1
|
Tang X, Wang Z, Wang J, Cui S, Xu R, Wang Y. Functions and regulatory mechanisms of resting hematopoietic stem cells: a promising targeted therapeutic strategy. Stem Cell Res Ther 2023; 14:73. [PMID: 37038215 PMCID: PMC10088186 DOI: 10.1186/s13287-023-03316-5] [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: 10/10/2022] [Accepted: 03/29/2023] [Indexed: 04/12/2023] Open
Abstract
Hematopoietic stem cells (HSCs) are the common and essential precursors of all blood cells, including immune cells, and they are responsible for the lifelong maintenance and damage repair of blood tissue homeostasis. The vast majority (> 95%) of HSCs are in a resting state under physiological conditions and are only activated to play a functional role under stress conditions. This resting state affects their long-term survival and is also closely related to the lifelong maintenance of hematopoietic function; however, abnormal changes may also be an important factor leading to the decline of immune function in the body and the occurrence of diseases in various systems. While the importance of resting HSCs has attracted increasing research attention, our current understanding of this topic remains insufficient, and the direction of clinical targeted treatments is unclear. Here, we describe the functions of HSCs, analyze the regulatory mechanisms that affect their resting state, and discuss the relationship between resting HSCs and different diseases, with a view to providing guidance for the future clinical implementation of related targeted treatments.
Collapse
Affiliation(s)
- Xinyu Tang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhenzhen Wang
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369 Jingshi Road, Lixia District, Jinan, 250014, China
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong Provincial Health Commission Key Laboratory of Hematology of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jingyi Wang
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369 Jingshi Road, Lixia District, Jinan, 250014, China
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong Provincial Health Commission Key Laboratory of Hematology of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Siyuan Cui
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369 Jingshi Road, Lixia District, Jinan, 250014, China
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong Provincial Health Commission Key Laboratory of Hematology of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ruirong Xu
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369 Jingshi Road, Lixia District, Jinan, 250014, China.
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Jinan, China.
- Shandong Provincial Health Commission Key Laboratory of Hematology of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Yan Wang
- Department of Hematology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 16369 Jingshi Road, Lixia District, Jinan, 250014, China.
- Institute of Hematology, Shandong University of Traditional Chinese Medicine, Jinan, China.
- Shandong Provincial Health Commission Key Laboratory of Hematology of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
2
|
Liu Z, Tian M, Ding K, Liu H, Wang Y, Fu R. High expression of PIM2 induces HSC proliferation in myelodysplastic syndromes via the IDH1/HIF1-α signaling pathway. Oncol Lett 2019; 17:5395-5402. [PMID: 31186757 PMCID: PMC6507299 DOI: 10.3892/ol.2019.10256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 03/15/2019] [Indexed: 12/12/2022] Open
Abstract
PIM2 proto-oncogene, serine/threonine kinase (PIM2) is a serine/threonine protein kinase that is upregulated in different types of cancer and serves essential roles in the regulation of signal transduction cascades, which promote cell survival and cell proliferation. The present study demonstrated that PIM2 was highly expressed in CD34+ cells derived from the bone marrow of patients with myelodysplastic syndromes (MDS)/acute myeloid leukemia. The mRNA expression level of PIM2 was quantified in MDS cell lines and mRNA expression was significantly decreased compared with that in KG-1 cells. In vitro, downregulation of PIM2 by short interfering RNA (siRNA) inhibited cell proliferation and delayed G0/G1 cell cycle progression in the MDS cell line SKM-1. Western blotting revealed that cyclin dependent kinase 2 was markedly downregulated and cyclin dependent kinase inhibitor 1A was markedly upregulated following transfection with PIM2 siRNA. Cell Counting Kit-8 analysis demonstrated that cell proliferation of si-PIM2-transfected cells was significantly decreased compared with control cells. Reverse-transcription quantitative polymerase chain reaction and western blotting revealed that PIM2 expression was negatively correlated with isocitrate dehydrogenase [NADP(+)]1 cytosolic (IDH1) and positively correlated with hypoxia inducible factor 1 subunit α (HIF1A) in CD34+ MDS cells. Collectively, these results suggested that the expression of PIM2 induced increased expression of HIF1A by decreasing the expression of IDH1, resulting in increased CD34+ cell proliferation. Therefore, PIM2 may be a potential biomarker for the diagnosis of MDS and AML or a target for novel therapeutic agents.
Collapse
Affiliation(s)
- Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Mengyue Tian
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Kai Ding
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Hui Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yangyang Wang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
3
|
Daw S, Law A, Law S. Myelodysplastic Syndrome related alterations of MAPK signaling in the bone marrow of experimental mice including stem/progenitor compartment. Acta Histochem 2019; 121:330-343. [PMID: 30808519 DOI: 10.1016/j.acthis.2019.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/17/2019] [Accepted: 02/18/2019] [Indexed: 12/30/2022]
Abstract
Myelodysplastic syndrome is considered globally as heterogenous group of neoplasm which often proclaims leukemic progression. The heterogeneity is reflected not only in clinical manifestations of the disease but also in salient causes of disease development. In spite of multiple therapeutic modalities, shortfall towards treatment of this disorder still persists. The focal point of tussle suggested toward defects, which are not confined to any unifying cellular signalling. The pathobiology of the disease often experiences an intriguing paradox involving 'hyperproliferative bone marrow with pancytopenic peripheral blood'. In our present study we have reported about MAPK signaling in the hematopoietic stem progenitor compartmental (HSPC) dysregulation during the course of alkylator(ENU) induced myelodysplasia. The phospho-protein status of RTK's(FLT3, PDGFR, EGFR) were markedly increased that activated MAPK signaling proteins which finally executed their tasks by transcription of c-Myc and Rb leading to uncontrolled cellular proliferation, simultaneously the activated c-Jun revealed stress related apoptosis. Altogether, the role of activated MAPK signaling in the HSPC's may have led to hyperproliferation and concurrent enhanced apoptosis of abnormal cells which gradually headed towards premalignant transformations during the course of disease. The phenotypic expression of the HSPC markers CD 150 and CD 90 also established a mechanistic correlation with MAPK signalling alterations and overall scenario.
Collapse
|
4
|
Masala E, Valencia-Martinez A, Pillozzi S, Rondelli T, Brogi A, Sanna A, Gozzini A, Arcangeli A, Sbarba PD, Santini V. Severe hypoxia selects hematopoietic progenitors with stem cell potential from primary Myelodysplastic syndrome bone marrow cell cultures. Oncotarget 2018; 9:10561-10571. [PMID: 29535827 PMCID: PMC5828219 DOI: 10.18632/oncotarget.24302] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 01/13/2018] [Indexed: 01/28/2023] Open
Abstract
Myelodysplastic Syndromes (MDS) are clonal neoplasms where stem/progenitor cells endowed with self-renewal and capable of perpetuating the disease have been demonstrated. It is known that oxygen tension plays a key role in driving normal hematopoiesis and that hematopoietic stem cells are maintained in hypoxic areas of the bone marrow (BM). Hypoxia could also regulate leukemic/dysplastic hematopoiesis. We evaluated the stem cell potential of MDS cells derived from the BM of 39 MDS patients and selected under severe hypoxia. MDS cells rescued from hypoxia-incubated cultures were subjected to stem and progenitor cell assays in vitro, as well as to hematopoietic reconstitution assay in NOD-SCID mice. Incubation in severe hypoxia of cells explanted from MDS patients selected a cell subset endowed with stem cell potential, as determined in vitro. This occurred only from the BM of patients classified as IPSS low/INT-1 risk. Transplantation into NOD-SCID mice confirmed using an in vivo model that severe hypoxia selects a cell subset endowed with stem cell potential from bone marrow mononuclear cells (BMMC). derived from patients belonging to the IPSS low/int-1 risk group. Data here reported show that cells endowed with stem cell potential and capable of adapting to hypoxia and escaping hypoxia-induced apoptosis exist within MDS cell populations.
Collapse
Affiliation(s)
- Erico Masala
- MDS UNIT, Hematology, AOU-Careggi University Hospital, Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| | - Ana Valencia-Martinez
- MDS UNIT, Hematology, AOU-Careggi University Hospital, Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| | | | - Alice Brogi
- MDS UNIT, Hematology, AOU-Careggi University Hospital, Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
- Department of Medical Biotechnologies, Università degli Studi di Siena, Siena, Italy
| | - Alessandro Sanna
- MDS UNIT, Hematology, AOU-Careggi University Hospital, Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| | - Antonella Gozzini
- Cellular Therapy and Transfusional Medicine Unit, Hematology, AOU-Careggi University Hospital, Florence, Italy
| | - Annarosa Arcangeli
- Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| | - Persio Dello Sbarba
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, Università degli Studi di Firenze, Florence, Italy
| | - Valeria Santini
- MDS UNIT, Hematology, AOU-Careggi University Hospital, Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| |
Collapse
|
5
|
Goulard M, Dosquet C, Chomienne C. [Towards a personalized pretransplantation conditioning in patients with myelodysplastic syndromes]. Med Sci (Paris) 2018; 34:9-11. [PMID: 29384085 DOI: 10.1051/medsci/20183401002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Marie Goulard
- Inserm UMR-S-1131, 1, avenue Claude Vellefaux, 75010 Paris, France
| | - Christine Dosquet
- Inserm UMR-S-1131, 1, avenue Claude Vellefaux, 75010 Paris, France - APHP, Hôpital Saint-Louis, unité de biologie cellulaire, 1, avenue Claude Vellefaux, 75010 Paris, France
| | - Christine Chomienne
- Inserm UMR-S-1131, 1, avenue Claude Vellefaux, 75010 Paris, France - APHP, Hôpital Saint-Louis, unité de biologie cellulaire, 1, avenue Claude Vellefaux, 75010 Paris, France
| |
Collapse
|
6
|
|
7
|
Chung YJ, Fry TJ, Aplan PD. Myeloablative hematopoietic stem cell transplantation improves survival but is not curative in a pre-clinical model of myelodysplastic syndrome. PLoS One 2017; 12:e0185219. [PMID: 28953912 PMCID: PMC5617185 DOI: 10.1371/journal.pone.0185219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 09/10/2017] [Indexed: 11/19/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (A-HSCT) remains the only curative option for patients with myelodysplastic syndrome (MDS). We used the NUP98-HOXD13 (NHD13) murine model for MDS to study HSCT in a pre-clinical setting. NHD13 recipients transplanted with syngeneic bone marrow (S-HSCT) following myeloablative irradiation showed disease remission, with normalization of peripheral blood parameters and marked decrease in circulating leukocytes derived from the MDS clone. Despite the disease remission and improved survival compared to non-transplanted NHD13 controls, all mice eventually relapsed, indicating persistence of a long-lived radio-resistant MDS clone. In an effort to induce a graft versus leukemia (GVL) effect, A-HSCT with donor bone marrow that was mismatched at minor histocompatibility loci was compared to S-HSCT. Although recipients in the A-HSCT showed a lower early relapse rate than in S-HSCT, all mice in both groups eventually relapsed and died by 54 weeks post-transplant. To obtain a more significant GVL effect, donor splenocytes containing reactive T-cells were transplanted with allogeneic bone marrow. Although the relapse rate was only 20% at post-transplantation week 38, suggesting a GVL effect, this was accompanied by a severe graft versus host disease (GVHD) Taken together, these findings indicate that a myeloablative dose of ionizing radiation is insufficient to eradicate the MDS initiating cell, and that transplantation of donor splenocytes leads to decreased relapse rates, at the cost of severe GVHD. We suggest that NHD13 mice represent a feasible pre-clinical model for the study of HSCT for MDS.
Collapse
Affiliation(s)
- Yang Jo Chung
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Terry J. Fry
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Peter D. Aplan
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
8
|
Li L, Sheng Y, Li W, Hu C, Mittal N, Tohyama K, Seba A, Zhao YY, Ozer H, Zhu T, Qian Z. β-Catenin Is a Candidate Therapeutic Target for Myeloid Neoplasms with del(5q). Cancer Res 2017; 77:4116-4126. [PMID: 28611040 DOI: 10.1158/0008-5472.can-17-0202] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 04/19/2017] [Accepted: 06/05/2017] [Indexed: 12/16/2022]
Abstract
Deletion of the chromosome 5q [del(5q)] is one of the most common cytogenetic abnormalities observed in patients with de novo myelodysplastic syndromes (MDS) and therapy-related MDS or acute myeloid leukemia (t-MDS/tAML). Emerging evidence indicates that activation of the Wnt/β-catenin pathway contributes to the development of myeloid neoplasms with del(5q). Whether β-catenin is a potential therapeutic target for myeloid neoplasms with del(5q) has yet to be evaluated. Here, we report that genetic deletion of a single allele of β-catenin rescues ineffective hematopoiesis in an Apc haploinsufficient mouse model, which recapitulates several characteristic features of the preleukemic stage of myeloid neoplasms with a -5/del(5q). In addition, loss of a single allele of β-catenin reversed the defective self-renewal capacity of Apc-haploinsufficient hematopoietic stem cells and reduced the frequency of apoptosis induced by Apc haploinsufficiency. Suppression of β-catenin by indomethacin or β-catenin shRNA reduced proliferation and survival of human leukemia cell lines with del(5q) but not of control leukemia cell lines in vitro; β-catenin inactivation also inhibited leukemia progression in vivo in xenograft mice reconstituted with del(5q) leukemia cell lines. Inhibition of β-catenin also stunted growth and colony-forming abilities of primary bone marrow cells from del(5q) AML patients in vitro Overall, our data support the idea that β-catenin could serve as a therapeutic target for the treatment of myeloid neoplasms with del(5q). Cancer Res; 77(15); 4116-26. ©2017 AACR.
Collapse
Affiliation(s)
- Liping Li
- Department of Medicine, Division of Hematology/Oncology, University of Illinois at Chicago, Chicago, Illinois.,Fudan University Zhong Shan Hospital, Shanghai, China
| | - Yue Sheng
- Department of Medicine, Division of Hematology/Oncology, University of Illinois at Chicago, Chicago, Illinois
| | - Wenshu Li
- College of Arts and Sciences, Shanghai New York University, Shanghai, China
| | - Chao Hu
- Department of Medicine, Division of Hematology/Oncology, University of Illinois at Chicago, Chicago, Illinois.,Fudan University Zhong Shan Hospital, Shanghai, China
| | - Nupur Mittal
- Department of Pediatrics, Division of Pediatric Hematology Oncology, University of Illinois at Chicago (Fellow, UIC-Rush-Stroger Fellowship Program, Chicago), Chicago, Illinois
| | - Kaoru Tohyama
- Department of Laboratory Medicine, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Amber Seba
- Department of Medicine, Division of Hematology/Oncology, University of Illinois at Chicago, Chicago, Illinois
| | - You-Yang Zhao
- Department of Pharmacology, University of Illinois at Chicago, Chicago, Illinois
| | - Howard Ozer
- Department of Medicine, Division of Hematology/Oncology, University of Illinois at Chicago, Chicago, Illinois
| | - Tongyu Zhu
- Fudan University Zhong Shan Hospital, Shanghai, China
| | - Zhijian Qian
- Department of Medicine, Division of Hematology/Oncology, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
9
|
Guezguez B, Almakadi M, Benoit YD, Shapovalova Z, Rahmig S, Fiebig-Comyn A, Casado FL, Tanasijevic B, Bresolin S, Masetti R, Doble BW, Bhatia M. GSK3 Deficiencies in Hematopoietic Stem Cells Initiate Pre-neoplastic State that Is Predictive of Clinical Outcomes of Human Acute Leukemia. Cancer Cell 2016; 29:61-74. [PMID: 26766591 DOI: 10.1016/j.ccell.2015.11.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 04/23/2015] [Accepted: 11/17/2015] [Indexed: 01/20/2023]
Abstract
Initial pathway alternations required for pathogenesis of human acute myeloid leukemia (AML) are poorly understood. Here we reveal that removal of glycogen synthase kinase-3α (GSK-3α) and GSK-3β dependency leads to aggressive AML. Although GSK-3α deletion alone has no effect, GSK-3β deletion in hematopoietic stem cells (HSCs) resulted in a pre-neoplastic state consistent with human myelodysplastic syndromes (MDSs). Transcriptome and functional studies reveal that each GSK-3β and GSK-3α uniquely contributes to AML by affecting Wnt/Akt/mTOR signaling and metabolism, respectively. The molecular signature of HSCs deleted for GSK-3β provided a prognostic tool for disease progression and survival of MDS patients. Our study reveals that GSK-3α- and GSK-3β-regulated pathways can be responsible for stepwise transition to MDS and subsequent AML, thereby providing potential therapeutic targets of disease evolution.
Collapse
Affiliation(s)
- Borhane Guezguez
- McMaster Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, Hamilton, ON L8N 3Z5, Canada
| | - Mohammed Almakadi
- McMaster Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, Hamilton, ON L8N 3Z5, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada; Department of Oncology, Juravinski Cancer Center, Faculty of Health Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Yannick D Benoit
- McMaster Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, Hamilton, ON L8N 3Z5, Canada
| | - Zoya Shapovalova
- McMaster Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, Hamilton, ON L8N 3Z5, Canada
| | - Susann Rahmig
- McMaster Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, Hamilton, ON L8N 3Z5, Canada
| | - Aline Fiebig-Comyn
- McMaster Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, Hamilton, ON L8N 3Z5, Canada
| | - Fanny L Casado
- McMaster Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, Hamilton, ON L8N 3Z5, Canada
| | - Borko Tanasijevic
- McMaster Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, Hamilton, ON L8N 3Z5, Canada
| | - Silvia Bresolin
- Department of Women's and Children's Health, University of Padova, Padua, Italy
| | - Riccardo Masetti
- Department of Pediatric Oncology and Hematology, University of Bologna, Bologna, Italy
| | - Bradley W Doble
- McMaster Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, Hamilton, ON L8N 3Z5, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Mickie Bhatia
- McMaster Stem Cell and Cancer Research Institute, Michael G. DeGroote School of Medicine, Hamilton, ON L8N 3Z5, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada; McMaster Stem Cell and Cancer Research Institute (SCC-RI), Michael G. DeGroote School of Medicine, McMaster University, 1280 Main Street West, MDCL 5029, Hamilton, ON L8S 4K1, Canada.
| |
Collapse
|
10
|
The transcription factor Foxm1 is essential for the quiescence and maintenance of hematopoietic stem cells. Nat Immunol 2015; 16:810-8. [PMID: 26147687 PMCID: PMC4509925 DOI: 10.1038/ni.3204] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 05/19/2015] [Indexed: 12/17/2022]
Abstract
Foxm1, a mammalian Forkhead Box M1 protein, is known as a typical proliferation-associated transcription factor. Here, we find that Foxm1 was essential for maintenance of hematopoietic stem cell (HSC) quiescence and self-renewal capacity in vivo in mice. Reducing expression of FOXM1 also decreased quiescence in human CD34+ HSCs and progenitor cells and its down-regulation was associated with a subset of myelodysplastic syndrome (MDS). Mechanistically, Foxm1 directly bound to the promoter region of Nurr1, inducing transcription while forced expression of Nurr1 reversed the loss of quiescence observed in Foxm1-deficient cells in vivo. Thus, our studies reveal a previously unrecognized role of Foxm1 as a critical regulator of HSC quiescence and self-renewal, mediated at least in part, by control of Nurr1 expression.
Collapse
|
11
|
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
|
12
|
Wang J, Xiao Z. Mesenchymal stem cells in pathogenesis of myelodysplastic syndromes. Stem Cell Investig 2014; 1:16. [PMID: 27358862 DOI: 10.3978/j.issn.2306-9759.2014.08.02] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 08/18/2014] [Indexed: 01/13/2023]
Abstract
Myelodysplastic syndromes (MDS) are clonal malignant stem cell disorders characterized by inefficient hematopoiesis. The role of the marrow microenvironment in the pathogenesis of the disease has been controversial. Emerging evidence indicated that mesenchymal stem cells (MSC) derived from MDS patients were cytogenetically abnormal, and they showed a deficient hematopoietic-supportive capacity and increased production of cytokine such as tumor necrosis factor α (TNF-α), interleukin 6 (IL-6), interferon γ (IFN-γ). From the point of some evidence, the abnormal microenvironment seems to participate in the progression of the disease by contributing to the selective expansion of the malignant clone. In this review, we will discuss the most recent progress related to identification of normal MSC and the importance of the stem cell niche in development and maintenance of MDS.
Collapse
Affiliation(s)
- Jingya Wang
- 1 MDS and MPN Centre, 2 State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Zhijian Xiao
- 1 MDS and MPN Centre, 2 State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| |
Collapse
|
13
|
Sashida G, Harada H, Matsui H, Oshima M, Yui M, Harada Y, Tanaka S, Mochizuki-Kashio M, Wang C, Saraya A, Muto T, Hayashi Y, Suzuki K, Nakajima H, Inaba T, Koseki H, Huang G, Kitamura T, Iwama A. Ezh2 loss promotes development of myelodysplastic syndrome but attenuates its predisposition to leukaemic transformation. Nat Commun 2014; 5:4177. [PMID: 24953053 DOI: 10.1038/ncomms5177] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 05/21/2014] [Indexed: 01/08/2023] Open
Abstract
Loss-of-function mutations of EZH2, a catalytic component of polycomb repressive complex 2 (PRC2), are observed in ~\n10% of patients with myelodysplastic syndrome (MDS), but are rare in acute myeloid leukaemia (AML). Recent studies have shown that EZH2 mutations are often associated with RUNX1 mutations in MDS patients, although its pathological function remains to be addressed. Here we establish an MDS mouse model by transducing a RUNX1S291fs mutant into hematopoietic stem cells and subsequently deleting Ezh2. Ezh2 loss significantly promotes RUNX1S291fs-induced MDS. Despite their compromised proliferative capacity of RUNX1S291fs/Ezh2-null MDS cells, MDS bone marrow impairs normal hematopoietic cells via selectively activating inflammatory cytokine responses, thereby allowing propagation of MDS clones. In contrast, loss of Ezh2 prevents the transformation of AML via PRC1-mediated repression of Hoxa9. These findings provide a comprehensive picture of how Ezh2 loss collaborates with RUNX1 mutants in the pathogenesis of MDS in both cell autonomous and non-autonomous manners.
Collapse
Affiliation(s)
- Goro Sashida
- 1] Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan [2] JST, CREST, 7 Gobancho, Chiyoda-ku, Tokyo 102-0076, Japan
| | - Hironori Harada
- 1] Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan [2]
| | - Hirotaka Matsui
- Division of Molecular Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Motohiko Oshima
- 1] Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan [2] JST, CREST, 7 Gobancho, Chiyoda-ku, Tokyo 102-0076, Japan
| | - Makiko Yui
- 1] Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan [2] JST, CREST, 7 Gobancho, Chiyoda-ku, Tokyo 102-0076, Japan
| | - Yuka Harada
- 1] Division of Radiation Information Registry, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan [2]
| | - Satomi Tanaka
- 1] Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan [2] Department of Hematology, Chiba University Hospital, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Makiko Mochizuki-Kashio
- 1] Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan [2] JST, CREST, 7 Gobancho, Chiyoda-ku, Tokyo 102-0076, Japan
| | - Changshan Wang
- 1] Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan [2] JST, CREST, 7 Gobancho, Chiyoda-ku, Tokyo 102-0076, Japan
| | - Atsunori Saraya
- 1] Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan [2] JST, CREST, 7 Gobancho, Chiyoda-ku, Tokyo 102-0076, Japan
| | - Tomoya Muto
- 1] Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan [2] Department of Hematology, Chiba University Hospital, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Yoshihiro Hayashi
- 1] Division of Pathology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229-3026, USA [2] Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229-3026, USA
| | - Kotaro Suzuki
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Hiroshi Nakajima
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Toshiya Inaba
- Division of Molecular Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8553, Japan
| | - Haruhiko Koseki
- 1] JST, CREST, 7 Gobancho, Chiyoda-ku, Tokyo 102-0076, Japan [2] Laboratory for Lymphocyte Development, RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Gang Huang
- 1] Division of Pathology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229-3026, USA [2] Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, Ohio 45229-3026, USA
| | - Toshio Kitamura
- Division of Cellular Therapy and Division of Stem Cell Signaling, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato, Tokyo 108-8639, Japan
| | - Atsushi Iwama
- 1] Department of Cellular and Molecular Medicine, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan [2] JST, CREST, 7 Gobancho, Chiyoda-ku, Tokyo 102-0076, Japan
| |
Collapse
|
14
|
Depletion of Sf3b1 impairs proliferative capacity of hematopoietic stem cells but is not sufficient to induce myelodysplasia. Blood 2014; 123:3336-43. [DOI: 10.1182/blood-2013-12-544544] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Key Points
The level of Sf3b1 expression is critical for the proliferative capacity of hematopoietic stem cells. Haploinsufficiency for Sf3b1 is not sufficient to induce a RARS-like phenotype in mice.
Collapse
|
15
|
Walenda T, Stiehl T, Braun H, Fröbel J, Ho AD, Schroeder T, Goecke TW, Rath B, Germing U, Marciniak-Czochra A, Wagner W. Feedback signals in myelodysplastic syndromes: increased self-renewal of the malignant clone suppresses normal hematopoiesis. PLoS Comput Biol 2014; 10:e1003599. [PMID: 24763223 PMCID: PMC3998886 DOI: 10.1371/journal.pcbi.1003599] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 03/18/2014] [Indexed: 12/20/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are triggered by an aberrant hematopoietic stem cell (HSC). It is, however, unclear how this clone interferes with physiologic blood formation. In this study, we followed the hypothesis that the MDS clone impinges on feedback signals for self-renewal and differentiation and thereby suppresses normal hematopoiesis. Based on the theory that the MDS clone affects feedback signals for self-renewal and differentiation and hence suppresses normal hematopoiesis, we have developed a mathematical model to simulate different modifications in MDS-initiating cells and systemic feedback signals during disease development. These simulations revealed that the disease initiating cells must have higher self-renewal rates than normal HSCs to outcompete normal hematopoiesis. We assumed that self-renewal is the default pathway of stem and progenitor cells which is down-regulated by an increasing number of primitive cells in the bone marrow niche – including the premature MDS cells. Furthermore, the proliferative signal is up-regulated by cytopenia. Overall, our model is compatible with clinically observed MDS development, even though a single mutation scenario is unlikely for real disease progression which is usually associated with complex clonal hierarchy. For experimental validation of systemic feedback signals, we analyzed the impact of MDS patient derived serum on hematopoietic progenitor cells in vitro: in fact, MDS serum slightly increased proliferation, whereas maintenance of primitive phenotype was reduced. However, MDS serum did not significantly affect colony forming unit (CFU) frequencies indicating that regulation of self-renewal may involve local signals from the niche. Taken together, we suggest that initial mutations in MDS particularly favor aberrant high self-renewal rates. Accumulation of primitive MDS cells in the bone marrow then interferes with feedback signals for normal hematopoiesis – which then results in cytopenia. Myelodysplastic syndromes are diseases which are characterized by ineffective blood formation. There is accumulating evidence that they are caused by an aberrant hematopoietic stem cell. However, it is yet unclear how this malignant clone suppresses normal hematopoiesis. To this end, we generated mathematical models under the assumption that feedback signals regulate self-renewal and proliferation of normal and diseased stem cells. The simulations demonstrate that the malignant cells must have particularly higher self-renewal rates than normal stem cells – rather than higher proliferation rates. On the other hand, down-regulation of self-renewal by the increasing number of malignant cells in the bone marrow niche can explain impairment of normal blood formation. In fact, we show that serum of patients with myelodysplastic syndrome, as compared to serum of healthy donors, stimulates proliferation and moderately impacts on maintenance of hematopoietic stem and progenitor cells in vitro. Thus, aberrant high self-renewal rates of the malignant clone seem to initiate disease development; suppression of normal blood formation is then caused by a rebound effect of feedback signals which down-regulate self-renewal of normal stem and progenitor cells as well.
Collapse
Affiliation(s)
- Thomas Walenda
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University Medical School, Aachen, Germany
| | - Thomas Stiehl
- Interdisciplinary Center of Scientific Computing (IWR), Institute of Applied Mathematics, University of Heidelberg, Heidelberg, Germany
| | - Hanna Braun
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University Medical School, Aachen, Germany
| | - Julia Fröbel
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Anthony D. Ho
- Department of Medicine V, Medical Center, University of Heidelberg, Heidelberg, Germany
| | - Thomas Schroeder
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Tamme W. Goecke
- Department of Obstetrics and Gynecology, RWTH Aachen University Medical School, Aachen, Germany
| | - Björn Rath
- Department for Orthopedics, RWTH Aachen University Medical School, Aachen, Germany
| | - Ulrich Germing
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Anna Marciniak-Czochra
- Interdisciplinary Center of Scientific Computing (IWR), Institute of Applied Mathematics, University of Heidelberg, Heidelberg, Germany
| | - Wolfgang Wagner
- Helmholtz Institute for Biomedical Engineering, RWTH Aachen University Medical School, Aachen, Germany
- * E-mail:
| |
Collapse
|
16
|
Ramsey H, Zhang Q, Brown DE, Steensma DP, Lin CP, Wu MX. Stress-induced hematopoietic failure in the absence of immediate early response gene X-1 (IEX-1, IER3). Haematologica 2013; 99:282-91. [PMID: 24056813 DOI: 10.3324/haematol.2013.092452] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Expression of the immediate early response gene X-1 (IEX-1, IER3) is diminished significantly in hematopoietic stem cells in a subgroup of patients with early stage myelodysplastic syndromes, but it is not clear whether the deregulation contributes to the disease. The current study demonstrates increased apoptosis and a concomitant decrease in the number of hematopoietic stem cells lacking this early response gene. Null mutation of the gene also impeded platelet differentiation and shortened a lifespan of red blood cells. When bone marrow cells deficient in the gene were transplanted into wild-type mice, the deficient stem cells produced significantly fewer circulating platelets and red blood cells, despite their enhanced repopulation capability. Moreover, after exposure to a non-myeloablative dose of radiation, absence of the gene predisposed to thrombocytopenia, a significant decline in red blood cells, and dysplastic bone marrow morphology, typical characteristics of myelodysplastic syndromes. These findings highlight a previously unappreciated role for this early response gene in multiple differentiation steps within hematopoiesis, including thrombopoiesis, erythropoiesis and in the regulation of hematopoietic stem cell quiescence. The deficient mice offer a novel model for studying the initiation and progression of myelodysplastic syndromes as well as strategies to prevent this disorder.
Collapse
|
17
|
Zhao JL, Rao DS, O'Connell RM, Garcia-Flores Y, Baltimore D. MicroRNA-146a acts as a guardian of the quality and longevity of hematopoietic stem cells in mice. eLife 2013; 2:e00537. [PMID: 23705069 PMCID: PMC3660742 DOI: 10.7554/elife.00537] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Accepted: 04/16/2013] [Indexed: 12/21/2022] Open
Abstract
During inflammation and infection, hematopoietic stem and progenitor cells are stimulated to proliferate and differentiate into mature immune cells, especially of the myeloid lineage. MicroRNA-146a (miR-146a) is a critical negative regulator of inflammation. Deletion of miR-146a produces effects that appear as dysregulated inflammatory hematopoiesis, leading to a decline in the number and quality of hematopoietic stem cells (HSCs), excessive myeloproliferation, and, ultimately, to HSC exhaustion and hematopoietic neoplasms. At the cellular level, the defects are attributable to both an intrinsic problem in the miR-146a-deficient HSCs and extrinsic effects of lymphocytes and nonhematopoietic cells. At the molecular level, this involves a molecular axis consisting of miR-146a, signaling protein TRAF6, transcriptional factor NF-κB, and cytokine IL-6. This study has identified miR-146a to be a critical regulator of HSC homeostasis during chronic inflammation in mice and provided a molecular connection between chronic inflammation and the development of bone marrow failure and myeloproliferative neoplasms. DOI:http://dx.doi.org/10.7554/eLife.00537.001.
Collapse
Affiliation(s)
- Jimmy L Zhao
- Department of Biology , California Institute of Technology , Pasadena , United States
| | | | | | | | | |
Collapse
|
18
|
Zhang W, Shao Z, Fu R, Wang H, Li L, Yue L. Effect of DLK1 on tumorigenesis in CD34 +CD38 - bone marrow cells in myelodysplastic syndromes. Oncol Lett 2013; 6:203-206. [PMID: 23946804 PMCID: PMC3742462 DOI: 10.3892/ol.2013.1346] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 05/03/2013] [Indexed: 11/23/2022] Open
Abstract
The myelodysplastic syndromes (MDSs) are a group of clonal stem cell disorders resulting from aberrations within hematopoietic stem cells (HSCs), which may lead to the onset of a number of diseases, including acute myeloid leukemia (AML). Recent studies have demonstrated that the expression levels of the DLK1 gene are increased in MDS. In order to determine whether the addition of DLK1 affects tumorigenesis, small interfering (si)RNAs were designed to target DLK1 in order to knockdown its expression in CD34+CD38− bone marrow cells in MDS. A lower proliferative rate was observed in the CD34+CD38− bone marrow cells following this knockdown of DLK1 expression. The suppression of DLK1 expression resulted in a less aggressive MDS phenotype, which suggests that the upregulation of DLK1 expression may play an oncogenic role in CD34+CD38− bone marrow cells.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Hematology, General Hospital of Tianjin Medical University, Tianjin 300052, P.R. China
| | | | | | | | | | | |
Collapse
|
19
|
IL10 inversely correlates with the percentage of CD8+ cells in MDS patients. Leuk Res 2013; 37:541-6. [DOI: 10.1016/j.leukres.2013.01.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 01/04/2013] [Accepted: 01/25/2013] [Indexed: 12/24/2022]
|
20
|
Das M, Chaudhuri S, Law S. Unveiling the paradoxical nature of myelodysplastic syndromes (MDS): why hypercellular marrow strongly favors accelerated apoptosis. Biochem Cell Biol 2013; 91:303-8. [PMID: 24032679 DOI: 10.1139/bcb-2012-0109] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The pathogenesis of bone marrow failure in myelodysplastic syndromes (MDS) is an unresolved mystery. MDS causes peripheral blood cytopenias and increased bone marrow cellularity. This apparent paradox has been interpreted as a sign of intramedullary destruction of a substantial portion of the developing hematopoietic cells by apoptosis. The present study aimed to delineate the exact mechanistic relationship between the bone marrow hypercellularity and the accelerated apoptosis in an N-ethyl-N-nitrosourea (ENU)-induced experimental MDS mouse model. The observations made so far clarify the quantitative and qualitative changes that occur in the bone marrow microenvironment through cell cycle analysis, especially involving the telomerase reverse transcriptase (TERT) and p53 expression patterns. The survival fate of the bone marrow cells were observed by measuring the expression level of some intracellular protein molecules like apoptosis signal-regulating kinase 1 (ASK-1), c-Jun N-terminal kinase (JNK), and cleaved caspase-3 of the extrinsic pathway toward apoptosis. We found myelodysplasia damage occurs within one or more multipotent progenitor populations resulting in uncontrolled cellular proliferation within the MDS bone marrow. Then, due to homeostatic balance, this high cellular burden is minimized by activating the apoptosis pathway. As a result, the peripheral blood suffers cellular deprivation. This study can throw some light on the mechanism of disease progression and also help to reveal the paradoxical nature of the disease.
Collapse
Affiliation(s)
- Madhurima Das
- a Stem Cell Research and Application Unit, Department of Biochemistry and Medical Biotechnology, Calcutta School of Tropical Medicine, 108 C R Avenue, Kolkata, West Bengal, India, 700073
| | | | | |
Collapse
|
21
|
Bone marrow dendritic cells are reduced in patients with high-risk myelodysplastic syndromes. Leuk Res 2013; 37:266-73. [DOI: 10.1016/j.leukres.2012.10.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2012] [Revised: 10/14/2012] [Accepted: 10/15/2012] [Indexed: 12/19/2022]
|
22
|
de Oliveira FM, Lucena-Araujo AR, Favarin MDC, Bonini Palma PV, Rego EM, Falcão RP, Covas DT, Fontes AM. Differential expression of AURKA and AURKB genes in bone marrow stromal mesenchymal cells of myelodysplastic syndrome: correlation with G-banding analysis and FISH. Exp Hematol 2013; 41:198-208. [DOI: 10.1016/j.exphem.2012.10.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 08/17/2012] [Accepted: 10/01/2012] [Indexed: 12/23/2022]
|
23
|
Khan C, Pathe N, Fazal S, Lister J, Rossetti JM. Azacitidine in the management of patients with myelodysplastic syndromes. Ther Adv Hematol 2012; 3:355-73. [PMID: 23606938 PMCID: PMC3627328 DOI: 10.1177/2040620712464882] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous group of clonal hematopoeitic disorders characterized by ineffective hematopoiesis and potential transformation to acute myeloid leukemia (AML). For decades, the mainstay of treatment for MDS was supportive care, including transfusion of blood products and growth factors. Further understanding of disease biology led to the discovery of a high prevalence of hypermethylation of tumor suppressor genes in high-risk MDS and secondary leukemias. Hence, the role of irreversible DNA methlytransferase inhibitors such as azacitidine was investigated with promising outcomes in the treatment of MDS. Azacitidine was initially approved in the USA by the Food and Drug Administration (FDA) in 2004 for the treatment of all subtypes of MDS and was granted expanded approval in 2009 to reflect new overall survival data demonstrated in the AZA-001 study of patients with higher-risk MDS. Azacitidine has demonstrated significant and clinically meaningful prolongation of survival in higher-risk patients with MDS and has changed the natural history of these disorders. The agent maintains a relatively safe toxicity profile, even in older patients. The role of azacitidine has been explored in the treatment of AML and chronic myelomonocytic leukemia and has also been studied in the peritransplant setting. Azacitidine has been combined with other novel agents such as lenalidomide, histone deacetylase inhibitors and growth factors in the hope of achieving improved outcomes. Currently, both intravenous and subcutaneous forms of azacitidine are approved for use in the USA with the oral form being granted fast track status by the FDA.
Collapse
Affiliation(s)
- Cyrus Khan
- Western Pennsylvania Cancer Institute, The Western Pennsylvania Hospital, Pittsburgh, PA, USA
| | | | | | | | | |
Collapse
|
24
|
Yang L, Mailloux A, Rollison DE, Painter JS, Maciejewski J, Paquette RL, Loughran TP, McGraw K, Makishima H, Radhakrishnan R, Wei S, Ren X, Komrokji R, List AF, Epling-Burnette PK. Naive T-cells in myelodysplastic syndrome display intrinsic human telomerase reverse transcriptase (hTERT) deficiency. Leukemia 2012; 27:897-906. [PMID: 23072779 PMCID: PMC4346223 DOI: 10.1038/leu.2012.300] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Telomeres are specialized structures providing chromosome integrity during cellular division along with protection against premature senescence and apoptosis. Accelerated telomere attrition in patients with myelodysplastic syndrome (MDS) occurs by an undefined mechanism. Although the MDS clone originates within the myeloid compartment, T-lymphocytes display repertoire contraction and loss of naive T-cells. The replicative lifespan of T-cells is stringently regulated by telomerase activity. In MDS cases, we show that purified CD3+ T-cells have significantly shorter telomere length and reduced proliferative capacity upon stimulation compared with controls. To understand the mechanism, telomerase enzymatic activity and telomerase reverse transcriptase (hTERT), gene expression were compared in MDS cases (n=35) and healthy controls (n=42) within different T-cell compartments. Telomerase activity is greatest in naive T-cells illustrating the importance of telomere repair in homeostatic repertoire regulation. Compared with healthy controls, MDS cases had lower telomerase induction (P<0.0001) that correlated with significantly lower hTERT mRNA (P<0.0001), independent of age and disease stratification. hTERT mRNA deficiency affected naive but not memory T-cells, and telomere erosion in MDS occurred without evidence of an hTERT-promoter mutation, copy number variation or deletion. Telomerase insufficiency may undermine homeostatic control within the hematopoietic compartment and promote a change in the T-cell repertoire in MDS.
Collapse
Affiliation(s)
- L Yang
- Immunology Program at the H Lee Moffitt Cancer Center, Tampa, FL, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Spinelli E, Caporale R, Buchi F, Masala E, Gozzini A, Sanna A, Sassolini F, Valencia A, Bosi A, Santini V. Distinct Signal Transduction Abnormalities and Erythropoietin Response in Bone Marrow Hematopoietic Cell Subpopulations of Myelodysplastic Syndrome Patients. Clin Cancer Res 2012; 18:3079-89. [PMID: 22496271 DOI: 10.1158/1078-0432.ccr-11-0686] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Elena Spinelli
- FU Hematology and Department of Laboratory Medicine, Azienda Ospedaliero Universitaria (AOU) Careggi, University of Florence, Florence, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
A new recurrent chromosomal translocation t(3;11)(q13;q14) in myelodysplastic syndromes associated with overexpression of the ILDR1 gene. Leuk Res 2012; 36:852-6. [PMID: 22365942 DOI: 10.1016/j.leukres.2012.01.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 01/24/2012] [Accepted: 01/30/2012] [Indexed: 11/22/2022]
Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous group of diseases characterized by ineffective hematopoiesis and an increased risk of evolution to acute myeloid leukemia (AML). In this study, the combination of conventional cytogenetic, FISH studies and molecular techniques allowed us to unveil a novel recurrent t(3;11)(q13;q14) causing the overexpression of the immunoglobulin-like domain-containing receptor (ILDR1) gene. The analysis of gene expression was extended to Refractory Anemia (RA) and Refractory Anemia with excess blasts (RAEB) cases revealing ILDR1 overexpression in 36% of RAEB subgroup. The biological implications of the ILDR1 overexpression in MDS pathogenesis and its potential prognostic significance should be further investigated.
Collapse
|
27
|
Antic D, Impera L, Fekete MD, Djordjevic V, Storlazzi CT, Elezovic I. Novel chromosomal translocation (17;22)(q12;q12) in a case of myelodisplastic syndrome characterized with signs of hemolytic anemia at presentation. Gene 2012; 493:161-4. [PMID: 22138479 DOI: 10.1016/j.gene.2011.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Accepted: 11/01/2011] [Indexed: 11/15/2022]
Abstract
Myelodysplastic syndromes (MDS) are clonal stem cell diseases that can result in cytopenias, dysplasia in one or more cell lineages, infective hematopoiesis, and increase the risk of progression to acute myeloid leukemia (AML). MDSs are characterized by several recurrent cytogenetic defects, which can affect diagnosis, prognosis, and treatment. Some of that chromosomal alterations are associated with very poor prognosis. Conventional cytogenetics cannot accurately define the rearranged karyotype. Instead, molecular cytogenetics analyses can provide important diagnostic and prognostic information for patients affected by MDS, allowing the characterization of the whole mutational spectrum and, mainly, novel chromosomal lesions. In this paper, we report a MDS case with a novel chromosomal translocation [t(17;22)(q12;q22)], described for the first time here. Following Giemsa-banding karyotyping, fluorescent in situ hybridization analyses, by using chromosome-specific probes, displayed the breakpoint regions at chromosomes 17 and 22, within which intra and inter-chromosomal segmental duplications (SD) are present. Because of the occurrence of SDs in breakpoint region, it was not possible to finely define the genomic regions where breaks fell. Further investigations could be required to better understand the molecular basis of the novel translocation t(17;22)(q12;q12) acting in MDS context and to explain if SDs could contribute to the pathogenesis of MDS.
Collapse
Affiliation(s)
- Darko Antic
- Clinic for hematology, Clinical Center Serbia, Koste Todorovica 2, 11 000 Belgrade, Serbia.
| | | | | | | | | | | |
Collapse
|
28
|
Myelodysplastic syndromes: revisiting the role of the bone marrow microenvironment in disease pathogenesis. Int J Hematol 2012; 95:17-25. [DOI: 10.1007/s12185-011-1001-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 12/12/2011] [Indexed: 12/11/2022]
|
29
|
Possibility of myelodysplastic syndromes screening using a complete blood automated cell count. Leuk Res 2011; 35:1623-7. [DOI: 10.1016/j.leukres.2011.06.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Revised: 06/07/2011] [Accepted: 06/14/2011] [Indexed: 11/19/2022]
|
30
|
Hopfer O, Nolte F, Mossner M, Komor M, Kmetsch A, Benslasfer O, Reißmann M, Nowak D, Hoelzer D, Thiel E, Hofmann WK. Epigenetic dysregulation of GATA1 is involved in myelodysplastic syndromes dyserythropoiesis. Eur J Haematol 2011; 88:144-53. [DOI: 10.1111/j.1600-0609.2011.01715.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
31
|
Ishibashi M, Tamura H, Ogata K. Disease progression mechanism in myelodysplastic syndromes: insight into the role of the microenvironment. Leuk Res 2011; 35:1449-52. [PMID: 21757231 DOI: 10.1016/j.leukres.2011.06.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2011] [Revised: 06/17/2011] [Accepted: 06/18/2011] [Indexed: 12/29/2022]
Abstract
The somatic mutation theory proposing that a sequential accumulation of genetic abnormalities plays a major role in cancer pathogenesis has not yet been confirmed for myelodysplastic syndromes (MDS). Meanwhile, recent data in some cancers has underscored the role of the microenvironment in tumor growth. MDS CD34+CD38- cells usually fail to repopulate after transplantation in mice, suggesting the importance of the microenvironment for MDS cells. Our recent data have provided a disease-progression model in which overproduction of interferon-γ and tumor necrosis factor-α in the microenvironment is the primary event. This causes B7-H1 molecule expression on MDS blasts, which generates a bifunctional signal inducing T-cell apoptosis and enhancing blast proliferation. The latter may provide more opportunity for developing secondary genetic changes.
Collapse
Affiliation(s)
- Mariko Ishibashi
- Division of Hematology, Department of Medicine, Nippon Medical School, Tokyo, Japan
| | | | | |
Collapse
|
32
|
Giachelia M, D’Alò F, Fabiani E, Saulnier N, Di Ruscio A, Guidi F, Hohaus S, Voso MT, Leone G. Gene expression profiling of myelodysplastic CD34+ hematopoietic stem cells treated in vitro with decitabine. Leuk Res 2011; 35:465-71. [DOI: 10.1016/j.leukres.2010.07.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Revised: 07/12/2010] [Accepted: 07/14/2010] [Indexed: 01/22/2023]
|
33
|
Bruserud Ø, Reikvam H. Therapeutic targeting of NF-κB in myelodysplastic syndromes and acute myeloid leukaemia - the biological heterogeneity. Expert Opin Ther Targets 2011; 14:1139-42. [PMID: 20942744 DOI: 10.1517/14728222.2010.525021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
NF-κB usually has antiapotptic effects and is involved in regulation of cell proliferation and intercellular communication. This is also true for the malignant cells in acute myeloid leukaemia (AML) and myelodysplastic syndromes (MDS), including the malignant stem cells. However, both AML and MDS patients are heterogeneous with regard to the effect of pharmacological NF-κB inhibition, and the final effect will probably also depend on the pharmacological agent used for the inhibition, e.g. proteasomal inhibitiors versus specific inhibitors. Even though initial studies suggest that NF-κB inhibitors have antileukemic effects, their future clinical use will also depend on their toxicity profile.
Collapse
Affiliation(s)
- Øystein Bruserud
- University of Bergen, Institute of Internal Medicine, Bergen, Norway.
| | | |
Collapse
|
34
|
Iwanaga M, Hsu WL, Soda M, Takasaki Y, Tawara M, Joh T, Amenomori T, Yamamura M, Yoshida Y, Koba T, Miyazaki Y, Matsuo T, Preston DL, Suyama A, Kodama K, Tomonaga M. Risk of Myelodysplastic Syndromes in People Exposed to Ionizing Radiation: A Retrospective Cohort Study of Nagasaki Atomic Bomb Survivors. J Clin Oncol 2011; 29:428-34. [PMID: 21149671 DOI: 10.1200/jco.2010.31.3080] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Purpose The risk of myelodysplastic syndromes (MDS) has not been fully investigated among people exposed to ionizing radiation. We investigate MDS risk and radiation dose-response in Japanese atomic bomb survivors. Patients and Methods We conducted a retrospective cohort study by using two databases of Nagasaki atomic bomb survivors: 64,026 people with known exposure distance in the database of Nagasaki University Atomic-Bomb Disease Institute (ABDI) and 22,245 people with estimated radiation dose in the Radiation Effects Research Foundation Life Span Study (LSS). Patients with MDS diagnosed from 1985 to 2004 were identified by record linkage between the cohorts and the Nagasaki Prefecture Cancer Registry. Cox and Poisson regression models were used to estimate relationships between exposure distance or dose and MDS risk. Results There were 151 patients with MDS in the ABDI cohort and 47 patients with MDS in the LSS cohort. MDS rate increased inversely with exposure distance, with an excess relative risk (ERR) decay per km of 1.2 (95% CI, 0.4 to 3.0; P < .001) for ABDI. MDS risk also showed a significant linear response to exposure dose level (P < .001) with an ERR per Gy of 4.3 (95% CI, 1.6 to 9.5; P < .001). After adjustment for sex, attained age, and birth year, the MDS risk was significantly greater in those exposed when young. Conclusion A significant linear radiation dose-response for MDS exists in atomic bomb survivors 40 to 60 years after radiation exposure. Clinicians should perform careful long-term follow-up of irradiated people to detect MDS as early as possible.
Collapse
Affiliation(s)
- Masako Iwanaga
- From the Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Science; Kwassui Women's College; Radiation Effects Research Foundation; Japanese Red Cross Nagasaki Genbaku Hospital; St. Francis Hospital; Nagasaki Municipal Hospital; Nagasaki Atomic Bomb Casualty Council Health Management Center; and Nagasaki Municipal Medical Center, Nagasaki; Radiation Effects Research Foundation, Hiroshima, Japan; and Hirosoft International, Seattle, WA
| | - Wan-Ling Hsu
- From the Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Science; Kwassui Women's College; Radiation Effects Research Foundation; Japanese Red Cross Nagasaki Genbaku Hospital; St. Francis Hospital; Nagasaki Municipal Hospital; Nagasaki Atomic Bomb Casualty Council Health Management Center; and Nagasaki Municipal Medical Center, Nagasaki; Radiation Effects Research Foundation, Hiroshima, Japan; and Hirosoft International, Seattle, WA
| | - Midori Soda
- From the Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Science; Kwassui Women's College; Radiation Effects Research Foundation; Japanese Red Cross Nagasaki Genbaku Hospital; St. Francis Hospital; Nagasaki Municipal Hospital; Nagasaki Atomic Bomb Casualty Council Health Management Center; and Nagasaki Municipal Medical Center, Nagasaki; Radiation Effects Research Foundation, Hiroshima, Japan; and Hirosoft International, Seattle, WA
| | - Yumi Takasaki
- From the Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Science; Kwassui Women's College; Radiation Effects Research Foundation; Japanese Red Cross Nagasaki Genbaku Hospital; St. Francis Hospital; Nagasaki Municipal Hospital; Nagasaki Atomic Bomb Casualty Council Health Management Center; and Nagasaki Municipal Medical Center, Nagasaki; Radiation Effects Research Foundation, Hiroshima, Japan; and Hirosoft International, Seattle, WA
| | - Masayuki Tawara
- From the Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Science; Kwassui Women's College; Radiation Effects Research Foundation; Japanese Red Cross Nagasaki Genbaku Hospital; St. Francis Hospital; Nagasaki Municipal Hospital; Nagasaki Atomic Bomb Casualty Council Health Management Center; and Nagasaki Municipal Medical Center, Nagasaki; Radiation Effects Research Foundation, Hiroshima, Japan; and Hirosoft International, Seattle, WA
| | - Tatsuro Joh
- From the Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Science; Kwassui Women's College; Radiation Effects Research Foundation; Japanese Red Cross Nagasaki Genbaku Hospital; St. Francis Hospital; Nagasaki Municipal Hospital; Nagasaki Atomic Bomb Casualty Council Health Management Center; and Nagasaki Municipal Medical Center, Nagasaki; Radiation Effects Research Foundation, Hiroshima, Japan; and Hirosoft International, Seattle, WA
| | - Tatsuhiko Amenomori
- From the Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Science; Kwassui Women's College; Radiation Effects Research Foundation; Japanese Red Cross Nagasaki Genbaku Hospital; St. Francis Hospital; Nagasaki Municipal Hospital; Nagasaki Atomic Bomb Casualty Council Health Management Center; and Nagasaki Municipal Medical Center, Nagasaki; Radiation Effects Research Foundation, Hiroshima, Japan; and Hirosoft International, Seattle, WA
| | - Masaomi Yamamura
- From the Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Science; Kwassui Women's College; Radiation Effects Research Foundation; Japanese Red Cross Nagasaki Genbaku Hospital; St. Francis Hospital; Nagasaki Municipal Hospital; Nagasaki Atomic Bomb Casualty Council Health Management Center; and Nagasaki Municipal Medical Center, Nagasaki; Radiation Effects Research Foundation, Hiroshima, Japan; and Hirosoft International, Seattle, WA
| | - Yoshiharu Yoshida
- From the Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Science; Kwassui Women's College; Radiation Effects Research Foundation; Japanese Red Cross Nagasaki Genbaku Hospital; St. Francis Hospital; Nagasaki Municipal Hospital; Nagasaki Atomic Bomb Casualty Council Health Management Center; and Nagasaki Municipal Medical Center, Nagasaki; Radiation Effects Research Foundation, Hiroshima, Japan; and Hirosoft International, Seattle, WA
| | - Takashi Koba
- From the Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Science; Kwassui Women's College; Radiation Effects Research Foundation; Japanese Red Cross Nagasaki Genbaku Hospital; St. Francis Hospital; Nagasaki Municipal Hospital; Nagasaki Atomic Bomb Casualty Council Health Management Center; and Nagasaki Municipal Medical Center, Nagasaki; Radiation Effects Research Foundation, Hiroshima, Japan; and Hirosoft International, Seattle, WA
| | - Yasushi Miyazaki
- From the Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Science; Kwassui Women's College; Radiation Effects Research Foundation; Japanese Red Cross Nagasaki Genbaku Hospital; St. Francis Hospital; Nagasaki Municipal Hospital; Nagasaki Atomic Bomb Casualty Council Health Management Center; and Nagasaki Municipal Medical Center, Nagasaki; Radiation Effects Research Foundation, Hiroshima, Japan; and Hirosoft International, Seattle, WA
| | - Tatsuki Matsuo
- From the Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Science; Kwassui Women's College; Radiation Effects Research Foundation; Japanese Red Cross Nagasaki Genbaku Hospital; St. Francis Hospital; Nagasaki Municipal Hospital; Nagasaki Atomic Bomb Casualty Council Health Management Center; and Nagasaki Municipal Medical Center, Nagasaki; Radiation Effects Research Foundation, Hiroshima, Japan; and Hirosoft International, Seattle, WA
| | - Dale L. Preston
- From the Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Science; Kwassui Women's College; Radiation Effects Research Foundation; Japanese Red Cross Nagasaki Genbaku Hospital; St. Francis Hospital; Nagasaki Municipal Hospital; Nagasaki Atomic Bomb Casualty Council Health Management Center; and Nagasaki Municipal Medical Center, Nagasaki; Radiation Effects Research Foundation, Hiroshima, Japan; and Hirosoft International, Seattle, WA
| | - Akihiko Suyama
- From the Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Science; Kwassui Women's College; Radiation Effects Research Foundation; Japanese Red Cross Nagasaki Genbaku Hospital; St. Francis Hospital; Nagasaki Municipal Hospital; Nagasaki Atomic Bomb Casualty Council Health Management Center; and Nagasaki Municipal Medical Center, Nagasaki; Radiation Effects Research Foundation, Hiroshima, Japan; and Hirosoft International, Seattle, WA
| | - Kazunori Kodama
- From the Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Science; Kwassui Women's College; Radiation Effects Research Foundation; Japanese Red Cross Nagasaki Genbaku Hospital; St. Francis Hospital; Nagasaki Municipal Hospital; Nagasaki Atomic Bomb Casualty Council Health Management Center; and Nagasaki Municipal Medical Center, Nagasaki; Radiation Effects Research Foundation, Hiroshima, Japan; and Hirosoft International, Seattle, WA
| | - Masao Tomonaga
- From the Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Science; Kwassui Women's College; Radiation Effects Research Foundation; Japanese Red Cross Nagasaki Genbaku Hospital; St. Francis Hospital; Nagasaki Municipal Hospital; Nagasaki Atomic Bomb Casualty Council Health Management Center; and Nagasaki Municipal Medical Center, Nagasaki; Radiation Effects Research Foundation, Hiroshima, Japan; and Hirosoft International, Seattle, WA
| |
Collapse
|
35
|
Bejar R, Levine R, Ebert BL. Unraveling the molecular pathophysiology of myelodysplastic syndromes. J Clin Oncol 2011; 29:504-15. [PMID: 21220588 DOI: 10.1200/jco.2010.31.1175] [Citation(s) in RCA: 238] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Somatically acquired genetic abnormalities lead to the salient features that define myelodysplastic syndromes (MDS): clonal hematopoiesis, aberrant differentiation, peripheral cytopenias, and risk of progression to acute myeloid leukemia. Although specific karyotypic abnormalities have been linked to MDS for decades, more recent findings have demonstrated the importance of mutations within individual genes, focal alterations that are not apparent by standard cytogenetics, and aberrant epigenetic regulation of gene expression. The spectrum of genetic abnormalities in MDS implicates a wide range of molecular mechanisms in the pathogenesis of these disorders, including activation of tyrosine kinase signaling, genomic instability, impaired differentiation, altered ribosome function, and changes in the bone marrow microenvironment. Specific alterations present in individual patients with MDS may explain much of the heterogeneity in clinical phenotype associated with this disease and can predict prognosis and response to therapy. Elucidation of the full complement of genetic causes of MDS promises profound insight into the biology of the disease, improved classification and prognostic scoring schemes, and the potential for novel targeted therapies with molecular predictors of response.
Collapse
Affiliation(s)
- Rafael Bejar
- Brigham and Women's Hospital, Karp Research Building, CHRB 05.211, 1 Blackfan Cir, Boston, MA 02115, USA
| | | | | |
Collapse
|
36
|
Xie W, Wang X, Du W, Liu W, Qin X, Huang S. Detection of molecular targets on the surface of CD34+CD38- bone marrow cells in myelodysplastic syndromes. Cytometry A 2010; 77:840-8. [PMID: 20662087 DOI: 10.1002/cyto.a.20929] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Myelodysplastic syndrome (MDS) is a kind of clonal stem-cell disorder in which aberration within a hematopoietic stem cell (HSC) gives rise to the entire disease as in acute myeloid leukemia (AML). Studies have showed that contrasting normal stem cells, AML stem cells express CD96 and CD123, but lack of CD90, although both of them reside within the CD34(+)CD38(-) population. So far, little is known about expression of the markers on MDS HSC. In this study, we analyzed the immunophenotypic characteristics of CD34(+)CD38(-) bone marrow (BM) cells by multicolor flow cytometry in 38 patients with MDS and 10 control patients. We found that CD34(+)CD38(-) BM cells coexpressed CD13, CD33, CD117, CD133, and HLA-DR almost in all patients, but in MDS they expressed higher amounts of CD13 (79% +/- 16% vs. 36% +/- 13%, P < 0.05) and CD133 (66% +/- 20% vs. 25% +/- 13%, P < 0.05). CD90 was expressed in all control patients but just in 63% of patients with MDS. No control patients had an expression of CD2, CD5, CD7, CD44, CD96, and CD123, which expressed variable amounts in 17-53% of patients with MDS. The level of CD13 in RCMD (89% +/- 7%), RAEB-1 (88% +/- 11%), and RAEB-2 (81% +/- 13%) were obviously higher than that of RA (63% +/- 16%, P < 0.05). CD2, CD5, and CD7 were more frequently observed in RAEB or INT and HIGH-R cases. Taken together, we demonstrate MDS stem cells display deranged phenotypic abnormalities that may make them particularly difficult to eradicate using therapies targeted against surface antigens, and the percentage of cells expressing CD13 is notably higher in patients with high-grade MDS that may be a potential prognostic indicator of MDS in the future.
Collapse
Affiliation(s)
- Wei Xie
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | |
Collapse
|
37
|
García-Fernández M, Kissel H, Brown S, Gorenc T, Schile AJ, Rafii S, Larisch S, Steller H. Sept4/ARTS is required for stem cell apoptosis and tumor suppression. Genes Dev 2010; 24:2282-93. [PMID: 20952537 DOI: 10.1101/gad.1970110] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Inhibitor of Apoptosis Proteins (IAPs) are frequently overexpressed in tumors and have become promising targets for developing anti-cancer drugs. IAPs can be inhibited by natural antagonists, but a physiological requirement of mammalian IAP antagonists remains to be established. Here we show that deletion of the mouse Sept4 gene, which encodes the IAP antagonist ARTS, promotes tumor development. Sept4-null mice have increased numbers of hematopoietic stem and progenitor cells, elevated XIAP protein, increased resistance to cell death, and accelerated tumor development in an Eμ-Myc background. These phenotypes are partially suppressed by inactivation of XIAP. Our results suggest that apoptosis plays an important role as a frontline defense against cancer by restricting the number of normal stem cells.
Collapse
Affiliation(s)
- María García-Fernández
- Laboratory of Apoptosis and Cancer Biology, Howard Hughes Medical Institute, The Rockefeller University, New York, New York 10065, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
DeWard AD, Eisenmann KM, Matheson SF, Alberts AS. The role of formins in human disease. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1803:226-33. [PMID: 19941910 DOI: 10.1016/j.bbamcr.2009.11.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2009] [Revised: 11/06/2009] [Accepted: 11/10/2009] [Indexed: 02/02/2023]
Abstract
Formins are a conserved family of proteins that play key roles in cytoskeletal remodeling. They nucleate and processively elongate non-branched actin filaments and also modulate microtubule dynamics. Despite their significant contributions to cell biology and development, few studies have directly implicated formins in disease pathogenesis. This review highlights the roles of formins in cell division, migration, immunity, and microvesicle formation in the context of human disease. In addition, we discuss the importance of controlling formin activity and protein expression to maintain cell homeostasis.
Collapse
Affiliation(s)
- Aaron D DeWard
- Laboratory of Cell Structure and Signal Integration, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | | | | | | |
Collapse
|
39
|
DeWard AD, Leali K, West RA, Prendergast GC, Alberts AS. Loss of RhoB expression enhances the myelodysplastic phenotype of mammalian diaphanous-related Formin mDia1 knockout mice. PLoS One 2009; 4:e7102. [PMID: 19768111 PMCID: PMC2740832 DOI: 10.1371/journal.pone.0007102] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2009] [Accepted: 08/18/2009] [Indexed: 12/21/2022] Open
Abstract
Myelodysplastic syndrome (MDS) is characterized by ineffective hematopoiesis and hyperplastic bone marrow. Complete loss or interstitial deletions of the long arm of chromosome 5 occur frequently in MDS. One candidate tumor suppressor on 5q is the mammalian Diaphanous (mDia)-related formin mDia1, encoded by DIAPH1 (5q31.3). mDia-family formins act as effectors for Rho-family small GTP-binding proteins including RhoB, which has also been shown to possess tumor suppressor activity. Mice lacking the Drf1 gene that encodes mDia1 develop age-dependent myelodysplastic features. We crossed mDia1 and RhoB knockout mice to test whether the additional loss of RhoB expression would compound the myelodysplastic phenotype. Drf1−/−RhoB−/− mice are fertile and develop normally. Relative to age-matched Drf1−/−RhoB+/− mice, the age of myelodysplasia onset was earlier in Drf1−/−RhoB−/− animals—including abnormally shaped erythrocytes, splenomegaly, and extramedullary hematopoiesis. In addition, we observed a statistically significant increase in the number of activated monocytes/macrophages in both the spleen and bone marrow of Drf1−/−RhoB−/− mice relative to Drf1−/−RhoB+/− mice. These data suggest a role for RhoB-regulated mDia1 in the regulation of hematopoietic progenitor cells.
Collapse
Affiliation(s)
- Aaron D. DeWard
- Laboratory of Cell Structure and Signal Integration, Van Andel Research Institute, Grand Rapids, Michigan, United States of America
- Program in Cell and Molecular Biology, Michigan State University, East Lansing, Michigan, United States of America
| | - Kellie Leali
- Laboratory of Cell Structure and Signal Integration, Van Andel Research Institute, Grand Rapids, Michigan, United States of America
- Flow Cytometry Core Facility, Van Andel Research Institute, Grand Rapids, Michigan, United States of America
| | - Richard A. West
- Laboratory of Cell Structure and Signal Integration, Van Andel Research Institute, Grand Rapids, Michigan, United States of America
- Flow Cytometry Core Facility, Van Andel Research Institute, Grand Rapids, Michigan, United States of America
| | - George C. Prendergast
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania, United States of America
| | - Arthur S. Alberts
- Laboratory of Cell Structure and Signal Integration, Van Andel Research Institute, Grand Rapids, Michigan, United States of America
- * E-mail:
| |
Collapse
|
40
|
Sips GJ, Amory CF, Delman BN, Kleinman GM, Lipsey LR, Tuhrim S. Intravascular lymphomatosis of the brain in a patient with myelodysplastic syndrome. Nat Rev Neurol 2009; 5:288-92. [PMID: 19488086 DOI: 10.1038/nrneurol.2009.36] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND A 77-year-old retired research pharmacologist with a long-standing history of anemia and a recent pathologically confirmed diagnosis of myelodysplastic syndrome was referred to a stroke unit for evaluation of slowly progressive cognitive deterioration, confusion and paroxysmal stroke-like episodes. A previous neurological work-up had revealed no noteworthy abnormalities except for chronic bilateral caudate infarctions seen on MRI and CT examinations of the brain. INVESTIGATIONS Physical examination, laboratory testing, brain MRI scanning, EEG, transesophageal echocardiography, cerebral angiography, CT scanning, and brain biopsy. DIAGNOSIS Intravascular lymphomatosis of the brain. MANAGEMENT Combined chemotherapy with CHOP (cyclophosphamide, doxorubicin, vincristine and prednisone) and rituximab.
Collapse
|
41
|
Eisenmann KM, Dykema KJ, Matheson SF, Kent NF, DeWard AD, West RA, Tibes R, Furge KA, Alberts AS. 5q– myelodysplastic syndromes: chromosome 5q genes direct a tumor-suppression network sensing actin dynamics. Oncogene 2009; 28:3429-41. [DOI: 10.1038/onc.2009.207] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|