1
|
Zhou G, Lu D. Proteomics screening uncovers HMGA1 as a promising negative regulator for γ-globin expression in response to decreased β-globin levels. J Proteomics 2023; 286:104957. [PMID: 37423548 DOI: 10.1016/j.jprot.2023.104957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/11/2023]
Abstract
Reactivation of fetal hemoglobin (HbF) is a critical goal for the treatment of patients with hemoglobinopathies. β-globin disorders can trigger stress erythropoiesis in red blood cells (RBCs). Cell-intrinsic erythroid stress signals promote erythroid precursors to express high levels of fetal hemoglobin, which is also known as γ-globin. However, the molecular mechanism underlying γ-globin production during cell-intrinsic erythroid stress remains to be elucidated. Here, we utilized CRISPR-Cas9 to model a stressed state caused by reduced levels of adult β-globin in HUDEP2 human erythroid progenitor cells. We found that a decrease in β-globin expression correlates with the upregulation of γ-globin expression. We also identified transcription factor high-mobility group A1 (HMGA1; formerly HMG-I/Y) as a potential γ-globin regulator that responds to reduced β-globin levels. Upon erythroid stress, there is a downregulation of HMGA1, which normally binds -626 to -610 base pairs upstream from the STAT3 promoter, to downregulate STAT3 expression. STAT3 is a known γ-globin repressor, so the downregulation of HMGA1 ultimately upregulates γ-globin expression. SIGNIFICANCE: This study demonstrated HMGA1 as a potential regulator in the poorly understood phenomenon of stress-induced globin compensation, and after further validation these results might inform new strategies to treat patients with sickle cell disease and β-thalassemia.
Collapse
Affiliation(s)
- Guoqiang Zhou
- Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Guangzhou, 511458, China
| | - Daru Lu
- Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Guangzhou, 511458, China; NHC Key Laboratory of Birth Defects and Reproductive Health, Chongqing Key Laboratory of Birth Defects and Reproductive Health, Chongqing Population and Family Planning, Science and Technology Research Institute, Chongqing, China.
| |
Collapse
|
2
|
Starlard-Davenport A, Gu Q, Pace BS. Targeting Genetic Modifiers of HBG Gene Expression in Sickle Cell Disease: The miRNA Option. Mol Diagn Ther 2022; 26:497-509. [PMID: 35553407 PMCID: PMC9098152 DOI: 10.1007/s40291-022-00589-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2022] [Indexed: 12/14/2022]
Abstract
Sickle cell disease (SCD) is one of the most common inherited hemoglobinopathy disorders that affects millions of people worldwide. Reactivation of HBG (HBG1, HBG2) gene expression and induction of fetal hemoglobin (HbF) is an important therapeutic strategy for ameliorating the clinical symptoms and severity of SCD. Hydroxyurea is the only US FDA-approved drug with proven efficacy to induce HbF in SCD patients, yet serious complications have been associated with its use. Over the last three decades, numerous additional pharmacological agents that reactivate HBG transcription in vitro have been investigated, but few have proceeded to FDA approval, with the exception of arginine butyrate and decitabine; however, neither drug met the requirements for routine clinical use due to difficulties with oral delivery and inability to achieve therapeutic levels. Thus, novel approaches that produce sufficient efficacy, specificity, and sustainable HbF induction with low adverse effects are desirable. More recently, microRNAs (miRNAs) have gained attention for their diagnostic and therapeutic potential to treat various diseases ranging from cancer to Alzheimer’s disease via targeting oncogenes and their gene products. Thus, it is plausible that miRNAs that target HBG regulatory genes may be useful for inducing HbF as a treatment for SCD. Our laboratory and others have documented the association of miRNAs with HBG activation or suppression via silencing transcriptional repressors and activators, respectively, of HBG expression. Herein, we review progress made in understanding molecular mechanisms of miRNA-mediated HBG regulation and discuss the extent to which molecular targets of HBG might be suitable prospects for development of SCD clinical therapy. Lastly, we discuss challenges with the application of miRNA delivery in vivo and provide potential strategies for overcoming barriers in the future.
Collapse
Affiliation(s)
- Athena Starlard-Davenport
- College of Medicine, Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| | - Qingqing Gu
- College of Medicine, Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.,Department of Cardiology, Affiliated Hospital of Nantong University, Jiangsu, 226001, China
| | - Betty S Pace
- Department of Pediatrics, Division of Hematology/Oncology, Augusta University, Augusta, GA, USA.,Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA
| |
Collapse
|
3
|
Sajjadi-Dokht M, Merza Mohamad TA, Rahman HS, Maashi MS, Danshina S, Shomali N, Solali S, Marofi F, Zeinalzadeh E, Akbari M, Adili A, Aslaminabad R, Hagh MF, Jarahian M. MicroRNAs and JAK/STAT3 signaling: A new promising therapeutic axis in blood cancers. Genes Dis 2021; 9:849-867. [PMID: 35685482 PMCID: PMC9170603 DOI: 10.1016/j.gendis.2021.10.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/16/2021] [Accepted: 10/22/2021] [Indexed: 11/27/2022] Open
Abstract
Blood disorders include a wide spectrum of blood-associated malignancies resulting from inherited or acquired defects. The ineffectiveness of existing therapies against blood disorders arises from different reasons, one of which is drug resistance, so different types of leukemia may show different responses to treatment. Leukemia occurs for a variety of genetic and acquired reasons, leading to uncontrolled proliferation in one or more cell lines. Regarding the genetic defects, oncogene signal transducer and activator of transcription (STAT) family transcription factor, especially STAT3, play an essential role in hematological disorders onset and progress upon mutations, dysfunction, or hyperactivity. Besides, microRNAs, as biological molecules, has been shown to play a dual role in either tumorigenesis and tumor suppression in various cancers. Besides, a strong association between STAT3 and miRNA has been reported. For example, miRNAs can regulate STAT3 via targeting its upstream mediators such as IL6, IL9, and JAKs or directly binding to the STAT3 gene. On the other hand, STAT3 can regulate miRNAs. In this review study, we aimed to determine the role of either microRNAs and STAT3 along with their effect on one another's activity and function in hematological malignancies.
Collapse
|
4
|
Giraud G, Kolovos P, Boltsis I, van Staalduinen J, Guyot B, Weiss-Gayet M, IJcken WV, Morlé F, Grosveld F. Interplay between FLI-1 and the LDB1 complex in murine erythroleukemia cells and during megakaryopoiesis. iScience 2021; 24:102210. [PMID: 33733070 PMCID: PMC7940982 DOI: 10.1016/j.isci.2021.102210] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/22/2020] [Accepted: 02/17/2021] [Indexed: 11/29/2022] Open
Abstract
Transcription factors are key players in a broad range of cellular processes such as cell-fate decision. Understanding how they act to control these processes is of critical importance for therapy purposes. FLI-1 controls several hematopoietic lineage differentiation including megakaryopoiesis and erythropoiesis. Its aberrant expression is often observed in cancer and is associated with poor prognosis. We showed that FLI-1 interacts with the LDB1 complex, which also plays critical roles in erythropoiesis and megakaryopoiesis. In this study, we aimed to unravel how FLI-1 and the LDB1 complex act together in murine erythroleukemia cells and in megakaryocyte. Combining omics techniques, we show that FLI-1 enables the recruitment of the LDB1 complex to regulatory sequences of megakaryocytic genes and to enhancers. We show as well for the first time that FLI-1 is able to modulate the 3D chromatin organization by promoting chromatin looping between enhancers and promoters most likely through the LDB1 complex. FLI-1 is important for the recruitment of the LDB1 complex FLI-1 is important for chromatin looping FLI-1 and the LDB1 complex co-regulate megakaryopoiesis
Collapse
Affiliation(s)
- Guillaume Giraud
- Department of Cell Biology, Erasmus Medical Centre, 3015CN Rotterdam, the Netherlands
| | - Petros Kolovos
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis 68100, Greece
| | - Ilias Boltsis
- Department of Cell Biology, Erasmus Medical Centre, 3015CN Rotterdam, the Netherlands
| | - Jente van Staalduinen
- Department of Cell Biology, Erasmus Medical Centre, 3015CN Rotterdam, the Netherlands
| | - Boris Guyot
- CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France.,Université de Lyon, Lyon, France.,Department of Immunity, Virus and Microenvironment, Lyon, France
| | - Michele Weiss-Gayet
- Institut NeuroMyoGène, CNRS UMR 5310 - INSERM U1217 - Université de Lyon - Université Claude Bernard Lyon 1, Lyon, France
| | - Wilfred van IJcken
- Biomics Center, Erasmus University Medical Center, 3015CN Rotterdam, the Netherlands
| | - François Morlé
- Institut NeuroMyoGène, CNRS UMR 5310 - INSERM U1217 - Université de Lyon - Université Claude Bernard Lyon 1, Lyon, France
| | - Frank Grosveld
- Department of Cell Biology, Erasmus Medical Centre, 3015CN Rotterdam, the Netherlands
| |
Collapse
|
5
|
Cannon M, Phillips H, Smith S, Williams K, Brinton L, Gregory C, Landes K, Desai P, Byrd J, Lapalombella R. Large-Scale Drug Screen Identifies FDA-Approved Drugs for Repurposing in Sickle-Cell Disease. J Clin Med 2020; 9:E2276. [PMID: 32708954 PMCID: PMC7408993 DOI: 10.3390/jcm9072276] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 07/07/2020] [Accepted: 07/15/2020] [Indexed: 12/26/2022] Open
Abstract
Sickle-cell disease (SCD) is a debilitating hematological disorder with very few approved treatment options. Therapeutic reactivation of fetal hemoglobin (HbF) is one of the most pursued methods for ameliorating the systemic manifestations of SCD. Despite this, very few pharmacological agents have advanced to clinical trials or marketing for use. In this study, we report the development of an HbF in situ intracellular immunoblot assay coupled to a high-throughput drug screen to identify Food and Drug Administration (FDA) approved drugs that can be repurposed clinically for treatment of SCD. Using this assay we evaluated the National Institute of Health (NIH) Clinical Collection (NCC), a publicly available library of 725 small molecules, and found nine candidates that can significantly re-express HbF in erythroid cell lines as well as primary erythroblasts derived from SCD patients. Furthermore, we show the strong effects on HbF expression of these candidates to occur with minimal cytotoxicity in 7 of the 9 drugs. Given these data and their proven history of use for other indications, we hypothesize that several of these candidate drugs warrant further investigation for use in SCD.
Collapse
Affiliation(s)
- Matthew Cannon
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA; (M.C.); (H.P.); (S.S.); (K.W.); (L.B.); (C.G.); (K.L.); (P.D.); (J.B.)
| | - Hannah Phillips
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA; (M.C.); (H.P.); (S.S.); (K.W.); (L.B.); (C.G.); (K.L.); (P.D.); (J.B.)
| | - Sidney Smith
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA; (M.C.); (H.P.); (S.S.); (K.W.); (L.B.); (C.G.); (K.L.); (P.D.); (J.B.)
| | - Katie Williams
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA; (M.C.); (H.P.); (S.S.); (K.W.); (L.B.); (C.G.); (K.L.); (P.D.); (J.B.)
| | - Lindsey Brinton
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA; (M.C.); (H.P.); (S.S.); (K.W.); (L.B.); (C.G.); (K.L.); (P.D.); (J.B.)
| | - Charles Gregory
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA; (M.C.); (H.P.); (S.S.); (K.W.); (L.B.); (C.G.); (K.L.); (P.D.); (J.B.)
| | - Kristina Landes
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA; (M.C.); (H.P.); (S.S.); (K.W.); (L.B.); (C.G.); (K.L.); (P.D.); (J.B.)
| | - Payal Desai
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA; (M.C.); (H.P.); (S.S.); (K.W.); (L.B.); (C.G.); (K.L.); (P.D.); (J.B.)
| | - John Byrd
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA; (M.C.); (H.P.); (S.S.); (K.W.); (L.B.); (C.G.); (K.L.); (P.D.); (J.B.)
- College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Rosa Lapalombella
- Division of Hematology, The Ohio State University, Columbus, OH 43210, USA; (M.C.); (H.P.); (S.S.); (K.W.); (L.B.); (C.G.); (K.L.); (P.D.); (J.B.)
| |
Collapse
|
6
|
Chen C, Lu M, Lin S, Qin W. The nuclear gene rpl18 regulates erythroid maturation via JAK2-STAT3 signaling in zebrafish model of Diamond-Blackfan anemia. Cell Death Dis 2020; 11:135. [PMID: 32075953 PMCID: PMC7031319 DOI: 10.1038/s41419-020-2331-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/04/2020] [Accepted: 02/04/2020] [Indexed: 11/09/2022]
Abstract
Diamond-Blackfan anemia (DBA) is a rare, inherited bone marrow failure syndrome, characterized by red blood cell aplasia, developmental abnormalities, and enhanced risk of malignancy. However, the underlying pathogenesis of DBA is yet to be understood. Recently, mutations in the gene encoding ribosomal protein (RP) L18 were identified in DBA patients. RPL18 is a crucial component of the ribosomal large subunit but its role in hematopoiesis remains unknown. To genetically model the ribosomal defect identified in DBA, we generated a rpl18 mutant line in zebrafish, using CRISPR/Cas9 system. Molecular characterization of this mutant line demonstrated that Rpl18 deficiency mirrored the erythroid defects of DBA, namely a lack of mature red blood cells. Rpl18 deficiency caused an increase in p53 activation and JAK2-STAT3 activity. Furthermore, we found inhibitors of JAK2 or STAT3 phosphorylation could rescue anemia in rpl18 mutants. Our research provides a new in vivo model of Rpl18 deficiency and suggests involvement of signal pathway of JAK2-STAT3 in the DBA pathogenesis.
Collapse
Affiliation(s)
- Cheng Chen
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Mengjia Lu
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China
| | - Shuo Lin
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA, 90095, USA
| | - Wei Qin
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, 518055, China.
| |
Collapse
|
7
|
Kuo SC, Li Y, Cheng KC, Niu CS, Cheng JT, Niu HS. Increase in renal erythropoietin receptors in diabetic rats is mainly mediated by hyperglycemia associated with the STAT3/GATA-1 signaling pathway. Biomed Pharmacother 2017; 96:1094-1102. [DOI: 10.1016/j.biopha.2017.11.115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 11/18/2017] [Accepted: 11/20/2017] [Indexed: 10/18/2022] Open
|
8
|
Ward CM, Li B, Pace BS. Original Research: Stable expression of miR-34a mediates fetal hemoglobin induction in K562 cells. Exp Biol Med (Maywood) 2016; 241:719-29. [PMID: 26940952 PMCID: PMC4950382 DOI: 10.1177/1535370216636725] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Sickle cell anemia is a common genetic disorder caused by a point mutation in the sixth codon of the β-globin gene affecting people of African descent worldwide. A wide variety of clinical phenotypes ranging from mild to severe symptoms and complications occur due to hemoglobin S polymerization, red blood cell sickling, and vaso-occlusion. Research efforts are ongoing to develop strategies of fetal hemoglobin (HbF; α2γ2) induction to inhibit sickle hemoglobin polymerization and improve clinical outcomes. Insights have been gained from investigating mutations in the β-globin locus or transcription factors involved in the mechanisms of hemoglobin switching. Recent efforts to expand molecular targets that modulate γ-globin expression involve microRNAs that work through posttranscriptional gene regulation. Therefore, the goal of our study was to identify novel microRNA genes involved in fetal hemoglobin expression. Using in silico analysis, we identified a miR-34a binding site in the γ-globin mRNA which was tested for functional relevance. Stable expression of the shMIMIC miR-34a lentivirus vector increased fetal hemoglobin levels in single cell K562 clones consistent with silencing of a γ-globin gene repressor. Furthermore, miR-34a promoted cell differentiation supported by increased expression of KLF1, glycophorin A, and the erythropoietin receptor. Western blot analysis of known negative regulators of γ-globin including YY1, histone deacetylase 1, and STAT3, which are regulated by miR-34a showed no change in YY1 and histone deacetylase 1 levels; however, total- and phosphorylated-STAT3 levels were decreased in single cell miR-34a K562 clones. These data support a mechanism of fetal hemoglobin activation by miR-34a involving STAT3 gene silencing.
Collapse
Affiliation(s)
- Christina M Ward
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA 30912, USA Department of Pediatrics, Augusta University, Augusta, GA 30912, USA
| | - Biaoru Li
- Department of Pediatrics, Augusta University, Augusta, GA 30912, USA
| | - Betty S Pace
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA 30912, USA Department of Pediatrics, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
9
|
Pace BS, Liu L, Li B, Makala LH. Cell signaling pathways involved in drug-mediated fetal hemoglobin induction: Strategies to treat sickle cell disease. Exp Biol Med (Maywood) 2015; 240:1050-64. [PMID: 26283707 DOI: 10.1177/1535370215596859] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The developmental regulation of globin gene expression has shaped research efforts to establish therapeutic modalities for individuals affected with sickle cell disease and β-thalassemia. Fetal hemoglobin has been shown to block sickle hemoglobin S polymerization to improve symptoms of sickle cell disease; moreover, fetal hemoglobin functions to replace inadequate hemoglobin A synthesis in β-thalassemia thus serving as an effective therapeutic target. In the perinatal period, fetal hemoglobin is synthesized at high levels followed by a decline to adult levels by one year of age. It is known that naturally occurring mutations in the γ-globin gene promoters and distant cis-acting transcription factors produce persistent fetal hemoglobin synthesis after birth to ameliorate clinical symptoms. Major repressor proteins that silence γ-globin during development have been targeted for gene therapy in β-hemoglobinopathies patients. In parallel effort, several classes of pharmacological agents that induce fetal hemoglobin expression through molecular and cell signaling mechanisms have been identified. Herein, we reviewed the progress made in the discovery of signaling molecules targeted by pharmacologic agents that enhance γ-globin expression and have the potential for future drug development to treat the β-hemoglobinopathies.
Collapse
Affiliation(s)
- Betty S Pace
- Department of Pediatrics, Georgia Regents University, Augusta, GA 30912, USA Department of Biochemistry and Molecular Biology, Georgia Regents University, Augusta, GA 30912, USA
| | - Li Liu
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX 75083, USA
| | - Biaoru Li
- Department of Pediatrics, Georgia Regents University, Augusta, GA 30912, USA
| | - Levi H Makala
- Department of Pediatrics, Georgia Regents University, Augusta, GA 30912, USA
| |
Collapse
|
10
|
Arlet JB, Ribeil JA, Guillem F, Negre O, Hazoume A, Marcion G, Beuzard Y, Dussiot M, Moura IC, Demarest S, de Beauchêne IC, Belaid-Choucair Z, Sevin M, Maciel TT, Auclair C, Leboulch P, Chretien S, Tchertanov L, Baudin-Creuza V, Seigneuric R, Fontenay M, Garrido C, Hermine O, Courtois G. HSP70 sequestration by free α-globin promotes ineffective erythropoiesis in β-thalassaemia. Nature 2014; 514:242-6. [PMID: 25156257 DOI: 10.1038/nature13614] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2013] [Accepted: 06/25/2014] [Indexed: 12/28/2022]
Abstract
β-Thalassaemia major (β-TM) is an inherited haemoglobinopathy caused by a quantitative defect in the synthesis of β-globin chains of haemoglobin, leading to the accumulation of free α-globin chains that form toxic aggregates. Despite extensive knowledge of the molecular defects causing β-TM, little is known of the mechanisms responsible for the ineffective erythropoiesis observed in the condition, which is characterized by accelerated erythroid differentiation, maturation arrest and apoptosis at the polychromatophilic stage. We have previously demonstrated that normal human erythroid maturation requires a transient activation of caspase-3 at the later stages of maturation. Although erythroid transcription factor GATA-1, the master transcriptional factor of erythropoiesis, is a caspase-3 target, it is not cleaved during erythroid differentiation. We have shown that, in human erythroblasts, the chaperone heat shock protein70 (HSP70) is constitutively expressed and, at later stages of maturation, translocates into the nucleus and protects GATA-1 from caspase-3 cleavage. The primary role of this ubiquitous chaperone is to participate in the refolding of proteins denatured by cytoplasmic stress, thus preventing their aggregation. Here we show in vitro that during the maturation of human β-TM erythroblasts, HSP70 interacts directly with free α-globin chains. As a consequence, HSP70 is sequestrated in the cytoplasm and GATA-1 is no longer protected, resulting in end-stage maturation arrest and apoptosis. Transduction of a nuclear-targeted HSP70 mutant or a caspase-3-uncleavable GATA-1 mutant restores terminal maturation of β-TM erythroblasts, which may provide a rationale for new targeted therapies of β-TM.
Collapse
Affiliation(s)
- Jean-Benoît Arlet
- 1] Laboratoire INSERM, unité mixte de recherche 1163, centre national de la recherche scientifique (CNRS) équipe de recherche labellisée 8254, 24 Boulevard de Montparnasse, 75015 Paris, France [2] Service de Médecine Interne, Faculté de médecine Paris Descartes, Sorbonne Paris-Cité et Assistance publique - Hôpitaux de Paris, Hôpital Européen Georges Pompidou, 15 rue Leblanc 75908 Paris, France [3] Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Assistance publique - Hôpitaux de Paris, Hôpital Necker, 24 Boulevard de Montparnasse, 75015 Paris, France [4] Laboratory of Excellence GR-Ex, 75015 Paris, France [5]
| | - Jean-Antoine Ribeil
- 1] Laboratoire INSERM, unité mixte de recherche 1163, centre national de la recherche scientifique (CNRS) équipe de recherche labellisée 8254, 24 Boulevard de Montparnasse, 75015 Paris, France [2] Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Assistance publique - Hôpitaux de Paris, Hôpital Necker, 24 Boulevard de Montparnasse, 75015 Paris, France [3] Laboratory of Excellence GR-Ex, 75015 Paris, France [4] Département de Biothérapie, Faculté de médecine Paris Descartes, Sorbonne Paris-Cité et Assistance publique - Hôpitaux de Paris, Hôpital Necker, 149 rue de Sèvres 75015 Paris, France [5]
| | - Flavia Guillem
- 1] Laboratoire INSERM, unité mixte de recherche 1163, centre national de la recherche scientifique (CNRS) équipe de recherche labellisée 8254, 24 Boulevard de Montparnasse, 75015 Paris, France [2] Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Assistance publique - Hôpitaux de Paris, Hôpital Necker, 24 Boulevard de Montparnasse, 75015 Paris, France [3] Laboratory of Excellence GR-Ex, 75015 Paris, France
| | - Olivier Negre
- Commissariat à l'énergie atomique (CEA), Institute of Emerging Diseases and Innovative Therapies (iMETI), 18 Route du Panorama, 92260 Fontenay-aux-Roses, France
| | - Adonis Hazoume
- 1] INSERM, unité mixte de recherche 866, Equipe labellisée Ligue contre le Cancer and Association pour la Recherche contre le Cancer, and Laboratoire d'Excellence Lipoprotéines et santé (LipSTIC), 21033 Dijon, France [2] University of Burgundy, Faculty of Medicine and Pharmacy, 7 boulevard Jeanne d'Arc, 21033 Dijon, France
| | - Guillaume Marcion
- 1] INSERM, unité mixte de recherche 866, Equipe labellisée Ligue contre le Cancer and Association pour la Recherche contre le Cancer, and Laboratoire d'Excellence Lipoprotéines et santé (LipSTIC), 21033 Dijon, France [2] University of Burgundy, Faculty of Medicine and Pharmacy, 7 boulevard Jeanne d'Arc, 21033 Dijon, France
| | - Yves Beuzard
- Commissariat à l'énergie atomique (CEA), Institute of Emerging Diseases and Innovative Therapies (iMETI), 18 Route du Panorama, 92260 Fontenay-aux-Roses, France
| | - Michaël Dussiot
- 1] Laboratoire INSERM, unité mixte de recherche 1163, centre national de la recherche scientifique (CNRS) équipe de recherche labellisée 8254, 24 Boulevard de Montparnasse, 75015 Paris, France [2] Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Assistance publique - Hôpitaux de Paris, Hôpital Necker, 24 Boulevard de Montparnasse, 75015 Paris, France [3] Laboratory of Excellence GR-Ex, 75015 Paris, France [4] INSERM, unité mixte de recherche 699, Hôpital Bichat, 46 rue Henri Huchard, 75018 Paris, France
| | - Ivan Cruz Moura
- 1] Laboratoire INSERM, unité mixte de recherche 1163, centre national de la recherche scientifique (CNRS) équipe de recherche labellisée 8254, 24 Boulevard de Montparnasse, 75015 Paris, France [2] Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Assistance publique - Hôpitaux de Paris, Hôpital Necker, 24 Boulevard de Montparnasse, 75015 Paris, France [3] Laboratory of Excellence GR-Ex, 75015 Paris, France [4] INSERM, unité mixte de recherche 699, Hôpital Bichat, 46 rue Henri Huchard, 75018 Paris, France [5] Faculté de médecine and Université Denis Diderot Paris VII, 5 Rue Thomas Mann, 75013 Paris, France
| | - Samuel Demarest
- Centre national de la recherche scientifique (CNRS), unité mixte de recherche 8113, Ecole Normale Supérieure de Cachan, 61 avenue du président Wilson, 94230 Cachan, France
| | - Isaure Chauvot de Beauchêne
- 1] Centre national de la recherche scientifique (CNRS), unité mixte de recherche 8113, Ecole Normale Supérieure de Cachan, 61 avenue du président Wilson, 94230 Cachan, France [2] Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Campus Paris Saclay, 5 rue Jean-Baptiste Clément 92296 Châtenay-Malabry, France
| | - Zakia Belaid-Choucair
- 1] Laboratoire INSERM, unité mixte de recherche 1163, centre national de la recherche scientifique (CNRS) équipe de recherche labellisée 8254, 24 Boulevard de Montparnasse, 75015 Paris, France [2] Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Assistance publique - Hôpitaux de Paris, Hôpital Necker, 24 Boulevard de Montparnasse, 75015 Paris, France [3] Laboratory of Excellence GR-Ex, 75015 Paris, France
| | - Margaux Sevin
- 1] INSERM, unité mixte de recherche 866, Equipe labellisée Ligue contre le Cancer and Association pour la Recherche contre le Cancer, and Laboratoire d'Excellence Lipoprotéines et santé (LipSTIC), 21033 Dijon, France [2] University of Burgundy, Faculty of Medicine and Pharmacy, 7 boulevard Jeanne d'Arc, 21033 Dijon, France
| | - Thiago Trovati Maciel
- 1] Laboratoire INSERM, unité mixte de recherche 1163, centre national de la recherche scientifique (CNRS) équipe de recherche labellisée 8254, 24 Boulevard de Montparnasse, 75015 Paris, France [2] Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Assistance publique - Hôpitaux de Paris, Hôpital Necker, 24 Boulevard de Montparnasse, 75015 Paris, France [3] Laboratory of Excellence GR-Ex, 75015 Paris, France [4] INSERM, unité mixte de recherche 699, Hôpital Bichat, 46 rue Henri Huchard, 75018 Paris, France [5] Faculté de médecine and Université Denis Diderot Paris VII, 5 Rue Thomas Mann, 75013 Paris, France
| | - Christian Auclair
- 1] Centre national de la recherche scientifique (CNRS), unité mixte de recherche 8113, Ecole Normale Supérieure de Cachan, 61 avenue du président Wilson, 94230 Cachan, France [2] Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Campus Paris Saclay, 5 rue Jean-Baptiste Clément 92296 Châtenay-Malabry, France
| | - Philippe Leboulch
- 1] Commissariat à l'énergie atomique (CEA), Institute of Emerging Diseases and Innovative Therapies (iMETI), 18 Route du Panorama, 92260 Fontenay-aux-Roses, France [2] Women's Hospital and Harvard Medical School, 25 Shattuck St, Boston, Massachusetts 02115, USA
| | - Stany Chretien
- Commissariat à l'énergie atomique (CEA), Institute of Emerging Diseases and Innovative Therapies (iMETI), 18 Route du Panorama, 92260 Fontenay-aux-Roses, France
| | - Luba Tchertanov
- 1] Centre national de la recherche scientifique (CNRS), unité mixte de recherche 8113, Ecole Normale Supérieure de Cachan, 61 avenue du président Wilson, 94230 Cachan, France [2] Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique (LERMIT), Campus Paris Saclay, 5 rue Jean-Baptiste Clément 92296 Châtenay-Malabry, France
| | | | - Renaud Seigneuric
- University of Burgundy, Faculty of Medicine and Pharmacy, 7 boulevard Jeanne d'Arc, 21033 Dijon, France
| | - Michaela Fontenay
- 1] Laboratory of Excellence GR-Ex, 75015 Paris, France [2] Institut Cochin, INSERM, unité mixte de recherche 1016, centre national de la recherche scientifique (CNRS), unité mixte de recherche 8104, Université Paris Descartes, and Assistance publique - Hôpitaux de Paris, Hôpitaux Universitaires Paris Centre, Hôpital Cochin, Service d'hématologie biologique, 27 rue du Faubourg Saitn-Jacques, 75014 Paris, France
| | - Carmen Garrido
- 1] INSERM, unité mixte de recherche 866, Equipe labellisée Ligue contre le Cancer and Association pour la Recherche contre le Cancer, and Laboratoire d'Excellence Lipoprotéines et santé (LipSTIC), 21033 Dijon, France [2] University of Burgundy, Faculty of Medicine and Pharmacy, 7 boulevard Jeanne d'Arc, 21033 Dijon, France [3] Centre anticancéreux George François Leclerc, 1 rue professeur Marion, 21079 Dijon, France [4]
| | - Olivier Hermine
- 1] Laboratoire INSERM, unité mixte de recherche 1163, centre national de la recherche scientifique (CNRS) équipe de recherche labellisée 8254, 24 Boulevard de Montparnasse, 75015 Paris, France [2] Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Assistance publique - Hôpitaux de Paris, Hôpital Necker, 24 Boulevard de Montparnasse, 75015 Paris, France [3] Laboratory of Excellence GR-Ex, 75015 Paris, France [4] Service d'hématologie, Faculté de médecine Paris Descartes, Sorbonne Paris-Cité et Assistance publique - Hôpitaux de Paris Hôpital Necker, 149 rue de Sèvres, 75015 Paris, France [5]
| | - Geneviève Courtois
- 1] Laboratoire INSERM, unité mixte de recherche 1163, centre national de la recherche scientifique (CNRS) équipe de recherche labellisée 8254, 24 Boulevard de Montparnasse, 75015 Paris, France [2] Paris Descartes-Sorbonne Paris Cité University, Imagine Institute, Assistance publique - Hôpitaux de Paris, Hôpital Necker, 24 Boulevard de Montparnasse, 75015 Paris, France [3] Laboratory of Excellence GR-Ex, 75015 Paris, France [4]
| |
Collapse
|
11
|
Ju J, Wang Y, Liu R, Zhang Y, Xu Z, Wang Y, Wu Y, Liu M, Cerruti L, Zou F, Ma C, Fang M, Tan R, Jane SM, Zhao Q. Human fetal globin gene expression is regulated by LYAR. Nucleic Acids Res 2014; 42:9740-52. [PMID: 25092918 PMCID: PMC4150809 DOI: 10.1093/nar/gku718] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Human globin gene expression during development is modulated by transcription factors in a stage-dependent manner. However, the mechanisms controlling the process are still largely unknown. In this study, we found that a nuclear protein, LYAR (human homologue of mouse Ly-1 antibody reactive clone) directly interacted with the methyltransferase PRMT5 which triggers the histone H4 Arg3 symmetric dimethylation (H4R3me2s) mark. We found that PRMT5 binding on the proximal γ-promoter was LYAR-dependent. The LYAR DNA-binding motif (GGTTAT) was identified by performing CASTing (cyclic amplification and selection of targets) experiments. Results of EMSA and ChIP assays confirmed that LYAR bound to a DNA region corresponding to the 5′-untranslated region of the γ-globin gene. We also found that LYAR repressed human fetal globin gene expression in both K562 cells and primary human adult erythroid progenitor cells. Thus, these data indicate that LYAR acts as a novel transcription factor that binds the γ-globin gene, and is essential for silencing the γ-globin gene.
Collapse
Affiliation(s)
- Junyi Ju
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Ying Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Ronghua Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Yichong Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Zhen Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Yadong Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Yupeng Wu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Ming Liu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Loretta Cerruti
- Department of Medicine, Monash University Central Clinical School, Prahran, VIC 3181, Australia
| | - Fengwei Zou
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
| | - Chi Ma
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Ming Fang
- Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Renxiang Tan
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| | - Stephen M Jane
- Department of Medicine, Monash University Central Clinical School, Prahran, VIC 3181, Australia
| | - Quan Zhao
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210093, China
| |
Collapse
|
12
|
Cao C, Zhao G, Yu W, Xie X, Wang W, Yang R, Lv X, Liu D. Activation of STAT3 stimulates AHSP expression in K562 cells. SCIENCE CHINA-LIFE SCIENCES 2014; 57:488-94. [DOI: 10.1007/s11427-014-4652-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 01/05/2014] [Indexed: 02/04/2023]
|
13
|
Bu Y, Su F, Wang X, Gao H, Lei L, Chang N, Wu Q, Hu K, Zhu X, Chang Z, Meng K, Xiong JW. Protein tyrosine phosphatase PTPN9 regulates erythroid cell development through STAT3 dephosphorylation in zebrafish. J Cell Sci 2014; 127:2761-70. [PMID: 24727614 DOI: 10.1242/jcs.145367] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Protein tyrosine phosphatases (PTPs) are involved in hematopoiesis, but the function of many PTPs is not well characterized in vivo. Here, we have identified Ptpn9a, an ortholog of human PTPN9, as a crucial regulator of erythroid cell development in zebrafish embryos. ptpn9a, but not ptpn9b, was expressed in the posterior lateral plate mesoderm and intermediate cell mass - two primitive hematopoietic sites during zebrafish embryogenesis. Morpholino-mediated knockdown of ptpn9a caused erythrocytes to be depleted by inhibiting erythroid cell maturation without affecting erythroid proliferation and apoptosis. Consistently, both dominant-negative PTPN9 (with mutation C515S) and siRNA against PTPN9 inhibited erythroid differentiation in human K562 cells. Mechanistically, depletion of ptpn9 in zebrafish embryos in vivo or in K562 cells in vitro increased phosphorylated STAT3, and the hyper-phosphorylated STAT3 entrapped and prevented the transcription factors GATA1 and ZBP-89 (also known as ZNF148) from regulating erythroid gene expression. These findings imply that PTPN9 plays an important role in erythropoiesis by disrupting an inhibitory complex of phosphorylated STAT3, GATA1 and ZBP-89, providing new cellular and molecular insights into the role of ptpn9a in developmental hematopoiesis.
Collapse
Affiliation(s)
- Ye Bu
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Institute of Molecular Medicine, Peking University, Beijing, 100871 China
| | - Fuqin Su
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, School of Life Sciences, Tsinghua University, Beijing, 100084 China
| | - Xu Wang
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Institute of Molecular Medicine, Peking University, Beijing, 100871 China
| | - Hai Gao
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Institute of Molecular Medicine, Peking University, Beijing, 100871 China Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Beijing, 100094 China
| | - Lei Lei
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Institute of Molecular Medicine, Peking University, Beijing, 100871 China
| | - Nannan Chang
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Institute of Molecular Medicine, Peking University, Beijing, 100871 China
| | - Qing Wu
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Institute of Molecular Medicine, Peking University, Beijing, 100871 China
| | - Keping Hu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Beijing, 100094 China
| | - Xiaojun Zhu
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Institute of Molecular Medicine, Peking University, Beijing, 100871 China
| | - Zhijie Chang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, School of Medicine, School of Life Sciences, Tsinghua University, Beijing, 100084 China
| | - Kun Meng
- Beijing Shenogen Biomedical Company Ltd, Beijing, 100085 China
| | - Jing-Wei Xiong
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine and State Key Laboratory of Natural and Biomimetic Drugs, Institute of Molecular Medicine, Peking University, Beijing, 100871 China
| |
Collapse
|
14
|
Regulation of Gγ-globin gene by ATF2 and its associated proteins through the cAMP-response element. PLoS One 2013; 8:e78253. [PMID: 24223142 PMCID: PMC3819381 DOI: 10.1371/journal.pone.0078253] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 09/10/2013] [Indexed: 01/12/2023] Open
Abstract
The upstream Gγ-globin cAMP-response element (G-CRE) plays an important role in regulating Gγ-globin expression through binding of ATF2 and its DNA-binding partners defined in this study. ATF2 knockdown resulted in a significant reduction of γ-globin expression accompanied by decreased ATF2 binding to the G-CRE. By contrast, stable ATF2 expression in K562 cells increased γ-globin transcription which was reduced by ATF2 knockdown. Moreover, a similar effect of ATF2 on γ-globin expression was observed in primary erythroid progenitors. To understand the role of ATF2 in γ-globin expression, chromatographically purified G-CRE/ATF2-interacting proteins were subjected to mass spectrometry analysis; major binding partners included CREB1, cJun, Brg1, and histone deacetylases among others. Immunoprecipitation assays demonstrated interaction of these proteins with ATF2 and in vivo GCRE binding in CD34+ cells undergoing erythroid differentiation which was correlated with γ-globin expression during development. These results suggest synergism between developmental stage-specific recruitments of the ATF2 protein complex and expression of γ-globin during erythropoiesis. Microarray studies in K562 cells support ATF2 plays diverse roles in hematopoiesis and chromatin remodeling.
Collapse
|
15
|
Katsumura KR, DeVilbiss AW, Pope NJ, Johnson KD, Bresnick EH. Transcriptional mechanisms underlying hemoglobin synthesis. Cold Spring Harb Perspect Med 2013; 3:a015412. [PMID: 23838521 PMCID: PMC3753722 DOI: 10.1101/cshperspect.a015412] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The physiological switch in expression of the embryonic, fetal, and adult β-like globin genes has garnered enormous attention from investigators interested in transcriptional mechanisms and the molecular basis of hemoglobinopathies. These efforts have led to the discovery of cell type-specific transcription factors, unprecedented mechanisms of transcriptional coregulator function, genome biology principles, unique contributions of nuclear organization to transcription and cell function, and promising therapeutic targets. Given the vast literature accrued on this topic, this article will focus on the master regulator of erythroid cell development and function GATA-1, its associated proteins, and its frontline role in controlling hemoglobin synthesis. GATA-1 is a crucial regulator of genes encoding hemoglobin subunits and heme biosynthetic enzymes. GATA-1-dependent mechanisms constitute an essential regulatory core that nucleates additional mechanisms to achieve the physiological control of hemoglobin synthesis.
Collapse
Affiliation(s)
- Koichi R Katsumura
- Department of Cell and Regenerative Biology, UW-Madison Blood Research Program, Wisconsin Institute for Medical Research, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin 53705
| | | | | | | | | |
Collapse
|
16
|
Bianchi N, Zuccato C, Finotti A, Lampronti I, Borgatti M, Gambari R. Involvement of miRNA in erythroid differentiation. Epigenomics 2012; 4:51-65. [PMID: 22332658 DOI: 10.2217/epi.11.104] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
miRNAs are a family of small ncRNAs that regulate gene expression by targeting mRNAs in a sequence-specific manner, inducing translational repression or mRNA degradation. In this review, we present and discuss the available literature on the expression of miRNAs in erythroid cells. There are several experimental systems that can be employed for studies focusing on the relationship between miRNAs and erythroid differentiation, including human embryonic stem cells forced to erythroid differentiation, K562 and UT-7 cells induced to hemoglobin production by chemical compounds, erythropoietin-treated erythroid precursor cells from normal subjects or patients affected by hematological disease and in vivo systems, such as zebrafish embryos. Several miRNAs were identified as deeply involved in the erythroid phenotype, including miR-15a, miR-16-1, miR-126, miR-144, miR-451 and miR-210. Several functions related with erythroid cells were demonstrated to be regulated by these miRNAs, including maturation and proliferation of early erythroid cells, expression of fetal γ-globin genes and enucleation. These identified erythroid specific miRNAs represent the starting point to develop new protocols for miRNA therapeutics, based on both anti-miR molecules or miRNA replacement.
Collapse
Affiliation(s)
- Nicoletta Bianchi
- BioPharmaNet, Department of Biochemistry & Molecular Biology, University of Ferrara, Ferrara, Italy
| | | | | | | | | | | |
Collapse
|
17
|
Li B, Ding L, Li W, Story MD, Pace BS. Characterization of the transcriptome profiles related to globin gene switching during in vitro erythroid maturation. BMC Genomics 2012; 13:153. [PMID: 22537182 PMCID: PMC3353202 DOI: 10.1186/1471-2164-13-153] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 04/26/2012] [Indexed: 12/14/2022] Open
Abstract
Background The fetal and adult globin genes in the human β-globin cluster on chromosome 11 are sequentially expressed to achieve normal hemoglobin switching during human development. The pharmacological induction of fetal γ-globin (HBG) to replace abnormal adult sickle βS-globin is a successful strategy to treat sickle cell disease; however the molecular mechanism of γ-gene silencing after birth is not fully understood. Therefore, we performed global gene expression profiling using primary erythroid progenitors grown from human peripheral blood mononuclear cells to characterize gene expression patterns during the γ-globin to β-globin (γ/β) switch observed throughout in vitro erythroid differentiation. Results We confirmed erythroid maturation in our culture system using cell morphologic features defined by Giemsa staining and the γ/β-globin switch by reverse transcription-quantitative PCR (RT-qPCR) analysis. We observed maximal γ-globin expression at day 7 with a switch to a predominance of β-globin expression by day 28 and the γ/β-globin switch occurred around day 21. Expression patterns for transcription factors including GATA1, GATA2, KLF1 and NFE2 confirmed our system produced the expected pattern of expression based on the known function of these factors in globin gene regulation. Subsequent gene expression profiling was performed with RNA isolated from progenitors harvested at day 7, 14, 21, and 28 in culture. Three major gene profiles were generated by Principal Component Analysis (PCA). For profile-1 genes, where expression decreased from day 7 to day 28, we identified 2,102 genes down-regulated > 1.5-fold. Ingenuity pathway analysis (IPA) for profile-1 genes demonstrated involvement of the Cdc42, phospholipase C, NF-Kβ, Interleukin-4, and p38 mitogen activated protein kinase (MAPK) signaling pathways. Transcription factors known to be involved in γ-and β-globin regulation were identified. The same approach was used to generate profile-2 genes where expression was up-regulated over 28 days in culture. IPA for the 2,437 genes with > 1.5-fold induction identified the mitotic roles of polo-like kinase, aryl hydrocarbon receptor, cell cycle control, and ATM (Ataxia Telangiectasia Mutated Protein) signaling pathways; transcription factors identified included KLF1, GATA1 and NFE2 among others. Finally, profile-3 was generated from 1,579 genes with maximal expression at day 21, around the time of the γ/β-globin switch. IPA identified associations with cell cycle control, ATM, and aryl hydrocarbon receptor signaling pathways. Conclusions The transcriptome analysis completed with erythroid progenitors grown in vitro identified groups of genes with distinct expression profiles, which function in metabolic pathways associated with cell survival, hematopoiesis, blood cells activation, and inflammatory responses. This study represents the first report of a transcriptome analysis in human primary erythroid progenitors to identify transcription factors involved in hemoglobin switching. Our results also demonstrate that the in vitro liquid culture system is an excellent model to define mechanisms of global gene expression and the DNA-binding protein and signaling pathways involved in globin gene regulation.
Collapse
Affiliation(s)
- Biaoru Li
- Department Pediatrics, Georgia Health Sciences University, 1120 15th St, CN-4112, Augusta, GA 30912, USA
| | | | | | | | | |
Collapse
|
18
|
Kalra IS, Alam MM, Choudhary PK, Pace BS. Krüppel-like Factor 4 activates HBG gene expression in primary erythroid cells. Br J Haematol 2011; 154:248-59. [PMID: 21539536 DOI: 10.1111/j.1365-2141.2011.08710.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The SP1/Krüppel-like Factor (SP1/KLF) family of transcription factors plays a role in diverse cellular processes, including proliferation, differentiation and control of gene transcription. The discovery of KLF1 (EKLF), a key regulator of HBB (β-globin) gene expression, expanded our understanding of the role of KLFs in erythropoiesis. In this study, we investigated a mechanism of HBG (γ-globin) regulation by KLF4. siRNA-mediated gene silencing and enforced expression of KLF4 in K562 cells substantiated the ability of KLF4 to positively regulate endogenous HBG gene transcription. The physiological significance of this finding was confirmed in primary erythroid cells, where KLF4 knockdown at day 11 significantly attenuated HBG mRNA levels and enforced expression at day 28 stimulated the silenced HBG genes. In vitro binding characterization using the γ-CACCC and β-CACCC probes demonstrated KLF4 preferentially binds the endogenous γ-CACCC, while CREB binding protein (CREBBP) binding was not selective. Co-immunoprecipitation studies confirmed protein-protein interaction between KLF4 and CREBBP. Furthermore, sequential chromatin immunoprecipitation assays showed co-localization of both factors in the γ-CACCC region. Subsequent luciferase reporter studies demonstrated that KLF4 trans-activated HBG promoter activity and that CREBBP enforced expression resulted in gene repression. Our data supports a model of antagonistic interaction of KLF4/CREBBP trans-factors in HBG regulation.
Collapse
Affiliation(s)
- Inderdeep S Kalra
- Department of Molecular and Cell Biology, University of Texas at Dallas, Richardson, TX, USA
| | | | | | | |
Collapse
|
19
|
Ramakrishnan V, Pace BS. Regulation of γ-globin gene expression involves signaling through the p38 MAPK/CREB1 pathway. Blood Cells Mol Dis 2011; 47:12-22. [PMID: 21497119 DOI: 10.1016/j.bcmd.2011.03.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Accepted: 03/11/2011] [Indexed: 10/18/2022]
Abstract
In response to sodium butyrate and trichostatin A treatment in erythroid cells, p38 mitogen activated protein kinase (MAPK) mediates fetal hemoglobin (HbF) induction by activating cAMP response element binding protein 1 (CREB1). To expand on this observation, we completed studies to determine the role of p38 MAPK in steady-state γ-globin regulation. We propose that p38 signaling regulates Gγ-globin transcription during erythroid maturation through its downstream effector CREB1 which binds the Gγ-globin cAMP response element (G-CRE). We demonstrated that a loss of p38 or CREB1 function by siRNA knockdown resulted in target gene silencing. Moreover, gain of p38 or CREB1 function augments γ-globin transcription. These regulatory effects were conserved under physiological conditions tested in primary erythroid cells. When the G-CRE was mutated in a stable chromatin environment Gγ-globin promoter activity was nearly abolished. Furthermore, introduction of mutations in the G-CRE abolished Gγ-globin activation via p38 MAPK/CREB1 signaling. Chromatin immunoprecipitation assays (ChIP) demonstrated that CREB1 and its binding partner CREB binding protein (CBP) co-localize at the G-CRE region. These data support the role of p38 MAPK/CREB1 signaling in Gγ-globin gene transcription under steady-state conditions.
Collapse
Affiliation(s)
- Valya Ramakrishnan
- Department of Molecular and Cell Biology, University of Texas at Dallas, Richardson, TX 75080, USA
| | | |
Collapse
|
20
|
Bottardi S, Zmiri FA, Bourgoin V, Ross J, Mavoungou L, Milot E. Ikaros interacts with P-TEFb and cooperates with GATA-1 to enhance transcription elongation. Nucleic Acids Res 2011; 39:3505-19. [PMID: 21245044 PMCID: PMC3089448 DOI: 10.1093/nar/gkq1271] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Ikaros is associated with both gene transcriptional activation and repression in lymphocytes. Ikaros acts also as repressor of human γ-globin (huγ-) gene transcription in fetal and adult erythroid cells. Whether and eventually, how Ikaros can function as a transcriptional activator in erythroid cells remains poorly understood. Results presented herein demonstrate that Ikaros is a developmental-specific activator of huγ-gene expression in yolk sac erythroid cells. Molecular analysis in primary cells revealed that Ikaros interacts with Gata-1 and favors Brg1 recruitment to the human β-globin Locus Control Region and the huγ-promoters, supporting long-range chromatin interactions between these regions. Additionally, we demonstrate that Ikaros contributes to transcription initiation and elongation of the huγ-genes, since it is not only required for TBP and RNA Polymerase II (Pol II) assembly at the huγ-promoters but also for conversion of Pol II into the elongation-competent phosphorylated form. In agreement with the latter, we show that Ikaros interacts with Cyclin-dependent kinase 9 (Cdk9), which contributes to efficient transcription elongation by phosphorylating the C-terminal domain of the large subunit of Pol II on Serine 2, and favours Cdk9 recruitment to huγ-promoters. Our results show that Ikaros exerts dual functionality during gene activation, by promoting efficient transcription initiation and elongation.
Collapse
Affiliation(s)
- Stefania Bottardi
- Maisonneuve-Rosemont Hospital Research Center, Maisonneuve-Rosemont Hospital and Faculty of Medicine, University of Montreal, 5415 boulevard l'Assomption, Montreal, Quebec, Canada H1T 2M4
| | | | | | | | | | | |
Collapse
|
21
|
Recombinant erythroid Kruppel-like factor fused to GATA1 up-regulates delta- and gamma-globin expression in erythroid cells. Blood 2011; 117:3045-52. [PMID: 21220744 DOI: 10.1182/blood-2010-07-294751] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The β-hemoglobinopathies sickle cell disease and β-thalassemia are among the most common human genetic disorders worldwide. Hemoglobin A2 (HbA2, α₂δ₂) and fetal hemoglobin (HbF, α₂γ₂) both inhibit the polymerization of hemoglobin S, which results in erythrocyte sickling. Expression of erythroid Kruppel-like factor (EKLF) and GATA1 is critical for transitioning hemoglobin from HbF to hemoglobin A (HbA, α₂β₂) and HbA2. The lower levels of δ-globin expression compared with β-globin expression seen in adulthood are likely due to the absence of an EKLF-binding motif in the δ-globin proximal promoter. In an effort to up-regulate δ-globin to increase HbA2 expression, we created a series of EKLF-GATA1 fusion constructs composed of the transactivation domain of EKLF and the DNA-binding domain of GATA1, and then tested their effects on hemoglobin expression. EKLF-GATA1 fusion proteins activated δ-, γ-, and β-globin promoters in K562 cells, and significantly up-regulated δ- and γ-globin RNA transcript and protein expression in K562 and/or CD34(+) cells. The binding of EKLF-GATA1 fusion proteins at the GATA1 consensus site in the δ-globin promoter was confirmed by chromatin immunoprecipitation assay. Our studies demonstrate that EKLF-GATA1 fusion proteins can enhance δ-globin expression through interaction with the δ-globin promoter, and may represent a new genetic therapeutic approach to β-hemoglobinopathies.
Collapse
|
22
|
Muralidhar SA, Ramakrishnan V, Kalra IS, Li W, Pace BS. Histone deacetylase 9 activates gamma-globin gene expression in primary erythroid cells. J Biol Chem 2010; 286:2343-53. [PMID: 21078662 DOI: 10.1074/jbc.m110.115725] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Strategies to induce fetal hemoglobin (HbF) synthesis for the treatment of β-hemoglobinopathies probably involve protein modifications by histone deacetylases (HDACs) that mediate γ-globin gene regulation. However, the role of individual HDACs in globin gene expression is not very well understood; thus, the focus of our study was to identify HDACs involved in γ-globin activation. K562 erythroleukemia cells treated with the HbF inducers hemin, trichostatin A, and sodium butyrate had significantly reduced mRNA levels of HDAC9 and its splice variant histone deacetylase-related protein. Subsequently, HDAC9 gene knockdown produced dose-dependent γ-globin gene silencing over an 80-320 nm range. Enforced expression with the pTarget-HDAC9 vector produced a dose-dependent 2.5-fold increase in γ-globin mRNA (p < 0.05). Furthermore, ChIP assays showed HDAC9 binding in vivo in the upstream Gγ-globin gene promoter region. To determine the physiological relevance of these findings, human primary erythroid progenitors were treated with HDAC9 siRNA; we observed 40 and 60% γ-globin gene silencing in day 11 (early) and day 28 (late) progenitors. Moreover, enforced HDAC9 expression increased γ-globin mRNA levels by 2.5-fold with a simultaneous 7-fold increase in HbF. Collectively, these data support a positive role for HDAC9 in γ-globin gene regulation.
Collapse
Affiliation(s)
- Shalini A Muralidhar
- Department of Molecular and Cell Biology, University of Texas at Dallas, Richardson, Texas 75080, USA
| | | | | | | | | |
Collapse
|
23
|
He D, Chen T, Yang M, Zhu X, Wang C, Cao X, Cai Z. Small Rab GTPase Rab7b promotes megakaryocytic differentiation by enhancing IL-6 production and STAT3-GATA-1 association. J Mol Med (Berl) 2010; 89:137-50. [PMID: 20953574 DOI: 10.1007/s00109-010-0689-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 09/17/2010] [Accepted: 09/27/2010] [Indexed: 11/28/2022]
Abstract
Induction of the differentiation of human leukemia cells is a useful strategy in treatment of human leukemia. However, the molecular mechanisms involved in leukemia cell differentiation have not been fully elucidated. Interleukin 6 (IL-6) is a pleiotropic cytokine acting on a variety of cell types, and plays important roles in hematopoiesis. GATA binding protein 1 (GATA-1) is an important transcription factor involved in either megakaryocytic or erythrocytic differentiation. Herein we report that Rab7b, a late endosome/lysosome-localized myeloid small GTPase, promotes phorbol-12-myristate-13-acetate (PMA)-induced megakaryocytic differentiation by increasing nuclear factor κB (NF-κB)-dependent IL-6 production and subsequently enhancing the association of activated signal transducer and activator of transcription 3 (STAT3) with GATA-1. By using PMA-induced megakaryocytic differentiation of leukemia cells as a model, we investigated the roles of Rab7b in megakaryocytic differentiation. We find that Rab7b can potentiate PMA-induced upregulation of megakaryocytic markers, production of IL-6, and activation of NF-κB. Inhibitor of NF-κB and neutralizing antibodies for IL-6 or the IL-6 signaling receptor gp130 can block the effects of Rab7b in megakaryocytic differentiation. In Rab7b-silenced cells, PMA-induced activation of NF-κB, IL-6 production, and megakaryocytic differentiation are impaired. Furthermore, we demonstrate that IL-6-induced activation of STAT3 and the subsequent association of STAT3 with GATA-1 may contribute to PMA-induced and Rab7b-mediated transcriptional upregulation of megakaryocytic differentiation markers. Therefore, our data suggest that Rab7b may play important roles in megakaryopoiesis by activating NF-κB and promoting IL-6 production. Our study also indicates that the IL-6-induced association of STAT3 with GATA-1 may regulate megakaryocytic differentiation.
Collapse
Affiliation(s)
- Donghua He
- Department of Hematology, First Affiliated Hospital, Zhejiang University School of Medicine, 38 Zheda Road, Hangzhou, 310027, China
| | | | | | | | | | | | | |
Collapse
|
24
|
The gene structure and promoter region of the vaccine target aminopeptidase H11 from the blood-sucking nematode parasite of ruminants, Haemonchus contortus. Funct Integr Genomics 2010; 10:589-601. [DOI: 10.1007/s10142-010-0172-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2010] [Revised: 03/29/2010] [Accepted: 04/01/2010] [Indexed: 12/17/2022]
|
25
|
Pro-inflammatory cytokine-mediated anemia: regarding molecular mechanisms of erythropoiesis. Mediators Inflamm 2010; 2009:405016. [PMID: 20204172 PMCID: PMC2830572 DOI: 10.1155/2009/405016] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2009] [Accepted: 12/17/2009] [Indexed: 12/26/2022] Open
Abstract
Anemia of cancer and chronic inflammatory diseases is a frequent complication affecting quality of life. For cancer patients it represents a particularly bad prognostic. Low level of erythropoietin is considered as one of the causes of anemia in these pathologies. The deficiency in erythropoietin production results from pro-inflammatory cytokines effect. However, few data is available concerning molecular mechanisms involved in cytokine-mediated anemia. Some recent publications have demonstrated the direct effect of pro-inflammatory cytokines on cell differentiation towards erythroid pathway, without erythropoietin defect. This suggested that pro-inflammatory cytokine-mediated signaling pathways affect erythropoietin activity. They could interfere with erythropoietin-mediated signaling pathways, inducing early apoptosis and perturbing the expression and regulation of specific transcription factors involved in the control of erythroid differentiation. In this review we summarize the effect of tumor necrosis factor (TNF)α, TNF-related apoptosis-inducing ligand (TRAIL), and interferon (IFN)-γ on erythropoiesis with a particular interest for molecular feature.
Collapse
|
26
|
Kodeboyina S, Balamurugan P, Liu L, Pace BS. cJun modulates Ggamma-globin gene expression via an upstream cAMP response element. Blood Cells Mol Dis 2009; 44:7-15. [PMID: 19861239 DOI: 10.1016/j.bcmd.2009.10.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Accepted: 09/28/2009] [Indexed: 02/08/2023]
Abstract
The upstream Ggamma-globin gene cAMP response element (G-CRE) was previously shown to play a role in drug-mediated fetal hemoglobin induction. This effect is achieved via p38 mitogen activated protein kinase (MAPK)-dependent CREB1 and ATF-2 phosphorylation and G-CRE transactivation. Since this motif is also a predicted consensus binding site for cJun we extended our analysis to determine the ability of cJun to transactivate gamma-globin through the G-CRE. Using chromatin immunoprecipitation assays we showed comparable in vivo cJun and CREB1 binding to the G-CRE region. Protein-protein interactions were confirmed between cJun/ATF-2 and CREB1/ATF-2 but not between CREB1 and cJun. However, we observed cJun and CREB1 binding to the G-CRE in vitro by electrophoretic mobility shift assay. Promoter pull-down assay followed by sequential western blot analysis confirmed co-localization of cJun, CREB1, and ATF-2 on the G-CRE. To show functional relevance, enforced expression studies with pLen-cJun and a Ggamma-promoter (-1500 to +36) luciferase reporter were completed; we observed a concentration-dependent increase in luciferase activity with pLen-cJun similar to that produced by CREB1 enforced expression. Moreover, the G/A mutation at -1225 in the G-CRE abolished cJun transactivation. Finally, enforced cJun expression in K562 cells and normal primary erythroid progenitors enhanced endogenous gamma-globin gene expression. We conclude that these data indicate that cJun activates the Ggamma-globin promoter via the G-CRE in a manner comparable with CREB1 and propose a model for gamma-globin activation based on DNA-protein interactions in the G-CRE.
Collapse
Affiliation(s)
- Sirisha Kodeboyina
- Department of Molecular and Cell Biology, University of Texas at Dallas, PO Box 830688, FO 3.1, Richardson, TX 75083, USA
| | | | | | | |
Collapse
|