mutations and pathways have provided opportunities for oncologists to target these patients with specific therapies. Several drugs which either target the spliceosome, oncogenic RAS signaling, or hypomethylating agents have been employed to successfully treat MDS patients.
Collapse
Affiliation(s)
- Suresh C Jhanwar
- Departments of Pathology and Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
24
|
Vassen L, Dührsen U, Kosan C, Zeng H, Möröy T. Growth factor independence 1 (Gfi1) regulates cell-fate decision of a bipotential granulocytic-monocytic precursor defined by expression of Gfi1 and CD48. AMERICAN JOURNAL OF BLOOD RESEARCH 2012; 2:228-242. [PMID: 23226623 PMCID: PMC3512177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 11/15/2012] [Indexed: 06/01/2023]
Abstract
The transcriptional repressor Gfi1 regulates the expression of genes important for survival, proliferation and differentiation of hematopoietic cells. Gfi1 deficient mice are severely neutropenic and accumulate ill-defined CD11b(+)GR1(int) myeloid cells. Here we show that Gfi1 expression levels determine mono- or granulocytic lineage choice in precursor cells. In addition, we identify CD48 as a cell surface marker which enables a better definition of monocytes and granulocytes in mouse bone marrow. Using the CD48/Gr1/Gfi1 marker combination we can show that the CD11b(+)GR1(int) cells accumulating in Gfi1 deficient mice are monocytes and not granulocyte precursors. Expression of CD48, Gr1 and Gfi1 define different bone marrow subpopulations that are either committed to the granulocytic lineage, or bipotential precursors of granulocytes or monocytes. Finally, a comparison of genes differentially expressed between murine Gfi1 high granulocytic precursors and mature granulocytes with gene expression changes from human myeloblasts versus neutrophils show a strong resemblance of human and mouse differentiation pathways. This underlines the value of the markers CD48 and Gfi1 identified here to study human and murine granulo-monocytic differentiation.
Collapse
Affiliation(s)
- Lothar Vassen
- Institut de recherches cliniques de Montréal IRCMMontreal, QC, Canada
| | - Ulrich Dührsen
- Westdeutsches Tumorzentrum Klinik für Hämatologie Universitätsklinikum Essen Hufelandstrasse 55 45122 EssenGermany
| | - Christian Kosan
- Institut de recherches cliniques de Montréal IRCMMontreal, QC, Canada
- present address: Center for Molecular Biomedicine (CMB), Department of Biochemistry, Friedrich-Schiller-University JenaHans-Knöll-Str. 2, D-07745 Jena
| | - Hui Zeng
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical UniversityBeijing, China
| | - Tarik Möröy
- Institut de recherches cliniques de Montréal IRCMMontreal, QC, Canada
- Département de Microbiologie et Immunologie, Université de MontréalMontréal, QC, H2W1R7 Canada
| |
Collapse
|
25
|
Beurlet S, Chomienne C, Padua RA. Engineering mouse models with myelodysplastic syndrome human candidate genes; how relevant are they? Haematologica 2012; 98:10-22. [PMID: 23065517 DOI: 10.3324/haematol.2012.069385] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Myelodysplastic syndromes represent particularly challenging hematologic malignancies that arise from a large spectrum of genetic events resulting in a disease characterized by a range of different presentations and outcomes. Despite efforts to classify and identify the key genetic events, little improvement has been made in therapies that will increase patient survival. Animal models represent powerful tools to model and study human diseases and are useful pre-clinical platforms. In addition to enforced expression of candidate oncogenes, gene inactivation has allowed the consequences of the genetic effects of human myelodysplastic syndrome to be studied in mice. This review aims to examine the animal models expressing myelodysplastic syndrome-associated genes that are currently available and to highlight the most appropriate model to phenocopy myelodysplastic syndrome disease and its risk of transformation to acute myelogenous leukemia.
Collapse
|
26
|
Dey A, Seshasayee D, Noubade R, French DM, Liu J, Chaurushiya MS, Kirkpatrick DS, Pham VC, Lill JR, Bakalarski CE, Wu J, Phu L, Katavolos P, LaFave LM, Abdel-Wahab O, Modrusan Z, Seshagiri S, Dong K, Lin Z, Balazs M, Suriben R, Newton K, Hymowitz S, Garcia-Manero G, Martin F, Levine RL, Dixit VM. Loss of the tumor suppressor BAP1 causes myeloid transformation. Science 2012; 337:1541-6. [PMID: 22878500 DOI: 10.1126/science.1221711] [Citation(s) in RCA: 333] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
De-ubiquitinating enzyme BAP1 is mutated in a hereditary cancer syndrome with increased risk of mesothelioma and uveal melanoma. Somatic BAP1 mutations occur in various malignancies. We show that mouse Bap1 gene deletion is lethal during embryogenesis, but systemic or hematopoietic-restricted deletion in adults recapitulates features of human myelodysplastic syndrome (MDS). Knockin mice expressing BAP1 with a 3xFlag tag revealed that BAP1 interacts with host cell factor-1 (HCF-1), O-linked N-acetylglucosamine transferase (OGT), and the polycomb group proteins ASXL1 and ASXL2 in vivo. OGT and HCF-1 levels were decreased by Bap1 deletion, indicating a critical role for BAP1 in stabilizing these epigenetic regulators. Human ASXL1 is mutated frequently in chronic myelomonocytic leukemia (CMML) so an ASXL/BAP1 complex may suppress CMML. A BAP1 catalytic mutation found in a MDS patient implies that BAP1 loss of function has similar consequences in mice and humans.
Collapse
Affiliation(s)
- Anwesha Dey
- Department of Physiological Chemistry, Genentech, 1 DNA Way, South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Zimmer SN, Lemieux ME, Karia BP, Day C, Zhou T, Zhou Q, Kung AL, Suresh U, Chen Y, Kinney MC, Bishop AJR, Rebel VI. Mice heterozygous for CREB binding protein are hypersensitive to γ-radiation and invariably develop myelodysplastic/myeloproliferative neoplasm. Exp Hematol 2011; 40:295-306.e5. [PMID: 22198154 DOI: 10.1016/j.exphem.2011.12.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2011] [Revised: 12/04/2011] [Accepted: 12/06/2011] [Indexed: 11/29/2022]
Abstract
Myelodysplastic syndrome is a complex family of preleukemic diseases in which hematopoietic stem cell defects lead to abnormal differentiation in one or more blood lineages. Disease progression is associated with increasing genomic instability and a large proportion of patients go on to develop acute myeloid leukemia. Primarily a disease of the elderly, it can also develop after chemotherapy. We have previously reported that CREB binding protein (Crebbp) heterozygous mice have an increased incidence of hematological malignancies, and others have shown that CREBBP is one of the genes altered by chromosomal translocations found in patients suffering from therapy-related myelodysplastic syndrome. This led us to investigate whether hematopoietic tumor development in Crebbp(+/-) mice is preceded by a myelodysplastic phase and whether we could uncover molecular mechanisms that might contribute to its development. We report here that Crebbp(+/-) mice invariably develop myelodysplastic/myeloproliferative neoplasm within 9 to 12 months of age. They are also hypersensitive to ionizing radiation and show a marked decrease in poly(ADP-ribose) polymerase-1 activity after irradiation. In addition, protein levels of XRCC1 and APEX1, key components of base excision repair machinery, are reduced in unirradiated Crebbp(+/-) cells or upon targeted knockdown of CREBBP levels. Our results provide validation of a novel myelodysplastic/myeloproliferative neoplasm mouse model and, more importantly, point to defective repair of DNA damage as a contributing factor to the pathogenesis of this currently incurable disease.
Collapse
Affiliation(s)
- Stephanie N Zimmer
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, TX 78229, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|