1
|
Herrera J, Bensussen A, García-Gómez ML, Garay-Arroyo A, Álvarez-Buylla ER. A system-level model reveals that transcriptional stochasticity is required for hematopoietic stem cell differentiation. NPJ Syst Biol Appl 2024; 10:145. [PMID: 39639033 PMCID: PMC11621455 DOI: 10.1038/s41540-024-00469-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024] Open
Abstract
HSCs differentiation has been difficult to study experimentally due to the high number of components and interactions involved, as well as the impact of diverse physiological conditions. From a 200-node network, that was grounded on experimental data, we derived a 21-node regulatory network by collapsing linear pathways and retaining the functional feedback loops. This regulatory network core integrates key nodes and interactions underlying HSCs differentiation, including transcription factors, metabolic, and redox signaling pathways. We used Boolean, continuous, and stochastic dynamic models to simulate the hypoxic conditions of the HSCs niche, as well as the patterns and temporal sequences of HSCs transitions and differentiation. Our findings indicate that HSCs differentiation is a plastic process in which cell fates can transdifferentiate among themselves. Additionally, we found that cell heterogeneity is fundamental for HSCs differentiation. Lastly, we found that oxygen activates ROS production, inhibiting quiescence and promoting growth and differentiation pathways of HSCs.
Collapse
Affiliation(s)
- Joel Herrera
- Instituto de Ecología, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Antonio Bensussen
- Departamento de Control Automático, Cinvestav-IPN, Ciudad de México, México
| | - Mónica L García-Gómez
- Theoretical Biology, Institute of Biodynamics and Biocomplexity; Experimental and Computational Plant Development, Institute of Environmental Biology, Department of Biology, Utrecht University, Utrecht, Netherlands
| | - Adriana Garay-Arroyo
- Instituto de Ecología, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Elena R Álvarez-Buylla
- Instituto de Ecología, Universidad Nacional Autónoma de México, Ciudad de México, México.
| |
Collapse
|
2
|
Tu X, Gomez HM, Kim RY, Brown AC, de Jong E, Galvao I, Faiz A, Bosco A, Horvat JC, Hansbro P, Donovan C. Airway and parenchyma transcriptomics in a house dust mite model of experimental asthma. Respir Res 2023; 24:32. [PMID: 36698141 PMCID: PMC9878882 DOI: 10.1186/s12931-022-02298-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 12/15/2022] [Indexed: 01/26/2023] Open
Abstract
Lung transcriptomics studies in asthma have provided valuable information in the whole lung context, however, deciphering the individual contributions of the airway and parenchyma in disease pathogenesis may expedite the development of novel targeted treatment strategies. In this study, we performed transcriptomics on the airway and parenchyma using a house dust mite (HDM)-induced model of experimental asthma that replicates key features of the human disease. HDM exposure increased the expression of 3,255 genes, of which 212 were uniquely increased in the airways, 856 uniquely increased in the parenchyma, and 2187 commonly increased in both compartments. Further interrogation of these genes using a combination of network and transcription factor enrichment analyses identified several transcription factors that regulate airway and/or parenchymal gene expression, including transcription factor EC (TFEC), transcription factor PU.1 (SPI1), H2.0-like homeobox (HLX), metal response element binding transcription factor-1 (MTF1) and E74-like factor 4 (ets domain transcription factor, ELF4) involved in controlling innate immune responses. We next assessed the effects of inhibiting lung SPI1 responses using commercially available DB1976 and DB2313 on key disease outcomes. We found that both compounds had no protective effects on airway inflammation, however DB2313 (8 mg/kg) decreased mucus secreting cell number, and both DB2313 (1 mg/kg) and DB1976 (2.5 mg/kg and 1 mg/kg) reduced small airway collagen deposition. Significantly, both compounds decreased airway hyperresponsiveness. This study demonstrates that SPI1 is important in HDM-induced experimental asthma and that its pharmacological inhibition reduces HDM-induced airway collagen deposition and hyperresponsiveness.
Collapse
Affiliation(s)
- Xiaofan Tu
- grid.266842.c0000 0000 8831 109XPriority Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW Australia
| | - Henry M. Gomez
- grid.266842.c0000 0000 8831 109XPriority Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW Australia
| | - Richard Y. Kim
- grid.266842.c0000 0000 8831 109XPriority Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW Australia ,grid.117476.20000 0004 1936 7611Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW Australia
| | - Alexandra C. Brown
- grid.266842.c0000 0000 8831 109XPriority Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW Australia
| | - Emma de Jong
- Centre for Health Research, Telethon Kids Institute, The University of Western Australia, Nedlands, WA Australia
| | - Izabela Galvao
- grid.117476.20000 0004 1936 7611Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW Australia
| | - Alen Faiz
- grid.117476.20000 0004 1936 7611Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW Australia
| | - Anthony Bosco
- grid.134563.60000 0001 2168 186XAsthma and Airway Disease Research Center, University of Arizona, Arizona, USA
| | - Jay C. Horvat
- grid.266842.c0000 0000 8831 109XPriority Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW Australia
| | - Philip Hansbro
- grid.266842.c0000 0000 8831 109XPriority Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW Australia ,grid.117476.20000 0004 1936 7611Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW Australia
| | - Chantal Donovan
- grid.266842.c0000 0000 8831 109XPriority Centre for Healthy Lungs, Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW Australia ,grid.117476.20000 0004 1936 7611Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW Australia
| |
Collapse
|
3
|
Zhao X, Bartholdy B, Yamamoto Y, Evans EK, Alberich-Jordà M, Staber PB, Benoukraf T, Zhang P, Zhang J, Trinh BQ, Crispino JD, Hoang T, Bassal MA, Tenen DG. PU.1-c-Jun interaction is crucial for PU.1 function in myeloid development. Commun Biol 2022; 5:961. [PMID: 36104445 PMCID: PMC9474506 DOI: 10.1038/s42003-022-03888-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 08/25/2022] [Indexed: 11/09/2022] Open
Abstract
The Ets transcription factor PU.1 is essential for inducing the differentiation of monocytes, macrophages, and B cells in fetal liver and adult bone marrow. PU.1 controls hematopoietic differentiation through physical interactions with other transcription factors, such as C/EBPα and the AP-1 family member c-Jun. We found that PU.1 recruits c-Jun to promoters without the AP-1 binding sites. To address the functional importance of this interaction, we generated PU.1 point mutants that do not bind c-Jun while maintaining normal DNA binding affinity. These mutants lost the ability to transactivate a target reporter that requires a physical PU.1-c-Jun interaction, and did not induce monocyte/macrophage differentiation of PU.1-deficient cells. Knock-in mice carrying these point mutations displayed an almost complete block in hematopoiesis and perinatal lethality. While the PU.1 mutants were expressed in hematopoietic stem and early progenitor cells, myeloid differentiation was severely blocked, leading to an almost complete loss of mature hematopoietic cells. Differentiation into mature macrophages could be restored by expressing PU.1 mutant fused to c-Jun, demonstrating that a physical PU.1-c-Jun interaction is crucial for the transactivation of PU.1 target genes required for myeloid commitment and normal PU.1 function in vivo during macrophage differentiation.
Collapse
Affiliation(s)
- Xinhui Zhao
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, 02115, USA
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Boris Bartholdy
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, 02115, USA
- Albert Einstein College of Medicine, New York, NY, USA
| | - Yukiya Yamamoto
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, 02115, USA
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Aichi, Japan
| | - Erica K Evans
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, 02115, USA
- MOMA Therapeutics, Cambridge, MA, USA
| | - Meritxell Alberich-Jordà
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, 02115, USA
- Department of Hematology-oncology, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská, Prague, Czech Republic
- Childhood Leukemia Investigation Prague, Department of Pediatric Haematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague, University Hospital Motol, Videnska, Czech Republic
| | - Philipp B Staber
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, 02115, USA
- Department of Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Touati Benoukraf
- Cancer Science Institute of Singapore, Singapore, Singapore
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Pu Zhang
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Junyan Zhang
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Bon Q Trinh
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - John D Crispino
- Department of Medicine, Northwestern University, Chicago, IL, USA
| | - Trang Hoang
- Institute for Research in Immunology and Cancer (IRIC), Department of Pharmacology and Physiology, Université de Montréal, Montréal, QC, H3C 3J7, Canada
| | - Mahmoud A Bassal
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, 02115, USA.
- Cancer Science Institute of Singapore, Singapore, Singapore.
| | - Daniel G Tenen
- Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, 02115, USA.
- Cancer Science Institute of Singapore, Singapore, Singapore.
| |
Collapse
|
4
|
NfκB signaling dynamics and their target genes differ between mouse blood cell types and induce distinct cell behavior. Blood 2022; 140:99-111. [PMID: 35468185 DOI: 10.1182/blood.2021012918] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 03/16/2022] [Indexed: 11/20/2022] Open
Abstract
Cells can use signaling pathway activity over time (i.e., dynamics) to control cell fates. However, little is known about the potential existence and function of signaling dynamics in primary hematopoietic stem and progenitor cells (HSPCs). Here, we use time-lapse imaging and tracking of single murine HSPCs from GFP-p65/H2BmCherry reporter mice to quantify their nuclear factor κB (NfκB) activity dynamics in response to TNFα and IL1β. We find response dynamics to be heterogeneous between individual cells, with cell type specific dynamics distributions. Transcriptome sequencing of single cells physically isolated after live dynamics quantification shows activation of different target gene programs in cells with different dynamics. Finally, artificial induction of oscillatory NfκB activity causes changes in GMP behavior. Thus, HSPC behavior can be influenced by signaling dynamics, which are tightly regulated during hematopoietic differentiation and enable cell type specific responses to the same signaling inputs.
Collapse
|
5
|
Gao T, Jernigan J, Raza SA, Dammer EB, Xiao H, Seyfried NT, Levey AI, Rangaraju S. Transcriptional regulation of homeostatic and disease-associated-microglial genes by IRF1, LXRβ, and CEBPα. Glia 2019; 67:1958-1975. [PMID: 31301160 PMCID: PMC7190149 DOI: 10.1002/glia.23678] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/23/2019] [Accepted: 06/25/2019] [Indexed: 01/08/2023]
Abstract
Microglia transform from homeostatic to disease-associated-microglia (DAM) profiles in neurodegeneration. Within DAM, we recently identified distinct pro-inflammatory and anti-inflammatory sub-profiles although transcriptional regulators of homeostatic and distinct DAM profiles remain unclear. Informed by these studies, we nominated CEBPα, IRF1, and LXRβ as likely regulators of homeostatic, pro-inflammatory and anti-inflammatory DAM states and performed in-vitro siRNA studies in primary microglia to identify roles of each transcriptional factor (TF) in regulating microglial activation, using an integrated transcriptomics, bioinformatics and experimental validation approach. Efficient (>70%) silencing of TFs in microglia revealed reciprocal regulation between each TF specifically following pro-inflammatory activation. Neuroinflammatory transcriptomic profiling of microglia coupled with qPCR validation revealed distinct gene clusters with unique patterns of regulation by each TF, which were independent of LPS stimulation. While all three TFs (especially IRF1 and LXRβ) positively regulated core DAM genes (Apoe, Axl, Clec7a, Tyrobp, and Trem2) as well as homeostatic and pro-inflammatory DAM genes, LPS, and IFNγ increased pro-inflammatory DAM but suppressed homeostatic and anti-inflammatory DAM gene expression via an Erk1/2-dependent signaling pathway. IRF1 and LXRβ silencing suppressed microglial phagocytic activity for polystyrene microspheres as well as fAβ42 while IRF1 silencing strongly suppressed production of pro-inflammatory cytokines in response to LPS. Our studies reveal complex transcriptional regulation of homeostatic and DAM profiles whereby IRF1, LXRβ, and CEBPα positively regulate both pro- and anti-inflammatory DAM genes while activating stimuli independently augment pro-inflammatory DAM responses and suppress homeostatic and anti-inflammatory responses via Erk signaling. This framework can guide development of therapeutic immuno-modulatory strategies for neurodegeneration.
Collapse
Affiliation(s)
- Tianwen Gao
- Department of Neurology, Emory University, Atlanta, GA, USA
- Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | | | - Syed Ali Raza
- Department of Neurology, Emory University, Atlanta, GA, USA
| | - Eric B Dammer
- Department of Biochemistry, Emory University, Atlanta, Georgia
| | - Hailian Xiao
- Department of Neurology, Emory University, Atlanta, GA, USA
| | | | - Allan I Levey
- Department of Neurology, Emory University, Atlanta, GA, USA
| | | |
Collapse
|
6
|
Jules J, Li YP, Chen W. C/EBPα and PU.1 exhibit different responses to RANK signaling for osteoclastogenesis. Bone 2018; 107:104-114. [PMID: 29032174 PMCID: PMC6240464 DOI: 10.1016/j.bone.2017.05.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Revised: 04/14/2017] [Accepted: 05/08/2017] [Indexed: 11/23/2022]
Abstract
The transcription factors C/EBPα and PU.1 are upregulated by RANKL through activation of its receptor RANK during osteoclastogenesis and are critical for osteoclast differentiation. Herein we investigated the mechanisms underlying how C/EBPα and PU.1 regulate osteoclast differentiation in response to RANK signaling. We showed that C/EBPα or PU.1 overexpression could initiate osteoclastogenesis and upregulate the expressions of the osteoclast genes encoding the nuclear factor of activated T-cells, C1, cathepsin K, and tartrate-resistant acid phosphatase independently of RANKL. However, while PU.1 upregulated C/EBPα, C/EBPα could not upregulate PU.1. RANK has a unique cytoplasmic domain, 535IVVY538 motif, which is crucial for osteoclast differentiation. We demonstrated that mutational inactivation of RANK IVVY motif blocked osteoclast differentiation and significantly attenuated C/EBPα, but not PU.1, expression, indicating that RANK-IVVY-induced signaling is dispensable to PU.1 upregulation during osteoclastogenesis. However, C/EBPα or PU.1 overexpression failed to promote osteoclastogenesis in cells expressing mutated RANK IVVY motif. We noted that RANK-IVVY-motif inactivation significantly repressed osteoclast genes as compared with a vector control, suggesting that IVVY motif might also negatively regulate osteoclast inhibitors during osteoclastogenesis. Consistently, IVVY-motif inactivation triggered upregulation of RBP-J, a potent osteoclast inhibitor, during osteoclastogenesis. Notably, C/EBPα or PU.1 overexpression in cells expressing mutated RANK IVVY motif failed to control the deregulated RBP-J expression, resulting in repression of osteoclast genes. Accordingly, RBP-J silencing in the mutant cells rescued osteoclastogenesis with C/EBPα or PU.1 overexpression. In conclusion, we revealed that while PU.1 and C/EBPα are critical for osteoclastogenesis, they respond differently to RANKL-induced activation of RANK IVVY motif.
Collapse
Affiliation(s)
- Joel Jules
- Department of Pathology, University of Alabama, Birmingham, AL 35294, United States
| | - Yi-Ping Li
- Department of Pathology, University of Alabama, Birmingham, AL 35294, United States.
| | - Wei Chen
- Department of Pathology, University of Alabama, Birmingham, AL 35294, United States.
| |
Collapse
|
7
|
Ishikawa Y, Nakayama K, Morimoto M, Mizutani A, Nakayama A, Toyoshima K, Hayashi A, Takagi S, Dairiki R, Miyashita H, Matsumoto S, Gamo K, Nomura T, Nakamura K. Synergistic anti-AML effects of the LSD1 inhibitor T-3775440 and the NEDD8-activating enzyme inhibitor pevonedistat via transdifferentiation and DNA rereplication. Oncogenesis 2017; 6:e377. [PMID: 28892104 PMCID: PMC5623902 DOI: 10.1038/oncsis.2017.76] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 06/18/2017] [Accepted: 07/11/2017] [Indexed: 12/12/2022] Open
Abstract
Lysine-specific demethylase 1A (LSD1, KDM1A) specifically demethylates di- and monomethylated histones H3K4 and K9, resulting in context-dependent transcriptional repression or activation. We previously identified an irreversible LSD1 inhibitor T-3775440, which exerts antileukemic activities in a subset of acute myeloid leukemia (AML) cell lines by inducing cell transdifferentiation. The NEDD8-activating enzyme inhibitor pevonedistat (MLN4924, TAK-924) is an investigational drug with antiproliferative activities in AML, and is also reported to induce cell differentiation. We therefore tested the combination of these two agents in AML models. The combination treatment resulted in synergistic growth inhibition of AML cells, accompanied by enhanced transdifferentiation of an erythroid leukemia lineage into granulomonocytic-like lineage cells. In addition, pevonedistat-induced rereplication stress during the S phase was greatly augmented by concomitant treatment with T-3775440, as reflected by the increased induction of apoptosis. We further demonstrated that the combination treatment was markedly effective in subcutaneous tumor xenograft models as well as in a disseminated model of AML, leading to tumor eradication or prolonged survival in T-3775440/pevonedistat cotreated mice. Our findings indicate the therapeutic potential of the combination of LSD1 inhibitors and pevonedistat for the treatment of AML.
Collapse
Affiliation(s)
- Y Ishikawa
- Oncology Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - K Nakayama
- Oncology Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - M Morimoto
- Oncology Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - A Mizutani
- Oncology Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - A Nakayama
- Oncology Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - K Toyoshima
- Oncology Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - A Hayashi
- Oncology Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - S Takagi
- Oncology Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - R Dairiki
- Oncology Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - H Miyashita
- Oncology Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - S Matsumoto
- Integrated Technology Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - K Gamo
- Oncology Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - T Nomura
- Oncology Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - K Nakamura
- Oncology Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| |
Collapse
|
8
|
Burda P, Vargova J, Curik N, Salek C, Papadopoulos GL, Strouboulis J, Stopka T. GATA-1 Inhibits PU.1 Gene via DNA and Histone H3K9 Methylation of Its Distal Enhancer in Erythroleukemia. PLoS One 2016; 11:e0152234. [PMID: 27010793 PMCID: PMC4807078 DOI: 10.1371/journal.pone.0152234] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 03/10/2016] [Indexed: 01/17/2023] Open
Abstract
GATA-1 and PU.1 are two important hematopoietic transcription factors that mutually inhibit each other in progenitor cells to guide entrance into the erythroid or myeloid lineage, respectively. PU.1 controls its own expression during myelopoiesis by binding to the distal URE enhancer, whose deletion leads to acute myeloid leukemia (AML). We herein present evidence that GATA-1 binds to the PU.1 gene and inhibits its expression in human AML-erythroleukemias (EL). Furthermore, GATA-1 together with DNA methyl Transferase I (DNMT1) mediate repression of the PU.1 gene through the URE. Repression of the PU.1 gene involves both DNA methylation at the URE and its histone H3 lysine-K9 methylation and deacetylation as well as the H3K27 methylation at additional DNA elements and the promoter. The GATA-1-mediated inhibition of PU.1 gene transcription in human AML-EL mediated through the URE represents important mechanism that contributes to PU.1 downregulation and leukemogenesis that is sensitive to DNA demethylation therapy.
Collapse
MESH Headings
- Cell Differentiation/genetics
- DNA (Cytosine-5-)-Methyltransferase 1
- DNA (Cytosine-5-)-Methyltransferases/genetics
- DNA (Cytosine-5-)-Methyltransferases/metabolism
- DNA Methylation/genetics
- Enhancer Elements, Genetic
- GATA1 Transcription Factor/genetics
- GATA1 Transcription Factor/metabolism
- Gene Expression Regulation, Leukemic
- Histones/genetics
- Humans
- Leukemia, Erythroblastic, Acute/genetics
- Leukemia, Erythroblastic, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Promoter Regions, Genetic
- Protein Binding
- Proto-Oncogene Proteins/biosynthesis
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Trans-Activators/biosynthesis
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Transcription, Genetic
Collapse
Affiliation(s)
- Pavel Burda
- Biocev and Pathological Physiology, 1st Faculty of Medicine, Charles University in Prague, Czech Republic
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Jarmila Vargova
- Biocev and Pathological Physiology, 1st Faculty of Medicine, Charles University in Prague, Czech Republic
| | - Nikola Curik
- Biocev and Pathological Physiology, 1st Faculty of Medicine, Charles University in Prague, Czech Republic
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Cyril Salek
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Giorgio Lucio Papadopoulos
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas, Heraklion, Crete, Greece
- Department of Biology, University of Crete, Heraklion, Crete, Greece
| | - John Strouboulis
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology-Hellas, Heraklion, Crete, Greece
| | - Tomas Stopka
- Biocev and Pathological Physiology, 1st Faculty of Medicine, Charles University in Prague, Czech Republic
- 1st Medical Department–Hematology, General Faculty Hospital, Prague, Czech Republic
- * E-mail:
| |
Collapse
|
9
|
Nouri M, Deezagi A, Ebrahimi M. Reprogramming of human peripheral blood monocytes to erythroid lineage by blocking of the PU-1 gene expression. Ann Hematol 2016; 95:549-56. [PMID: 26758270 DOI: 10.1007/s00277-015-2583-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 12/23/2015] [Indexed: 11/26/2022]
Abstract
In hematopoietic system development, PU.1 and GATA-1 as lineage-specific transcription factors (TF) are expressed in common myeloid progenitors. The cross antagonism between them ascertains gene expression programs of monocytic and erythroid cells, respectively. This concept in transdifferentiation approaches has not been well considered yet, especially in intralineage conversion systems. To demonstrate whether PU.1 suppression induces monocyte lineage conversion into red blood cells, a combination of three PU.1-specific siRNAs was implemented to knock down PU.1 gene expression and generate the balance in favor of GATA-1 expression to induce erythroid differentiation. For this purpose, monocytes were isolated from human peripheral blood and transfected by PU.1 siRNAs. In transfected monocytes, the rate of PU.1 expression in mRNA level was significantly decreased until 0.38 ± 0.118 when compared to untreated monocytes at 72 h (p value ≤0.05) which resulted in significant overexpression of GATA1 of 16.1 ± 0.343-fold compared to the untreated group (p value ≤0.01). Subsequently, overexpression of hemoglobin (α 13.26 ± 1.34-fold; p value≤0.0001) and β-globin (37.55 ± 16.56-fold; p value≤0.0001) was observed when compared to control groups. The results of western immunoblotting confirm those findings too. While, reduced expression of monocyte, CD14 gene, was observed in qRT-PCR and flow cytometry results. Our results suggest that manipulating the ratio of the two TFs in bifurcation differentiation pathways via applying siRNA technology can possibly change the cells' fate as a safe way for therapeutics application.
Collapse
Affiliation(s)
- Masoumeh Nouri
- Department of Molecular Medicine and Biochemistry, National Institute of Genetic Engineering and Biotechnology Km. 17, Karaj-Tehran freeway Pajouhesh Blvd., P.O.Box 14155-6343, Tehran, Iran
| | - Abdolkhalegh Deezagi
- Department of Molecular Medicine and Biochemistry, National Institute of Genetic Engineering and Biotechnology Km. 17, Karaj-Tehran freeway Pajouhesh Blvd., P.O.Box 14155-6343, Tehran, Iran.
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
10
|
Seipel K, Marques MT, Bozzini MA, Meinken C, Mueller BU, Pabst T. Inactivation of the p53-KLF4-CEBPA Axis in Acute Myeloid Leukemia. Clin Cancer Res 2015; 22:746-56. [PMID: 26408402 DOI: 10.1158/1078-0432.ccr-15-1054] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 09/20/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE In acute myeloid leukemia (AML), the transcription factors CEBPA and KLF4 as well as the universal tumor suppressor p53 are frequently deregulated. Here, we investigated the extent of dysregulation, the molecular interactions, and the mechanisms involved. EXPERIMENTAL DESIGN One hundred ten AML patient samples were analyzed for protein levels of CEBPA, KLF4, p53, and p53 modulators. Regulation of CEBPA gene expression by KLF4 and p53 or by chemical p53 activators was characterized in AML cell lines. RESULTS We found that CEBPA gene transcription can be directly activated by p53 and KLF4, suggesting a p53-KLF4-CEBPA axis. In AML patient cells, we observed a prominent loss of p53 function and concomitant reduction of KLF4 and CEBPA protein levels. Assessment of cellular p53 modulator proteins indicated that p53 inactivation in leukemic cells correlated with elevated levels of the nuclear export protein XPO1/CRM1 and increase of the p53 inhibitors MDM2 and CUL9/PARC in the cytoplasm. Finally, restoring p53 function following treatment with cytotoxic chemotherapy compounds and p53 restoring non-genotoxic agents induced CEBPA gene expression, myeloid differentiation, and cell-cycle arrest in AML cells. CONCLUSIONS The p53-KLF4-CEBPA axis is deregulated in AML but can be functionally restored by conventional chemotherapy and novel p53 activating treatments.
Collapse
Affiliation(s)
- Katja Seipel
- Department of Clinical Research, University and University Hospital of Berne, Berne, Switzerland. Department of Medical Oncology, University and University Hospital of Berne, Berne, Switzerland
| | - Miguel Teixera Marques
- Department of Clinical Research, University and University Hospital of Berne, Berne, Switzerland
| | - Marie-Ange Bozzini
- Department of Clinical Research, University and University Hospital of Berne, Berne, Switzerland
| | - Christina Meinken
- Department of Clinical Research, University and University Hospital of Berne, Berne, Switzerland
| | - Beatrice U Mueller
- Department of Clinical Research, University and University Hospital of Berne, Berne, Switzerland
| | - Thomas Pabst
- Department of Medical Oncology, University and University Hospital of Berne, Berne, Switzerland.
| |
Collapse
|
11
|
Dluhosova M, Curik N, Vargova J, Jonasova A, Zikmund T, Stopka T. Epigenetic control of SPI1 gene by CTCF and ISWI ATPase SMARCA5. PLoS One 2014; 9:e87448. [PMID: 24498324 PMCID: PMC3911986 DOI: 10.1371/journal.pone.0087448] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 12/24/2013] [Indexed: 01/08/2023] Open
Abstract
CCCTC-binding factor (CTCF) can both activate as well as inhibit transcription by forming chromatin loops between regulatory regions and promoters. In this regard, Ctcf binding on non-methylated DNA and its interaction with the Cohesin complex results in differential regulation of the H19/Igf2 locus. Similarly, a role for CTCF has been established in normal hematopoietic development; however its involvement in leukemia remains elusive. Here, we show that Ctcf binds to the imprinting control region of H19/Igf2 in AML blasts. We also demonstrate that Smarca5, which also associates with the Cohesin complex, facilitates Ctcf binding to its target sites on DNA. Furthermore, Smarca5 supports Ctcf functionally and is needed for enhancer-blocking effect at ICR. We next asked whether CTCF and SMARCA5 control the expression of key hematopoiesis regulators. In normally differentiating myeloid cells both CTCF and SMARCA5 together with members of the Cohesin complex are recruited to the SPI1 gene, a key hematopoiesis regulator and leukemia suppressor. Due to DNA methylation, CTCF binding to the SPI1 gene is blocked in AML blasts. Upon AZA-mediated DNA demethylation of human AML blasts, CTCF and SMARCA5 are recruited to the −14.4 Enhancer of SPI1 gene and block its expression. Our data provide new insight into complex SPI1 gene regulation now involving additional key epigenetic factors, CTCF and SMARCA5 that control PU.1 expression at the −14.4 Enhancer.
Collapse
MESH Headings
- Acute Disease
- Adenosine Triphosphatases/genetics
- Adenosine Triphosphatases/metabolism
- Animals
- Azacitidine/pharmacology
- CCCTC-Binding Factor
- Cell Line, Tumor
- Chromosomal Proteins, Non-Histone/genetics
- Chromosomal Proteins, Non-Histone/metabolism
- DNA Methylation/drug effects
- Epigenesis, Genetic
- Gene Expression Regulation, Neoplastic
- Genomic Imprinting
- HeLa Cells
- Humans
- Immunoblotting
- Insulin-Like Growth Factor II/genetics
- Insulin-Like Growth Factor II/metabolism
- K562 Cells
- Leukemia, Erythroblastic, Acute/genetics
- Leukemia, Erythroblastic, Acute/metabolism
- Leukemia, Erythroblastic, Acute/pathology
- Leukemia, Myeloid/genetics
- Leukemia, Myeloid/metabolism
- Leukemia, Myeloid/pathology
- Microscopy, Confocal
- Protein Binding
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- RNA Interference
- RNA, Long Noncoding/genetics
- RNA, Long Noncoding/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Trans-Activators/genetics
- Trans-Activators/metabolism
Collapse
Affiliation(s)
- Martina Dluhosova
- Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Nikola Curik
- Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Jarmila Vargova
- Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Anna Jonasova
- Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
- Department of Medicine - Hematology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Tomas Zikmund
- Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - Tomas Stopka
- Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
- Department of Medicine - Hematology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
- * E-mail:
| |
Collapse
|
12
|
Basova P, Pospisil V, Savvulidi F, Burda P, Vargova K, Stanek L, Dluhosova M, Kuzmova E, Jonasova A, Steidl U, Laslo P, Stopka T. Aggressive acute myeloid leukemia in PU.1/p53 double-mutant mice. Oncogene 2013; 33:4735-45. [PMID: 24121269 DOI: 10.1038/onc.2013.414] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 08/23/2013] [Accepted: 09/02/2013] [Indexed: 01/02/2023]
Abstract
PU.1 downregulation within hematopoietic stem and progenitor cells (HSPCs) is the primary mechanism for the development of acute myeloid leukemia (AML) in mice with homozygous deletion of the upstream regulatory element (URE) of PU.1 gene. p53 is a well-known tumor suppressor that is often mutated in human hematologic malignancies including AML and adds to their aggressiveness; however, its genetic deletion does not cause AML in mouse. Deletion of p53 in the PU.1(ure/ure) mice (PU.1(ure/ure)p53(-/-)) results in more aggressive AML with shortened overall survival. PU.1(ure/ure)p53(-/-) progenitors express significantly lower PU.1 levels. In addition to URE deletion we searched for other mechanisms that in the absence of p53 contribute to decreased PU.1 levels in PU.1(ure/ure)p53(-/-) mice. We found involvement of Myb and miR-155 in downregulation of PU.1 in aggressive murine AML. Upon inhibition of either Myb or miR-155 in vitro the AML progenitors restore PU.1 levels and lose leukemic cell growth similarly to PU.1 rescue. The MYB/miR-155/PU.1 axis is a target of p53 and is activated early after p53 loss as indicated by transient p53 knockdown. Furthermore, deregulation of both MYB and miR-155 coupled with PU.1 downregulation was observed in human AML, suggesting that MYB/miR-155/PU.1 mechanism may be involved in the pathogenesis of AML and its aggressiveness characterized by p53 mutation.
Collapse
Affiliation(s)
- P Basova
- 1] Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic [2] Department of Experimental Biomodels, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - V Pospisil
- Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - F Savvulidi
- Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - P Burda
- Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - K Vargova
- Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - L Stanek
- 1] Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic [2] Department of Pathology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - M Dluhosova
- Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - E Kuzmova
- Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - A Jonasova
- 1] Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic [2] Department of Medicine-Haematology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | - U Steidl
- Albert Einstein College of Medicine, Yeshiva University, Bronx, NY, USA
| | - P Laslo
- Section of Experimental Haematology, Leeds Institute of Cancer and Pathology, St James's University Hospital, University of Leeds, Leeds, UK
| | - T Stopka
- 1] Department of Pathophysiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic [2] Department of Medicine-Haematology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| |
Collapse
|
13
|
Kar A, Gutierrez-Hartmann A. Molecular mechanisms of ETS transcription factor-mediated tumorigenesis. Crit Rev Biochem Mol Biol 2013; 48:522-43. [PMID: 24066765 DOI: 10.3109/10409238.2013.838202] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The E26 transformation-specific (ETS) family of transcription factors is critical for development, differentiation, proliferation and also has a role in apoptosis and tissue remodeling. Changes in expression of ETS proteins therefore have a significant impact on normal physiology of the cell. Transcriptional consequences of ETS protein deregulation by overexpression, gene fusion, and modulation by RAS/MAPK signaling are linked to alterations in normal cell functions, and lead to unlimited increased proliferation, sustained angiogenesis, invasion and metastasis. Existing data show that ETS proteins control pathways in epithelial cells as well as stromal compartments, and the crosstalk between the two is essential for normal development and cancer. In this review, we have focused on ETS factors with a known contribution in cancer development. Instead of focusing on a prototype, we address cancer associated ETS proteins and have highlighted the diverse mechanisms by which they affect carcinogenesis. Finally, we discuss strategies for ETS factor targeting as a potential means for cancer therapeutics.
Collapse
|
14
|
5-azacitidine in aggressive myelodysplastic syndromes regulates chromatin structure at PU.1 gene and cell differentiation capacity. Leukemia 2012; 26:1804-11. [PMID: 22343522 DOI: 10.1038/leu.2012.47] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Epigenetic 5-azacitidine (AZA) therapy of high-risk myelodysplastic syndromes (MDS) and acute myelogenous leukemia (AML) represents a promising, albeit not fully understood, approach. Hematopoietic transcription factor PU.1 is dynamically regulated by upstream regulatory element (URE), whose deletion causes downregulation of PU.1 leading to AML in mouse. In this study a significant group of the high-risk MDS patients, as well as MDS cell lines, displayed downregulation of PU.1 expression within CD34+ cells, which was associated with DNA methylation of the URE. AZA treatment in vitro significantly demethylated URE, leading to upregulation of PU.1 followed by derepression of its transcriptional targets and onset of myeloid differentiation. Addition of colony-stimulating factors (CSFs; granulocyte-CSF, granulocyte-macrophage-CSF and macrophage-CSF) modulated AZA-mediated effects on reprogramming of histone modifications at the URE and cell differentiation outcome. Our data collectively support the importance of modifying the URE chromatin structure as a regulatory mechanism of AZA-mediated activation of PU.1 and induction of the myeloid program in MDS.
Collapse
|
15
|
Increasing the folic acid content of maternal or post-weaning diets induces differential changes in phosphoenolpyruvate carboxykinase mRNA expression and promoter methylation in rats. Br J Nutr 2011; 108:852-7. [PMID: 22136740 DOI: 10.1017/s0007114511006155] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Environmental exposures throughout the life course, including nutrition, may induce phenotypic and epigenetic changes. There is limited information about how timing affects the nature of such effects induced by a specific nutritional exposure. We investigated the effect of increased exposure to folic acid before birth or during the juvenile-pubertal period in rats on the epigenetic regulation of glucose homeostasis. Rats were fed either a folic acid-adequate (AF; 1 mg/kg feed) or a folic acid-supplemented (FS; 5 mg/kg feed) diet from conception until delivery and then an AF diet during lactation. Juvenile rats were fed either the AF or the FS diet from weaning for 28 d and then an AF diet. Liver and blood were collected after a 12 h fast between postnatal days 84 and 90. Maternal FS diet increased plasma glucose concentration significantly (P < 0·05) in females, but not in males. Post-weaning FS diet decreased glucose concentration significantly in females, but increased glucose concentration in males. There were no effects of the FS diet on phosphoenolpyruvate carboxykinase (PEPCK) mRNA expression in males, while the pattern of expression was related to plasma glucose concentration in females. The FS diet induced specific changes in the methylation of individual CpG in females, but not in males, which were related to the time of exposure. Methylation of CpG - 248 increased the binding of CCAAT-enhancer-binding protein β to the PEPCK promoter. Together, these findings show that both the period during the life course and sex influence the effect of increased exposure to folic acid on the epigenetic regulation of PEPCK and glucose homeostasis.
Collapse
|
16
|
PU.1 and Haematopoietic Cell Fate: Dosage Matters. Int J Cell Biol 2011; 2011:808524. [PMID: 21845190 PMCID: PMC3154517 DOI: 10.1155/2011/808524] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Accepted: 06/22/2011] [Indexed: 11/17/2022] Open
Abstract
The ETS family transcription factor PU.1 is a key regulator of haematopoietic differentiation. Its expression is dynamically controlled throughout haematopoiesis in order to direct appropriate lineage specification. Elucidating the biological role of PU.1 has proved challenging. This paper will discuss how a range of experiments in cell lines and mutant and transgenic mouse models have enhanced our knowledge of the mechanisms by which PU.1 drives lineage-specific differentiation during haematopoiesis.
Collapse
|
17
|
Wontakal SN, Guo X, Will B, Shi M, Raha D, Mahajan MC, Weissman S, Snyder M, Steidl U, Zheng D, Skoultchi AI. A large gene network in immature erythroid cells is controlled by the myeloid and B cell transcriptional regulator PU.1. PLoS Genet 2011; 7:e1001392. [PMID: 21695229 PMCID: PMC3111485 DOI: 10.1371/journal.pgen.1001392] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2010] [Accepted: 05/10/2011] [Indexed: 01/17/2023] Open
Abstract
PU.1 is a hematopoietic transcription factor that is required for the development of myeloid and B cells. PU.1 is also expressed in erythroid progenitors, where it blocks erythroid differentiation by binding to and inhibiting the main erythroid promoting factor, GATA-1. However, other mechanisms by which PU.1 affects the fate of erythroid progenitors have not been thoroughly explored. Here, we used ChIP-Seq analysis for PU.1 and gene expression profiling in erythroid cells to show that PU.1 regulates an extensive network of genes that constitute major pathways for controlling growth and survival of immature erythroid cells. By analyzing fetal liver erythroid progenitors from mice with low PU.1 expression, we also show that the earliest erythroid committed cells are dramatically reduced in vivo. Furthermore, we find that PU.1 also regulates many of the same genes and pathways in other blood cells, leading us to propose that PU.1 is a multifaceted factor with overlapping, as well as distinct, functions in several hematopoietic lineages.
Collapse
Affiliation(s)
- Sandeep N. Wontakal
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Xingyi Guo
- The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Britta Will
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Minyi Shi
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut, United States of America
- Department of Genetics, Stanford University, Stanford, California, United States of America
| | - Debasish Raha
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut, United States of America
| | - Milind C. Mahajan
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Sherman Weissman
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Michael Snyder
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut, United States of America
- Department of Genetics, Stanford University, Stanford, California, United States of America
| | - Ulrich Steidl
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Deyou Zheng
- The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Departments of Genetics and Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
- * E-mail: (AI Skoultchi); (D Zheng)
| | - Arthur I. Skoultchi
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- * E-mail: (AI Skoultchi); (D Zheng)
| |
Collapse
|
18
|
Cao N, Yao ZX. The hemangioblast: from concept to authentication. Anat Rec (Hoboken) 2011; 294:580-8. [PMID: 21370498 DOI: 10.1002/ar.21360] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Accepted: 01/13/2011] [Indexed: 11/06/2022]
Abstract
The hemangioblast hypothesis has been hotly debated for over a century. Hemangioblasts are defined as multipotent cells that can give rise to both hematopoietic cells and endothelial cells. The existence of hemangioblasts has now been confirmed and many important molecules and several signaling pathways are involved in their generation and differentiation. Fibroblast growth factor, renin-angiotensin system and runt-related transcription factor 1 (Runx1) direct the formation of hemangioblasts through highly selective gene expression patterns. On the other hand, the hemogenic endothelium theory and a newly discovered pattern of hematopoietic/endothelial differentiation make the genesis of hemangioblasts more complicated. But how hemangioblasts are formed and how hematopoietic cells or endothelial cells are derived from remains largely unknown. Here we summarize the current knowledge of the signaling pathways and molecules involved in hemangioblast development and suggest some future clinical applications.
Collapse
Affiliation(s)
- Nian Cao
- Department of Physiology, Third Military Medical University, Chongqing, China
| | | |
Collapse
|
19
|
MYB transcriptionally regulates the miR-155 host gene in chronic lymphocytic leukemia. Blood 2011; 117:3816-25. [PMID: 21296997 DOI: 10.1182/blood-2010-05-285064] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Elevated levels of microRNA miR-155 represent a candidate pathogenic factor in chronic B-lymphocytic leukemia (B-CLL). In this study, we present evidence that MYB (v-myb myeloblastosis viral oncogene homolog) is overexpressed in a subset of B-CLL patients. MYB physically associates with the promoter of miR-155 host gene (MIR155HG, also known as BIC, B-cell integration cluster) and stimulates its transcription. This coincides with the hypermethylated histone H3K4 residue and spread hyperacetylation of H3K9 at MIR155HG promoter. Our data provide evidence of oncogenic activities of MYB in B-CLL that include its stimulatory role in MIR155HG transcription.
Collapse
|
20
|
Brown G, Hughes PJ, Michell RH, Ceredig R. The versatility of haematopoietic stem cells: implications for leukaemia. Crit Rev Clin Lab Sci 2010; 47:171-80. [DOI: 10.3109/10408363.2010.530150] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Geoffrey Brown
- School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| | | | | | | |
Collapse
|
21
|
The role of PU.1 and GATA-1 transcription factors during normal and leukemogenic hematopoiesis. Leukemia 2010; 24:1249-57. [DOI: 10.1038/leu.2010.104] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|