51
|
Wang SA, Tam W, Tsai AG, Arber DA, Hasserjian RP, Geyer JT, George TI, Czuchlewski DR, Foucar K, Rogers HJ, Hsi ED, Bryan Rea B, Bagg A, Dal Cin P, Zhao C, Kelley TW, Verstovsek S, Bueso-Ramos C, Orazi A. Targeted next-generation sequencing identifies a subset of idiopathic hypereosinophilic syndrome with features similar to chronic eosinophilic leukemia, not otherwise specified. Mod Pathol 2016; 29:854-64. [PMID: 27174585 DOI: 10.1038/modpathol.2016.75] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 03/07/2016] [Accepted: 03/08/2016] [Indexed: 01/07/2023]
Abstract
The distinction between chronic eosinophilic leukemia, not otherwise specified and idiopathic hypereosinophilic syndrome largely relies on clonality assessment. Prior to the advent of next-generation sequencing, clonality was usually determined by cytogenetic analysis. We applied targeted next-generation sequencing panels designed for myeloid neoplasms to bone marrow specimens from a cohort of idiopathic hypereosinophilic syndrome patients (n=51), and assessed the significance of mutations in conjunction with clinicopathological features. The findings were further compared with those of 17 chronic eosinophilic leukemia, not otherwise specified patients defined by their abnormal cytogenetics and/or increased blasts. Mutations were detected in 14/51 idiopathic hypereosinophilic syndrome patients (idiopathic hypereosinophilic syndrome/next-generation sequencing-positive) (28%), involving single gene in 7 and ≥2 in 7 patients. The more frequently mutated genes included ASXL1 (43%), TET2 (36%), EZH2 (29%), SETBP1 (22%), CBL (14%), and NOTCH1 (14%). Idiopathic hypereosinophilic syndrome/next-generation sequencing-positive patients showed a number of clinical features and bone marrow findings resembling chronic eosinophilic leukemia, not otherwise specified. Chronic eosinophilic leukemia, not otherwise specified patients showed a disease-specific survival of 14.4 months, markedly inferior to idiopathic hypereosinophilic syndrome/next-generation sequencing-negative (P<0.001), but not significantly different from idiopathic hypereosinophilic syndrome/next-generation sequencing-positive (P=0.117). These data suggest that targeted next-generation sequencing helps to establish clonality in a subset of patients with hypereosinophilia that would otherwise be classified as idiopathic hypereosinophilic syndrome. In conjunction with other diagnostic features, mutation data can be used to establish a diagnosis of chronic eosinophilic leukemia, not otherwise specified in patients presenting with hypereosinophilia.
Collapse
Affiliation(s)
- Sa A Wang
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Wayne Tam
- Department of Pathology, Weill Cornell Medical College, New York, NY, USA
| | - Albert G Tsai
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Daniel A Arber
- Department of Pathology, Stanford University, Stanford, CA, USA
| | | | - Julia T Geyer
- Department of Pathology, Weill Cornell Medical College, New York, NY, USA
| | - Tracy I George
- Department of Pathology, University of New Mexico, Albuquerque, NM, USA
| | | | - Kathryn Foucar
- Department of Pathology, University of New Mexico, Albuquerque, NM, USA
| | - Heesun J Rogers
- Department of Laboratory Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Eric D Hsi
- Department of Laboratory Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - B Bryan Rea
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adam Bagg
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paola Dal Cin
- Department of Pathology, Brigham and Women Hospital, Boston, MA, USA
| | - Chong Zhao
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Todd W Kelley
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Srdan Verstovsek
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Carlos Bueso-Ramos
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Attilio Orazi
- Department of Pathology, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
52
|
Mangaru Z, Shetty S, Visconte V, Liu HD, Rogers HJ. Acute myeloid leukemia with inv(16)(p13.1q22), abnormal eosinophils, and absence of peripheral blood and bone marrow blasts. Am J Hematol 2016; 91:E273-4. [PMID: 26719044 DOI: 10.1002/ajh.24289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 12/18/2015] [Accepted: 12/22/2015] [Indexed: 11/10/2022]
Affiliation(s)
- Zareema Mangaru
- Department of Laboratory Medicine; Cleveland Clinic; Cleveland Ohio
| | | | - Valeria Visconte
- Department of Translational Hematology & Oncology Research; Cleveland Clinic; Cleveland Ohio
| | - Hien D. Liu
- Department of Hematology & Medical Oncology; Cleveland Clinic; Cleveland Ohio
| | - Heesun J. Rogers
- Department of Laboratory Medicine; Cleveland Clinic; Cleveland Ohio
| |
Collapse
|
53
|
LIN XIAOCONG, XU YONG, SUN GUOPING, WEN JINLI, LI NING, ZHANG YUMING, YANG ZHIGANG, ZHANG HAITAO, DAI YONG. Molecular dysfunctions in acute myeloid leukemia revealed by integrated analysis of microRNA and transcription factor. Int J Oncol 2016; 48:2367-80. [DOI: 10.3892/ijo.2016.3489] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Accepted: 01/19/2016] [Indexed: 11/05/2022] Open
|
54
|
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
|
55
|
Gao L, Liu F, Zhang H, Sun J, Ma Y. CHFRhypermethylation, a frequent event in acute myeloid leukemia, is independently associated with an adverse outcome. Genes Chromosomes Cancer 2015; 55:158-68. [PMID: 26542416 DOI: 10.1002/gcc.22322] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 09/22/2015] [Accepted: 09/23/2015] [Indexed: 12/19/2022] Open
Affiliation(s)
- Li Gao
- Department of Hematology, China-Japan Friendship Hospital, Beijing, China
| | - Fang Liu
- Department of Hematology and Oncology, The First Affiliated Hospital of Chinese PLA General Hospital, Beijing, China
- Department of Oncology, Chinese PLA General Hospital, Beijing, China
| | - Hui Zhang
- Department of Hematology, China-Japan Friendship Hospital, Beijing, China
| | - Junzhong Sun
- Department of Hematology and Oncology, The First Affiliated Hospital of Chinese PLA General Hospital, Beijing, China
| | - Yigai Ma
- Department of Hematology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
56
|
Gross SA, Fedak KM. Applying a Weight-of-Evidence Approach to Evaluate Relevance of Molecular Landscapes in the Exposure-Disease Paradigm. BIOMED RESEARCH INTERNATIONAL 2015; 2015:515798. [PMID: 26339619 PMCID: PMC4538402 DOI: 10.1155/2015/515798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 03/16/2015] [Indexed: 12/04/2022]
Abstract
Information on polymorphisms, mutations, and epigenetic events has become increasingly important in our understanding of molecular mechanisms associated with exposures-disease outcomes. Molecular landscapes can be developed to illustrate the molecular characteristics for environmental carcinogens as well as associated disease outcomes, although comparison of these molecular landscapes can often be difficult to navigate. We developed a method to organize these molecular data that uses a weight-of-evidence approach to rank overlapping molecular events by relative importance for susceptibility to an exposure-disease paradigm. To illustrate the usefulness of this approach, we discuss the example of benzene as an environmental carcinogen and myelodysplastic syndrome (MDS) as a causative disease endpoint. Using this weight-of-evidence method, we found overlapping polymorphisms in the genes for the metabolic enzymes GST and NQO1, both of which may infer risk of benzene-induced MDS. Polymorphisms in the tumor suppressor gene, TP53, and the inflammatory cytokine gene, TNF-α, were also noted, albeit inferring opposing outcomes. The alleles identified in the DNA repair gene RAD51 indicated an increased risk for MDS in MDS patients and low blood cell counts in benzene-exposed workers. We propose the weight-of-evidence approach as a tool to assist in organizing the sea of emerging molecular data in exposure-disease paradigms.
Collapse
Affiliation(s)
- Sherilyn A. Gross
- Cardno ChemRisk, 4840 Pearl East Circle 300 W., Boulder, CO 80304, USA
| | - Kristen M. Fedak
- Department of Environmental & Radiological Health Sciences, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
57
|
Chigaev A. Does aberrant membrane transport contribute to poor outcome in adult acute myeloid leukemia? Front Pharmacol 2015; 6:134. [PMID: 26191006 PMCID: PMC4489100 DOI: 10.3389/fphar.2015.00134] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 06/15/2015] [Indexed: 12/31/2022] Open
Abstract
Acute myeloid leukemia in adults is a highly heterogeneous disease. Gene expression profiling performed using unsupervised algorithms can be used to distinguish specific groups of patients within a large patient cohort. The identified gene expression signatures can offer insights into underlying physiological mechanisms of disease pathogenesis. Here, the analysis of several related gene expression clusters associated with poor outcome, worst overall survival and highest rates of resistant disease and obtained from the patients at the time of diagnosis or from previously untreated individuals is presented. Surprisingly, these gene clusters appear to be enriched for genes corresponding to proteins involved in transport across membranes (transporters, carriers and channels). Several ideas describing the possible relationship of membrane transport activity and leukemic cell biology, including the "Warburg effect," the specific role of chloride ion transport, direct "import" of metabolic energy through uptake of creatine phosphate, and modification of the bone marrow niche microenvironment are discussed.
Collapse
Affiliation(s)
- Alexandre Chigaev
- Department of Pathology and Cancer Center, University of New Mexico Health Sciences Center, University of New Mexico Albuquerque, NM, USA
| |
Collapse
|
58
|
Colla S, Ong DST, Ogoti Y, Marchesini M, Mistry NA, Clise-Dwyer K, Ang SA, Storti P, Viale A, Giuliani N, Ruisaard K, Ganan Gomez I, Bristow CA, Estecio M, Weksberg DC, Ho YW, Hu B, Genovese G, Pettazzoni P, Multani AS, Jiang S, Hua S, Ryan MC, Carugo A, Nezi L, Wei Y, Yang H, D'Anca M, Zhang L, Gaddis S, Gong T, Horner JW, Heffernan TP, Jones P, Cooper LJN, Liang H, Kantarjian H, Wang YA, Chin L, Bueso-Ramos C, Garcia-Manero G, DePinho RA. Telomere dysfunction drives aberrant hematopoietic differentiation and myelodysplastic syndrome. Cancer Cell 2015; 27:644-57. [PMID: 25965571 PMCID: PMC4596059 DOI: 10.1016/j.ccell.2015.04.007] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 01/31/2015] [Accepted: 04/13/2015] [Indexed: 12/14/2022]
Abstract
Myelodysplastic syndrome (MDS) risk correlates with advancing age, therapy-induced DNA damage, and/or shorter telomeres, but whether telomere erosion directly induces MDS is unknown. Here, we provide the genetic evidence that telomere dysfunction-induced DNA damage drives classical MDS phenotypes and alters common myeloid progenitor (CMP) differentiation by repressing the expression of mRNA splicing/processing genes, including SRSF2. RNA-seq analyses of telomere dysfunctional CMP identified aberrantly spliced transcripts linked to pathways relevant to MDS pathogenesis such as genome stability, DNA repair, chromatin remodeling, and histone modification, which are also enriched in mouse CMP haploinsufficient for SRSF2 and in CD34(+) CMML patient cells harboring SRSF2 mutation. Together, our studies establish an intimate link across telomere biology, aberrant RNA splicing, and myeloid progenitor differentiation.
Collapse
Affiliation(s)
- Simona Colla
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Derrick Sek Tong Ong
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yamini Ogoti
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Matteo Marchesini
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nipun A Mistry
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Karen Clise-Dwyer
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sonny A Ang
- Department of Pediatric Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Paola Storti
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Hematology, Department of Clinical and Experimental Medicine, University of Parma, 43126 Parma, Italy
| | - Andrea Viale
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nicola Giuliani
- Hematology, Department of Clinical and Experimental Medicine, University of Parma, 43126 Parma, Italy
| | - Kathryn Ruisaard
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Irene Ganan Gomez
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Christopher A Bristow
- Institute for Applied Cancer Science, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Marcos Estecio
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA
| | - David C Weksberg
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yan Wing Ho
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Baoli Hu
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Giannicola Genovese
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Piergiorgio Pettazzoni
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Asha S Multani
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shan Jiang
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sujun Hua
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Alessandro Carugo
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Luigi Nezi
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yue Wei
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hui Yang
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Marianna D'Anca
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Li Zhang
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sarah Gaddis
- Department of Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA
| | - Ting Gong
- Department of Molecular Carcinogenesis, University of Texas MD Anderson Cancer Center, Smithville, TX 78957, USA
| | - James W Horner
- Institute for Applied Cancer Science, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Timothy P Heffernan
- Institute for Applied Cancer Science, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Philip Jones
- Institute for Applied Cancer Science, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Laurence J N Cooper
- Department of Pediatric Research, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Han Liang
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hagop Kantarjian
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Y Alan Wang
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lynda Chin
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Carlos Bueso-Ramos
- Department of Hematopathology, University of Texas MD Cancer Center, Houston, TX 77030, USA
| | - Guillermo Garcia-Manero
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ronald A DePinho
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
59
|
Shi J, Pabon K, Scotto KW. Methylxanthines Increase Expression of the Splicing Factor SRSF2 by Regulating Multiple Post-transcriptional Mechanisms. J Biol Chem 2015; 290:14986-5003. [PMID: 25818199 DOI: 10.1074/jbc.m114.624254] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Indexed: 01/20/2023] Open
Abstract
We have previously reported that the methylxanthine caffeine increases expression of the splicing factor SRSF2, the levels of which are normally controlled by a negative autoregulatory loop. In the present study we have investigated the mechanisms by which methylxanthines induce this aberrant overexpression. RT-PCR analyses suggested little impact of caffeine on SRSF2 total mRNA levels. Instead, caffeine induced changes in the levels of SRSF2 3' UTR splice variants. Although some of these variants were substrates for nonsense-medicated decay (NMD), and could potentially have been stabilized by caffeine-mediated inhibition of NMD, down-regulation of NMD by a genetic approach was not sufficient to reproduce the phenotype. Furthermore, cell-based assays demonstrated that some of the caffeine-induced variants were intrinsically more efficiently translated than others; the addition of caffeine increased the translational efficiency of most SRSF2 transcripts. MicroRNA array analyses revealed a significant caffeine-mediated decrease in the expression of two SRSF2-targeting miRs, both of which were shown to repress translation of specific SRSF2 splice variants. These data support a complex model whereby caffeine down-regulates SRSF2-targeting microRNAs, leading to an increase in SRSF2 translation, which in turn induces SRSF2 splicing. SRSF2 splice variants are then stabilized by caffeine-mediated NMD inhibition, breaking the normal negative feedback loop and allowing the aberrant increase in SRSF2 protein levels. These findings highlight the complexity of SRSF2 gene regulation, and suggest ways in which SRSF2 expression may be dysregulated in disease.
Collapse
Affiliation(s)
- Jia Shi
- From the Rutgers Cancer Institute of New Jersey, the Robert Wood Johnson Medical School, and the Graduate School of Biomedical Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08903
| | - Kirk Pabon
- From the Rutgers Cancer Institute of New Jersey, the Robert Wood Johnson Medical School, and the Graduate School of Biomedical Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08903
| | - Kathleen W Scotto
- From the Rutgers Cancer Institute of New Jersey, the Robert Wood Johnson Medical School, and the Graduate School of Biomedical Sciences, Rutgers, The State University of New Jersey, New Brunswick, New Jersey 08903
| |
Collapse
|
60
|
Yang Y, Yu B, Chen Y. Blood disorders typically associated with renal transplantation. Front Cell Dev Biol 2015; 3:18. [PMID: 25853131 PMCID: PMC4365751 DOI: 10.3389/fcell.2015.00018] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 03/03/2015] [Indexed: 12/11/2022] Open
Abstract
Renal transplantation has become one of the most common surgical procedures performed to replace a diseased kidney with a healthy kidney from a donor. It can help patients with kidney failure live decades longer. However, renal transplantation also faces a risk of developing various blood disorders. The blood disorders typically associated with renal transplantation can be divided into two main categories: (1) Common disorders including post-transplant anemia (PTA), post-transplant lymphoproliferative disorder (PTLD), post-transplant erythrocytosis (PTE), and post-transplant cytopenias (PTC, leukopenia/neutropenia, thrombocytopenia, and pancytopenia); and (2) Uncommon but serious disorders including hemophagocytic syndrome (HPS), thrombotic microangiopathy (TMA), therapy-related myelodysplasia (t-MDS), and therapy-related acute myeloid leukemia (t-AML). Although many etiological factors involve the development of post-transplant blood disorders, immunosuppressive agents, and viral infections could be the two major contributors to most blood disorders and cause hematological abnormalities and immunodeficiency by suppressing hematopoietic function of bone marrow. Hematological abnormalities and immunodeficiency will result in severe clinical outcomes in renal transplant recipients. Understanding how blood disorders develop will help cure these life-threatening complications. A potential therapeutic strategy against post-transplant blood disorders should focus on tapering immunosuppression or replacing myelotoxic immunosuppressive drugs with lower toxic alternatives, recognizing and treating promptly the etiological virus, bacteria, or protozoan, restoring both hematopoietic function of bone marrow and normal blood counts, and improving kidney graft survival.
Collapse
Affiliation(s)
- Yu Yang
- Department of Urology, First Affiliated Hospital of PLA General Hospital Beijing, China
| | - Bo Yu
- Department of Urology, First Affiliated Hospital of PLA General Hospital Beijing, China
| | - Yun Chen
- BrightstarTech, Inc. Clarksburg, MD, USA
| |
Collapse
|
61
|
Tao YF, Xu LX, Lu J, Hu SY, Fang F, Cao L, Xiao PF, Du XJ, Sun LC, Li ZH, Wang NN, Su GH, Li YH, Li G, Zhao H, Li YP, Xu YY, Zhou HT, Wu Y, Jin MF, Liu L, Zhu XM, Ni J, Wang J, Xing F, Zhao WL, Pan J. Early B-cell factor 3 (EBF3) is a novel tumor suppressor gene with promoter hypermethylation in pediatric acute myeloid leukemia. J Exp Clin Cancer Res 2015; 34:4. [PMID: 25609158 PMCID: PMC4311429 DOI: 10.1186/s13046-014-0118-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 11/27/2014] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Pediatric acute myeloid leukemia (AML) comprises up to 20% of all childhood leukemia. Recent research shows that aberrant DNA methylation patterning may play a role in leukemogenesis. The epigenetic silencing of the EBF3 locus is very frequent in glioblastoma. However, the expression profiles and molecular function of EBF3 in pediatric AML is still unclear. METHODS Twelve human acute leukemia cell lines, 105 pediatric AML samples and 30 normal bone marrow/idiopathic thrombocytopenic purpura (NBM/ITP) control samples were analyzed. Transcriptional level of EBF3 was evaluated by semi-quantitative and real-time PCR. EBF3 methylation status was determined by methylation specific PCR (MSP) and bisulfite genomic sequencing (BGS). The molecular mechanism of EBF3 was investigated by apoptosis assays and PCR array analysis. RESULTS EBF3 promoter was hypermethylated in 10/12 leukemia cell lines. Aberrant EBF3 methylation was observed in 42.9% (45/105) of the pediatric AML samples using MSP analysis, and the BGS results confirmed promoter methylation. EBF3 expression was decreased in the AML samples compared with control. Methylated samples revealed similar survival outcomes by Kaplan-Meier survival analysis. EBF3 overexpression significantly inhibited cell proliferation and increased apoptosis. Real-time PCR array analysis revealed 93 dysregulated genes possibly implicated in the apoptosis of EBF3-induced AML cells. CONCLUSION In this study, we firstly identified epigenetic inactivation of EBF3 in both AML cell lines and pediatric AML samples for the first time. Our findings also showed for the first time that transcriptional overexpression of EBF3 could inhibit proliferation and induce apoptosis in AML cells. We identified 93 dysregulated apoptosis-related genes in EBF3-overexpressing, including DCC, AIFM2 and DAPK1. Most of these genes have never been related with EBF3 over expression. These results may provide new insights into the molecular mechanism of EBF3-induced apoptosis; however, further research will be required to determine the underlying details. Our findings suggest that EBF3 may act as a putative tumor suppressor gene in pediatric AML.
Collapse
MESH Headings
- Adolescent
- Age Factors
- Apoptosis/genetics
- Cell Line, Tumor
- Child
- Child, Preschool
- Cluster Analysis
- DNA Methylation
- Epigenesis, Genetic
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic
- Genes, Tumor Suppressor
- HL-60 Cells
- Humans
- Kaplan-Meier Estimate
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Male
- Prognosis
- Promoter Regions, Genetic
- Signal Transduction
- Transcription Factors/genetics
Collapse
Affiliation(s)
- Yan-Fang Tao
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - Li-Xiao Xu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - Jun Lu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - Shao-Yan Hu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - Fang Fang
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - Lan Cao
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - Pei-Fang Xiao
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - Xiao-Juan Du
- Department of Gastroenterology, the 5th Hospital of Chinese PLA, Yin chuan, China.
| | - Li-Chao Sun
- Department of Cell and Molecular Biology, Cancer Institute (Hospital), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.
| | - Zhi-Heng Li
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - Na-Na Wang
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - Guang-Hao Su
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - Yan-Hong Li
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - Gang Li
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - He Zhao
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - Yi-Ping Li
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - Yun-Yun Xu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - Hui-Ting Zhou
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - Yi Wu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - Mei-Fang Jin
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - Lin Liu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - Xue-Ming Zhu
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - Jian Ni
- Translational Research Center, Second Hospital, The Second Clinical School, Nanjing Medical University, Nanjing, China.
| | - Jian Wang
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - Feng Xing
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - Wen-Li Zhao
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| | - Jian Pan
- Department of Hematology and Oncology, Children's Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
62
|
Progress in RNAi-mediated Molecular Therapy of Acute and Chronic Myeloid Leukemia. MOLECULAR THERAPY. NUCLEIC ACIDS 2015; 4:e240. [DOI: 10.1038/mtna.2015.13] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 03/26/2015] [Indexed: 02/08/2023]
|
63
|
Haran M, Gross A. Balancing glycolysis and mitochondrial OXPHOS: lessons from the hematopoietic system and exercising muscles. Mitochondrion 2014; 19 Pt A:3-7. [PMID: 25264322 DOI: 10.1016/j.mito.2014.09.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2014] [Revised: 09/14/2014] [Accepted: 09/17/2014] [Indexed: 01/14/2023]
Abstract
Living organisms require a constant supply of safe and efficient energy to maintain homeostasis and to allow locomotion of single cells, tissues and the entire organism. The source of energy can be glycolysis, a simple series of enzymatic reactions in the cytosol, or a much more complex process in the mitochondria, oxidative phosphorylation (OXPHOS). In this review we will examine how does the organism balance its source of energy in two seemingly distinct and unrelated processes: hematopoiesis and exercise. In both processes we will show the importance of the metabolic program and its regulation. We will also discuss the importance of oxygen availability not as a sole determinant, but in the context of the nutrient and cellular state, and address the emerging role of lactate as an energy source and signaling molecule in health and disease.
Collapse
Affiliation(s)
- Michal Haran
- Hematology Institute, Kaplan Medical Center, Rehovot, Israel.
| | - Atan Gross
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
64
|
Karoopongse E, Yeung C, Byon J, Ramakrishnan A, Holman ZJ, Jiang PYZ, Yu Q, Deeg HJ, Marcondes AM. The KDM2B- let-7b -EZH2 axis in myelodysplastic syndromes as a target for combined epigenetic therapy. PLoS One 2014; 9:e107817. [PMID: 25225797 PMCID: PMC4166605 DOI: 10.1371/journal.pone.0107817] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 08/14/2014] [Indexed: 12/02/2022] Open
Abstract
Both DNA and histone methylation are dysregulated in the myelodysplastic syndromes (MDS). Based on preliminary data we hypothesized that dysregulated interactions of KDM2B, let-7b and EZH2 signals lead to an aberrant epigenetic landscape. Gene expression in CD34+ cells from MDS marrows was analyzed by NanoString miR array and validated by real-time polymerase chain reaction (PCR). The functions of KDM2B, let-7b and EZH2 were characterized in myeloid cell lines and in primary MDS cells. Let-7b levels were significantly higher, and KDM2B and EZH2 expression was lower in primary CD34+ MDS marrow cells (n = 44) than in healthy controls (n = 21; p<0.013, and p<0.0001, respectively). Overexpression of let-7b reduced EZH2 and KDM2B protein levels, and decreased cells in S-phase while increasing G0/G1 cells (p = 0.0005), accompanied by decreased H3K27me3 and cyclin D1. Silencing of KDM2B increased let-7b expression. Treatment with the cyclopentanyl analog of 3-deazaadenosine, DZNep, combined with the DNA hypomethylating agent 5-azacitidine, decreased levels of EZH2, suppressed methylation of di- and tri-methylated H3K27, and increased p16 expression, associated with cell proliferation. Thus, KDM2B, via let-7b/EZH2, promotes transcriptional repression. DZNep bypassed the inhibitory KDM2B/let-7b/EZH2 axis by preventing H3K27 methylation and reducing cell proliferation. DZNep might be able to enhance the therapeutic effects of DNA hypomethylating agents such as 5-azacitidine, currently considered standard therapy for patients with MDS.
Collapse
Affiliation(s)
- Ekapun Karoopongse
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Cecilia Yeung
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Anatomic Pathology, University of Washington, Seattle, Washington, United States of America
| | - John Byon
- Department of Hematology, University of Washington, Seattle, Washington, United States of America
| | - Aravind Ramakrishnan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - Zaneta J. Holman
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Peter Y. Z. Jiang
- Medical Oncology, Providence Regional Cancer Partnership and the Everett Clinic, Everett, Washington, United States of America
| | - Qiang Yu
- Cancer Biology and Pharmacology, Genome Institute of Singapore, A*STAR (Agency for Science, Technology and Research), Biopolis, Singapore, China
| | - H. Joachim Deeg
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
| | - A. Mario Marcondes
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
- Department of Medicine, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
65
|
Reghunathan R, Bi C, Liu SC, Loong KT, Chung TH, Huang G, Chng WJ. Clonogenic multiple myeloma cells have shared stemness signature associated with patient survival. Oncotarget 2014; 4:1230-40. [PMID: 23985559 PMCID: PMC3787153 DOI: 10.18632/oncotarget.1145] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Multiple myeloma is the abnormal clonal expansion of post germinal B cells in the bone marrow. It was previously reported that clonogenic myeloma cells are CD138−. Human MM cell lines RPMI8226 and NCI H929 contained 2-5% of CD138− population. In this study, we showed that CD138− cells have increased ALDH1 activity, a hallmark of normal and neoplastic stem cells. CD138−ALDH+ cells were more clonogenic than CD138+ALDH− cells and only CD138− cells differentiated into CD138+ population. In vivo tumor initiation and clonogenic potentials of the CD138− population was confirmed using NOG mice. We derived a gene expression signature from functionally validated and enriched CD138− clonogenic population from MM cell lines and validated these in patient samples. This data showed that CD138− cells had an enriched expression of genes that are expressed in normal and malignant stem cells. Differentially expressed genes included components of the polycomb repressor complex (PRC) and their targets. Inhibition of PRC by DZNep showed differential effect on CD138− and CD138+ populations. The ‘stemness’ signature derived from clonogenic CD138− cells overlap significantly with signatures of common progenitor cells, hematopoietic stem cells, and Leukemic stem cells and is associated with poorer survival in different clinical datasets.
Collapse
|
66
|
Daver N, Naqvi K, Jabbour E, Kadia T, DiNardo C, Cardenas-Turanzas M, Pierce S, Nguyen KTT, Bueso-Ramos C, Kantarjian H, Garcia-Manero G. Impact of comorbidities by ACE-27 in the revised-IPSS for patients with myelodysplastic syndromes. Am J Hematol 2014; 89:509-16. [PMID: 24458781 DOI: 10.1002/ajh.23675] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 01/18/2014] [Indexed: 01/21/2023]
Abstract
Comorbidities significantly affect the prognosis and outcomes of patients with hematological malignancies. We have previously reported the impact of comorbidities on the International Prognostic Scoring System (IPSS) score. The aim of this study was to determine whether comorbidities continued to have a significant impact when patients were reclassified according to the Revised-IPSS (IPSS-R). The medical records of 600 consecutive myelodysplastic syndrome patients who presented to MD Anderson Cancer Center between January 2002 and June 2004 were reviewed. The Adult Comorbidity Evaluation-27 (ACE-27) was used to assess the severity of comorbid conditions. Four hundred and two (67%) patients were male. Median age at presentation was 66.6 years (17-94). Mean duration of follow-up was 54 months (1-100). Five hundred and two (84%) patients died, and 54 (9%) patients underwent stem cell transplantation. Overall median survival was 16.8 months (1-100). Median survival by IPSS-R was 47, 34, 21, 16, and 6 months for patients in very low, low, intermediate, high, and very high-risk groups, respectively (P < 0.001). The ACE-27 comorbidity score significantly impacted the median survival of patients in the intermediate (P < 0.001), high (P = 0.045), and very high (P = 0.004) IPSS-R groups; but did not significantly impact the median survival in the low (P = 0.11) and very low (P = 0.49) IPSS-R groups. The ACE-27 comorbidity score significantly impacted the median survival of patients ≤65 years (P < 0.001) but did not significantly impact those >65 years (P = 0.18). Assessment of comorbidity may enhance the prognostic ability of the IPSS-R.
Collapse
Affiliation(s)
- Naval Daver
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Kiran Naqvi
- Department of Hematology/Oncology; Baylor College of Medicine; Houston Texas
| | - Elias Jabbour
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Tapan Kadia
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Courtney DiNardo
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | | | - Sherry Pierce
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Khanh Thi-Thuy Nguyen
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Carlos Bueso-Ramos
- Department of Hematopathology; The University of Texas MD Anderson Cancer Center; Houston Texas
| | - Hagop Kantarjian
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston Texas
| | | |
Collapse
|
67
|
Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood 2013; 122:3616-27; quiz 3699. [PMID: 24030381 DOI: 10.1182/blood-2013-08-518886] [Citation(s) in RCA: 1444] [Impact Index Per Article: 120.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous group of chronic hematological malignancies characterized by dysplasia, ineffective hematopoiesis and a variable risk of progression to acute myeloid leukemia. Sequencing of MDS genomes has identified mutations in genes implicated in RNA splicing, DNA modification, chromatin regulation, and cell signaling. We sequenced 111 genes across 738 patients with MDS or closely related neoplasms (including chronic myelomonocytic leukemia and MDS-myeloproliferative neoplasms) to explore the role of acquired mutations in MDS biology and clinical phenotype. Seventy-eight percent of patients had 1 or more oncogenic mutations. We identify complex patterns of pairwise association between genes, indicative of epistatic interactions involving components of the spliceosome machinery and epigenetic modifiers. Coupled with inferences on subclonal mutations, these data suggest a hypothesis of genetic "predestination," in which early driver mutations, typically affecting genes involved in RNA splicing, dictate future trajectories of disease evolution with distinct clinical phenotypes. Driver mutations had equivalent prognostic significance, whether clonal or subclonal, and leukemia-free survival deteriorated steadily as numbers of driver mutations increased. Thus, analysis of oncogenic mutations in large, well-characterized cohorts of patients illustrates the interconnections between the cancer genome and disease biology, with considerable potential for clinical application.
Collapse
|