1
|
Ok SM, Jo JH, Cho HJ, Jang SM. RepID depletion enhances TWS119-induced erythropoiesis through chromatin reprogramming and transcription factor recruitment. Genes Genomics 2025:10.1007/s13258-025-01627-w. [PMID: 40100582 DOI: 10.1007/s13258-025-01627-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/18/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Erythrocytes, derived from hematopoietic stem cells, are essential for oxygen transport, ensuring survival in all vertebrate animals. The process of erythropoiesis is associated with gene expression changes, but many key regulatory factors that govern erythroid differentiation remain to be fully understood. OBJECTIVE This study investigates the role of TWS119, a known GSK3β inhibitor, in inducing erythropoiesis in K562 erythroleukemia cells and explores the impact of Replication initiation determinant protein (RepID) depletion on the process. METHODS K562 cells were treated with TWS119 and erythropoiesis markers including various erythrocytic phenotypes were assessed. Chromatin-immunoprecipitation analysis was employed to examine the changes in chromatin structure and gene expression regulation. The impact of RepID depletion on TWS119-induced erythropoiesis was also evaluated by analyzing globin promoter euchromatinization and NRF2 binding. RESULTS TWS119 treatment led to erythrocytic phenotypes in K562 cells, such as red pellet formation, enucleation, and nucleus condensation, along with the upregulation of erythropoiesis markers. Furthermore, RepID depletion accelerated TWS119-mediated erythropoiesis. Chromatin-immunoprecipitation analysis revealed euchromatinization of the globin promoter and enhanced NRF2 binding in RepID-depleted cells, suggesting a mechanism of gene expression regulation during erythropoiesis. CONCLUSION This study demonstrates that TWS119 can induce erythropoiesis in K562 cells, and that RepID depletion enhances this process by modulating chromatin structure and facilitating transcription factor binding. These findings highlight a RepID-dependent mechanism in the regulation of gene expression during erythropoiesis.
Collapse
Affiliation(s)
- Seon-Mi Ok
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Jae-Hyun Jo
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Hyo Je Cho
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, 28644, Republic of Korea.
- Department of Biochemistry, Chungbuk National University, Cheongju, 28644, Republic of Korea.
| | - Sang-Min Jang
- Department of Biological Sciences and Biotechnology, Chungbuk National University, Cheongju, 28644, Republic of Korea.
- Department of Biochemistry, Chungbuk National University, Cheongju, 28644, Republic of Korea.
| |
Collapse
|
2
|
Zhang YW, Schönberger K, Cabezas‐Wallscheid N. Bidirectional interplay between metabolism and epigenetics in hematopoietic stem cells and leukemia. EMBO J 2023; 42:e112348. [PMID: 38010205 PMCID: PMC10711668 DOI: 10.15252/embj.2022112348] [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: 08/11/2022] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 11/29/2023] Open
Abstract
During the last decades, remarkable progress has been made in further understanding the complex molecular regulatory networks that maintain hematopoietic stem cell (HSC) function. Cellular and organismal metabolisms have been shown to directly instruct epigenetic alterations, and thereby dictate stem cell fate, in the bone marrow. Epigenetic regulatory enzymes are dependent on the availability of metabolites to facilitate DNA- and histone-modifying reactions. The metabolic and epigenetic features of HSCs and their downstream progenitors can be significantly altered by environmental perturbations, dietary habits, and hematological diseases. Therefore, understanding metabolic and epigenetic mechanisms that regulate healthy HSCs can contribute to the discovery of novel metabolic therapeutic targets that specifically eliminate leukemia stem cells while sparing healthy HSCs. Here, we provide an in-depth review of the metabolic and epigenetic interplay regulating hematopoietic stem cell fate. We discuss the influence of metabolic stress stimuli, as well as alterations occurring during leukemic development. Additionally, we highlight recent therapeutic advancements toward eradicating acute myeloid leukemia cells by intervening in metabolic and epigenetic pathways.
Collapse
Affiliation(s)
- Yu Wei Zhang
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | | | | |
Collapse
|
3
|
Tewari AB, Saini A, Sharma D. Extirpating the cancer stem cell hydra: Differentiation therapy and Hyperthermia therapy for targeting the cancer stem cell hierarchy. Clin Exp Med 2023; 23:3125-3145. [PMID: 37093450 DOI: 10.1007/s10238-023-01066-5] [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: 12/09/2022] [Accepted: 04/02/2023] [Indexed: 04/25/2023]
Abstract
Ever since the discovery of cancer stem cells (CSCs), they have progressively attracted more attention as a therapeutic target. Like the mythical hydra, this subpopulation of cells seems to contribute to cancer immortality, spawning more cells each time that some components of the cancer cell hierarchy are destroyed. Traditional modalities focusing on cancer treatment have emphasized apoptosis as a route to eliminate the tumor burden. A major problem is that cancer cells are often in varying degrees of dedifferentiation contributing to what is known as the CSCs hierarchy and cells which are known to be resistant to conventional therapy. Differentiation therapy is an experimental therapeutic modality aimed at the conversion of malignant phenotype to a more benign one. Hyperthermia therapy (HT) is a modality exploiting the changes induced in cells by the application of heat produced to aid in cancer therapy. While differentiation therapy has been successfully employed in the treatment of acute myeloid leukemia, it has not been hugely successful for other cancer types. Mounting evidence suggests that hyperthermia therapy may greatly augment the effects of differentiation therapy while simultaneously overcoming many of the hard-to-treat facets of recurrent tumors. This review summarizes the progress made so far in integrating hyperthermia therapy with existing modules of differentiation therapy. The focus is on studies related to the successful application of both hyperthermia and differentiation therapy when used alone or in conjunction for hard-to-treat cancer cell niche with emphasis on combined approaches to target the CSCs hierarchy.
Collapse
Affiliation(s)
- Amit B Tewari
- Institute of Nano Science and Technology (INST), Knowledge City, Sector 81, Mohali, Punjab, 140306, India
| | - Anamika Saini
- Institute of Nano Science and Technology (INST), Knowledge City, Sector 81, Mohali, Punjab, 140306, India
| | - Deepika Sharma
- Institute of Nano Science and Technology (INST), Knowledge City, Sector 81, Mohali, Punjab, 140306, India.
| |
Collapse
|
4
|
Miljkovic M, Seguin A, Jia X, Cox JE, Catrow JL, Bergonia H, Phillips JD, Stephens WZ, Ward DM. Loss of the mitochondrial protein Abcb10 results in altered arginine metabolism in MEL and K562 cells and nutrient stress signaling through ATF4. J Biol Chem 2023; 299:104877. [PMID: 37269954 PMCID: PMC10316008 DOI: 10.1016/j.jbc.2023.104877] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/11/2023] [Accepted: 05/25/2023] [Indexed: 06/05/2023] Open
Abstract
Abcb10 is a mitochondrial membrane protein involved in hemoglobinization of red cells. Abcb10 topology and ATPase domain localization suggest it exports a substrate, likely biliverdin, out of mitochondria that is necessary for hemoglobinization. In this study, we generated Abcb10 deletion cell lines in both mouse murine erythroleukemia and human erythroid precursor human myelogenous leukemia (K562) cells to better understand the consequences of Abcb10 loss. Loss of Abcb10 resulted in an inability to hemoglobinize upon differentiation in both K562 and mouse murine erythroleukemia cells with reduced heme and intermediate porphyrins and decreased levels of aminolevulinic acid synthase 2 activity. Metabolomic and transcriptional analyses revealed that Abcb10 loss gave rise to decreased cellular arginine levels, increased transcripts for cationic and neutral amino acid transporters with reduced levels of the citrulline to arginine converting enzymes argininosuccinate synthetase and argininosuccinate lyase. The reduced arginine levels in Abcb10-null cells gave rise to decreased proliferative capacity. Arginine supplementation improved both Abcb10-null proliferation and hemoglobinization upon differentiation. Abcb10-null cells showed increased phosphorylation of eukaryotic translation initiation factor 2 subunit alpha, increased expression of nutrient sensing transcription factor ATF4 and downstream targets DNA damage inducible transcript 3 (Chop), ChaC glutathione specific gamma-glutamylcyclotransferase 1 (Chac1), and arginyl-tRNA synthetase 1 (Rars). These results suggest that when the Abcb10 substrate is trapped in the mitochondria, the nutrient sensing machinery is turned on remodeling transcription to block protein synthesis necessary for proliferation and hemoglobin biosynthesis in erythroid models.
Collapse
Affiliation(s)
- Marisa Miljkovic
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Alexandra Seguin
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Xuan Jia
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - James E Cox
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah, USA; Metabolomics Core Research Facility, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Jonathan Leon Catrow
- Metabolomics Core Research Facility, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Hector Bergonia
- Iron and Heme Core Research Facility, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - John D Phillips
- Division of Hematology, Department of Medicine, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - W Zac Stephens
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Diane M Ward
- Division of Microbiology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, USA.
| |
Collapse
|
5
|
Lomovskaya YV, Kobyakova MI, Senotov AS, Fadeeva IS, Lomovsky AI, Krasnov KS, Shtatnova DY, Akatov VS, Fadeev RS. Myeloid Differentiation Increases Resistance of Leukemic Cells to TRAIL-Induced Death by Reducing the Expression of DR4 and DR5 Receptors. BIOCHEMISTRY (MOSCOW), SUPPLEMENT SERIES A: MEMBRANE AND CELL BIOLOGY 2023. [DOI: 10.1134/s1990747822060101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
|
6
|
Abbasalipour M, Khosravi MA, Zeinali S, Khanahmad H, Azadmanesh K, Karimipoor M. Lentiviral vector containing beta-globin gene for beta thalassemia gene therapy. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
7
|
Kaur J, Rawat Y, Sood V, Periwal N, Rathore DK, Kumar S, Kumar N, Bhattacharyya S. Replication of Dengue Virus in K562-Megakaryocytes Induces Suppression in the Accumulation of Reactive Oxygen Species. Front Microbiol 2022; 12:784070. [PMID: 35087488 PMCID: PMC8787197 DOI: 10.3389/fmicb.2021.784070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/07/2021] [Indexed: 11/13/2022] Open
Abstract
Dengue virus can infect human megakaryocytes leading to decreased platelet biogenesis. In this article, we report a study of Dengue replication in human K562 cells undergoing PMA-induced differentiation into megakaryocytes. PMA-induced differentiation in these cells recapitulates steps of megakaryopoiesis including gene activation, expression of CD41/61 and CD61 platelet surface markers and accumulation of intracellular reactive oxygen species (ROS). Our results show differentiating megakaryocyte cells to support higher viral replication without any apparent increase in virus entry. Further, Dengue replication suppresses the accumulation of ROS in differentiating cells, probably by only augmenting the activity of the transcription factor NFE2L2 without influencing the expression of the coding gene. Interestingly pharmacological modulation of NFE2L2 activity showed a simultaneous but opposite effect on intracellular ROS and virus replication suggesting the former to have an inhibitory effect on the later. Also cells that differentiated while supporting intracellular virus replication showed reduced level of surface markers compared to uninfected differentiated cells.
Collapse
Affiliation(s)
- Jaskaran Kaur
- Translational Health Science and Technology Institute, National Capital Region (NCR) Biotech Science Cluster, Faridabad, India
| | - Yogita Rawat
- Translational Health Science and Technology Institute, National Capital Region (NCR) Biotech Science Cluster, Faridabad, India
| | - Vikas Sood
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Neha Periwal
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard (Hamdard University), New Delhi, India
| | - Deepak Kumar Rathore
- Translational Health Science and Technology Institute, National Capital Region (NCR) Biotech Science Cluster, Faridabad, India
| | - Shrikant Kumar
- Translational Health Science and Technology Institute, National Capital Region (NCR) Biotech Science Cluster, Faridabad, India
| | - Niraj Kumar
- Translational Health Science and Technology Institute, National Capital Region (NCR) Biotech Science Cluster, Faridabad, India
| | - Sankar Bhattacharyya
- Translational Health Science and Technology Institute, National Capital Region (NCR) Biotech Science Cluster, Faridabad, India
| |
Collapse
|
8
|
A rapid spectrophotometric method to identify inhibitors of human erythropoiesis. J Pharmacol Toxicol Methods 2021; 113:107134. [PMID: 34798285 DOI: 10.1016/j.vascn.2021.107134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/05/2021] [Accepted: 11/12/2021] [Indexed: 11/22/2022]
Abstract
Erythropoiesis is a complex physiological process by which erythroid progenitors proliferate and differentiate into nonnucleated red blood cells. Several methods can be used to monitor in vitro the differentiation of erythroid precursors, and hence the toxic effects of drugs, chemicals, or pollutants. One of the most commonly available assay of erythropoiesis is the microscopic observation of differentiated cells after benzidine staining, which forms a blue complex with hemoglobin. However, this method is laborious and does not provide accurate results since it heavily relies on the reader's interpretation. Moreover, benzidine is a carcinogen and a highly reactive molecule which forces the reader to microscopically count differentiated and non-differentiated cells within a short time frame (5 min). Here we have developed a simple, inexpensive, in-vitro spectrophotometric assay to measure erythroid differentiation using K562 cell line as a model. Materials needed included 96-well round-bottomed microplates and a microplate reader. Remarkably, carcinogenic benzidine was replaced by its isomeric tetramethyl derivative, the 3,3', 5,5'- tetramethylbenzidine (TMB), which presents several advantages: it is cheap, not mutagenic and a ready-to-use chromogenic substrate. A small volume (50 μl) of TMB added to the samples forms a blue complex in 15 min, and the reaction can be easily stopped and stabilized by the addition of H2SO4. The yellow precipitate is then solubilized, and the absorbance is measured at 450 nm. In addition, the suitability of the assay to determine the effects of compounds on erythroid differentiation was further tested with known inhibitors (artemisinin derivatives) of K562 differentiation. Overall, the reported methodology permits to measure in an accurate and reproducible manner the K562 differentiation and can be used for medium throughput screenings (MTS) of compounds or environmental toxics with potential erythro-toxicity and ability to inhibit erythroid differentiation.
Collapse
|
9
|
Lu Q, Guo P, Wang X, Ares I, Lopez-Torres B, Martínez-Larrañaga MR, Li T, Zhang Y, Wang X, Anadón A, Martínez MA. MS4A3-HSP27 target pathway reveals potential for haematopoietic disorder treatment in alimentary toxic aleukia. Cell Biol Toxicol 2021; 39:201-216. [PMID: 34581912 DOI: 10.1007/s10565-021-09639-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 07/22/2021] [Indexed: 12/17/2022]
Abstract
Alimentary toxic aleukia (ATA) is correlated with consuming grains contaminated by Fusarium species, particularly T-2 toxin, which causes serious hurt to human and animal health, chiefly in disorders of the haematopoietic system. However, the mechanism of haematopoietic dysfunction induced by T-2 toxin and the possible target pathway for the treatment of T-2 toxin-induced haematopoietic disorder of ATA remains unclear. In this study, genomes and proteomics were used for the first time to investigate the key differential genes and proteins that inhibit erythroid differentiation of K562 cells caused by T-2 toxin, and it was found that heat shock protein 27 (HSP27) and membrane-spanning 4-domains, subfamily A, member 3 (MS4A3) may play an important role in erythroid differentiation. Meanwhile, MS4A3 interference can inhibit the occurrence of erythroid differentiation of K562 cells and promote the phosphorylation of HSP27. Moreover, the binding of HSP27 to MS4A3 in natural state can activate the phosphorylation site of HSP27 (Ser-83), while T-2 toxin can abolish the activation of phosphorylation site by inhibiting the expression of MS4A3. These findings for the first time demonstrated that the MS4A3-HSP27 pathway may function an efficient therapeutic target pathway for treating T-2 toxin elicited haematopoietic disorders of ATA.
Collapse
Affiliation(s)
- Qirong Lu
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Pu Guo
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Xiaohui Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Irma Ares
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Bernardo Lopez-Torres
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - María-Rosa Martínez-Larrañaga
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, 28040, Madrid, Spain
| | - Tingting Li
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Yuanyuan Zhang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, 430070, Hubei, China. .,MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Wuhan, 430070, Hubei, China.
| | - Arturo Anadón
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, 28040, Madrid, Spain.
| | - María-Aránzazu Martínez
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Universidad Complutense de Madrid, 28040, Madrid, Spain
| |
Collapse
|
10
|
Ali H, Khan F, Musharraf SG. Cilostazol-mediated reversion of γ-globin silencing is associated with a high level of HbF production: A potential therapeutic candidate for β-globin disorders. Biomed Pharmacother 2021; 142:112058. [PMID: 34426256 DOI: 10.1016/j.biopha.2021.112058] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/09/2021] [Accepted: 08/13/2021] [Indexed: 11/30/2022] Open
Abstract
Reversal of fetal hemoglobin (HbF) silencing is an attractive therapeutic intervention for β-thalassemia and sickle cell anemia. The current study proposes the therapeutic of repurposing of cilostazol, an FDA-approved antithrombotic agent, as a promising HbF inducer. Preliminary, we report that cilostazol induced erythroid differentiation and hemoglobinization of human erythroleukemia K562 cells. The erythroid differentiation was accompanied by increased expression of γ-globin mRNA transcripts and HbF production. Cilostazol induced erythroid differentiation and HbF production, without significantly affecting proliferation and viability of hemoglobin producing cells at maximum erythroid inducing concentration. Moreover, we investigated the effect of cilostazol on human β- and γ-globin transgenes in in vivo β-YAC transgenic mice, harboring human β-locus along with β-LCR. A good in vitro correlation was found with substantial up-regulation in fetal globin mRNA; whereas, the β-globin gene expression was not significantly changed. F-cells, analysis in the peripheral blood of cilostazol-treated mice, revealed a significant increase in the F-cells population as compared with sham control groups. Together, these findings support the potential of cilostazol as an HbF inducer, which can be evaluated further to develop a new HbF inducer.
Collapse
Affiliation(s)
- Hamad Ali
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Faisal Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Syed Ghulam Musharraf
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| |
Collapse
|
11
|
Su H, Jiang M, Senevirathne C, Aluri S, Zhang T, Guo H, Xavier-Ferrucio J, Jin S, Tran NT, Liu SM, Sun CW, Zhu Y, Zhao Q, Chen Y, Cable L, Shen Y, Liu J, Qu CK, Han X, Klug CA, Bhatia R, Chen Y, Nimer SD, Zheng YG, Iancu-Rubin C, Jin J, Deng H, Krause DS, Xiang J, Verma A, Luo M, Zhao X. Methylation of dual-specificity phosphatase 4 controls cell differentiation. Cell Rep 2021; 36:109421. [PMID: 34320342 PMCID: PMC9110119 DOI: 10.1016/j.celrep.2021.109421] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 02/17/2021] [Accepted: 06/28/2021] [Indexed: 12/11/2022] Open
Abstract
Mitogen-activated protein kinases (MAPKs) are inactivated by dual-specificity phosphatases (DUSPs), the activities of which are tightly regulated during cell differentiation. Using knockdown screening and single-cell transcriptional analysis, we demonstrate that DUSP4 is the phosphatase that specifically inactivates p38 kinase to promote megakaryocyte (Mk) differentiation. Mechanistically, PRMT1-mediated methylation of DUSP4 triggers its ubiquitinylation by an E3 ligase HUWE1. Interestingly, the mechanistic axis of the DUSP4 degradation and p38 activation is also associated with a transcriptional signature of immune activation in Mk cells. In the context of thrombocytopenia observed in myelodysplastic syndrome (MDS), we demonstrate that high levels of p38 MAPK and PRMT1 are associated with low platelet counts and adverse prognosis, while pharmacological inhibition of p38 MAPK or PRMT1 stimulates megakaryopoiesis. These findings provide mechanistic insights into the role of the PRMT1-DUSP4-p38 axis on Mk differentiation and present a strategy for treatment of thrombocytopenia associated with MDS.
Collapse
Affiliation(s)
- Hairui Su
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ming Jiang
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; Program of Pharmacology, Weill Cornell Medical College of Cornell University, New York, NY 10021, USA
| | - Chamara Senevirathne
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Srinivas Aluri
- Department of Oncology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Tuo Zhang
- Genomics and Epigenomics Core Facility, Weill Cornell Medical College of Cornell University, New York, NY 10021, USA
| | - Han Guo
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Juliana Xavier-Ferrucio
- Department of Laboratory Medicine, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Shuiling Jin
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ngoc-Tung Tran
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Szu-Mam Liu
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Chiao-Wang Sun
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Yongxia Zhu
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA
| | - Qing Zhao
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Yuling Chen
- Department of School of Life Sciences, Tsinghua University, Beijing 100084, China
| | | | - Yudao Shen
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jing Liu
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Cheng-Kui Qu
- Aflac Cancer and Blood Disorders Center, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| | - Xiaosi Han
- Department of Neurology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Christopher A Klug
- Department of Microbiology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ravi Bhatia
- Division of Hematology and Oncology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Yabing Chen
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA; Veterans Affairs Birmingham Medical Center, Research Department, Birmingham, AL 35294, USA
| | - Stephen D Nimer
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33146 USA
| | - Y George Zheng
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, USA
| | - Camelia Iancu-Rubin
- Department of Medicine, Hematology and Oncology Division, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Haiteng Deng
- Department of School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Diane S Krause
- Department of Laboratory Medicine, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Jenny Xiang
- Genomics and Epigenomics Core Facility, Weill Cornell Medical College of Cornell University, New York, NY 10021, USA
| | - Amit Verma
- Department of Oncology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA.
| | - Minkui Luo
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10021, USA; Program of Pharmacology, Weill Cornell Medical College of Cornell University, New York, NY 10021, USA.
| | - Xinyang Zhao
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
12
|
Fu YH, Ou DL, Yang YR, Su KW, Chen CY, Tien HF, Lai ZS, Shen CKJ, Chien HF, Lin LI. Cabozantinib promotes erythroid differentiation in K562 erythroleukemia cells through global changes in gene expression and JNK activation. Cancer Gene Ther 2021; 29:784-792. [PMID: 34117374 DOI: 10.1038/s41417-021-00358-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 05/12/2021] [Accepted: 05/25/2021] [Indexed: 11/09/2022]
Abstract
Cabozantinib is a potent tyrosine kinase inhibitor with multiple targets including MET, VEGFR2, RET, KIT, and FLT3. Cabozantinib is widely used for the treatment of medullary thyroid cancer and renal cell carcinoma. We recently suggested cabozantinib as a potential therapeutic alternative for acute myeloid leukemia (AML) patients with FLT3-internal tandem duplication (FLT3-ITD). Here, we report that cabozantinib can promote differentiation in erythroid leukemia cells. We found that K562 erythroid leukemia cells treated with 1 μM cabozantinib for 72 h underwent erythroid lineage differentiation. Transcriptomic analysis revealed that various pathways associated with heme biosynthesis, hemoglobin production, and GATA1 targets were upregulated, whereas cell survival pathways were downregulated. Further examination revealed that cabozantinib-induced erythroid differentiation is at least in part regulated by JNK activation and phosphorylation. Levels of phosphorylated BCR-ABL, AKT, STAT5, ERK, and p38 also decreased following cabozantinib treatment. Therefore, we indicate that cabozantinib has dual functions. First, it induces K562 cell differentiation toward the erythroid lineage by upregulating heme biosynthesis, globin synthesis, and erythroid-associated reactions. Second, cabozantinib inhibits K562 cell proliferation by inhibiting the phosphorylation of BCR-ABL and the downstream MAPK, PI3K-AKT, and JAK-STAT signaling pathways.
Collapse
Affiliation(s)
- Yu-Hsuan Fu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Da-Liang Ou
- Department of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yi-Ru Yang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Kuan-Wei Su
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan
| | - Chien-Yuan Chen
- Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Hwei-Fan Tien
- Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Zheng-Sheng Lai
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Che-Kun James Shen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan.,The Ph.D. Program for Neural Regenerative Medicine, Taipei Medical University, Taipei, Taiwan
| | - Hsiung-Fei Chien
- Division of Plastic Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei, Taiwan. .,TMU Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Liang-In Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University, Taipei, Taiwan. .,Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
13
|
Papayannopoulou T. Control of fetal globin expression in man: new opportunities to challenge past discoveries. Exp Hematol 2020; 92:43-50. [PMID: 32976950 DOI: 10.1016/j.exphem.2020.09.195] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/18/2020] [Accepted: 09/19/2020] [Indexed: 01/01/2023]
Abstract
Decades-old findings supporting origin of F cells in adult life from adult-type progenitors and the in vitro and in vivo enhancement of fetal globin under stress conditions have been juxtaposed against recent mechanistic underpinnings. An updated molecular interrogation did not debunk prior conclusions on the origin of F cells. Although fetal globin reactivation by widely diverse approaches in vitro and in response to anemic stresses in vivo is a work in progress, accumulating evidence converges toward an integrated stress response pathway. The newly uncovered developmental regulators of globin gene switching not only have provided answers to the long-awaited quest of transregulation of switching, they are also reaching a clinical threshold. Although the effect of fetal globin silencers has been robustly validated in adult cells, the response of cells at earlier developmental stages has been unclear and inadequately studied.
Collapse
|
14
|
Sobh A, Loguinov A, Zhou J, Jenkitkasemwong S, Zeidan R, El Ahmadie N, Tagmount A, Knutson M, Fraenkel PG, Vulpe CD. Genetic screens reveal CCDC115 as a modulator of erythroid iron and heme trafficking. Am J Hematol 2020; 95:1085-1098. [PMID: 32510613 DOI: 10.1002/ajh.25899] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 05/27/2020] [Accepted: 05/31/2020] [Indexed: 12/26/2022]
Abstract
Transferrin-bound iron (TBI), the physiological circulating iron form, is acquired by cells through the transferrin receptor (TfR1) by endocytosis. In erythroid cells, most of the acquired iron is incorporated into heme in the mitochondria. Cellular trafficking of heme is indispensable for erythropoiesis and many other essential biological processes. Comprehensive elucidation of molecular pathways governing and regulating cellular iron acquisition and heme trafficking is required to better understand physiological and pathological processes affecting erythropoiesis. Here, we report the first genome-wide clustered regularly interspaced short palindromic repeats (CRISPR) screens in human erythroid cells to identify determinants of iron and heme uptake, as well as heme-mediated erythroid differentiation. We identified several candidate modulators of TBI acquisition including TfR1, indicating that our approach effectively revealed players mechanistically relevant to the process. Interestingly, components of the endocytic pathway were also revealed as potential determinants of transferrin acquisition. We deciphered a role for the vacuolar-type H+ - ATPase (V- ATPase) assembly factor coiled-coil domain containing 115 (CCDC115) in TBI uptake and validated this role in CCDC115 deficient K562 cells. Our screen in hemin-treated cells revealed perturbations leading to cellular adaptation to heme, including those corresponding to trafficking mechanisms and transcription factors potentiating erythroid differentiation. Pathway analysis indicated that endocytosis and vesicle acidification are key processes for heme trafficking in erythroid precursors. Furthermore, we provided evidence that CCDC115, which we identified as required for TBI uptake, is also involved in cellular heme distribution. This work demonstrates a previously unappreciated common intersection in trafficking of transferrin iron and heme in the endocytic pathway of erythroid cells.
Collapse
Affiliation(s)
- Amin Sobh
- Department of Nutritional Sciences & Toxicology, Comparative Biochemistry Program University of California Berkeley Berkeley California
- Department of Physiological Sceinces University of Florida Gainesville Florida
| | - Alex Loguinov
- Department of Physiological Sceinces University of Florida Gainesville Florida
| | - Jie Zhou
- Department of Physiological Sceinces University of Florida Gainesville Florida
- Department of Food Science and Human Nutrition University of Florida Gainesville Florida
| | - Supak Jenkitkasemwong
- Department of Food Science and Human Nutrition University of Florida Gainesville Florida
| | - Rola Zeidan
- Department of Physiological Sceinces University of Florida Gainesville Florida
| | - Nader El Ahmadie
- Department of Physiological Sceinces University of Florida Gainesville Florida
| | | | - Mitchell Knutson
- Department of Food Science and Human Nutrition University of Florida Gainesville Florida
| | - Paula G. Fraenkel
- Division of Hematology/Oncology and Cancer Research Institute Beth Israel Deaconess Medical Center Boston Massachusetts
- Department of Medicine Harvard Medical School Boston Massachusetts
- Oncology Research and Development, Sanofi Cambridge Massachusetts
| | | |
Collapse
|
15
|
Jaiswal AK, Makhija S, Stahr N, Sandey M, Suryawanshi A, Saxena A, Dagur PK, McCoy JP, Levine SJ, Mishra A. Dendritic Cell-Restricted Progenitors Contribute to Obesity-Associated Airway Inflammation via Adam17-p38 MAPK-Dependent Pathway. Front Immunol 2020; 11:363. [PMID: 32184787 PMCID: PMC7058657 DOI: 10.3389/fimmu.2020.00363] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/14/2020] [Indexed: 01/08/2023] Open
Abstract
Proliferation of dendritic cell (DC)—restricted progenitor cells in bone marrow compartment is tightly regulated at steady state and responds to multiple tissue-specific triggers during disturbed homeostasis such as obesity. DCs in the lung stem from a rapidly dividing DC-restricted progenitor cells and are effective at generating adaptive immune responses in allergic airway inflammation. Precisely, how DC-restricted progenitor expansion and differentiation are influenced by airway inflammation to maintain constant supply of myeloid DCs is poorly understood. Here we show that a high fat diet (HFD) induces oxidative stress and accelerates the expansion of DC- restricted progenitor cells in bone marrow and correlates with persistent induction of p38 mitogen activated protein kinase (MAPK), which is blocked with a selective p38α/β MAPK inhibitor. Mice fed a HFD and sensitized to inhaled allergen house dust mite (HDM) led to alterations of DC- restricted progenitor cells that were characterized by increased expansion and seeding of lung DCs in airway inflammation. Mechanistically, we establish that the expansion induced by HFD dysregulates the expression of a disintegrin and metallopeptidase domain 17 (Adam17) and is required for p38 MAPK activation in DC-restricted progenitors. These results demonstrates that obesity produces persistent changes in DC precursors and that elevation of Adam17 expression is tightly coupled to p38 MAPK and is a key driver of proliferation. Altogether, these data provide phenotypic and mechanistic insight into dendritic cell supply chain in obesity-associated airway inflammation.
Collapse
Affiliation(s)
- Anil Kumar Jaiswal
- Laboratory of Lung Inflammation, College of Veterinary Medicine, Auburn University, Auburn, AL, United States.,Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Sangeet Makhija
- Laboratory of Lung Inflammation, College of Veterinary Medicine, Auburn University, Auburn, AL, United States.,Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Natalie Stahr
- Laboratory of Lung Inflammation, College of Veterinary Medicine, Auburn University, Auburn, AL, United States.,Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Maninder Sandey
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Amol Suryawanshi
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Ankit Saxena
- Flow Cytometry Core Facility, Division of Intramural Research, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Pradeep K Dagur
- Flow Cytometry Core Facility, Division of Intramural Research, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - J Philip McCoy
- Flow Cytometry Core Facility, Division of Intramural Research, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Stewart J Levine
- Laboratory of Asthma and Lung Inflammation, Division of Intramural Research, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Amarjit Mishra
- Laboratory of Lung Inflammation, College of Veterinary Medicine, Auburn University, Auburn, AL, United States.,Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| |
Collapse
|
16
|
Theodorou A, Phylactides M, Katsantoni E, Vougas K, Garbis SD, Fanis P, Sitarou M, Thein SL, Kleanthous M. Proteomic Studies for the Investigation of γ-Globin Induction by Decitabine in Human Primary Erythroid Progenitor Cultures. J Clin Med 2020; 9:jcm9010134. [PMID: 31947809 PMCID: PMC7019605 DOI: 10.3390/jcm9010134] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/17/2019] [Accepted: 12/26/2019] [Indexed: 11/16/2022] Open
Abstract
Reactivation of γ-globin is considered a promising approach for the treatment of β-thalassemia and sickle cell disease. Therapeutic induction of γ-globin expression, however, is fraught with lack of suitable therapeutic targets. The aim of this study was to investigate the effects that treatment with decitabine has on the proteome of human primary erythroid cells from healthy and thalassemic volunteers, as a means of identifying new potential pharmacological targets. Decitabine is a known γ-globin inducer, which is not, however, safe enough for clinical use. A proteomic approach utilizing isobaric tags for relative and absolute quantitation (iTRAQ) analysis, in combination with high-pH reverse phase peptide fractionation followed by liquid chromatography-tandem mass spectrometry (LC-MS/MS), was employed to investigate the effects of decitabine treatment. Bioinformatics analysis making use of the Database for Annotation, Visualization and Integrated Discovery (DAVID) was employed for functional annotation of the 192 differentially expressed proteins identified. The data are available via ProteomeXchange with identifier PXD006889. The proteins fall into various biological pathways, such as the NF-κB signaling pathway, and into many functional categories including regulation of cell proliferation, transcription factor and DNA binding, protein stabilization, chromatin modification and organization, and oxidative stress proteins.
Collapse
Affiliation(s)
- Andria Theodorou
- Molecular Genetics Thalassaemic Department, Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus
| | - Marios Phylactides
- Molecular Genetics Thalassaemic Department, Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus
- Correspondence: ; Tel.: +357-22-392657
| | - Eleni Katsantoni
- Basic Research Center, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
| | - Kostas Vougas
- Basic Research Center, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
| | - Spyros D. Garbis
- Basic Research Center, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
- Division for Cancer Sciences, Southampton General Hospital, University of Southampton, Southampton SO16 6YD, UK
- Centre for Proteomics Research, Institute for Life Sciences, Highfield Campus, University of Southampton, Southampton SO17 1BJ, UK
| | - Pavlos Fanis
- Molecular Genetics Thalassaemic Department, Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus
- Molecular Genetics Function and Therapy Department, Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus
| | - Maria Sitarou
- Thalassaemia Centre, Larnaca General Hospital, Larnaca 6043, Cyprus
| | - Swee Lay Thein
- Sickle cell branch, National Heart, Lung and Blood Institute, The National Institutes of Health, Bethesda, MD 20814, USA
| | - Marina Kleanthous
- Molecular Genetics Thalassaemic Department, Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus
| |
Collapse
|
17
|
Paul BT, Tesfay L, Winkler CR, Torti FM, Torti SV. Sideroflexin 4 affects Fe-S cluster biogenesis, iron metabolism, mitochondrial respiration and heme biosynthetic enzymes. Sci Rep 2019; 9:19634. [PMID: 31873120 PMCID: PMC6928202 DOI: 10.1038/s41598-019-55907-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 12/03/2019] [Indexed: 12/12/2022] Open
Abstract
Sideroflexin4 (SFXN4) is a member of a family of nuclear-encoded mitochondrial proteins. Rare germline mutations in SFXN4 lead to phenotypic characteristics of mitochondrial disease including impaired mitochondrial respiration and hematopoetic abnormalities. We sought to explore the function of this protein. We show that knockout of SFXN4 has profound effects on Fe-S cluster formation. This in turn diminishes mitochondrial respiratory chain complexes and mitochondrial respiration and causes a shift to glycolytic metabolism. SFXN4 knockdown reduces the stability and activity of cellular Fe-S proteins, affects iron metabolism by influencing the cytosolic aconitase-IRP1 switch, redistributes iron from the cytosol to mitochondria, and impacts heme synthesis by reducing levels of ferrochelatase and inhibiting translation of ALAS2. We conclude that SFXN4 is essential for normal functioning of mitochondria, is necessary for Fe-S cluster biogenesis and iron homeostasis, and plays a critical role in mitochondrial respiration and synthesis of heme.
Collapse
Affiliation(s)
- Bibbin T Paul
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Lia Tesfay
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - C R Winkler
- Institute for Critical Technology and Applied Science, Nanoscale Characterization and Fabrication Laboratory, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Frank M Torti
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Suzy V Torti
- Department of Molecular Biology and Biophysics, University of Connecticut Health Center, Farmington, CT, 06030, USA.
| |
Collapse
|
18
|
Oseghale AR, Zhu X, Li B, Peterson KR, Nudelman A, Rephaeli A, Xu H, Pace BS. Conjugate prodrug AN-233 induces fetal hemoglobin expression in sickle erythroid progenitors and β-YAC transgenic mice. Blood Cells Mol Dis 2019; 79:102345. [PMID: 31351219 DOI: 10.1016/j.bcmd.2019.102345] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 07/08/2019] [Indexed: 12/24/2022]
Abstract
Pharmacologic induction of fetal hemoglobin (HbF) is an effective strategy for treating sickle cell disease (SCD) by ameliorating disease severity. Hydroxyurea is the only FDA-approved agent that induces HbF, but significant non-responders and requirement for frequent monitoring of blood counts for drug toxicity limit clinical usefulness. Therefore, we investigated a novel prodrug conjugate of butyric acid (BA) and δ-aminolevulinate (ALA) as a potential HbF inducing agent, using erythroid precursors and a preclinical β-YAC mouse model. We observed significantly increased γ-globin gene transcription and HbF expression mediated by AN-233 in K562 cells. Moreover, AN-233 stimulated mild heme biosynthesis and inhibited expression of heme-regulated eIF2α kinase involved in silencing γ-globin expression. Studies using primary erythroid precursors generated from sickle peripheral blood mononuclear cells verified the ability of AN-233 to induce HbF, increase histone H3 and H4 acetylation levels at the γ-globin promoter and reduce erythroid precursor sickling by 50%. Subsequent drug treatment of β-YAC transgenic mice confirmed HbF induction in vivo by AN-233 through an increase in the percentage of HbF positive red blood cells and HbF levels measured by flow cytometry. These data support the potential development of AN-233 for the treatment of SCD.
Collapse
Affiliation(s)
| | - Xingguo Zhu
- Department of Pediatrics, Augusta University, Augusta, GA, USA
| | - Biaoru Li
- Department of Pediatrics, Augusta University, Augusta, GA, USA
| | - Kenneth R Peterson
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | | | - Ada Rephaeli
- Felsenstein Medical Research Center, Sackler Medical School, Tel Aviv University, Petach Tikva, Israel
| | - Hongyan Xu
- Department of Population Health Sciences, Augusta University, Augusta, GA, USA
| | - Betty S Pace
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Department of Pediatrics, Augusta University, Augusta, GA, USA; Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA, USA.
| |
Collapse
|
19
|
Ali H, Iftikhar F, Shafi S, Siddiqui H, Khan IA, Choudhary MI, Musharraf SG. Thiourea derivatives induce fetal hemoglobin production in-vitro: A new class of potential therapeutic agents for β-thalassemia. Eur J Pharmacol 2019; 855:285-293. [PMID: 31100414 DOI: 10.1016/j.ejphar.2019.05.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/10/2019] [Accepted: 05/13/2019] [Indexed: 12/23/2022]
Abstract
Fetal hemoglobin (HbF) induction is a cost-effective therapeutic approach for the treatment of β-hemoglobinopathies like β-thalassemia and sickle cell anemia. The present study discusses the potential of thiourea derivatives as new class of compounds that induce the fetal hemoglobin production. HbF inducing effect of thiourea derivatives was studied using experimental cell system, the human erythroleukemic K562 cell line. Erythroid induction of K562 cells was studied by the benzidine/H2O2 reaction, total hemoglobin production was estimated by plasma hemoglobin assay kit, and γ-globin gene expression by RT-qPCR, whereas fetal hemoglobin production was estimated by flow cytometry and immunofluorescence microscopy. The results indicated that newly synthesized thiourea derivative are potent inducers of erythroid differentiation of K562 cells with an increased γ-globin gene expression and fetal hemoglobin production. Moreover, these compounds showed no cytotoxic effect and inhibition on K562 cells at HbF inducing concentrations. It is important to note that hydroxyurea is a cytotoxic chemotherapeutic agent and have deleterious side effects, reflecting the need to identify new safe and effective HbF induces. These results signify thiourea derivatives as promising HbF inducers, with the potential to be studied against hematological disorders, including β-thalassemia and sickle cell anemia.
Collapse
Affiliation(s)
- Hamad Ali
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Fizza Iftikhar
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Sarah Shafi
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Hina Siddiqui
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - Ishtiaq Ahmad Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan
| | - M Iqbal Choudhary
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan; H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan; Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, 21452, Saudi Arabia
| | - Syed Ghulam Musharraf
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan; H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, 75270, Pakistan.
| |
Collapse
|
20
|
Kikuchi G, Kurita R, Ogasawara K, Isa K, Tsuneyama H, Nakamura Y, Yabe R, Shiba M, Tadokoro K, Nagai T, Satake M. Application of immortalized human erythroid progenitor cell line in serologic tests to detect red blood cell alloantibodies. Transfusion 2018; 58:2675-2682. [DOI: 10.1111/trf.14840] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 05/17/2018] [Accepted: 05/23/2018] [Indexed: 12/16/2022]
Affiliation(s)
- Go Kikuchi
- Department of Research and Development, Central Blood Institute, Blood Service Headquarters; Japanese Red Cross Society; Tokyo Japan
| | - Ryo Kurita
- Department of Research and Development, Central Blood Institute, Blood Service Headquarters; Japanese Red Cross Society; Tokyo Japan
| | - Kenichi Ogasawara
- Department of Research and Development, Central Blood Institute, Blood Service Headquarters; Japanese Red Cross Society; Tokyo Japan
| | - Kazumi Isa
- Department of Research and Development, Central Blood Institute, Blood Service Headquarters; Japanese Red Cross Society; Tokyo Japan
| | - Hatsue Tsuneyama
- Blood Testing Division, Kanto-Koshinetsu Block Blood Center; Japanese Red Cross Society; Tokyo Japan
| | - Yukio Nakamura
- Cell Engineering Division; RIKEN BioResource Center; Ibaraki Japan
| | - Ryuichi Yabe
- Blood Testing Division, Kanto-Koshinetsu Block Blood Center; Japanese Red Cross Society; Tokyo Japan
| | - Masayuki Shiba
- Department of Research and Development, Central Blood Institute, Blood Service Headquarters; Japanese Red Cross Society; Tokyo Japan
| | - Kenji Tadokoro
- Department of Research and Development, Central Blood Institute, Blood Service Headquarters; Japanese Red Cross Society; Tokyo Japan
| | - Tadashi Nagai
- Department of Research and Development, Central Blood Institute, Blood Service Headquarters; Japanese Red Cross Society; Tokyo Japan
| | - Masahiro Satake
- Department of Research and Development, Central Blood Institute, Blood Service Headquarters; Japanese Red Cross Society; Tokyo Japan
| |
Collapse
|
21
|
Zhong L, Zhou S, Tong R, Shi J, Bai L, Zhu Y, Duan X, Liu W, Bao J, Su L, Peng Q. Preclinical assessment of histone deacetylase inhibitor quisinostat as a therapeutic agent against esophageal squamous cell carcinoma. Invest New Drugs 2018; 37:616-624. [DOI: 10.1007/s10637-018-0651-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 07/24/2018] [Indexed: 12/19/2022]
|
22
|
Takahashi K, Umebayashi C, Numata T, Honda A, Ichikawa J, Hu Y, Yamaura K, Inoue R. TRPM7-mediated spontaneous Ca 2+ entry regulates the proliferation and differentiation of human leukemia cell line K562. Physiol Rep 2018; 6:e13796. [PMID: 30033625 PMCID: PMC6055029 DOI: 10.14814/phy2.13796] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/20/2018] [Accepted: 06/24/2018] [Indexed: 11/30/2022] Open
Abstract
Continuous Ca2+ influx is essential to maintain intracellular Ca2+ homeostasis and its dysregulation leads to a variety of cellular dysfunctions. In this study, we explored the functional roles of spontaneous Ca2+ influx for the proliferation and differentiation of a human erythromyeloid leukemia cell line K562. mRNA/protein expressions were assessed by the real-time RT-PCR, western blotting, and immunocytochemical staining. Intracellular Ca2+ concentration ([Ca2+ ]i ) and ionic currents were measured by fluorescent imaging and patch clamping techniques, respectively. Cell counting/viability and colorimetric assays were applied to assess proliferation rate and hemoglobin synthesis, respectively. Elimination of extracellular Ca2+ decreased basal [Ca2+ ]i in proliferating K562 cells. Cation channel blockers such as SK&F96365, 2-APB, Gd3+ , and FTY720 dose dependently decreased basal [Ca2+ ]i . A spontaneously active inward current (Ispont ) contributive to basal [Ca2+ ]i was identified by the nystatin-perforated whole-cell recording. Ispont permeated Ca2+ comparably to Na+ , and was greatly eliminated by siRNA targeting TRPM7, a melastatin member of the transient receptor potential (TRP) superfamily. Consistent with these findings, TRPM7 immune reactivity was detected by western blotting, and immunofluorescence representing TRPM7 was found localized to the K562 cell membrane. Strikingly, all these procedures, that is, Ca2+ removal, TRPM7 blockers and siRNA-mediated TRPM7 knockdown significantly retarded the growth and suppressed hemin-induced γ-globin and hemoglobin syntheses in K562 cells, respectively, both of which appeared associated with the inhibition of ERK activation. These results collectively suggest that spontaneous Ca2+ influx through constitutively active TRPM7 channels may critically regulate both proliferative and erythroid differentiation potentials of K562 cells.
Collapse
Affiliation(s)
- Kiriko Takahashi
- Department of PhysiologyFukuoka University School of MedicineFukuokaJapan
- Department of AnesthesiologyFukuoka University School of MedicineFukuokaJapan
| | - Chisato Umebayashi
- Department of PhysiologyFukuoka University School of MedicineFukuokaJapan
| | - Tomohiro Numata
- Department of PhysiologyFukuoka University School of MedicineFukuokaJapan
| | - Akira Honda
- Department of PhysiologyFukuoka University School of MedicineFukuokaJapan
| | - Jun Ichikawa
- Department of PhysiologyFukuoka University School of MedicineFukuokaJapan
| | - Yaopeng Hu
- Department of PhysiologyFukuoka University School of MedicineFukuokaJapan
| | - Ken Yamaura
- Department of AnesthesiologyFukuoka University School of MedicineFukuokaJapan
| | - Ryuji Inoue
- Department of PhysiologyFukuoka University School of MedicineFukuokaJapan
| |
Collapse
|
23
|
Yang Z, Yao H, Fei F, Li Y, Qu J, Li C, Zhang S. Generation of erythroid cells from polyploid giant cancer cells: re-thinking about tumor blood supply. J Cancer Res Clin Oncol 2018; 144:617-627. [PMID: 29417259 DOI: 10.1007/s00432-018-2598-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 01/29/2018] [Indexed: 12/12/2022]
Abstract
INTRODUCTION During development and tumor progression, cells need a sufficient blood supply to maintain development and rapid growth. It is reported that there are three patterns of blood supply for tumor growth: endothelium-dependent vessels, mosaic vessels, and vasculogenic mimicry (VM). VM was first reported in highly aggressive uveal melanomas, with tumor cells mimicking the presence and function of endothelial cells forming the walls of VM vessels. The walls of mosaic vessels are randomly lined with both endothelial cells and tumor cells. We previously proposed a three-stage process, beginning with VM, progressing to mosaic vessels, and eventually leading to endothelium-dependent vessels. However, many phenomena unique to VM channel formation remain to be elucidated, such as the origin of erythrocytes before VM vessels connect with endothelium-dependent vessels. RESULTS In adults, erythroid cells are generally believed to be generated from hematopoietic stem cells in the bone marrow. In contrast, embryonic tissue obtains oxygen through formation of blood islands, which are largely composed of embryonic hemoglobin with a higher affinity with oxygen, in the absence of mature erythrocytes. Recent data from our laboratory suggest that embryonic blood-forming mechanisms also exist in cancer tissue, particularly when these tissues are under environmental stress such as hypoxia. We review the evidence from induced pluripotent stem cells in vitro and in vivo to support this previously underappreciated cell functionality in normal and cancer cells, including the ability to generate erythroid cells. We will also summarize the current understanding of tumor angiogenesis, VM, and our recent work on polyploid giant cancer cells, with emphasis on their ability to generate erythroid cells and their association with tumor growth under hypoxia. CONCLUSION An alternative embryonic pathway to obtain oxygen in cancer cells exists, particularly when they are under hypoxic conditions.
Collapse
Affiliation(s)
- Zhigang Yang
- Departments of Pathology, Baodi Traditional Chinese Medicine Hospital, Baodi District, Tianjin, 300121, People's Republic of China
| | - Hong Yao
- Department of thoracic Surgery, Tianjin Union Medical Center, Tianjin, 300121, People's Republic of China
| | - Fei Fei
- Nankai University School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China
- Department of Pathology, Tianjin Union Medical Center, Jieyuan Road, Hongqiao District, Tianjin, 300121, People's Republic of China
| | - Yuwei Li
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, 300121, People's Republic of China
| | - Jie Qu
- Nankai University School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China
- Department of Pathology, Tianjin Union Medical Center, Jieyuan Road, Hongqiao District, Tianjin, 300121, People's Republic of China
| | - Chunyuan Li
- Nankai University School of Medicine, Nankai University, Tianjin, 300071, People's Republic of China
- Department of Pathology, Tianjin Union Medical Center, Jieyuan Road, Hongqiao District, Tianjin, 300121, People's Republic of China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Jieyuan Road, Hongqiao District, Tianjin, 300121, People's Republic of China.
| |
Collapse
|
24
|
Sheng L, Jena PK, Hu Y, Liu HX, Nagar N, Kalanetra KM, French SW, French SW, Mills DA, Wan YJY. Hepatic inflammation caused by dysregulated bile acid synthesis is reversible by butyrate supplementation. J Pathol 2017; 243:431-441. [PMID: 28892150 DOI: 10.1002/path.4983] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 08/14/2017] [Accepted: 09/06/2017] [Indexed: 12/16/2022]
Abstract
Dysregulated bile acid (BA) synthesis or reduced farnesoid X receptor (FXR) levels are found in patients having metabolic diseases, autoimmune hepatitis, and liver cirrhosis or cancer. The objective of this study was to establish the relationship between butyrate and dysregulated BA synthesis-induced hepatitis as well as the effect of butyrate in reversing the liver pathology. Wild-type (WT) and FXR knockout (KO) male mice were placed on a control (CD) or western diet (WD) for 15 months. In the presence or absence of butyrate supplementation, feces obtained from 15-month-old WD-fed FXR KO mice, which had severe hepatitis and liver tumors, were transplanted to 7-month-old WD-fed FXR KO for 3 months. Hepatic phenotypes, microbiota profile, and BA composition were analyzed. Butyrate-generating bacteria and colonic butyrate concentration were reduced due to FXR inactivation and further reduced by WD intake. In addition, WD-fed FXR KO male mice had the highest concentration of hepatic β-muricholic acid (β-MCA) and bacteria-generated deoxycholic acid (DCA) accompanied by serious hepatitis. Moreover, dysregulated BA and reduced SCFA signaling co-existed in both human liver cancers and WD-fed FXR KO mice. Microbiota transplantation using butyrate-deficient feces derived from 15-month-old WD-fed FXR KO mice increased hepatic lymphocyte numbers as well as hepatic β-MCA and DCA concentrations. Furthermore, butyrate supplementation reduced hepatic β-MCA as well as DCA and eliminated hepatic lymphocyte infiltration. In conclusion, reduced butyrate contributes to the development of hepatitis in the FXR KO mouse model. In addition, butyrate reverses dysregulated BA synthesis and its associated hepatitis. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Lili Sheng
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA
| | - Prasant Kumar Jena
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA
| | - Ying Hu
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA
| | - Hui-Xin Liu
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA
| | - Nidhi Nagar
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA.,Department of Biological Sciences, California State University, East Bay, Hayward, CA, USA
| | - Karen M Kalanetra
- Department of Food Science and Technology, Department of Viticulture and Enology, University of California, Davis, CA, USA
| | | | - Samuel Wheeler French
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - David A Mills
- Department of Food Science and Technology, Department of Viticulture and Enology, University of California, Davis, CA, USA
| | - Yu-Jui Yvonne Wan
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Sacramento, CA, USA
| |
Collapse
|
25
|
Ha SH, Kang SK, Choi H, Kwak CH, Abekura F, Park JY, Kwon KM, Chang HW, Lee YC, Ha KT, Hou BK, Chung TW, Kim CH. Induction of GD3/α1-adrenergic receptor/transglutaminase 2-mediated erythroid differentiation in chronic myelogenous leukemic K562 cells. Oncotarget 2017; 8:72205-72219. [PMID: 29069780 PMCID: PMC5641123 DOI: 10.18632/oncotarget.20080] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 07/18/2017] [Indexed: 11/25/2022] Open
Abstract
The disialic acid-containing glycosphingolipid GD3 recruited membrane transglutaminase 2 (TG2) as a signaling molecule for erythroid differentiation in human chronic myelogenous leukemia (CML) K562 cells. The α1-adrenergic receptor (α1-AR)/TG2-mediated signaling pathway regulated GD3 functions, including gene expression and production, to differentiate CML K562 cells into erythroid lineage cells. Epinephrine, an AR agonist, increased membrane recruitment as well as GTP-photoaffinity of TG2, inducing GD3 synthase gene expression. Epinephrine activated PI3K/Akt signaling and GTPase downstream of TG2 activated Akt. The coupling of TG2 and GD3 production was specifically suppressed by prazosin (α1-AR antagonist), but not by propranolol (β-AR antagonist) or rauwolscine (α2-AR antagonist), indicating α1-AR specificity. Small interfering RNA (siRNA) experiment results indicated that the α1-AR/TG2-mediated signaling pathway activated PKCs α and δ to induce GD3 synthase gene expression. Transcription factors CREB, AP-1, and NF-κB regulated GD3 synthase gene expression during α1-AR-induced differentiation in CML K562 cells. In addition, GD3 synthase gene expression was upregulated in TG2-transfected cells via α1-AR with expression of erythroid lineage markers and benzidine-positive staining. α1-AR/TG2 signaling pathway-directed GD3 production is a crucial step in erythroid differentiation of K562 cells and GD3 interacts with α1-AR/TG2, inducing GD3/α1-AR/TG2-mediated erythroid differentiation. These results suggest that GD3, which acts as a membrane mediator of erythroid differentiation in CML cells, provides a therapeutic avenue for leukemia treatment.
Collapse
Affiliation(s)
- Sun-Hyung Ha
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Seoburo, Jangan-Gu, Kyunggi-Do, Korea
| | - Sung-Koo Kang
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Seoburo, Jangan-Gu, Kyunggi-Do, Korea
| | - Hyunju Choi
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Seoburo, Jangan-Gu, Kyunggi-Do, Korea
| | - Choong-Hwan Kwak
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Seoburo, Jangan-Gu, Kyunggi-Do, Korea
| | - Fukushi Abekura
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Seoburo, Jangan-Gu, Kyunggi-Do, Korea
| | - Jun-Young Park
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Seoburo, Jangan-Gu, Kyunggi-Do, Korea
| | - Kyung-Min Kwon
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Seoburo, Jangan-Gu, Kyunggi-Do, Korea
- Research Institute, Davinch-K Co., Ltd., Geumcheon-gu, Seoul, Korea
| | | | - Young-Choon Lee
- Faculty of Medicinal Biotechnology, Dong-A University, Busan, Korea
| | - Ki-Tae Ha
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan City, Gyeongsangnam-Do, Korea
| | - Bo Kyeng Hou
- Korean Bioinformation Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Tae-Wook Chung
- Division of Applied Medicine, School of Korean Medicine, Pusan National University, Yangsan City, Gyeongsangnam-Do, Korea
| | - Cheorl-Ho Kim
- Molecular and Cellular Glycobiology Unit, Department of Biological Sciences, SungKyunKwan University, Seoburo, Jangan-Gu, Kyunggi-Do, Korea
- Department of Medical Device Management and Research, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
26
|
Kurano M, Nishikawa M, Kuma H, Jona M, Yatomi Y. Involvement of Band3 in the efflux of sphingosine 1-phosphate from erythrocytes. PLoS One 2017; 12:e0177543. [PMID: 28494002 PMCID: PMC5426782 DOI: 10.1371/journal.pone.0177543] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 04/28/2017] [Indexed: 12/15/2022] Open
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive lipid mediator that is thought to be involved in various diseases. Although the main source of S1P in the plasma is erythrocytes, how S1P is exported from erythrocytes has not been elucidated. When we differentiated K562 cells into erythroblast-like cells with sodium butyrate, we observed that the efflux of S1P was increased without increased expression of previously proposed S1P transporters, while the expression levels of Band3 were increased. Therefore, in this study, we investigated the involvement of Band 3, the most characteristic membranous transporter for erythrocytes, in S1P efflux, using 4,4'-diisothiocyanatodihydrostilbene-2,2'-disulfonic acid, disodium salt (H2DIDS), which is an inhibitor of Band3. First, we treated human washed erythrocytes with H2DIDS and found that H2DIDS decreased the S1P levels in the supernatant, while it increased the cellular S1P contents. Next, when we injected H2DIDS into mice, the plasma S1P level was significantly decreased. Finally, when we overexpressed or suppressed Band3 in K562 cells, S1P efflux was enhanced or decreased, respectively, while the overexpression of Band3 in HEK293 cells did not modulate S1P efflux. These results suggested the possible involvement of Band3 in the transport of S1P, a multi-functional bioactive phospholipid, from erythrocytes.
Collapse
Affiliation(s)
- Makoto Kurano
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masako Nishikawa
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Kuma
- Department of Clinical Chemistry, Faculty of Pharmaceutical Sciences, Nagasaki International University, Nagasaki, Japan
| | - Masahiro Jona
- Department of Clinical Laboratory, The University of Tokyo Hospital, Tokyo, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- * E-mail:
| |
Collapse
|
27
|
Chen JL, Ping YH, Tseng MJ, Chang YI, Lee HC, Hsieh RH, Yeh TS. Notch1-promoted TRPA1 expression in erythroleukemic cells suppresses erythroid but enhances megakaryocyte differentiation. Sci Rep 2017; 7:42883. [PMID: 28220825 PMCID: PMC5318885 DOI: 10.1038/srep42883] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 01/16/2017] [Indexed: 01/09/2023] Open
Abstract
The Notch1 pathway plays important roles in modulating erythroid and megakaryocyte differentiation. To screen the Notch1-related genes that regulate differentiation fate of K562 and HEL cells, the expression of transient receptor potential ankyrin 1 (TRPA1) was induced by Notch1 receptor intracellular domain (N1IC), the activated form of Notch1 receptor. N1IC and v-ets erythroblastosis virus E26 oncogene homolog 1 (Ets-1) bound to TRPA1 promoter region to regulate transcription in K562 cells. Transactivation of TRPA1 promoter by N1IC depended on the methylation status of TRPA1 promoter. N1IC and Ets-1 suppressed the DNA methyltransferase 3B (DNMT3B) level in K562 cells. Inhibition of TRPA1 expression after Notch1 knockdown could be attenuated by nanaomycin A, an inhibitor of DNMT3B, in K562 and HEL cells. Functionally, hemin-induced erythroid differentiation could be suppressed by TRPA1, and the reduction of erythroid differentiation of both cells by N1IC and Ets-1 occurred via TRPA1. However, PMA-induced megakaryocyte differentiation could be enhanced by TRPA1, and the surface markers of megakaryocytes could be elevated by nanaomycin A. Megakaryocyte differentiation could be reduced by Notch1 or Ets-1 knockdown and relieved by TRPA1 overexpression. The results suggest that Notch1 and TRPA1 might be critical modulators that control the fate of erythroid and megakaryocyte differentiation.
Collapse
Affiliation(s)
- Ji-Lin Chen
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Yueh-Hsin Ping
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Min-Jen Tseng
- Department of Life Science, National Chung Cheng University, Chia-Yi 621, Taiwan
| | - Yuan-I Chang
- Department and Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Hsin-Chen Lee
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Rong-Hong Hsieh
- School of Nutrition and Health Sciences, College of Nutrition, Taipei Medical University, Taipei 110, Taiwan
| | - Tien-Shun Yeh
- Institute of Anatomy and Cell Biology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
- Genome Research Center, National Yang-Ming University, Taipei 112, Taiwan
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
28
|
Takahashi Y, Kubo R, Sano R, Nakajima T, Takahashi K, Kobayashi M, Handa H, Tsukada J, Kominato Y. Histone deacetylase inhibitors suppress ABO transcription in vitro, leading to reduced expression of the antigens. Transfusion 2016; 57:554-562. [PMID: 28019030 DOI: 10.1111/trf.13958] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 10/17/2016] [Accepted: 11/04/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND The ABO system is of fundamental importance in the fields of transfusion and transplantation and has apparent associations with certain diseases, including cardiovascular disorders. ABO expression is reduced in the late phase of erythroid differentiation in vitro, whereas histone deacetylase inhibitors (HDACIs) are known to promote cell differentiation. Therefore, whether or not HDACIs could reduce the amount of ABO transcripts and A or B antigens is an intriguing issue. STUDY DESIGN AND METHODS Quantitative polymerase chain reactions were carried out for the ABO transcripts in erythroid-lineage K562 and epithelial-lineage KATOIII cells after incubation with HDACIs, such as sodium butyrate, panobinostat, vorinostat, and sodium valproate. Flow cytometric analysis was conducted to evaluate the amounts of antigen in KATOIII cells treated with panobinostat. Quantitative chromatin immunoprecipitation (ChIP) assays and luciferase assays were performed on both cell types to examine the mechanisms of ABO suppression. RESULTS HDACIs reduced the ABO transcripts in both K562 and KATOIII cells, with panobinostat exerting the most significant effect. Flow cytometric analysis demonstrated a decrease in B-antigen expression on panobinostat-treated KATOIII cells. ChIP assays indicated that panobinostat altered the modification of histones in the transcriptional regulatory regions of ABO, and luciferase assays demonstrated reduced activity of these elements. CONCLUSION ABO transcription seems to be regulated by an epigenetic mechanism. Panobinostat appears to suppress ABO transcription, reducing the amount of antigens on the surface of cultured cells.
Collapse
Affiliation(s)
- Yoichiro Takahashi
- Department of Legal Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Rieko Kubo
- Department of Legal Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Rie Sano
- Department of Legal Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Tamiko Nakajima
- Department of Legal Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Keiko Takahashi
- Department of Legal Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Momoko Kobayashi
- Department of Legal Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Hiroshi Handa
- Department of Medicine and Clinical Science, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Junichi Tsukada
- Department of Hematology, University of Occupational and Environmental Health, Kitakyushu City, Japan
| | - Yoshihiko Kominato
- Department of Legal Medicine, Gunma University Graduate School of Medicine, Maebashi, Japan
| |
Collapse
|
29
|
Piel RB, Shiferaw MT, Vashisht AA, Marcero JR, Praissman JL, Phillips JD, Wohlschlegel JA, Medlock AE. A Novel Role for Progesterone Receptor Membrane Component 1 (PGRMC1): A Partner and Regulator of Ferrochelatase. Biochemistry 2016; 55:5204-17. [PMID: 27599036 DOI: 10.1021/acs.biochem.6b00756] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Heme is an iron-containing cofactor essential for multiple cellular processes and fundamental activities such as oxygen transport. To better understand the means by which heme synthesis is regulated during erythropoiesis, affinity purification coupled with mass spectrometry (MS) was performed to identify putative protein partners interacting with ferrochelatase (FECH), the terminal enzyme in the heme biosynthetic pathway. Both progesterone receptor membrane component 1 (PGRMC1) and progesterone receptor membrane component 2 (PGRMC2) were identified in these experiments. These interactions were validated by reciprocal affinity purification followed by MS analysis and immunoblotting. The interaction between PGRMC1 and FECH was confirmed in vitro and in HEK 293T cells, a non-erythroid cell line. When cells that are recognized models for erythroid differentiation were treated with a small molecule inhibitor of PGRMC1, AG-205, there was an observed decrease in the level of hemoglobinization relative to that of untreated cells. In vitro heme transfer experiments showed that purified PGRMC1 was able to donate heme to apo-cytochrome b5. In the presence of PGRMC1, in vitro measured FECH activity decreased in a dose-dependent manner. Interactions between FECH and PGRMC1 were strongest for the conformation of FECH associated with product release, suggesting that PGRMC1 may regulate FECH activity by controlling heme release. Overall, the data illustrate a role for PGRMC1 in regulating heme synthesis via interactions with FECH and suggest that PGRMC1 may be a heme chaperone or sensor.
Collapse
Affiliation(s)
- Robert B Piel
- Department of Biochemistry and Molecular Biology, Biomedical and Health Sciences Institute, AU/UGA Medical Partnership, University of Georgia , Athens, Georgia 30602, United States
| | - Mesafint T Shiferaw
- Department of Biochemistry and Molecular Biology, Biomedical and Health Sciences Institute, AU/UGA Medical Partnership, University of Georgia , Athens, Georgia 30602, United States
| | - Ajay A Vashisht
- Department of Biological Chemistry, University of California , Los Angeles, California 90095-1737, United States
| | - Jason R Marcero
- Department of Biochemistry and Molecular Biology, Biomedical and Health Sciences Institute, AU/UGA Medical Partnership, University of Georgia , Athens, Georgia 30602, United States
| | - Jeremy L Praissman
- Department of Biochemistry and Molecular Biology, Biomedical and Health Sciences Institute, AU/UGA Medical Partnership, University of Georgia , Athens, Georgia 30602, United States
| | - John D Phillips
- Hematology Division, University of Utah School of Medicine , Salt Lake City, Utah 84132, United States
| | - James A Wohlschlegel
- Department of Biological Chemistry, University of California , Los Angeles, California 90095-1737, United States
| | - Amy E Medlock
- Department of Biochemistry and Molecular Biology, Biomedical and Health Sciences Institute, AU/UGA Medical Partnership, University of Georgia , Athens, Georgia 30602, United States
| |
Collapse
|
30
|
Liao YJ, Lee YH, Chang FL, Ho H, Huang CH, Twu YC. The SHP2-ERK2 signaling pathway regulates branched I antigen formation by controlling the binding of CCAAT/enhancer binding protein α to the IGnTC promoter region during erythroid differentiation. Transfusion 2016; 56:2691-2702. [PMID: 27600951 DOI: 10.1111/trf.13796] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 06/20/2016] [Accepted: 06/25/2016] [Indexed: 11/29/2022]
Abstract
BACKGROUND Phosphorylation status of the transcription factor CCAAT/enhancer binding protein α (C/EBPα) has been demonstrated in a human hematopoietic cell model to regulate the formation of branched I antigen by affecting its binding affinity to the promoter region of the IGnTC gene during erythroid and granulocytic differentiation. STUDY DESIGN AND METHODS The K-562 cell line was induced to differentiate into red blood cells (RBCs) or granulocytes by sodium butyrate or retinoic acid, respectively, to study the involvement of three MAP kinase pathways in I antigen synthesis. The regulatory effects of the extracellular signal-regulated kinase (ERK)2-Src homology region 2 domain-containing phosphatase 2 (SHP2) pathway on phosphorylation status and binding affinities of C/EBPα as well as the subsequent activation of IGnTC and synthesis of surface I formation were studied in wild-type K-562 cells and in mutant cells that overexpress ERK2 and SHP2. RESULTS We found that SHP2-ERK2 signaling regulates the phosphorylation status of C/EBPα to alter its binding affinity onto the IGnTC promoter region, thereby activating the synthesis of cell surface I antigen formation during erythropoiesis. CONCLUSION SHP2-ERK2 signaling acts upstream of C/EBPα as a regulator of cell surface I antigen synthesis. Such regulation is specific for RBC but not for granulocyte differentiation.
Collapse
Affiliation(s)
- Yi-Jen Liao
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University
| | - Yen-Hua Lee
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei, Taiwan.,Department of Medical Research, Mackay Memorial Hospital, New Taipei City, Taiwan
| | - Fu-Ling Chang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hsun Ho
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chin-Han Huang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yuh-Ching Twu
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
31
|
Shariati L, Modaress M, Khanahmad H, Hejazi Z, Tabatabaiefar MA, Salehi M, Modarressi MH. Comparison of different methods for erythroid differentiation in the K562 cell line. Biotechnol Lett 2016; 38:1243-1250. [PMID: 27075690 DOI: 10.1007/s10529-016-2101-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 04/05/2016] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To compare methods for erythroid differentiation of K562 cells that will be promising in the treatment of beta-thalassemia by inducing γ-globin synthesis. RESULTS Cells were treated separately with: RPMI 1640 medium without glutamine, RPMI 1640 medium without glutamine supplemented with 1 mM sodium butyrate, RPMI 1640 medium supplemented with 1 mM sodium butyrate, 25 µg cisplatin/ml, 0.1 µg cytosine arabinoside/ml. The highest differentiation (84 %) with minimum toxicity was obtained with cisplatin at 15 µg /ml. Real-time RT-PCR showed that expression of the γ-globin gene was significantly higher in the cells differentiated with cisplatin compared to undifferentiated cells (P < 0.001). CONCLUSIONS Cisplatin is useful in the experimental therapy of ß-globin gene defects and can be considered for examining the basic mechanism of γ-reactivation.
Collapse
Affiliation(s)
- Laleh Shariati
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehran Modaress
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Khanahmad
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Hejazi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Amin Tabatabaiefar
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mansoor Salehi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Hossein Modarressi
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
32
|
Pulliam SR, Pellom ST, Shanker A, Adunyah SE. Butyrate regulates the expression of inflammatory and chemotactic cytokines in human acute leukemic cells during apoptosis. Cytokine 2016; 84:74-87. [PMID: 27253488 DOI: 10.1016/j.cyto.2016.05.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 05/12/2016] [Accepted: 05/13/2016] [Indexed: 02/06/2023]
Abstract
Butyrate is a histone deacetylase inhibitor implicated in many studies as a potential therapy for various forms of cancer. High concentrations of butyrate (>1.5mM) have been shown to activate apoptosis in several cancer cell lines including prostate, breast, and leukemia. Butyrate is also known to influence multiple signaling pathways that are mediators of cytokine production. The purpose of this study was to evaluate the impact of high concentrations of butyrate on the cancer microenvironment vis-à-vis apoptosis, cellular migration, and capacity to modulate cytokine expression in cancer cells. The results indicate that high concentrations of butyrate induced a 2-fold activation of caspase-3 and reduced cell viability by 60% in U937 leukemia cells. Within 24h, butyrate significantly decreased the levels of chemokines CCL2 and CCL5 in HL-60 and U937 cells, and decreased CCL5 in THP-1 leukemia cells. Differential effects were observed in treatments with valproic acid for CCL2 and CCL5 indicating butyrate-specificity. Many of the biological effects examined in this study are linked to activation of the AKT and MAPK signaling pathways; therefore, we investigated whether butyrate alters the levels of phosphorylated forms of these signaling proteins and how it correlated with the expression of chemokines. The results show that butyrate may partially regulate CCL5 production via p38 MAPK. The decrease in p-ERK1/2 and p-AKT levels correlated with the decrease in CCL2 production. These data suggest that while promoting apoptosis, butyrate has the potential to influence the cancer microenvironment by inducing differential expression of cytokines.
Collapse
Affiliation(s)
- Stephanie R Pulliam
- School of Graduate Studies and Research, Meharry Medical College, 1005 Dr. DB Todd, Jr. Blvd., Nashville, TN 37208, USA; Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, 1005 Dr. DB Todd, Jr. Blvd., Nashville, TN 37208, USA
| | - Samuel T Pellom
- School of Graduate Studies and Research, Meharry Medical College, 1005 Dr. DB Todd, Jr. Blvd., Nashville, TN 37208, USA; Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, 1005 Dr. DB Todd, Jr. Blvd., Nashville, TN 37208, USA; Department of Microbiology and Immunology, School of Medicine, Meharry Medical College, 1005 Dr. DB Todd, Jr. Blvd., Nashville, TN 37208, USA
| | - Anil Shanker
- School of Graduate Studies and Research, Meharry Medical College, 1005 Dr. DB Todd, Jr. Blvd., Nashville, TN 37208, USA; Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, 1005 Dr. DB Todd, Jr. Blvd., Nashville, TN 37208, USA; Host-Tumor Interactions Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Samuel E Adunyah
- School of Graduate Studies and Research, Meharry Medical College, 1005 Dr. DB Todd, Jr. Blvd., Nashville, TN 37208, USA; Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, 1005 Dr. DB Todd, Jr. Blvd., Nashville, TN 37208, USA.
| |
Collapse
|
33
|
Frank CL, Manandhar D, Gordân R, Crawford GE. HDAC inhibitors cause site-specific chromatin remodeling at PU.1-bound enhancers in K562 cells. Epigenetics Chromatin 2016; 9:15. [PMID: 27087856 PMCID: PMC4833939 DOI: 10.1186/s13072-016-0065-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 04/05/2016] [Indexed: 12/25/2022] Open
Abstract
Background Small molecule inhibitors of histone deacetylases (HDACi) hold promise as anticancer agents for particular malignancies. However, clinical use is often confounded by toxicity, perhaps due to indiscriminate hyperacetylation of cellular proteins. Therefore, elucidating the mechanisms by which HDACi trigger differentiation, cell cycle arrest, or apoptosis of cancer cells could inform development of more targeted therapies. We used the myelogenous leukemia line K562 as a model of HDACi-induced differentiation to investigate chromatin accessibility (DNase-seq) and expression (RNA-seq) changes associated with this process. Results We identified several thousand specific regulatory elements [~10 % of total DNase I-hypersensitive (DHS) sites] that become significantly more or less accessible with sodium butyrate or suberanilohydroxamic acid treatment. Most of the differential DHS sites display hallmarks of enhancers, including being enriched for non-promoter regions, associating with nearby gene expression changes, and increasing luciferase reporter expression in K562 cells. Differential DHS sites were enriched for key hematopoietic lineage transcription factor motifs, including SPI1 (PU.1), a known pioneer factor. We found PU.1 increases binding at opened DHS sites with HDACi treatment by ChIP-seq, but PU.1 knockdown by shRNA fails to block the chromatin accessibility and expression changes. A machine-learning approach indicates H3K27me3 initially marks PU.1-bound sites that open with HDACi treatment, suggesting these sites are epigenetically poised. Conclusions We find HDACi treatment of K562 cells results in site-specific chromatin remodeling at epigenetically poised regulatory elements. PU.1 shows evidence of a pioneer role in this process by marking poised enhancers but is not required for transcriptional activation. Electronic supplementary material The online version of this article (doi:10.1186/s13072-016-0065-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christopher L Frank
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC 27708 USA ; Center for Genomic and Computational Biology, Duke University, Durham, NC 27708 USA
| | - Dinesh Manandhar
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708 USA ; Program in Computational Biology and Bioinformatics, Duke University, Durham, NC 27708 USA
| | - Raluca Gordân
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708 USA ; Department of Biostatistics and Bioinformatics, Duke University, Durham, NC 27708 USA
| | - Gregory E Crawford
- Center for Genomic and Computational Biology, Duke University, Durham, NC 27708 USA ; Division of Medical Genetics, Department of Pediatrics, Duke University, Durham, NC 27708 USA
| |
Collapse
|
34
|
Voltage-Gated K+ Channel, Kv3.3 Is Involved in Hemin-Induced K562 Differentiation. PLoS One 2016; 11:e0148633. [PMID: 26849432 PMCID: PMC4743930 DOI: 10.1371/journal.pone.0148633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 01/20/2016] [Indexed: 12/31/2022] Open
Abstract
Voltage-gated K+ (Kv) channels are well known to be involved in cell proliferation. However, even though cell proliferation is closely related to cell differentiation, the relationship between Kv channels and cell differentiation remains poorly investigated. This study demonstrates that Kv3.3 is involved in K562 cell erythroid differentiation. Down-regulation of Kv3.3 using siRNA-Kv3.3 increased hemin-induced K562 erythroid differentiation through decreased activation of signal molecules such as p38, cAMP response element-binding protein, and c-fos. Down-regulation of Kv3.3 also enhanced cell adhesion by increasing integrin β3 and this effect was amplified when the cells were cultured with fibronectin. The Kv channels, or at least Kv3.3, appear to be associated with cell differentiation; therefore, understanding the mechanisms of Kv channel regulation of cell differentiation would provide important information regarding vital cellular processes.
Collapse
|
35
|
A novel miR-200b-3p/p38IP pair regulates monocyte/macrophage differentiation. Cell Discov 2016; 2:15043. [PMID: 27462440 PMCID: PMC4860955 DOI: 10.1038/celldisc.2015.43] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 11/05/2015] [Indexed: 01/13/2023] Open
Abstract
Monocyte/macrophage differentiation represents a major branch of hematopoiesis and is a central event in the immune response, but the molecular mechanisms underlying are not fully delineated. Here we show that p38 mitogen-activated protein kinase (MAPK) interacting protein (p38IP) is downregulated during monocyte/macrophage differentiation in vitro. Overexpression of p38IP halted monocyte/macrophage differentiation, whereas forward knockdown of p38IP by RNA interference induced G1/S arrest and promoted monocyte differentiation into macrophages and the maturation of macrophages as well. Moreover, we found that miR-200b-3p was upregulated during monocyte/macrophage differentiation and mediated the downregulation of p38IP by binding to the 3′ untranslated terminal region of p38IP mRNA. Overexpression of a miR-200b-3p mimic resembled the effect of p38IP knockdown, whereas a miR-200b-3p inhibitor blocked monocyte/macrophage differentiation by enhancing p38IP expression. Further western blotting analysis revealed that p38IP downregulation enhanced the activity of p38 MAPK and the subsequent accumulation of cyclin-dependent kinase inhibitor p21, thus promoting G1/S arrest and monocyte/macrophage differentiation. Moreover, the decline of GCN5 acetyltransferase caused by p38IP downregulation was required but was not sufficient for monocyte/macrophage differentiation. This study demonstrated a new role for p38IP and a novel miR-200b-3p/p38IP pair in the regulation of monocyte/macrophage differentiation.
Collapse
|
36
|
Manteniotis S, Wojcik S, Brauhoff P, Möllmann M, Petersen L, Göthert JR, Schmiegel W, Dührsen U, Gisselmann G, Hatt H. Functional characterization of the ectopically expressed olfactory receptor 2AT4 in human myelogenous leukemia. Cell Death Discov 2016; 2:15070. [PMID: 27551494 PMCID: PMC4979481 DOI: 10.1038/cddiscovery.2015.70] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 11/26/2015] [Accepted: 12/04/2015] [Indexed: 12/24/2022] Open
Abstract
The olfactory receptor (OR) family was found to be expressed mainly in the nasal epithelium. In the last two decades members of the OR family were detected to be functional expressed in different parts of the human body such as in liver, prostate or intestine cancer cells. Here, we detected the expression of several ORs in the human chronic myelogenous leukemia (CML) cell line K562 and in white blood cells of clinically diagnosed acute myeloid leukemia (AML) patients by RT-PCR and next-generation sequencing. With calcium-imaging, we characterized in greater detail the cell biological role of one OR (OR2AT4) in leukemia. In both cell systems, the OR2AT4 agonist Sandalore-evoked strong Ca2+ influx via the adenylate cyclase-cAMP-mediated pathway. The OR2AT4 antagonist Phenirat prevented the Sandalore-induced intracellular Ca2+ increase. Western blot and flow cytometric experiments revealed that stimulation of OR2AT4 reduced the proliferation by decreasing p38-MAPK phosphorylation and induced apoptosis via phosphorylation of p44/42-MAPK. Furthermore, Sandalore increased the number of hemoglobin-containing cells in culture. We described for the first time an OR-mediated pathway in CML and AML that can regulate proliferation, apoptosis and differentiation after activation. This mechanism offers novel therapeutic options for the treatment of AML.
Collapse
Affiliation(s)
- S Manteniotis
- Department of Cell Physiology, Ruhr-University Bochum , Bochum, Germany
| | - S Wojcik
- Department of Cell Physiology, Ruhr-University Bochum , Bochum, Germany
| | - P Brauhoff
- Department of Cell Physiology, Ruhr-University Bochum , Bochum, Germany
| | - M Möllmann
- Department of Hematology, University Hospital Essen , Essen, Germany
| | - L Petersen
- Department of Hematology, University Hospital Knappschaftskrankenhaus Bochum , Bochum, Germany
| | - J R Göthert
- Department of Hematology, University Hospital Essen , Essen, Germany
| | - W Schmiegel
- Department of Hematology, University Hospital Knappschaftskrankenhaus Bochum , Bochum, Germany
| | - U Dührsen
- Department of Hematology, University Hospital Essen , Essen, Germany
| | - G Gisselmann
- Department of Cell Physiology, Ruhr-University Bochum , Bochum, Germany
| | - H Hatt
- Department of Cell Physiology, Ruhr-University Bochum , Bochum, Germany
| |
Collapse
|
37
|
MiR-218 Inhibits Erythroid Differentiation and Alters Iron Metabolism by Targeting ALAS2 in K562 Cells. Int J Mol Sci 2015; 16:28156-68. [PMID: 26703568 PMCID: PMC4691035 DOI: 10.3390/ijms161226088] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 11/13/2015] [Accepted: 11/17/2015] [Indexed: 11/16/2022] Open
Abstract
microRNAs (miRNAs) are involved in a variety of biological processes. The regulatory function and potential role of miRNAs targeting the mRNA of the 5'-aminolevulinate synthase 2 (ALAS2) in erythropoiesis were investigated in order to identify miRNAs which play a role in erythroid iron metabolism and differentiation. Firstly, the role of ALAS2 in erythroid differentiation and iron metabolism in human erythroid leukemia cells (K562) was confirmed by ALAS2 knockdown. Through a series of screening strategies and experimental validations, it was identified that hsa-miR-218 (miR-218) targets and represses the expression of ALAS2 by binding to the 3'-untranslated region (UTR). Overexpression of miR-218 repressed erythroid differentiation and altered iron metabolism in K562 cells similar to that seen in the ALAS2 knockdown in K562 cells. In addition to iron metabolism and erythroid differentiation, miR-218 was found to be responsible for a reduction in K562 cell growth. Taken together, our results show that miR-218 inhibits erythroid differentiation and alters iron metabolism by targeting ALAS2 in K562 cells.
Collapse
|
38
|
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: 1.7] [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
|
39
|
Chaman N, Iqbal MA, Siddiqui FA, Gopinath P, Bamezai RNK. ERK2-Pyruvate Kinase Axis Permits Phorbol 12-Myristate 13-Acetate-induced Megakaryocyte Differentiation in K562 Cells. J Biol Chem 2015; 290:23803-15. [PMID: 26269597 DOI: 10.1074/jbc.m115.657411] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Indexed: 11/06/2022] Open
Abstract
Metabolic changes that contribute to differentiation are not well understood. Overwhelming evidence shows the critical role of glycolytic enzyme pyruvate kinase (PK) in directing metabolism of proliferating cells. However, its role in metabolism of differentiating cells is unclear. Here we studied the role of PK in phorbol 12-myristate 13-acetate (PMA)-induced megakaryocytic differentiation in human leukemia K562 cells. We observed that PMA treatment decreased cancer-type anabolic metabolism but increased ATP production, along with up-regulated expression of two PK isoforms (PKM2 and PKR) in an ERK2-dependent manner. Interestingly, silencing of PK (PKM2 and PKR) inhibited PMA-induced megakaryocytic differentiation, as revealed by decreased expression of megakaryocytic differentiation marker CD61 and cell cycle behavior. Further, PMA-induced ATP production reduced greatly upon PK silencing, suggesting that PK is required for ATP synthesis. In addition to metabolic effects, PMA treatment also translocated PKM2, but not PKR, into nucleus. ERK1/2 knockdowns independently and together suggested the role of ERK2 in the up-regulation of both the isoforms of PK, proposing a role of ERK2-PK isoform axis in differentiation. Collectively, our findings unravel ERK2 guided PK-dependent metabolic changes during PMA induction, which are important in megakaryocytic differentiation.
Collapse
Affiliation(s)
- Noor Chaman
- From the National Centre of Applied Human Genetics, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Mohammad Askandar Iqbal
- From the National Centre of Applied Human Genetics, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Farid Ahmad Siddiqui
- From the National Centre of Applied Human Genetics, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Prakasam Gopinath
- From the National Centre of Applied Human Genetics, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Rameshwar N K Bamezai
- From the National Centre of Applied Human Genetics, School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| |
Collapse
|
40
|
Pharmacological Induction of Human Fetal Globin Gene in Hydroxyurea-Resistant Primary Adult Erythroid Cells. Mol Cell Biol 2015; 35:2541-53. [PMID: 25986606 DOI: 10.1128/mcb.00035-15] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 05/06/2015] [Indexed: 11/20/2022] Open
Abstract
Pharmacological induction of the fetal γ globin gene and the consequent formation of HbF (α2/γ2) in adult erythroid cells are one feasible therapeutic strategy for sickle cell disease (SCD) and severe β-thalassemias. Hydroxyurea (HU) is the current drug of choice for SCD, but serious side effects limit its clinical use. Moreover, 30 to 50% of patients are irresponsive to HU treatment. We have used high-throughput screening to identify benzo[de]benzo[4,5]imidazo[2,1-a]isoquinolin-7-one and its derivatives (compounds I to VI) as potent γ globin inducers. Of the compounds, I to V exert superior γ globin induction and have better therapeutic potential than HU, likely because of their activation of the p38 mitogen-activated protein kinase (MAPK) signaling pathway and modulation of expression levels and/or chromosome binding of γ globin gene regulators, including BCL11A, and chromatin structure over the γ globin promoter. Unlike sodium butyrate (NaB), the global levels of acetylated histones H3 and H4 are not changed by compound II treatment. Remarkably, compound II induces the γ globin gene in HU-resistant primary human adult erythroid cells, the p38 signaling pathway of which appears to be irresponsive to HU and NaB as well as compound II. This study provides a new framework for the development of new and superior compounds for treating SCD and severe β-thalassemias.
Collapse
|
41
|
Inaguma Y, Akatsuka Y, Hosokawa K, Maruyama H, Okamoto A, Katagiri T, Shiraishi K, Murayama Y, Tsuzuki-Iba S, Mizutani Y, Nishii C, Yamamoto N, Demachi-Okamura A, Kuzushima K, Ogawa S, Emi N, Nakao S. Induction of HLA-B*40:02-restricted T cells possessing cytotoxic and suppressive functions against haematopoietic progenitor cells from a patient with severe aplastic anaemia. Br J Haematol 2015; 172:131-4. [PMID: 25929998 DOI: 10.1111/bjh.13464] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Yoko Inaguma
- Department of Haematology, Fujita Health University School of Medicine, Aichi, Japan
| | - Yoshiki Akatsuka
- Department of Haematology, Fujita Health University School of Medicine, Aichi, Japan.
| | - Kohei Hosokawa
- Cellular Transplantation Biology, Kanazawa University Graduate School, Ishikawa, Japan
| | - Hiroyuki Maruyama
- Cellular Transplantation Biology, Kanazawa University Graduate School, Ishikawa, Japan
| | - Akinao Okamoto
- Department of Haematology, Fujita Health University School of Medicine, Aichi, Japan
| | - Takamasa Katagiri
- Cellular Transplantation Biology, Kanazawa University Graduate School, Ishikawa, Japan
| | - Keiko Shiraishi
- Division of Immunology, Aichi Cancer Center Research Institute, Aichi, Japan
| | - Yuko Murayama
- Department of Haematology, Fujita Health University School of Medicine, Aichi, Japan
| | - Sachiko Tsuzuki-Iba
- Department of Haematology, Fujita Health University School of Medicine, Aichi, Japan
| | - Yuuki Mizutani
- Department of Clinical Laboratory, Fujita Health University School of Medicine, Aichi, Japan
| | - Chikako Nishii
- Department of Clinical Laboratory, Fujita Health University School of Medicine, Aichi, Japan
| | - Naoki Yamamoto
- Laboratory of Molecular Biology and Histochemistry, Fujita Health University Joint Research Laboratory, Aichi, Japan
| | | | - Kiyotaka Kuzushima
- Division of Immunology, Aichi Cancer Center Research Institute, Aichi, Japan
| | - Seishi Ogawa
- Departments of Pathology and Tumour Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Nobuhiko Emi
- Department of Haematology, Fujita Health University School of Medicine, Aichi, Japan
| | - Shinji Nakao
- Cellular Transplantation Biology, Kanazawa University Graduate School, Ishikawa, Japan
| |
Collapse
|
42
|
Natural Remedies for the Treatment of Beta-Thalassemia and Sickle Cell Anemia-Current Status and Perspectives in Fetal Hemoglobin Reactivation. INTERNATIONAL SCHOLARLY RESEARCH NOTICES 2014; 2014:123257. [PMID: 27350962 PMCID: PMC4897541 DOI: 10.1155/2014/123257] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 07/21/2014] [Accepted: 07/25/2014] [Indexed: 11/18/2022]
Abstract
For the treatment of β-thalassemia and sickle cell disease (SCD), pharmacological induction of fetal hemoglobin (HbF) production may be a promising approach. To date, numerous studies have been done on identifying the novel HbF-inducing agents and understanding the underlying mechanism for stimulating the HbF production. In this review, we have summarized the identified HbF-inducing agents by far. By examining the action mechanisms of the HbF-inducing agents, various studies have suggested that despite the ability of stimulating HbF production, the chemotherapeutic agents could not be practically applied for treating β-hemoglobinopathies, especially β-thalassemia, due to the their cytotoxicity and growth-inhibitory effect. Owing to this therapeutic obstacle, much effort has been put on identifying new HbF-inducing agents from the natural world with the combination of efficacy, safety, and ease of use. Therefore, this review aims to (i) reveal the novel screening platforms for identifying potential inducers with high efficiency and accuracy and to (ii) summarize the new identified natural remedies for stimulating HbF production. Hopefully, this review can provide a new insight into the current status and future perspectives in fetal hemoglobin reactivation for treating β-thalassaemia and SCD.
Collapse
|
43
|
Lai YJ, Wu WY, Yang CM, Yang LR, Chu CC, Chan YS, Lin M, Yu LC. A systematic study of single-nucleotide polymorphisms in theA4GALTgene suggests a molecular genetic basis for the P1/P2blood groups. Transfusion 2014; 54:3222-31. [DOI: 10.1111/trf.12771] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 05/14/2014] [Accepted: 05/20/2014] [Indexed: 01/03/2023]
Affiliation(s)
- Yin-Ju Lai
- Institute of Biochemical Sciences, College of Life Science; National Taiwan University; Taipei Taiwan
- Institute of Biological Chemistry; Academia Sinica; Taipei Taiwan
- Department of Medical Research; Mackay Memorial Hospital; Taipei Taiwan
- Blood Bank; Mackay Memorial Hospital; Taipei Taiwan
| | - Wan-Yi Wu
- Institute of Biochemical Sciences, College of Life Science; National Taiwan University; Taipei Taiwan
- Institute of Biological Chemistry; Academia Sinica; Taipei Taiwan
- Department of Medical Research; Mackay Memorial Hospital; Taipei Taiwan
- Blood Bank; Mackay Memorial Hospital; Taipei Taiwan
| | - Chen-Ming Yang
- Institute of Biochemical Sciences, College of Life Science; National Taiwan University; Taipei Taiwan
- Institute of Biological Chemistry; Academia Sinica; Taipei Taiwan
- Department of Medical Research; Mackay Memorial Hospital; Taipei Taiwan
- Blood Bank; Mackay Memorial Hospital; Taipei Taiwan
| | - Li-Rong Yang
- Institute of Biochemical Sciences, College of Life Science; National Taiwan University; Taipei Taiwan
- Institute of Biological Chemistry; Academia Sinica; Taipei Taiwan
- Department of Medical Research; Mackay Memorial Hospital; Taipei Taiwan
- Blood Bank; Mackay Memorial Hospital; Taipei Taiwan
| | - Chen-Chung Chu
- Institute of Biochemical Sciences, College of Life Science; National Taiwan University; Taipei Taiwan
- Institute of Biological Chemistry; Academia Sinica; Taipei Taiwan
- Department of Medical Research; Mackay Memorial Hospital; Taipei Taiwan
- Blood Bank; Mackay Memorial Hospital; Taipei Taiwan
| | - Yung-Syu Chan
- Institute of Biochemical Sciences, College of Life Science; National Taiwan University; Taipei Taiwan
- Institute of Biological Chemistry; Academia Sinica; Taipei Taiwan
- Department of Medical Research; Mackay Memorial Hospital; Taipei Taiwan
- Blood Bank; Mackay Memorial Hospital; Taipei Taiwan
| | - Marie Lin
- Institute of Biochemical Sciences, College of Life Science; National Taiwan University; Taipei Taiwan
- Institute of Biological Chemistry; Academia Sinica; Taipei Taiwan
- Department of Medical Research; Mackay Memorial Hospital; Taipei Taiwan
- Blood Bank; Mackay Memorial Hospital; Taipei Taiwan
| | - Lung-Chih Yu
- Institute of Biochemical Sciences, College of Life Science; National Taiwan University; Taipei Taiwan
- Institute of Biological Chemistry; Academia Sinica; Taipei Taiwan
- Department of Medical Research; Mackay Memorial Hospital; Taipei Taiwan
- Blood Bank; Mackay Memorial Hospital; Taipei Taiwan
| |
Collapse
|
44
|
Chu HC, Lee HY, Huang YS, Tseng WL, Yen CJ, Cheng JC, Tseng CP. Erythroid differentiation is augmented in Reelin-deficient K562 cells and homozygous reeler mice. FEBS Lett 2013; 588:58-64. [PMID: 24239537 DOI: 10.1016/j.febslet.2013.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 10/08/2013] [Accepted: 11/04/2013] [Indexed: 02/06/2023]
Abstract
Reelin is an extracellular glycoprotein that is highly conserved in mammals. In addition to its expression in the nervous system, Reelin is present in erythroid cells but its function there is unknown. We report in this study that Reelin is up-regulated during erythroid differentiation of human erythroleukemic K562 cells and is expressed in the erythroid progenitors of murine bone marrow. Reelin deficiency promotes erythroid differentiation of K562 cells and augments erythroid production in murine bone marrow. In accordance with these findings, Reelin deficiency attenuates AKT phosphorylation of the Ter119(+)CD71(+) erythroid progenitors and alters the cell number and frequency of the progenitors at different erythroid differentiation stages. A regulatory role of Reelin in erythroid differentiation is thus defined.
Collapse
Affiliation(s)
- Hui-Chun Chu
- Graduate Institute of Biomedical Science, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 333, Taiwan, ROC
| | - Hsing-Ying Lee
- Graduate Institute of Biomedical Science, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 333, Taiwan, ROC
| | - Yen-Shu Huang
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 333, Taiwan, ROC
| | - Wei-Lien Tseng
- Graduate Institute of Biomedical Science, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 333, Taiwan, ROC
| | - Ching-Ju Yen
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 333, Taiwan, ROC
| | - Ju-Chien Cheng
- Department of Medical Laboratory Sciences and Biotechnology, China Medical University, Taichung 404, Taiwan, ROC.
| | - Ching-Ping Tseng
- Graduate Institute of Biomedical Science, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 333, Taiwan, ROC; Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 333, Taiwan, ROC; Molecular Medicine Research Center, Chang Gung University, Kwei-Shan, Taoyuan 333, Taiwan, ROC.
| |
Collapse
|
45
|
Lee YL, Chen CW, Liu FH, Huang YW, Huang HM. Aclacinomycin A sensitizes K562 chronic myeloid leukemia cells to imatinib through p38MAPK-mediated erythroid differentiation. PLoS One 2013; 8:e61939. [PMID: 23613979 PMCID: PMC3629111 DOI: 10.1371/journal.pone.0061939] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 03/15/2013] [Indexed: 12/31/2022] Open
Abstract
Expression of oncogenic Bcr-Abl inhibits cell differentiation of hematopoietic stem/progenitor cells in chronic myeloid leukemia (CML). Differentiation therapy is considered to be a new strategy for treating this type of leukemia. Aclacinomycin A (ACM) is an antitumor antibiotic. Previous studies have shown that ACM induced erythroid differentiation of CML cells. In this study, we investigate the effect of ACM on the sensitivity of human CML cell line K562 to Bcr-Abl specific inhibitor imatinib (STI571, Gleevec). We first determined the optimal concentration of ACM for erythroid differentiation but not growth inhibition and apoptosis in K562 cells. Then, pretreatment with this optimal concentration of ACM followed by a minimally toxic concentration of imatinib strongly induced growth inhibition and apoptosis compared to that with simultaneous co-treatment, indicating that ACM-induced erythroid differentiation sensitizes K562 cells to imatinib. Sequential treatment with ACM and imatinib induced Bcr-Abl down-regulation, cytochrome c release into the cytosol, and caspase-3 activation, as well as decreased Mcl-1 and Bcl-xL expressions, but did not affect Fas ligand/Fas death receptor and procaspase-8 expressions. ACM/imatinib sequential treatment-induced apoptosis was suppressed by a caspase-9 inhibitor and a caspase-3 inhibitor, indicating that the caspase cascade is involved in this apoptosis. Furthermore, we demonstrated that ACM induced erythroid differentiation through the p38 mitogen-activated protein kinase (MAPK) pathway. The inhibition of erythroid differentiation by p38MAPK inhibitor SB202190, p38MAPK dominant negative mutant or p38MAPK shRNA knockdown, reduced the ACM/imatinib sequential treatment-mediated growth inhibition and apoptosis. These results suggest that differentiated K562 cells induced by ACM-mediated p38MAPK pathway become more sensitive to imatinib and result in down-regulations of Bcr-Abl and anti-apoptotic proteins, growth inhibition and apoptosis. These results provided a potential management by which ACM might have a crucial impact on increasing sensitivity of CML cells to imatinib in the differentiation therapeutic approaches.
Collapse
Affiliation(s)
- Yueh-Lun Lee
- Department of Microbiology and Immunology, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
46
|
Qian X, Chen J, Zhao D, Guo L, Qian X. Plastrum testudinis induces γ-globin gene expression through epigenetic histone modifications within the γ-globin gene promoter via activation of the p38 MAPK signaling pathway. Int J Mol Med 2013; 31:1418-28. [PMID: 23588991 DOI: 10.3892/ijmm.2013.1338] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 03/15/2013] [Indexed: 11/06/2022] Open
Abstract
The pharmacologically-induced expression of the γ-globin gene, to increase fetal hemoglobin (HbF) production, is a therapeutic strategy used for the treatment of β-thalassemia and sickle cell anemia (SCA). The aim of this study was to investigate the effects of Plastrum testudinis (PT) on differentiation, proliferation, γ-globin gene expression and HbF synthesis in human erythroid cells. For this purpose, we used the K562 human leukemia cell line and human erythroid progenitor cells from normal donors and patients with β-thalassemia cultured using the two-phase liquid culture system. The effects of PT on erythroid differentiation, proliferation, γ-globin gene expression and HbF synthesis, as well as the involvement of epigenetic histone modifications within the γ-globin gene promoter via activation of the p38 mitogen-activated protein kinase (MAPK) signaling pathway, were assessed by benzidine staining, trypan-blue dye exclusion, quantitative real-time RT-PCR (qRT-PCR), western blot analysis and chromatin immunoprecipitation (ChIP). PT promoted the erythroid differentiation of K562 cells, and increased γ-globin mRNA accumulation and HbF synthesis without inhibiting cell proliferation in K562 cells and human erythroid progenitors. PT exerted no effect on α- and β-globin gene expression. In human erythroid cells, PT activated the p38 MAPK signaling pathway, and enhanced the acetylation of histone H3 and H4, the phosphorylation of histone H3 within the Gγ- and Aγ-globin gene promoter regions, γ-globin mRNA accumulation and HbF synthesis. These effects were suppressed by pre-treatment with the p38 MAPK inhibitor, SB203580. Epigenetic histone modifications within γ-globin gene promoter regions, via activation of the p38 MAPK signaling pathway, are important for the induction of γ-globin gene expression in human erythroid cells by PT. PT may be a novel potential therapeutic agent for β-hemoglobinopathies, including β-thalassemia and SCA.
Collapse
Affiliation(s)
- Xinhua Qian
- Department of Neonatology, Southern Medical University, Guangzhou, Guangdong, People's Republic of China.
| | | | | | | | | |
Collapse
|
47
|
Song L, Li X, Jayandharan GR, Wang Y, Aslanidi GV, Ling C, Zhong L, Gao G, Yoder MC, Ling C, Tan M, Srivastava A. High-efficiency transduction of primary human hematopoietic stem cells and erythroid lineage-restricted expression by optimized AAV6 serotype vectors in vitro and in a murine xenograft model in vivo. PLoS One 2013; 8:e58757. [PMID: 23516552 PMCID: PMC3597592 DOI: 10.1371/journal.pone.0058757] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 02/06/2013] [Indexed: 11/19/2022] Open
Abstract
We have observed that of the 10 AAV serotypes, AAV6 is the most efficient in transducing primary human hematopoietic stem cells (HSCs), and that the transduction efficiency can be further increased by specifically mutating single surface-exposed tyrosine (Y) residues on AAV6 capsids. In the present studies, we combined the two mutations to generate a tyrosine double-mutant (Y705+731F) AAV6 vector, with which >70% of CD34+ cells could be transduced. With the long-term objective of developing recombinant AAV vectors for the potential gene therapy of human hemoglobinopathies, we generated the wild-type (WT) and tyrosine-mutant AAV6 vectors containing the following erythroid cell-specific promoters: β-globin promoter (βp) with the upstream hyper-sensitive site 2 (HS2) enhancer from the β-globin locus control region (HS2-βbp), and the human parvovirus B19 promoter at map unit 6 (B19p6). Transgene expression from the B19p6 was significantly higher than that from the HS2-βp, and increased up to 30-fold and up to 20-fold, respectively, following erythropoietin (Epo)-induced differentiation of CD34+ cells in vitro. Transgene expression from the B19p6 or the HS2-βp was also evaluated in an immuno-deficient xenograft mouse model in vivo. Whereas low levels of expression were detected from the B19p6 in the WT AAV6 capsid, and that from the HS2-βp in the Y705+731F AAV6 capsid, transgene expression from the B19p6 promoter in the Y705+731F AAV6 capsid was significantly higher than that from the HS2-βp, and was detectable up to 12 weeks post-transplantation in primary recipients, and up to 6 additional weeks in secondary transplanted animals. These data demonstrate the feasibility of the use of the novel Y705+731F AAV6-B19p6 vectors for high-efficiency transduction of HSCs as well as expression of the b-globin gene in erythroid progenitor cells for the potential gene therapy of human hemoglobinopathies such as β-thalassemia and sickle cell disease.
Collapse
Affiliation(s)
- Liujiang Song
- Experimental Hematology Laboratory, Department of Physiology, School of Basic Medical Sciences, Central South University, Changsha, China
- Shenzhen Institute of Xiangya Biomedicine, Shenzhen, China
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Genetics Institute, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Xiaomiao Li
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Genetics Institute, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Giridhara R. Jayandharan
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Genetics Institute, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Department of Haematology, Christian Medical College, Vellore, Tamil Nadu, India
- Center for Stem Cell Research, Christian Medical College, Vellore, Tamil Nadu, India
| | - Yuan Wang
- Department of Traditional Chinese Medicine, Changhai Hospital, Second Military Medical University, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - George V. Aslanidi
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Genetics Institute, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Chen Ling
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Genetics Institute, University of Florida College of Medicine, Gainesville, Florida, United States of America
| | - Li Zhong
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Genetics Institute, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Guangping Gao
- Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Department of Microbiology & Physiology Systems, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Mervin C. Yoder
- Herman B Well Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Changquan Ling
- Department of Traditional Chinese Medicine, Changhai Hospital, Second Military Medical University, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mengqun Tan
- Experimental Hematology Laboratory, Department of Physiology, School of Basic Medical Sciences, Central South University, Changsha, China
- Shenzhen Institute of Xiangya Biomedicine, Shenzhen, China
- * E-mail: (MT); (AS)
| | - Arun Srivastava
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Powell Gene Therapy Center, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Genetics Institute, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, Florida, United States of America
- Shands Cancer Center, University of Florida College of Medicine, Gainesville, Florida, United States of America
- * E-mail: (MT); (AS)
| |
Collapse
|
48
|
Saito T, Nishida K, Furumatsu T, Yoshida A, Ozawa M, Ozaki T. Histone deacetylase inhibitors suppress mechanical stress-induced expression of RUNX-2 and ADAMTS-5 through the inhibition of the MAPK signaling pathway in cultured human chondrocytes. Osteoarthritis Cartilage 2013; 21:165-74. [PMID: 23017871 DOI: 10.1016/j.joca.2012.09.003] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 07/18/2012] [Accepted: 09/19/2012] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To investigate the inhibitory effects and the regulatory mechanisms of histone deacetylase (HDAC) inhibitors on mechanical stress-induced gene expression of runt-related transcription factor (RUNX)-2 and adisintegrin and metalloproteinase with thrombospondin motif (ADAMTS)-5 in human chondrocytes. METHODS Human chondrocytes were seeded in stretch chambers at a concentration of 5 × 10(4)cells/chamber. Cells were pre-incubated with or without HDAC inhibitors (MS-275 or trichostatin A; TSA) for 12h, followed by uniaxial cyclic tensile strain (CTS) (0.5Hz, 10% elongation), which was applied for 30 min using the ST-140-10 system (STREX, Osaka, Japan). Total RNA was extracted and the expression of RUNX-2, ADAMTS-5, matrix metalloproteinase (MMP)-3, and MMP-13 at the mRNA and protein levels were examined by real-time polymerase chain reaction (PCR) and immunocytochemistry, respectively. The activation of diverse mitogen-activated protein kinase (MAPK) pathways with or without HDAC inhibitors during CTS was examined by western blotting. RESULTS HDAC inhibitors (TSA: 10 nM, MS-275: 100 nM) suppressed CTS-induced expression of RUNX-2, ADAMTS-5, and MMP-3 at both the mRNA and protein levels within 1h. CTS-induced activation of p38 MAPK (p38), extracellular signal-regulated kinase (ERK), and c-Jun N-terminal kinase (JNK) MAPKs was downregulated by both HDAC inhibitors. CONCLUSION The CTS-induced expression of RUNX-2 and ADAMTS-5 was suppressed by HDAC inhibitors via the inhibition of the MAPK pathway activation in human chondrocytes. The results of the current study suggested a novel therapeutic role for HDAC inhibitors against degenerative joint disease such as osteoarthritis.
Collapse
Affiliation(s)
- T Saito
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Density and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama City, Okayama 700-8558, Japan
| | | | | | | | | | | |
Collapse
|
49
|
Fyrberg A, Skoglund K, Wolk M, Lotfi K. A potential role of fetal hemoglobin in the development of multidrug resistance. Biochem Biophys Res Commun 2012; 427:456-60. [PMID: 22910410 DOI: 10.1016/j.bbrc.2012.07.129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 07/23/2012] [Indexed: 10/28/2022]
Abstract
Our previous data from a human leukemic cell line made resistant to the nucleoside analog (NA) 9-β-D-arabinofuranosylguanine (AraG) revealed a massive upregulation of fetal hemoglobin (HbF) genes and the ABCB1 gene coding for the multidrug resistance P-glycoprotein (P-gp). The expression of these genes is regulated through the same mechanisms, with activation of the p38-MAPK pathway and inhibition of methylation making transcription factors more accessible to activate these genes. We could show that AraG, as well as other NAs, and P-gp substrates could induce global DNA demethylation and induction of Hbγ and P-gp both at the mRNA and protein expression level. We speculate that the expression of HbF prior to drug exposure or in drug-resistant cell lines is a strategy of the cancer to gain more oxygen, and thereby survival benefits. We also believe that P-gp may be induced in order to excrete Hb degradation products from the cells that would otherwise be toxic. By using Hbγ siRNA and pharmacological inhibitors of HbF production we here present a possible relationship between HbF induction and multi-drug resistance in a human leukemia cell line model.
Collapse
Affiliation(s)
- A Fyrberg
- Centre for Biomedical Resources, Faculty of Health Sciences, Linköping University, Linköping, Sweden.
| | | | | | | |
Collapse
|
50
|
Canh Hiep N, Kinohira S, Furuyama K, Taketani S. Depletion of glutamine enhances sodium butyrate-induced erythroid differentiation of K562 cells. J Biochem 2012; 152:509-19. [DOI: 10.1093/jb/mvs097] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|