1
|
Han X, He W, Liang D, Liu X, Zhou J, de Thé H, Zhu J, Yuan H. Creg1 Regulates Erythroid Development via TGF-β/Smad2-Klf1 Axis in Zebrafish. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402804. [PMID: 38953462 PMCID: PMC11434009 DOI: 10.1002/advs.202402804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/04/2024] [Indexed: 07/04/2024]
Abstract
Understanding the regulation of normal erythroid development will help to develop new potential therapeutic strategies for disorders of the erythroid lineage. Cellular repressor of E1A-stimulated genes 1 (CREG1) is a glycoprotein that has been implicated in the regulation of tissue homeostasis. However, its role in erythropoiesis remains largely undefined. In this study, it is found that CREG1 expression increases progressively during erythroid differentiation. In zebrafish, creg1 mRNA is preferentially expressed within the intermediate cell mass (ICM)/peripheral blood island (PBI) region where primitive erythropoiesis occurs. Loss of creg1 leads to anemia caused by defective erythroid differentiation and excessive apoptosis of erythroid progenitors. Mechanistically, creg1 deficiency results in reduced activation of TGF-β/Smad2 signaling pathway. Treatment with an agonist of the Smad2 pathway (IDE2) could significantly restore the defective erythroid development in creg1-/- mutants. Further, Klf1, identified as a key target gene downstream of the TGF-β/Smad2 signaling pathway, is involved in creg1 deficiency-induced aberrant erythropoiesis. Thus, this study reveals a previously unrecognized role for Creg1 as a critical regulator of erythropoiesis, mediated at least in part by the TGF-β/Smad2-Klf1 axis. This finding may contribute to the understanding of normal erythropoiesis and the pathogenesis of erythroid disorders.
Collapse
Affiliation(s)
- Xiao Han
- Shanghai Institute of HematologyState Key Laboratory of Medical GenomicsNational Research Center for Translational Medicine at ShanghaiRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- CNRS‐LIA Hematology and CancerSino‐French Research Center for Life Sciences and GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Wenxin He
- Shanghai Institute of HematologyState Key Laboratory of Medical GenomicsNational Research Center for Translational Medicine at ShanghaiRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- CNRS‐LIA Hematology and CancerSino‐French Research Center for Life Sciences and GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Dongguo Liang
- Shanghai Institute of HematologyState Key Laboratory of Medical GenomicsNational Research Center for Translational Medicine at ShanghaiRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Xiaohui Liu
- Shanghai Institute of HematologyState Key Laboratory of Medical GenomicsNational Research Center for Translational Medicine at ShanghaiRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- CNRS‐LIA Hematology and CancerSino‐French Research Center for Life Sciences and GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Jun Zhou
- Shanghai Institute of HematologyState Key Laboratory of Medical GenomicsNational Research Center for Translational Medicine at ShanghaiRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- CNRS‐LIA Hematology and CancerSino‐French Research Center for Life Sciences and GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Hugues de Thé
- CNRS‐LIA Hematology and CancerSino‐French Research Center for Life Sciences and GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- Université de Paris 7/INSERM/CNRS UMR 944/7212Equipe Labellisée Ligue Nationale Contre le CancerHôpital St. LouisParis75010France
| | - Jun Zhu
- CNRS‐LIA Hematology and CancerSino‐French Research Center for Life Sciences and GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- Université de Paris 7/INSERM/CNRS UMR 944/7212Equipe Labellisée Ligue Nationale Contre le CancerHôpital St. LouisParis75010France
| | - Hao Yuan
- Shanghai Institute of HematologyState Key Laboratory of Medical GenomicsNational Research Center for Translational Medicine at ShanghaiRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
- CNRS‐LIA Hematology and CancerSino‐French Research Center for Life Sciences and GenomicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghai200025China
| |
Collapse
|
2
|
Lu Y, Ma WB, Ren GM, Li YT, Wang T, Zhan YQ, Xiang SS, Chen H, Gao HY, Zhao K, Yu M, Li CY, Yang XM, Yin RH. GPS2 promotes erythroid differentiation in K562 erythroleukemia cells primarily via NCOR1. Int J Hematol 2024; 120:157-166. [PMID: 38814500 DOI: 10.1007/s12185-024-03797-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 05/10/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024]
Abstract
G protein pathway suppressor 2 (GPS2) has been shown to play a pivotal role in human and mouse definitive erythropoiesis in an EKLF-dependent manner. However, whether GPS2 affects human primitive erythropoiesis is still unknown. This study demonstrated that GPS2 positively regulates erythroid differentiation in K562 cells, which have a primitive erythroid phenotype. Overexpression of GPS2 promoted hemin-induced hemoglobin synthesis in K562 cells as assessed by the increased percentage of benzidine-positive cells and the deeper red coloration of the cell pellets. In contrast, knockdown of GPS2 inhibited hemin-induced erythroid differentiation of K562 cells. GPS2 overexpression also enhanced erythroid differentiation of K562 cells induced by cytosine arabinoside (Ara-C). GPS2 induced hemoglobin synthesis by increasing the expression of globin and ALAS2 genes, either under steady state or upon hemin treatment. Promotion of erythroid differentiation of K562 cells by GPS2 mainly relies on NCOR1, as knockdown of NCOR1 or lack of the NCOR1-binding domain of GPS2 potently diminished the promotive effect. Thus, our study revealed a previously unknown role of GPS2 in regulating human primitive erythropoiesis in K562 cells.
Collapse
Affiliation(s)
- Ying Lu
- College of Life Science and Bioengineering, Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, China
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Wen-Bing Ma
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Guang-Ming Ren
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Ya-Ting Li
- College of Life Science and Bioengineering, Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, China
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Ting Wang
- College of Life Science and Bioengineering, Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, China
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yi-Qun Zhan
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Shen-Si Xiang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Hui Chen
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Hui-Ying Gao
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Ke Zhao
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Miao Yu
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Chang-Yan Li
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Xiao-Ming Yang
- College of Life Science and Bioengineering, Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, China.
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Rong-Hua Yin
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| |
Collapse
|
3
|
Yu Z, Qiao X, Yu S, Gu X, Jin Y, Tang C, Niu J, Wang L, Song L. The involvement of interferon regulatory factor 8 in regulating the proliferation of haemocytes in oyster Crassostrea gigas. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 156:105172. [PMID: 38537730 DOI: 10.1016/j.dci.2024.105172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/18/2024] [Accepted: 03/24/2024] [Indexed: 05/03/2024]
Abstract
Interferon regulatory factor 8 (IRF8) is an important transcriptional regulatory factor involving in multiple biological process, such as the antiviral immune response, immune cell proliferation and differentiation. In the present study, the involvement of a previously identified IRF8 homologue (CgIRF8) in regulating haemocyte proliferation of oyster were further investigated. CgIRF8 mRNA transcripts were detectable in all the stages of C. gigas larvae with the highest level in D-veliger (1.76-fold of that in zygote, p < 0.05). Its mRNA transcripts were also detected in all the three haemocyte subpopulations of adult oysters with the highest expression in granulocytes (2.79-fold of that in agranulocytes, p < 0.01). After LPS stimulation, the mRNA transcripts of CgIRF8 in haemocytes significantly increased at 12 h and 48 h, which were 2.04-fold and 1.65-fold (p < 0.05) of that in control group, respectively. Meanwhile, the abundance of CgIRF8 protein in the haemocytes increased significantly at 12 h after LPS stimulation (1.71-fold of that in seawater, p < 0.05). The immunofluorescence assay and Western blot showed that LPS stimulation induced an obvious nucleus translocation of CgIRF8 protein in haemocytes. After the expression of CgIRF8 was inhibited by the injection of CgIRF8 siRNA, the percentage of EdU positive haemocytes, the proportion of granulocytes, and the mRNA expression levels of CgGATA and CgSCL all declined significantly at 12 h after LPS stimulation, which was 0.64-fold (p < 0.05), 0.7-fold (p < 0.05), 0.31-fold and 0.54-fold (p < 0.001) of that in the NC group, respectively. While the expression level of cell proliferation-related protein CgCDK2, CgCDC6, CgCDC45 and CgPCNA were significantly increased (1.99-fold, and 2.41-fold, 3.76-fold and 4.79-fold compared to that in the NC group respectively, p < 0.001). Dual luciferase reporter assay demonstrated that CgIRF8 was able to activate the CgGATA promoter in HEK293T cells after transfection of CgGATA and CgIRF8. These results collectively indicated that CgIRF8 promoted haemocyte proliferation by regulating the expression of CgGATA and other related genes in the immune response of oyster.
Collapse
Affiliation(s)
- Zhuo Yu
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Xue Qiao
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Simiao Yu
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Xiaoyu Gu
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Yuhao Jin
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Chunyu Tang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Jixiang Niu
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Southern Laboratory of Ocean Science and Engineering (Guangdong, Zhuhai), Zhuhai, 519000, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology and Disease Control, Dalian Ocean University, Dalian, 116023, China; Southern Laboratory of Ocean Science and Engineering (Guangdong, Zhuhai), Zhuhai, 519000, China; Laboratory of Marine Fisheries Science and Food Production Process, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Dalian Key Laboratory of Aquatic Animal Disease Prevention and Control, Dalian Ocean University, Dalian, 116023, China.
| |
Collapse
|
4
|
Deleuze V, Soler E, Andrieu-Soler C. Protocol for efficient CRISPR-Cas9-mediated fluorescent tag knockin in hard-to-transfect erythroid cell lines. STAR Protoc 2024; 5:103016. [PMID: 38640065 PMCID: PMC11044133 DOI: 10.1016/j.xpro.2024.103016] [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: 01/16/2024] [Revised: 03/01/2024] [Accepted: 03/27/2024] [Indexed: 04/21/2024] Open
Abstract
Precise insertion of fluorescent tags by CRISPR-Cas9-mediated homologous recombination (HR) in mammalian genes is a powerful tool allowing to study gene function and protein gene products. Here, we present a protocol for efficient HR-mediated targeted insertion of fluorescent markers in the genome of hard-to-transfect erythroid cell lines MEL (mouse erythroleukemic) and MEDEP (mouse ES cell-derived erythroid progenitor line). We describe steps for plasmid construction, electroporation, amplification, and verification of genome editing. We then detail procedures for isolating positive clones and validating knockin clones. For complete details on the use and execution of this protocol, please refer to Deleuze et al.1.
Collapse
Affiliation(s)
- Virginie Deleuze
- IGMM University Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Université' de Paris, Paris, France
| | - Eric Soler
- IGMM University Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Université' de Paris, Paris, France.
| | - Charlotte Andrieu-Soler
- IGMM University Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Université' de Paris, Paris, France.
| |
Collapse
|
5
|
Ju H, Sohn Y, Nam Y, Rim YA. Progresses in overcoming the limitations of in vitro erythropoiesis using human induced pluripotent stem cells. Stem Cell Res Ther 2024; 15:142. [PMID: 38750578 PMCID: PMC11094930 DOI: 10.1186/s13287-024-03754-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/04/2024] [Indexed: 05/19/2024] Open
Abstract
Researchers have attempted to generate transfusable oxygen carriers to mitigate RBC supply shortages. In vitro generation of RBCs using stem cells such as hematopoietic stem and progenitor cells (HSPCs), embryonic stem cells (ESCs), and induced pluripotent stem cells (iPSCs) has shown promise. Specifically, the limited supplies of HSPCs and ethical issues with ESCs make iPSCs the most promising candidate for in vitro RBC generation. However, researchers have encountered some major challenges when using iPSCs to produce transfusable RBC products, such as enucleation and RBC maturation. In addition, it has proven difficult to manufacture these products on a large scale. In this review, we provide a brief overview of erythropoiesis and examine endeavors to recapitulate erythropoiesis in vitro using various cell sources. Furthermore, we explore the current obstacles and potential solutions aimed at enabling the large-scale production of transfusable RBCs in vitro.
Collapse
Affiliation(s)
- Hyeonwoo Ju
- Department of Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Yeowon Sohn
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea
| | - Yoojun Nam
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea.
- YiPSCELL Inc., L2 Omnibus Park, Banpo-dearo 222, Seocho-gu, Seoul, 06591, Republic of Korea.
| | - Yeri Alice Rim
- YiPSCELL Inc., L2 Omnibus Park, Banpo-dearo 222, Seocho-gu, Seoul, 06591, Republic of Korea.
- CiSTEM laboratory, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| |
Collapse
|
6
|
Guo C, Lv X, Zhang Q, Yi L, Ren Y, Li Z, Yan J, Zheng S, Sun M, Liu S. CRKL but not CRKII contributes to hemin-induced erythroid differentiation of CML. J Cell Mol Med 2024; 28:e18308. [PMID: 38683131 PMCID: PMC11057422 DOI: 10.1111/jcmm.18308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/22/2023] [Accepted: 03/26/2024] [Indexed: 05/01/2024] Open
Abstract
Destruction of erythropoiesis process leads to various diseases, including thrombocytopenia, anaemia, and leukaemia. miR-429-CT10 regulation of kinase-like (CRKL) axis involved in development, progression and metastasis of cancers. However, the exact role of miR-429-CRKL axis in leukaemic cell differentiation are still unknown. The current work aimed to uncover the effect of miR-429-CRKL axis on erythropoiesis. In the present study, CRKL upregulation was negatively correlated with miR-429 downregulation in both chronic myeloid leukaemia (CML) patient and CR patient samples. Moreover, CRKL expression level was significantly decreased while miR-429 expression level was increased during the erythroid differentiation of K562 cells following hemin treatment. Functional investigations revealed that overexpression and knockdown of CRKL was remarkably effective in suppressing and promoting hemin-induced erythroid differentiation of K562 cells, whereas, miR-429 exhibited opposite effects to CRKL. Mechanistically, miR-429 regulates erythroid differentiation of K562 cells by downregulating CRKL via selectively targeting CRKL-3'-untranslated region (UTR) through Raf/MEK/ERK pathway. Conversely, CRKII had no effect on erythroid differentiation of K562 cells. Taken together, our data demonstrated that CRKL (but not CRKII) and miR-429 contribute to development, progression and erythropoiesis of CML, miR-429-CRKL axis regulates erythropoiesis of K562 cells via Raf/MEK/ERK pathway, providing novel insights into effective diagnosis and therapy for CML patients.
Collapse
MESH Headings
- Humans
- 3' Untranslated Regions
- Adaptor Proteins, Signal Transducing/metabolism
- Adaptor Proteins, Signal Transducing/genetics
- Cell Differentiation/drug effects
- Erythroid Cells/metabolism
- Erythroid Cells/drug effects
- Erythroid Cells/pathology
- Erythroid Cells/cytology
- Erythropoiesis/genetics
- Erythropoiesis/drug effects
- Gene Expression Regulation, Leukemic/drug effects
- Hemin/pharmacology
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- MAP Kinase Signaling System/drug effects
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Proto-Oncogene Proteins c-crk/metabolism
- Proto-Oncogene Proteins c-crk/genetics
Collapse
Affiliation(s)
- Chunmei Guo
- Department of Biotechnology & Liaoning Key Laboratory of Cancer Stem Cell Research, College of Basic Medical SciencesDalian Medical UniversityDalianLiaoningChina
| | - Xinxin Lv
- Department of Biotechnology & Liaoning Key Laboratory of Cancer Stem Cell Research, College of Basic Medical SciencesDalian Medical UniversityDalianLiaoningChina
| | - Qiuling Zhang
- Department of Biochemistry, College of Basic Medical SciencesDalian Medical UniversityDalianLiaoningChina
| | - Lina Yi
- Department of Biochemistry, College of Basic Medical SciencesDalian Medical UniversityDalianLiaoningChina
| | - Yingying Ren
- Department of Biotechnology & Liaoning Key Laboratory of Cancer Stem Cell Research, College of Basic Medical SciencesDalian Medical UniversityDalianLiaoningChina
| | - Zhaopeng Li
- Department of Biochemistry, College of Basic Medical SciencesDalian Medical UniversityDalianLiaoningChina
| | - Jinsong Yan
- Department of Hematology, The Second Affiliated Hospital of Dalian Medical UniversityInstitute of Stem Cell Transplantation of Dalian Medical UniversityDalianLiaoningChina
| | - Shanliang Zheng
- Department of Biochemistry, College of Basic Medical SciencesDalian Medical UniversityDalianLiaoningChina
| | - Ming‐Zhong Sun
- Department of Biotechnology & Liaoning Key Laboratory of Cancer Stem Cell Research, College of Basic Medical SciencesDalian Medical UniversityDalianLiaoningChina
| | - Shuqing Liu
- Department of Biochemistry, College of Basic Medical SciencesDalian Medical UniversityDalianLiaoningChina
| |
Collapse
|
7
|
Yu HC, Cui R, Chen MY, Du XY, Bai QR, Zhang SL, Guo JJ, Tong FC, Wu J. Regulation of Erythroid Differentiation via the HIF1α-NFIL3-PIM1 Signaling Axis Under Hypoxia. Antioxid Redox Signal 2024. [PMID: 38573002 DOI: 10.1089/ars.2023.0508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/05/2024]
Abstract
Aims: Erythropoiesis is controlled by several factors, including oxygen level under different circumstances. However, the role of hypoxia in erythroid differentiation and the underlying mechanisms are poorly understood. We studied the effect and mechanism of hypoxia on erythroid differentiation of K562 cells and observed the effect of hypoxia on early erythropoiesis of zebrafish. Results: Compared with normal oxygen culture, both hemin-induced erythroid differentiation of K562 cells and the early erythropoiesis of zebrafish were inhibited under hypoxic treatment conditions. Hypoxia-inducible factor 1 alpha (HIF1α) plays a major role in the response to hypoxia. Here, we obtained a stable HIF1α knockout K562 cell line using the CRISPR-Cas9 technology and further demonstrated that HIF1α knockout promoted hemin-induced erythroid differentiation of K562 cells under hypoxia. We demonstrated an HIF1-mediated induction of the nuclear factor interleukin-3 (NFIL3) regulated in K562 cells under hypoxia. Interestingly, a gradual decrease in NFIL3 expression was detected during erythroid differentiation of erythropoietin-induced CD34+ hematopoietic stem/progenitor cells (HSPCs) and hemin-induced K562 cells. Notably, erythroid differentiation was inhibited by enforced expression of NFIL3 under normoxia and was promoted by the knockdown of NFIL3 under hypoxia in hemin-treated K562 cells. In addition, a target of NFIL3, pim-1 proto-oncogene, serine/threonine kinase (PIM1), was obtained by RNA microarray after NFIL3 knockdown. PIM1 can rescue the inhibitory effect of NFIL3 on hemin-induced erythroid differentiation of K562 cells. Innovation and Conclusion: Our findings demonstrate that the HIF1α-NFIL3-PIM1 signaling axis plays an important role in erythroid differentiation under hypoxia. These results will provide useful clues for preventing the damage of acute hypoxia to erythropoiesis.
Collapse
Affiliation(s)
- Hai-Chuan Yu
- School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| | - Rui Cui
- School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| | - Meng-Yao Chen
- School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| | - Xiao-Yan Du
- School of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Qi-Rong Bai
- School of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Shuang-Ling Zhang
- School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| | - Jiao-Jie Guo
- School of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Fang-Chao Tong
- School of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Jiao Wu
- School of Pharmacy, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
8
|
Cong X, Li X, Xu K, Yin L, Liang G, Sun R, Pu Y, Zhang J. HIF-1α/m 6A/NF-κB/CCL3 axis-mediated immunosurveillance participates in low level benzene-related erythrohematopoietic development toxicity. ENVIRONMENT INTERNATIONAL 2024; 184:108493. [PMID: 38350257 DOI: 10.1016/j.envint.2024.108493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 02/15/2024]
Abstract
Defective erythropoiesis is one of the causes of anemia and leukemia. However, the mechanisms underlying defective erythropoiesis under a low-dose environment of benzene are poorly understood. In the present study, multiple omics (transcriptomics and metabolomics) and methods from epidemiology to experimental biology (e.g., benzene-induced (WT and HIF-1α + ) mouse, hiPSC-derived HSPCs) were used. Here, we showed that erythropoiesis is more easily impacted than other blood cells, and the process is reversible, which involves HIF-1 and NF-kB signaling pathways in low-level benzene exposure workers. Decreased HIF-1α expression in benzene-induced mouse bone marrow resulted in DNA damage, senescence, and apoptosis in BMCs and HSCs, causing disturbances in iron homeostasis and erythropoiesis. We further revealed that HIF-1α mediates CCL3/macrophage-related immunosurveillance against benzene-induced senescent and damaged cells and contributes to iron homeostasis. Mechanistically, we showed that m6A modification is essential in this process. Benzene-induced depletion of m6A promotes the mRNA stability of gene NFKBIA and regulates the NF-κB/CCL3 pathway, which is regulated by HIF-1α/METTL3/YTHDF2. Overall, our results identified an unidentified role for HIF-1α, m6A, and the NF-kB signaling machinery in erythroid progenitor cells, suggesting that HIF-1α/METTL3/YTHDF2-m6A/NF-κB/CCL3 axis may be a potential prevention and therapeutic target for chronic exposure of humans to benzene-associated anemia and leukemia.
Collapse
Affiliation(s)
- Xiaowei Cong
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China
| | - Xiaoqin Li
- Yangzhou Center for Disease Control and Prevention, Yangzhou 225100, Jiangsu, China
| | - Kai Xu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China
| | - Lihong Yin
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China
| | - Geyu Liang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China
| | - Rongli Sun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China
| | - Yuepu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China.
| | - Juan Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, Jiangsu, China.
| |
Collapse
|
9
|
Zhao B, Che H, Li L, Hu L, Yi W, Xiao L, Liu S, Hou Z. Asperuloside regulates the proliferation, apoptosis, and differentiation of chronic myeloid leukemia cell line K562 through the RAS/MEK/ERK pathway. Heliyon 2024; 10:e23580. [PMID: 38226258 PMCID: PMC10788273 DOI: 10.1016/j.heliyon.2023.e23580] [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: 08/13/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 01/17/2024] Open
Abstract
Context Chronic myeloid leukemia (CML) is a malignant hematopoietic stem cell disease caused by excessive proliferation and abnormal differentiation of hematopoietic stem cells. Asperuloside (ASP) is considered to have good biological activity and may be a good anti-CML drug. Objective This study aimed to explore the effects and possible mechanisms of ASP on the biological behavior of K562 cells based on RNA-seq. Materials and methods The IC50 of ASP in K562 cells was calculated by the concentration-effect curve. Cell viability, apoptosis, and differentiation were detected by CCK8, flow cytometry, benzidine staining, and WB analysis, respectively. Further, RNA-seq was used to analyze the possible mechanism of ASP regulating K562 cells. Results ASP significantly inhibited the proliferation, and promoted apoptosis and differentiation of K562 cells. A total of 117 differentially expressed genes were screened by RNA-seq, mainly involved in the RAS/MEK/ERK pathway. PD98059 was used to inhibit the RAS/MEK/ERK pathway in K562 cells, and results confirmed that PD98059 could not only inhibit the RAS/MEK/ERK pathway, but also inhibit the regulation of ASP on the proliferation and differentiation of K562 cells. Conclusion ASP inhibited the proliferation, promoted apoptosis and differentiation of K562 cells by regulating the RAS/MEK/ERK pathway, and played a good anti-CML role.
Collapse
Affiliation(s)
| | | | - Linlin Li
- Department of Hematology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Lian Hu
- Department of Hematology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Wenjing Yi
- Department of Hematology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Li Xiao
- Department of Hematology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Songshan Liu
- Department of Hematology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Zhufa Hou
- Department of Hematology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| |
Collapse
|
10
|
Iftikhar F, Khan MBN, Tehreem S, Kanwal N, Musharraf SG. BCL11A-targeted γ-globin gene induction by triterpenoid glycosides of Fagonia indica: A preclinical scientific validation of indigenous herb for the treatment of β-hemoglobinopathies. Bioorg Chem 2023; 140:106768. [PMID: 37586133 DOI: 10.1016/j.bioorg.2023.106768] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/18/2023]
Abstract
Pharmacological induction of fetal hemoglobin has proven to be a promising therapeutic intervention in β-hemoglobinopathies by reducing the globin chain imbalance and inhibiting sickle cell polymerization. Fagonia indica has shown therapeutic relevance to β-thalassemia. Therefore, we study the ethnopharmacological potential of Fagonia indica and its biomarker compounds for their HbF induction ability for the treatment of β-thalassemia. Here, we identify, compound 8 (triterpenoid glycosides) of F. indica. as a prominent HbF inducer in-vitro and in-vivo. Compound 8 showed potent erythroid differentiation, enhanced cellular proliferation, ample accumulation of total hemoglobin, and a strong notion of γ-globin gene expression in K562 cultures. Compound 8 treatment also revealed strong induction of erythroid differentiation and fetal hemoglobin mRNA and protein in adult erythroid precursor cells. This induction was associated with simultaneous downregulation of BCL11A and SOX6, and overexpression of the GATA-1 gene, suggesting a compound 8-mediated partial mechanism involved in the reactivation of fetal-like globin genes. The in vivo study with compound 8 (10 mg/kg) in β-YAC mice resulted in significant HbF synthesis demonstrated by the enhanced level of F-cells (84.14 %) and an 8.85-fold increase in the γ-globin gene. Overall, the study identifies compound 8 as a new HbF-inducing entity and provides an early "proof-of-concept" to enable the initiation of preclinical and clinical studies in the development of this HbF-inducing agent for β-thalassemia.
Collapse
Affiliation(s)
- Fizza Iftikhar
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Muhammad Behroz Naeem Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Syeda Tehreem
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Nayab Kanwal
- H.E.J. Research Institute of Chemistry, 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
|
Pegka F, Ben-Califa N, Neumann D, Jäkel H, Hengst L. EpoR Activation Stimulates Erythroid Precursor Proliferation by Inducing Phosphorylation of Tyrosine-88 of the CDK-Inhibitor p27 Kip1. Cells 2023; 12:1704. [PMID: 37443738 PMCID: PMC10340229 DOI: 10.3390/cells12131704] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/07/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Erythrocyte biogenesis needs to be tightly regulated to secure oxygen transport and control plasma viscosity. The cytokine erythropoietin (Epo) governs erythropoiesis by promoting cell proliferation, differentiation, and survival of erythroid precursor cells. Erythroid differentiation is associated with an accumulation of the cyclin-dependent kinase inhibitor p27Kip1, but the regulation and role of p27 during erythroid proliferation remain largely unknown. We observed that p27 can bind to the erythropoietin receptor (EpoR). Activation of EpoR leads to immediate Jak2-dependent p27 phosphorylation of tyrosine residue 88 (Y88). This modification is known to impair its CDK-inhibitory activity and convert the inhibitor into an activator and assembly factor of CDK4,6. To investigate the physiological role of p27-Y88 phosphorylation in erythropoiesis, we analyzed p27Y88F/Y88F knock-in mice, where tyrosine-88 was mutated to phenylalanine. We observed lower red blood cell counts, lower hematocrit levels, and a reduced capacity for colony outgrowth of CFU-Es (colony-forming unit-erythroid), indicating impaired cell proliferation of early erythroid progenitors. Compensatory mechanisms of reduced p27 and increased Epo expression protect from stronger dysregulation of erythropoiesis. These observations suggest that p27-Y88 phosphorylation by EpoR pathway activation plays an important role in the stimulation of erythroid progenitor proliferation during the early stages of erythropoiesis.
Collapse
Affiliation(s)
- Fragka Pegka
- Institute of Medical Biochemistry, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Nathalie Ben-Califa
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel (D.N.)
| | - Drorit Neumann
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel (D.N.)
| | - Heidelinde Jäkel
- Institute of Medical Biochemistry, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Ludger Hengst
- Institute of Medical Biochemistry, Biocenter, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
12
|
Tsiftsoglou SA. Heme Interactions as Regulators of the Alternative Pathway Complement Responses and Implications for Heme-Associated Pathologies. Curr Issues Mol Biol 2023; 45:5198-5214. [PMID: 37367079 DOI: 10.3390/cimb45060330] [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: 05/07/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/28/2023] Open
Abstract
Heme (Fe2+-protoporphyrin IX) is a pigment of life, and as a prosthetic group in several hemoproteins, it contributes to diverse critical cellular processes. While its intracellular levels are tightly regulated by networks of heme-binding proteins (HeBPs), labile heme can be hazardous through oxidative processes. In blood plasma, heme is scavenged by hemopexin (HPX), albumin and several other proteins, while it also interacts directly with complement components C1q, C3 and factor I. These direct interactions block the classical pathway (CP) and distort the alternative pathway (AP). Errors or flaws in heme metabolism, causing uncontrolled intracellular oxidative stress, can lead to several severe hematological disorders. Direct interactions of extracellular heme with alternative pathway complement components (APCCs) may be implicated molecularly in diverse conditions at sites of abnormal cell damage and vascular injury. In such disorders, a deregulated AP could be associated with the heme-mediated disruption of the physiological heparan sulphate-CFH coat of stressed cells and the induction of local hemostatic responses. Within this conceptual frame, a computational evaluation of HBMs (heme-binding motifs) aimed to determine how heme interacts with APCCs and whether these interactions are affected by genetic variation within putative HBMs. Combined computational analysis and database mining identified putative HBMs in all of the 16 APCCs examined, with 10 exhibiting disease-associated genetic (SNPs) and/or epigenetic variation (PTMs). Overall, this article indicates that among the pleiotropic roles of heme reviewed, the interactions of heme with APCCs could induce differential AP-mediated hemostasis-driven pathologies in certain individuals.
Collapse
Affiliation(s)
- Stefanos A Tsiftsoglou
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
13
|
Lu Y, Ma WB, Ren GM, Liu X, Li YT, Wang T, Zhan YQ, Xiang SS, Yu M, Li CY, Yang XM, Yin RH. VPS37C facilitates erythroid differentiation by promoting EKLF stability. Biochem Biophys Res Commun 2023; 671:229-235. [PMID: 37307706 DOI: 10.1016/j.bbrc.2023.06.023] [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: 05/15/2023] [Revised: 05/26/2023] [Accepted: 06/06/2023] [Indexed: 06/14/2023]
Abstract
The process of erythroid differentiation is orchestrated at the molecular level by a complex network of transcription factors. Erythroid Krüppel-like factor (EKLF/KLF1) is a master erythroid gene regulator that directly regulates most aspects of terminal erythroid differentiation. However, the underlying regulatory mechanisms of EKLF protein stability are still largely unknown. In this study, we identified Vacuolar protein sorting 37 C (VPS37C), a core subunit of the Endosomal sorting complex required for transport-I (ESCRT-I) complex, as an essential regulator of EKLF stability. Our study showed that VPS37C interacts with EKLF and prevents K48-linked polyubiquitination of EKLF and proteasome-mediated EKLF degradation, thus enhancing EKLF protein stability and transcriptional activity. VPS37C overexpression in murine erythroleukemia (MEL) cells promotes hexamethylene bisacetamide (HMBA)-induced erythroid differentiation manifested by up-regulating erythroid-specific EKLF target genes and increasing benzidine-positive cells. In contrast, VPS37C knockdown inhibits HMBA-induced MEL cell erythroid differentiation. Particularly, the restoration of EKLF expression in VPS37C-knockdown MEL cells reverses erythroid-specific gene expression and hemoglobin production. Collectively, our study demonstrated VPS37C is a novel regulator of EKLF ubiquitination and degradation, which plays a positive role in erythroid differentiation of MEL cells by enhancing EKLF protein stability.
Collapse
Affiliation(s)
- Ying Lu
- College of Life Science and Bioengineering, Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, China; State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Wen-Bing Ma
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Guang-Ming Ren
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Xian Liu
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Ya-Ting Li
- College of Life Science and Bioengineering, Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, China; State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Ting Wang
- College of Life Science and Bioengineering, Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, China; State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yi-Qun Zhan
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Shen-Si Xiang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Miao Yu
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Chang-Yan Li
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Xiao-Ming Yang
- College of Life Science and Bioengineering, Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, China; State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Rong-Hua Yin
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| |
Collapse
|
14
|
Maslova A, Plotnikov V, Nuriddinov M, Gridina M, Fishman V, Krasikova A. Hi-C analysis of genomic contacts revealed karyotype abnormalities in chicken HD3 cell line. BMC Genomics 2023; 24:66. [PMID: 36750787 PMCID: PMC9906895 DOI: 10.1186/s12864-023-09158-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/31/2023] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND Karyotype abnormalities are frequent in immortalized continuous cell lines either transformed or derived from primary tumors. Chromosomal rearrangements can cause dramatic changes in gene expression and affect cellular phenotype and behavior during in vitro culture. Structural variations of chromosomes in many continuous mammalian cell lines are well documented, but chromosome aberrations in cell lines from other vertebrate models often remain understudied. The chicken LSCC-HD3 cell line (HD3), generated from erythroid precursors, was used as an avian model for erythroid differentiation and lineage-specific gene expression. However, karyotype abnormalities in the HD3 cell line were not assessed. In the present study, we applied high-throughput chromosome conformation capture to analyze 3D genome organization and to detect chromosome rearrangements in the HD3 cell line. RESULTS We obtained Hi-C maps of genomic interactions for the HD3 cell line and compared A/B compartments and topologically associating domains between HD3 and several other cell types. By analysis of contact patterns in the Hi-C maps of HD3 cells, we identified more than 25 interchromosomal translocations of regions ≥ 200 kb on both micro- and macrochromosomes. We classified most of the observed translocations as unbalanced, leading to the formation of heteromorphic chromosomes. In many cases of microchromosome rearrangements, an entire microchromosome together with other macro- and microchromosomes participated in the emergence of a derivative chromosome, resembling "chromosomal fusions'' between acrocentric microchromosomes. Intrachromosomal inversions, deletions and duplications were also detected in HD3 cells. Several of the identified simple and complex chromosomal rearrangements, such as between GGA2 and GGA1qter; GGA5, GGA4p and GGA7p; GGA4q, GGA6 and GGA19; and duplication of the sex chromosome GGAW, were confirmed by FISH. CONCLUSIONS In the erythroid progenitor HD3 cell line, in contrast to mature and immature erythrocytes, the genome is organized into distinct topologically associating domains. The HD3 cell line has a severely rearranged karyotype with most of the chromosomes engaged in translocations and can be used in studies of genome structure-function relationships. Hi-C proved to be a reliable tool for simultaneous assessment of the spatial genome organization and chromosomal aberrations in karyotypes of birds with a large number of microchromosomes.
Collapse
Affiliation(s)
- A. Maslova
- grid.15447.330000 0001 2289 6897Saint Petersburg State University, Saint Petersburg, Russia
| | - V. Plotnikov
- grid.15447.330000 0001 2289 6897Saint Petersburg State University, Saint Petersburg, Russia
| | - M. Nuriddinov
- grid.418953.2Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | - M. Gridina
- grid.418953.2Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | - V. Fishman
- grid.418953.2Institute of Cytology and Genetics SB RAS, Novosibirsk, Russia
| | - A. Krasikova
- grid.15447.330000 0001 2289 6897Saint Petersburg State University, Saint Petersburg, Russia
| |
Collapse
|
15
|
Feldman TP, Egan ES. Uncovering a Cryptic Site of Malaria Pathogenesis: Models to Study Interactions Between Plasmodium and the Bone Marrow. Front Cell Infect Microbiol 2022; 12:917267. [PMID: 35719356 PMCID: PMC9201243 DOI: 10.3389/fcimb.2022.917267] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/03/2022] [Indexed: 12/21/2022] Open
Abstract
The bone marrow is a critical site of host-pathogen interactions in malaria infection. The discovery of Plasmodium asexual and transmission stages in the bone marrow has renewed interest in the tissue as a niche for cellular development of both host and parasite. Despite its importance, bone marrow in malaria infection remains largely unexplored due to the challenge of modeling the complex hematopoietic environment in vitro. Advancements in modeling human erythropoiesis ex-vivo from primary human hematopoietic stem and progenitor cells provide a foothold to study the host-parasite interactions occurring in this understudied site of malaria pathogenesis. This review focuses on current in vitro methods to recapitulate and assess bone marrow erythropoiesis and their potential applications in the malaria field. We summarize recent studies that leveraged ex-vivo erythropoiesis to shed light on gametocyte development in nucleated erythroid stem cells and begin to characterize host cell responses to Plasmodium infection in the hematopoietic niche. Such models hold potential to elucidate mechanisms of disordered erythropoiesis, an underlying contributor to malaria anemia, as well as understand the biological determinants of parasite sexual conversion. This review compares the advantages and limitations of the ex-vivo erythropoiesis approach with those of in vivo human and animal studies of the hematopoietic niche in malaria infection. We highlight the need for studies that apply single cell analyses to this complex system and incorporate physical and cellular components of the bone marrow that may influence erythropoiesis and parasite development.
Collapse
Affiliation(s)
- Tamar P. Feldman
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, United States
| | - Elizabeth S. Egan
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, United States
- *Correspondence: Elizabeth S. Egan,
| |
Collapse
|
16
|
Feng Y, Borosha S, Ratri A, Lee EB, Wang H, Fields TA, Kinsey WH, Vivian JL, Rumi MAK, Fields PE. DOT1L Methyltransferase Regulates Calcium Influx in Erythroid Progenitor Cells in Response to Erythropoietin. Int J Mol Sci 2022; 23:5137. [PMID: 35563527 PMCID: PMC9099724 DOI: 10.3390/ijms23095137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/02/2022] [Accepted: 05/03/2022] [Indexed: 02/01/2023] Open
Abstract
Erythropoietin (EPO) signaling plays a vital role in erythropoiesis by regulating proliferation and lineage-specific differentiation of murine hematopoietic progenitor cells (HPCs). An important downstream response of EPO signaling is calcium (Ca2+) influx, which is regulated by transient receptor potential channel (TRPC) proteins, particularly TRPC2 and TRPC6. While EPO induces Ca2+ influx through TRPC2, TRPC6 inhibits the function of TRPC2. Thus, interactions between TRPC2 and TRPC6 regulate the rate of Ca2+ influx in EPO-induced erythropoiesis. In this study, we observed that the expression of TRPC6 in KIT-positive erythroid progenitor cells was regulated by DOT1L. DOT1L is a methyltransferase that plays an important role in many biological processes during embryonic development including early erythropoiesis. We previously reported that Dot1l knockout (Dot1lKO) HPCs in the yolk sac failed to develop properly, which resulted in lethal anemia. In this study, we detected a marked downregulation of Trpc6 gene expression in Dot1lKO progenitor cells in the yolk sac compared to the wild type (WT). The promoter and the proximal regions of the Trpc6 gene locus exhibited an enrichment of H3K79 methylation, which is mediated solely by DOT1L. However, the expression of Trpc2, the positive regulator of Ca2+ influx, remained unchanged, resulting in an increased TRPC2/TRPC6 ratio. As the loss of DOT1L decreased TRPC6, which inhibited Ca2+ influx by TRPC2, Dot1lKO HPCs in the yolk sac exhibited accelerated and sustained elevated levels of Ca2+ influx. Such heightened Ca2+ levels might have detrimental effects on the growth and proliferation of HPCs in response to EPO.
Collapse
Affiliation(s)
- Yi Feng
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (Y.F.); (S.B.); (A.R.); (E.B.L.); (T.A.F.); (J.L.V.); (M.A.K.R.)
| | - Shaon Borosha
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (Y.F.); (S.B.); (A.R.); (E.B.L.); (T.A.F.); (J.L.V.); (M.A.K.R.)
| | - Anamika Ratri
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (Y.F.); (S.B.); (A.R.); (E.B.L.); (T.A.F.); (J.L.V.); (M.A.K.R.)
| | - Eun Bee Lee
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (Y.F.); (S.B.); (A.R.); (E.B.L.); (T.A.F.); (J.L.V.); (M.A.K.R.)
| | - Huizhen Wang
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.W.); (W.H.K.)
| | - Timothy A. Fields
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (Y.F.); (S.B.); (A.R.); (E.B.L.); (T.A.F.); (J.L.V.); (M.A.K.R.)
| | - William H. Kinsey
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA; (H.W.); (W.H.K.)
| | - Jay L. Vivian
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (Y.F.); (S.B.); (A.R.); (E.B.L.); (T.A.F.); (J.L.V.); (M.A.K.R.)
| | - M. A. Karim Rumi
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (Y.F.); (S.B.); (A.R.); (E.B.L.); (T.A.F.); (J.L.V.); (M.A.K.R.)
| | - Patrick E. Fields
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (Y.F.); (S.B.); (A.R.); (E.B.L.); (T.A.F.); (J.L.V.); (M.A.K.R.)
| |
Collapse
|
17
|
Medina S, Bolt AM, Zhou X, Wan G, Xu H, Lauer FT, Liu KJ, Burchiel SW. Arsenite and monomethylarsonous acid disrupt erythropoiesis through combined effects on differentiation and survival pathways in early erythroid progenitors. Toxicol Lett 2021; 350:111-120. [PMID: 34274428 PMCID: PMC8487637 DOI: 10.1016/j.toxlet.2021.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/23/2021] [Accepted: 07/12/2021] [Indexed: 10/20/2022]
Abstract
Strong epidemiological evidence demonstrates an association between chronic arsenic exposure and anemia. We recently found that As+3 impairs erythropoiesis by disrupting the function of GATA-1; however the downstream pathways impacted by the loss of GATA-1 function have not been evaluated. Additionally, our previous findings indicate that the predominant arsenical in the bone marrow of mice exposed to As+3 in their drinking water for 30 days was MMA+3, but the impacts of this arsenical on erythorpoisis also remain largely unknown. The goal of this study was to address these critical knowledge gaps by evaluating the comparative effects of arsenite (As+3) and the As+3 metabolite, monomethyarsonous acid (MMA+3) on two critical regulatory pathways that control the differentiation and survival of early erythroid progenitor cells. We found that 500 nM As+3 and 100 and 500 nM MMA+3 suppress erythropoiesis by impairing the differentiation of early stage erythroid progenitors. The suppression of early erythroid progenitor cell development was attributed to combined effects on differentiation and survival pathways mediated by disruption of GATA-1 and STAT5. Our results show that As+3 primarily disrupted GATA-1 function; whereas, MMA+3 suppressed both GATA-1 and STAT5 activity. Collectively, these findings provide novel mechanistic insights into arsenic-induced dyserythropoiesis and suggest that MMA+3 may be more toxic than As+3 to early developing erythroid cells.
Collapse
Affiliation(s)
- Sebastian Medina
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, Albuquerque, NM, 87131, USA; New Mexico Highlands University, Department of Biology, Las Vegas, NM, 87701, USA
| | - Alicia M Bolt
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, Albuquerque, NM, 87131, USA
| | - Xixi Zhou
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, Albuquerque, NM, 87131, USA
| | - Guanghua Wan
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, Albuquerque, NM, 87131, USA
| | - Huan Xu
- East China University of Science and Technology, School of Pharmacy, Shanghai, 200237, China
| | - Fredine T Lauer
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, Albuquerque, NM, 87131, USA
| | - Ke Jian Liu
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, Albuquerque, NM, 87131, USA
| | - Scott W Burchiel
- The University of New Mexico College of Pharmacy, Department of Pharmaceutical Sciences, Albuquerque, NM, 87131, USA.
| |
Collapse
|
18
|
Tsiftsoglou AS. Erythropoietin (EPO) as a Key Regulator of Erythropoiesis, Bone Remodeling and Endothelial Transdifferentiation of Multipotent Mesenchymal Stem Cells (MSCs): Implications in Regenerative Medicine. Cells 2021; 10:cells10082140. [PMID: 34440909 PMCID: PMC8391952 DOI: 10.3390/cells10082140] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/15/2021] [Accepted: 08/17/2021] [Indexed: 02/06/2023] Open
Abstract
Human erythropoietin (EPO) is an N-linked glycoprotein consisting of 166 aa that is produced in the kidney during the adult life and acts both as a peptide hormone and hematopoietic growth factor (HGF), stimulating bone marrow erythropoiesis. EPO production is activated by hypoxia and is regulated via an oxygen-sensitive feedback loop. EPO acts via its homodimeric erythropoietin receptor (EPO-R) that increases cell survival and drives the terminal erythroid maturation of progenitors BFU-Es and CFU-Es to billions of mature RBCs. This pathway involves the activation of multiple erythroid transcription factors, such as GATA1, FOG1, TAL-1, EKLF and BCL11A, and leads to the overexpression of genes encoding enzymes involved in heme biosynthesis and the production of hemoglobin. The detection of a heterodimeric complex of EPO-R (consisting of one EPO-R chain and the CSF2RB β-chain, CD131) in several tissues (brain, heart, skeletal muscle) explains the EPO pleotropic action as a protection factor for several cells, including the multipotent MSCs as well as cells modulating the innate and adaptive immunity arms. EPO induces the osteogenic and endothelial transdifferentiation of the multipotent MSCs via the activation of EPO-R signaling pathways, leading to bone remodeling, induction of angiogenesis and secretion of a large number of trophic factors (secretome). These diversely unique properties of EPO, taken together with its clinical use to treat anemias associated with chronic renal failure and other blood disorders, make it a valuable biologic agent in regenerative medicine for the treatment/cure of tissue de-regeneration disorders.
Collapse
Affiliation(s)
- Asterios S Tsiftsoglou
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
19
|
Kim HJ, Park JW, Kang JY, Seo SB. Negative Regulation of Erythroid Differentiation via the CBX8-TRIM28 Axis. Mol Cells 2021; 44:444-457. [PMID: 34253692 PMCID: PMC8334346 DOI: 10.14348/molcells.2021.0012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/25/2021] [Accepted: 04/29/2021] [Indexed: 12/26/2022] Open
Abstract
Although the mechanism of chronic myeloid leukemia (CML) initiation through BCR/ABL oncogene has been well characterized, CML cell differentiation into erythroid lineage cells remains poorly understood. Using CRISPR-Cas9 screening, we identify Chromobox 8 (CBX8) as a negative regulator of K562 cell differentiation into erythrocytes. CBX8 is degraded via proteasomal pathway during K562 cell differentiation, which activates the expression of erythroid differentiation-related genes that are repressed by CBX8 in the complex of PRC1. During the differentiation process, the serine/threonine-protein kinase PIM1 phosphorylates serine 196 on CBX8, which contributes to CBX8 reduction. When CD235A expression levels are analyzed, the result reveals that the knockdown of PIM1 inhibits K562 cell differentiation. We also identify TRIM28 as another interaction partner of CBX8 by proteomic analysis. Intriguingly, TRIM28 maintains protein stability of CBX8 and TRIM28 loss significantly induces proteasomal degradation of CBX8, resulting in an acceleration of erythroid differentiation. Here, we demonstrate the involvement of the CBX8-TRIM28 axis during CML cell differentiation, suggesting that CBX8 and TRIM28 are promising novel targets for CML research.
Collapse
Affiliation(s)
- Hyun Jeong Kim
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul 06974, Korea
| | - Jin Woo Park
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul 06974, Korea
| | - Joo-Young Kang
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul 06974, Korea
| | - Sang-Beom Seo
- Department of Life Science, College of Natural Sciences, Chung-Ang University, Seoul 06974, Korea
| |
Collapse
|
20
|
Li M, Sun X, Yao H, Chen W, Zhang F, Gao S, Zou X, Chen J, Qiu S, Wei H, Hu Z, Chen W. Genomic methylation variations predict the susceptibility of six chemotherapy related adverse effects and cancer development for Chinese colorectal cancer patients. Toxicol Appl Pharmacol 2021; 427:115657. [PMID: 34332992 DOI: 10.1016/j.taap.2021.115657] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/08/2021] [Accepted: 07/26/2021] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) remains a major concern with high morbidity and mortality worldwide. Despite the positive influence of chemotherapy on the decline in CRC mortality, the negative influence of chemotherapy-related adverse effects (CRAEs) caused by capecitabine (Cap) remains a challenging problem. DNA methylation alteration plays a pivotal role in gene expression regulation. Here, we aimed to screen reliable and novel biomarkers for CRC diagnosis and CRAE prediction using the advanced Illumina Infinium MethylationEPIC (850 K) BeadChip. Paired tumor and normal tissues from 21 Chinese CRC patients who received Cap-based adjuvant chemotherapy were analyzed. CRC-related methylation was characterized by hypermethylated promoter islands and hypomethylated intragenic openseas; CRAE-related methylation was characterized by hyper- (or hypo-) methylated intragenic (or intergenic) regions. Based on three types of methylation profiles (differentially methylated probes, differentially methylated regions, and gene-function-differentially methylated regions), pathway enrichment analyses revealed that CRC-related genes were significantly enriched in the neuronal system, metabolism of RNA, and extracellular matrix organization; CRAE-related genes were abundantly enriched in pathways controlling regeneration functions and immune response. Finally, based on genes within the mostly related pathways and LASSO logistic regression selection, the integrated-methylation-marker systems developed here demonstrated high discriminative accuracy in both CRC diagnosis (AUROC = 1) and CRAE prediction (AUROC = 0.817-1). In conclusion, we conducted a comprehensive DNA methylation analysis of CRC patients with chemotherapy, which provided new insights into the formation of CRC and CRAEs. Most importantly, our findings identified potentially CRAE-related metabolic pathways and markers, providing a valuable reference for personalized medicine promising better safety. Trail registration:ClinicalTrials.gov,NCT03030508, Registered 25 January 2017,https://www.clinicaltrials.gov/ct2/show/NCT03030508?term=NCT03030508&draw=2&rank=1.
Collapse
Affiliation(s)
- Mingming Li
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Xiaomeng Sun
- Research Institute, GloriousMed Clinical Laboratory Co., Ltd., Shanghai 201318, China
| | - Houshan Yao
- Department of General Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Wei Chen
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Feng Zhang
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Shouhong Gao
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Xun Zou
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Jiani Chen
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Shi Qiu
- Traditional Chinese Medicine Resource and Technology Center, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hua Wei
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China; Department of Pharmacy, 905th Hospital of PLA Navy, Naval Medical University, Shanghai 200052, China.
| | - Zhiqian Hu
- Department of General Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China; Department of Gastrointestinal Surgery, Tongji Hospital, Medical College of Tongji University, Shanghai 200065, China.
| | - Wansheng Chen
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China; Traditional Chinese Medicine Resource and Technology Center, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
21
|
Shan M, Ji X, Janssen K, Silverman IM, Humenik J, Garcia BA, Liebhaber SA, Gregory BD. Dynamic changes in RNA-protein interactions and RNA secondary structure in mammalian erythropoiesis. Life Sci Alliance 2021; 4:4/9/e202000659. [PMID: 34315813 PMCID: PMC8321672 DOI: 10.26508/lsa.202000659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 11/24/2022] Open
Abstract
Two features of eukaryotic RNA molecules that regulate their post-transcriptional fates are RNA secondary structure and RNA-binding protein (RBP) interaction sites. However, a comprehensive global overview of the dynamic nature of these sequence features during erythropoiesis has never been obtained. Here, we use our ribonuclease-mediated structure and RBP-binding site mapping approach to reveal the global landscape of RNA secondary structure and RBP-RNA interaction sites and the dynamics of these features during this important developmental process. We identify dynamic patterns of RNA secondary structure and RBP binding throughout the process and determine a set of corresponding protein-bound sequence motifs along with their dynamic structural and RBP-binding contexts. Finally, using these dynamically bound sequences, we identify a number of RBPs that have known and putative key functions in post-transcriptional regulation during mammalian erythropoiesis. In total, this global analysis reveals new post-transcriptional regulators of mammalian blood cell development.
Collapse
Affiliation(s)
- Mengge Shan
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA.,Genomics and Computational Biology Graduate Group, Perelman School of Medicine, Epigenetics Institute, University of Pennsylvania, PA, USA
| | - Xinjun Ji
- Department of Genetics, Perelman School of Medicine, Epigenetics Institute, University of Pennsylvania, PA, USA
| | - Kevin Janssen
- Department of Biochemistry and Biophysics, Perelman School of Medicine, Epigenetics Institute, University of Pennsylvania, PA, USA
| | - Ian M Silverman
- Department of Genetics, Perelman School of Medicine, Epigenetics Institute, University of Pennsylvania, PA, USA
| | - Jesse Humenik
- Department of Genetics, Perelman School of Medicine, Epigenetics Institute, University of Pennsylvania, PA, USA
| | - Ben A Garcia
- Department of Biochemistry and Biophysics, Perelman School of Medicine, Epigenetics Institute, University of Pennsylvania, PA, USA
| | - Stephen A Liebhaber
- Department of Genetics, Perelman School of Medicine, Epigenetics Institute, University of Pennsylvania, PA, USA .,Department of Medicine, Perelman School of Medicine, Epigenetics Institute, University of Pennsylvania, PA, USA
| | - Brian D Gregory
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA .,Genomics and Computational Biology Graduate Group, Perelman School of Medicine, Epigenetics Institute, University of Pennsylvania, PA, USA
| |
Collapse
|
22
|
Expression Silencing of Glutathione Peroxidase 4 in Mouse Erythroleukemia Cells Delays In Vitro Erythropoiesis. Int J Mol Sci 2021; 22:ijms22157795. [PMID: 34360557 PMCID: PMC8345999 DOI: 10.3390/ijms22157795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/20/2021] [Accepted: 07/20/2021] [Indexed: 01/31/2023] Open
Abstract
Among the eight human glutathione peroxidase isoforms, glutathione peroxidase 4 (GPX4) is the only enzyme capable of reducing complex lipid peroxides to the corresponding alcohols. In mice, corruption of the Gpx4 gene leads to embryonic lethality and more detailed expression silencing studies have implicated the enzyme in several physiological processes (e.g., embryonal cerebrogenesis, neuronal function, male fertility). Experiments with conditional knockout mice, in which expression of the Gpx4 gene was silenced in erythroid precursors, indicated a role of Gpx4 in erythropoiesis. To test this hypothesis in a cellular in vitro model we transfected mouse erythroleukemia cells with a Gpx4 siRNA construct and followed the expression kinetics of erythropoietic gene products. Our data indicate that Gpx4 is expressed at high levels in mouse erythroleukemia cells and that expression silencing of the Gpx4 gene delays in vitro erythropoiesis. However, heterozygous expression of a catalytically inactive Gpx4 mutant (Gpx4+/Sec46Ala) did not induce a defective erythropoietic phenotype in different in vivo and ex vivo models. These data suggest that Gpx4 plays a role in erythroid differentiation of mouse erythroleukemia cells but that heterozygous expression of a catalytically inactive Gpx4 is not sufficient to compromise in vivo and ex vivo erythropoiesis.
Collapse
|
23
|
miR-16-5p Promotes Erythroid Maturation of Erythroleukemia Cells by Regulating Ribosome Biogenesis. Pharmaceuticals (Basel) 2021; 14:ph14020137. [PMID: 33572085 PMCID: PMC7915806 DOI: 10.3390/ph14020137] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/05/2021] [Accepted: 02/05/2021] [Indexed: 12/20/2022] Open
Abstract
miRNAs constitute a class of non-coding RNA that act as powerful epigenetic regulators in animal and plant cells. In order to identify putative tumor-suppressor miRNAs we profiled the expression of various miRNAs during differentiation of erythroleukemia cells. RNA was purified before and after differentiation induction and subjected to quantitative RT-PCR. The majority of the miRNAs tested were found upregulated in differentiated cells with miR-16-5p showing the most significant increase. Functional studies using gain- and loss-of-function constructs proposed that miR-16-5p has a role in promoting the erythroid differentiation program of murine erythroleukemia (MEL) cells. In order to identify the underlying mechanism of action, we utilized bioinformatic in-silico platforms that incorporate predictions for the genes targeted by miR-16-5p. Interestingly, ribosome constituents, as well as ribosome biogenesis factors, were overrepresented among the miR-16-5p predicted gene targets. Accordingly, biochemical experiments showed that, indeed, miR-16-5p could modulate the levels of independent ribosomal proteins, and the overall ribosomal levels in cultured cells. In conclusion, miR-16-5p is identified as a differentiation-promoting agent in erythroleukemia cells, demonstrating antiproliferative activity, likely as a result of its ability to target the ribosomal machinery and restore any imbalanced activity imposed by the malignancy and the blockade of differentiation.
Collapse
|
24
|
The histone methyltransferase inhibitor A-366 enhances hemoglobin expression in erythroleukemia cells upon co-exposure with chemical inducers in culture. ACTA ACUST UNITED AC 2021; 28:2. [PMID: 33407944 PMCID: PMC7788816 DOI: 10.1186/s40709-020-00132-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/20/2020] [Accepted: 12/23/2020] [Indexed: 01/02/2023]
Abstract
Background Erythroleukemia is caused by the uncontrolled multiplication of immature erythroid progenitor cells which fail to differentiate into erythrocytes. By directly targeting this class of malignant cells, the induction of terminal erythroid differentiation represents a vital therapeutic strategy for this disease. Erythroid differentiation involves the execution of a well-orchestrated gene expression program in which epigenetic enzymes play critical roles. In order to identify novel epigenetic mediators of differentiation, this study explores the effects of multiple, highly specific, epigenetic enzyme inhibitors, in murine and human erythroleukemia cell lines. Results We used a group of compounds designed to uniquely target the following epigenetic enzymes: G9a/GLP, EZH1/2, SMYD2, PRMT3, WDR5, SETD7, SUV420H1 and DOT1L. The majority of the probes had a negative impact on both cell proliferation and differentiation. On the contrary, one of the compounds, A-366, demonstrated the opposite effect by promoting erythroid differentiation of both cell models. A-366 is a selective inhibitor of the G9a methyltransferase and the chromatin reader Spindlin1. Investigation of the molecular mechanism of action revealed that A-366 forced cells to exit from the cell cycle, a fact that favored erythroid differentiation. Further analysis led to the identification of a group of genes that mediate the A-366 effects and include CDK2, CDK4 and CDK6. Conclusions A-366, a selective inhibitor of G9a and Spindlin1, demonstrates a compelling role in the erythroid maturation process by promoting differentiation, a fact that is highly beneficial for patients suffering from erythroleukemia. In conclusion, this data calls for further investigation towards the delivery of epigenetic drugs and especially A-366 in hematopoietic disorders.
Collapse
|
25
|
Hirano T, Tsuruda T, Tanaka Y, Harada H, Yamazaki T, Ishida A. Long noncoding RNA CCDC26 as a modulator of transcriptional switching between fetal and embryonic globins. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1868:118931. [PMID: 33340546 DOI: 10.1016/j.bbamcr.2020.118931] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/04/2020] [Accepted: 12/11/2020] [Indexed: 01/04/2023]
Abstract
The CCDC26 gene is considered to encode a functional noncoding RNA associated with acute myeloid leukemia and other cancers. However, investigations into the physiological roles of CCDC26 are rare. Previously, we reported that CCDC26 regulated proliferation and cell death of leukemia cells through KIT, a receptor tyrosine kinase, by using K562 leukemia cells and their derivative CCDC26-knockdown (KD) cells. Here we propose a new role of CCDC26 in the differentiation of erythroid cells. We showed that expression of embryonic (ε- and ζ-) globins was markedly upregulated in CCDC26-KD cells compared with K562 control cells during hemin-induced differentiation. In contrast, expression of fetal-type γ-globin, a major globin expressed in original K562 cells, was decreased. These changes in the expression of globin genes mainly took place at the transcriptional level, with significant suppression of transcription of adult (β-, δ-) globins in CCDC26-KD cells. Re-introduction of exogenous CCDC26 into the CCDC26-KD cells recovered low-level expression of the embryonal globins. These results suggest CCDC26 has a role in switching transcription of globin genes in the differentiation of erythroid cells. The expression profile of the CCDC26-KD cells and control cells suggests FOG-2, a transcriptional modulator, as a candidate for a mediator of the CCDC26-associated regulation. We showed that both embryonic globins were transcriptionally activated in FOG-2-KD K562 cells. The KIT inhibitor ISCK03 suppressed the production of hemoglobin in K562 cells but did not affect transcription of globin genes. To summarize, FOG-2, but not KIT, is responsible for globin transcriptional regulation by CCDC26.
Collapse
Affiliation(s)
- Tetsuo Hirano
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-7-1 Kagamiyama, Higashihiroshima, Hiroshima 739-8521, Japan.
| | - Tomomi Tsuruda
- School of Integrated Arts and Sciences, Hiroshima University, 1-7-1 Kagamiyama, Higashihiroshima, Hiroshima 739-8521, Japan
| | - Yuka Tanaka
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-7-1 Kagamiyama, Higashihiroshima, Hiroshima 739-8521, Japan
| | - Hironori Harada
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan.
| | - Takeshi Yamazaki
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-7-1 Kagamiyama, Higashihiroshima, Hiroshima 739-8521, Japan.
| | - Atsuhiko Ishida
- Graduate School of Integrated Sciences for Life, Hiroshima University, 1-7-1 Kagamiyama, Higashihiroshima, Hiroshima 739-8521, Japan.
| |
Collapse
|
26
|
Ebrahimi M, Forouzesh M, Raoufi S, Ramazii M, Ghaedrahmati F, Farzaneh M. Differentiation of human induced pluripotent stem cells into erythroid cells. Stem Cell Res Ther 2020; 11:483. [PMID: 33198819 PMCID: PMC7667818 DOI: 10.1186/s13287-020-01998-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 10/25/2020] [Indexed: 02/07/2023] Open
Abstract
During the last years, several strategies have been made to obtain mature erythrocytes or red blood cells (RBC) from the bone marrow or umbilical cord blood (UCB). However, UCB-derived hematopoietic stem cells (HSC) are a limited source and in vitro large-scale expansion of RBC from HSC remains problematic. One promising alternative can be human pluripotent stem cells (PSCs) that provide an unlimited source of cells. Human PSCs, including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), are self-renewing progenitors that can be differentiated to lineages of ectoderm, mesoderm, and endoderm. Several previous studies have revealed that human ESCs can differentiate into functional oxygen-carrying erythrocytes; however, the ex vivo expansion of human ESC-derived RBC is subjected to ethical concerns. Human iPSCs can be a suitable therapeutic choice for the in vitro/ex vivo manufacture of RBCs. Reprogramming of human somatic cells through the ectopic expression of the transcription factors (OCT4, SOX2, KLF4, c-MYC, LIN28, and NANOG) has provided a new avenue for disease modeling and regenerative medicine. Various techniques have been developed to generate enucleated RBCs from human iPSCs. The in vitro production of human iPSC-derived RBCs can be an alternative treatment option for patients with blood disorders. In this review, we focused on the generation of human iPSC-derived erythrocytes to present an overview of the current status and applications of this field.
Collapse
Affiliation(s)
- Mohsen Ebrahimi
- Neonatal and Children's Health Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mehdi Forouzesh
- Legal Medicine Organization of Iran, Legal Medicine Research Center, Legal Medicine organization, Tehran, Iran
| | - Setareh Raoufi
- Faculty of Medical Sciences and Technologies, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Ramazii
- Kerman University of Medical Sciences, University of Kerman, Kerman, Iran
| | - Farhoodeh Ghaedrahmati
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Farzaneh
- Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
27
|
Deng Y, Yao H, Chen W, Wei H, Li X, Zhang F, Gao S, Man H, Chen J, Tao X, Li M, Chen W. Profiling of polar urine metabolite extracts from Chinese colorectal cancer patients to screen for potential diagnostic and adverse-effect biomarkers. J Cancer 2020; 11:6925-6938. [PMID: 33123283 PMCID: PMC7592006 DOI: 10.7150/jca.47631] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/27/2020] [Indexed: 02/07/2023] Open
Abstract
Background: Metabolomics has demonstrated its potential in the early diagnosis, drug safety evaluation and personalized toxicology research of various cancers. Objectives: We aim to screen for potential diagnostic and capecitabine-related adverse effect (CRAE) biomarkers from urinary endogenous metabolites in Chinese colorectal cancer (CRC) patients. Methods: The metabolic profiles of 139 CRC patients and 50 non-neoplastic controls were analyzed using ultra-high-performance liquid chromatography combined with quadrupole time-of-flight mass spectrometry. Results: There were 41 metabolites identified between the CRC patients and the non-neoplastic controls, and 19 metabolites were identified between CRC patients with and without CRAE. Based on these identified metabolites, bioinformatic analysis and prediction model construction were completed. Most of these differential metabolites have important roles in cell proliferation and differentiation and the immune system. Based on binary logistic regression, a CRC prediction model, composed of 3-methylhistidine, N-heptanoylglycine, N1,N12-diacetylspermine and hippurate, was established, with an area under curve (AUC) of 0.980 (95% CI: 0.953-1.000; sensitivity: 94.3%; specificity: 92.0%) in the training set, and an AUC of 0.968 (95% CI: 0.933-1.000; sensitivity: 89.9%; specificity: 92.0%) in the testing set. In addition, methionine and 4-pyridoxic acid can be combined to predict hand foot syndrome, with an AUC of 0.884; ubiquinone-1 and 4-pyridoxic acid can be combined to predict anemia, with an AUC of 0.889; and 5-acetamidovalerate and 3,4-methylenesebacic acid can be combined to predict neutropenia, with an AUC of 0.882. Conclusion: The profiling of urine polar metabolites has great potential in the early detection of CRC and the prediction of CRAE.
Collapse
Affiliation(s)
- Yi Deng
- Department of Pharmacy, Changzheng Hospital, Secondary Military Medical University, Shanghai, China, 200003
| | - Houshan Yao
- Department of Surgery, Changzheng Hospital, Secondary Military Medical University, Shanghai, China, 200003
| | - Wei Chen
- Department of Pharmacy, Changzheng Hospital, Secondary Military Medical University, Shanghai, China, 200003
| | - Hua Wei
- Department of Pharmacy, Changzheng Hospital, Secondary Military Medical University, Shanghai, China, 200003
| | - Xinxing Li
- Department of Surgery, Changzheng Hospital, Secondary Military Medical University, Shanghai, China, 200003
| | - Feng Zhang
- Department of Pharmacy, Changzheng Hospital, Secondary Military Medical University, Shanghai, China, 200003
| | - Shouhong Gao
- Department of Pharmacy, Changzheng Hospital, Secondary Military Medical University, Shanghai, China, 200003
| | - Huan Man
- Department of Pharmacy, Changzheng Hospital, Secondary Military Medical University, Shanghai, China, 200003
- College of Chemical and Biological Engineering, Yichun University, Jiangxi Province, China, 336000
| | - Jing Chen
- Department of Pharmacy, Changzheng Hospital, Secondary Military Medical University, Shanghai, China, 200003
- College of Chemical and Biological Engineering, Yichun University, Jiangxi Province, China, 336000
| | - Xia Tao
- Department of Pharmacy, Changzheng Hospital, Secondary Military Medical University, Shanghai, China, 200003
| | - Mingming Li
- Department of Pharmacy, Changzheng Hospital, Secondary Military Medical University, Shanghai, China, 200003
| | - Wansheng Chen
- Department of Pharmacy, Changzheng Hospital, Secondary Military Medical University, Shanghai, China, 200003
- Research and Development Center of Chinese Medicine Resources and Biotechnology, Shanghai University of Traditional Chinese Medicine, Shanghai, China, 201203
| |
Collapse
|
28
|
Tougan T, Itagaki S, Toya Y, Uchihashi K, Horii T. Implementation of a red blood cell-optical (RBO) channel for detection of latent iron deficiency anaemia by automated measurement of autofluorescence-emitting red blood cells. Sci Rep 2020; 10:15605. [PMID: 32973246 PMCID: PMC7518259 DOI: 10.1038/s41598-020-72382-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 06/15/2020] [Indexed: 11/30/2022] Open
Abstract
Iron deficiency is the most common and widespread nutritional disorder worldwide. The automated haematology analyser XN-30 (Sysmex, Kobe, Japan) was developed to detect malaria-infected red blood cells (RBCs) in human blood samples using flow cytometry. The optical system of the analyser detects autofluorescence (AF)-emitting RBCs containing iron-deficient haem groups and would aid in the diagnosis of anaemia resulting from iron deficiency. Here, an RBC-optical (RBO) channel was devised and implemented on the analyser. In vitro analyses showed that the analyser detected AF-emitting RBCs treated with 5-aminolevulinic acid. Furthermore, the analyser detected AF-emitting RBCs in mice fed a low iron diet and infected with a rodent malaria parasite; it could also be effectively used in humans. This study demonstrates that the analyser can quantitatively and reproducibly detect AF-emitting RBCs and measure other haematological parameters, suggesting its usefulness for the initial evaluation of latent iron deficiency anaemia in conjunction with the diagnosis of malaria.
Collapse
Affiliation(s)
- Takahiro Tougan
- Research Centre for Infectious Disease Control, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Sawako Itagaki
- Department of Malaria Vaccine Development, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yuji Toya
- Sysmex Corporation, 4-4-4 Takatsukadai Nishiku, Kobe, Hyogo, 651-2271, Japan
| | - Kinya Uchihashi
- Sysmex Corporation, 4-4-4 Takatsukadai Nishiku, Kobe, Hyogo, 651-2271, Japan
| | - Toshihiro Horii
- Department of Malaria Vaccine Development, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
29
|
A Unique Epigenomic Landscape Defines Human Erythropoiesis. Cell Rep 2020; 28:2996-3009.e7. [PMID: 31509757 PMCID: PMC6863094 DOI: 10.1016/j.celrep.2019.08.020] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/28/2019] [Accepted: 08/02/2019] [Indexed: 12/15/2022] Open
Abstract
Mammalian erythropoiesis yields a highly specialized cell type, the mature erythrocyte, evolved to meet the organismal needs of increased oxygen-carrying capacity. To better understand the regulation of erythropoiesis, we performed genome-wide studies of chromatin accessibility, DNA methylation, and transcriptomics using a recently developed strategy to obtain highly purified populations of primary human erythroid cells. The integration of gene expression, DNA methylation, and chromatin state dynamics reveals that stage-specific gene regulation during erythropoiesis is a stepwise and hierarchical process involving many cis-regulatory elements. Erythroid-specific, nonpromoter sites of chromatin accessibility are linked to erythroid cell phenotypic variation and inherited disease. Comparative analyses of stage-specific chromatin accessibility indicate that there is limited early chromatin priming of erythroid genes during hematopoiesis. The epigenome of terminally differentiating erythroid cells defines a distinct subset of highly specialized cells that are vastly dissimilar from other hematopoietic and nonhematopoietic cell types. These epigenomic and transcriptome data are powerful tools to study human erythropoiesis. Schulz et al. use genome-wide studies of chromatin accessibility, DNA methylation, and transcriptomes in primary human erythroid cells to reveal important characteristics of erythropoiesis. Chromatin accessibility of terminal erythroid differentiation is markedly dissimilar from other hematopoietic cell types. Epigenomic changes are linked to erythroid cell traits and disease genes.
Collapse
|
30
|
Rademacher M, Kuhn H, Borchert A. Systemic deficiency of mouse arachidonate 15-lipoxygenase induces defective erythropoiesis and transgenic expression of the human enzyme rescues this phenotype. FASEB J 2020; 34:14318-14335. [PMID: 32918502 DOI: 10.1096/fj.202000408rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 07/29/2020] [Accepted: 08/07/2020] [Indexed: 02/06/2023]
Abstract
Arachidonic acid 15-lipoxygenases (ALOX15) are lipid peroxidizing enzymes, which has previously been implicated in the maturational breakdown of intracellular organelles and plasma membrane remodeling during reticulocyte-erythrocyte transition. Conventional Alox15-/- mice are viable, develop normally but do not exhibit a major defective erythropoietic phenotype. To characterize the putative in vivo relevance of Alox15 for red blood cell development, we explored the impact of systemic inactivation of the Alox15 gene on mouse erythropoiesis. We found that Alox15-/- mice exhibited reduced erythrocyte counts, elevated reticulocyte counts and red cell hyperchromia. The structure of the plasma membrane of Alox15-/- erythrocytes is altered and a significant share of the red cells was present as echinocytes and/or acanthocytes. An increased share of the Alox15-/- erythrocytes cells were annexin V positive, which indicates a loss of plasma membrane asymmetry. Erythrocytes of Alox15-/- mice were more susceptible to osmotic hemolysis and exhibited a reduced ex vivo life span. When we transgenically expressed human ALOX15 in Alox15-/- mice under the control of the aP2 promoter the defective erythropoietic system was rescued and the impaired osmotic resistance was normalized. Together these data suggest the involvement Alox15 in the maturational remodeling of the plasma membrane during red cell development.
Collapse
Affiliation(s)
- Marlena Rademacher
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Biochemistry, Berlin, Germany
| | - Hartmut Kuhn
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Biochemistry, Berlin, Germany
| | - Astrid Borchert
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Biochemistry, Berlin, Germany
| |
Collapse
|
31
|
Holshouser S, Cafiero R, Robinson M, Kirkpatrick J, Casero RA, Hyacinth HI, Woster PM. Epigenetic Reexpression of Hemoglobin F Using Reversible LSD1 Inhibitors: Potential Therapies for Sickle Cell Disease. ACS OMEGA 2020; 5:14750-14758. [PMID: 32596612 PMCID: PMC7315572 DOI: 10.1021/acsomega.0c01585] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 05/26/2020] [Indexed: 06/11/2023]
Abstract
Sickle cell disease (SCD) is caused by a single nucleotide polymorphism on chromosome 11 in the β-globin gene. The resulting mutant hemoglobin S (HbS) is a poor oxygen transporter and causes a variety of vascular symptoms and organ failures. At birth, the DRED epigenetic complex forms and silences the γ-globin gene, and fetal hemoglobin (HbF, 2 α-, and 2 γ-subunits) is replaced by adult HbA (α2β2) or HbS (α2βs 2) in SCD patients. HbF is a potent inhibitor of HbS polymerization, thus alleviating the symptoms of SCD. The current therapy, hydroxyurea (HU), increases γ-globin and the HbF content in sickle cells but is highly underutilized due to concern for adverse effects and other complications. The DRED complex contains the epigenetic eraser lysine-specific demethylase 1 (LSD1), which appears to serve as a scaffolding protein. Our recently discovered 1,2,4-triazole derivatives and cyclic peptide LSD1 inhibitors promote the upregulation of γ-globin production in vitro without significant toxicity. Herein, we demonstrate that these LSD1 inhibitors can be used to disrupt the DRED complex and increase the cellular HbF content in vitro and in vivo. This approach could lead to an innovative and effective treatment for SCD.
Collapse
Affiliation(s)
- Steven Holshouser
- Department
of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, 70 President St., Charleston, South Carolina 29414, United States
| | - Rebecca Cafiero
- Department
of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, 70 President St., Charleston, South Carolina 29414, United States
| | - Mayra Robinson
- Department
of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, 70 President St., Charleston, South Carolina 29414, United States
| | - Joy Kirkpatrick
- Department
of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, 70 President St., Charleston, South Carolina 29414, United States
| | - Robert A. Casero
- Sidney
Kimmel Comprehensive Cancer Center, Johns
Hopkins School of Medicine, 1650 Orleans St. Room 551, Baltimore, Maryland 21287, United States
| | - Hyacinth I. Hyacinth
- Department
of Pediatrics, School of Medicine, Emory
University, 2015 Uppergate Dr., Atlanta, Georgia 30322, United
States
| | - Patrick M. Woster
- Department
of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, 70 President St., Charleston, South Carolina 29414, United States
| |
Collapse
|
32
|
Huang S, Kong A, Cao Q, Tong Z, Wang X. The role of blood vessels in broiler chickens with tibial dyschondroplasia. Poult Sci 2020; 98:6527-6532. [PMID: 31433842 PMCID: PMC8913930 DOI: 10.3382/ps/pez497] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 08/07/2019] [Indexed: 12/11/2022] Open
Abstract
Tibial dyschondroplasia (TD) is an intractable tibiotarsal bone disorder of rapid growing avian species, which leads to huge economic losses and compromised poultry welfare. However, the exact pathogenesis and treatment of TD remain largely unknown. Based on continuous research findings, we propose the TD pathogenesis hypothesis: during skeletal development of TD chickens, due to the absence of vasculature of proximal tibial growth plates (TGP), hypertrophic chondrocytes of the TGP are unable to complete calcification in normal bone development and less dead chondrocytes in the corresponding area can be timely transported through the blood vessels. Moreover, recent studies demonstrate that the TD formation mechanism gradually tends to a large number of dead chondrocytes in the TGP region or apoptosis occur due to various factors (such as, reduction of vascular invasion and blood cells, and increased weight or mechanical force of the tibia), while the reduction of blood vessels is insufficient to remove these chondrocytes and eventually leads to the TD formation. Recognizing the possible role of the blood vessels in the incidence of TD and can propose that the improvement in vasculature might be a novel therapeutic approach for ending TD in chickens.
Collapse
Affiliation(s)
- Shucheng Huang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Anan Kong
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Qinqin Cao
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Zongxi Tong
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| | - Xuebing Wang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, China
| |
Collapse
|
33
|
Park KT, Han JK, Kim SJ, Lim YH. Gamma-Aminobutyric Acid Increases Erythropoietin by Activation of Citrate Cycle and Stimulation of Hypoxia-Inducible Factors Expression in Rats. Biomolecules 2020; 10:E595. [PMID: 32290638 PMCID: PMC7226430 DOI: 10.3390/biom10040595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/04/2020] [Accepted: 04/09/2020] [Indexed: 02/07/2023] Open
Abstract
Erythropoietin (EPO) is the primary regulator of erythropoiesis in the mammalian fetus and adult. Deficiency of EPO induces anemia. In this study, we investigated the effect of gamma-aminobutyric acid (GABA) on serum EPO levels and erythropoiesis in rats. Expression levels of Epo-related genes were measured by quantitative real-time PCR (qPCR) and expression of Epo and Epo receptor (Epor) proteins were measured by immunohistochemistry. The gene and protein expression profiles of kidney tissue in GABA-treated rats were evaluated by ribonucleic acid (RNA) sequencing and two-dimensional electrophoresis (2-DE), respectively. GABA significantly increased serum EPO levels and expression levels of Epo and Epor. GABA increased expression levels of hypoxia-inducible factor (Hif)-1 and Hif-2. Seven proteins with expression levels showing >2-fold change were identified by 2-DE followed by MALDI-TOF MS in GABA-treated rat kidney. The top KEGG pathway from the identified proteins was the tricarboxylic acid cycle, and nicotinamide adenine dinucleotide (NADH) dehydrogenase, succinate dehydrogenase, and isocitrate dehydrogenase were identified as key proteins. GABA treatment significantly increased ATP levels and NADH dehydrogenase activity in a dose-dependent manner. In conclusion, GABA shows a new physiological role in EPO production, and it can thus can contribute to the prevention of anemia when used alone or in combination with other anemia treating drugs.
Collapse
Affiliation(s)
- Keun-Tae Park
- Research and Development Center, Milae Bioresources Co., Ltd., Seoul 05836, Korea; (K.-T.P.); (J.-K.H.); (S.J.K.)
- Department of Integrated Biomedical and Life Sciences, College of Health Science, Korea University, Seoul 02841, Korea
| | - Jong-Kwon Han
- Research and Development Center, Milae Bioresources Co., Ltd., Seoul 05836, Korea; (K.-T.P.); (J.-K.H.); (S.J.K.)
| | - Seong Jin Kim
- Research and Development Center, Milae Bioresources Co., Ltd., Seoul 05836, Korea; (K.-T.P.); (J.-K.H.); (S.J.K.)
| | - Young-Hee Lim
- Department of Integrated Biomedical and Life Sciences, College of Health Science, Korea University, Seoul 02841, Korea
- Department of Public Health Science (BK21 PLUS Program), Graduate School, Korea University, Seoul 02841, Korea
- Department of Laboratory Medicine, Korea University Guro Hospital, Seoul 08308, Korea
| |
Collapse
|
34
|
Wang W, Zhao H, Yang Y, Chi Y, Lv X, Zhang L. Interferon-γ exerts dual functions on human erythropoiesis via interferon regulatory factor 1 signal pathway. Biochem Biophys Res Commun 2019; 521:326-332. [PMID: 31668371 DOI: 10.1016/j.bbrc.2019.10.068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/06/2019] [Accepted: 10/06/2019] [Indexed: 12/21/2022]
Abstract
Hematopoiesis is systematically regulated by microenvironmental factors. The positive and negative factors coordinated together to yield a complicated blood system. Interferon-γ (IFNγ) has been identified as a common cause of various hematopoietic abnormalities, such as aplastic anemia. However, its impact on monolineage development, especially erythropoiesis, has not been fully elucidated from the cellular angle. In this study, we investigated the behavior of IFNγ and found that IFNγ plays dual functions on erythropoiesis; it not only blocks the erythroid lineage commitment but also accelerates the erythroid differentiation process, ultimately leading to the erythropoietic window clearance. IFNγ can even powerfully initiate early differentiation without the existence of erythropoietin (EPO). Interferon regulatory factor 1 (IRF1) was confirmed as the essential downstream effector, and its ectopic overexpression can also have the same effect as that of IFNγ. These results reveal that the IFNγ-IRF1 axis plays a bidirectional role on erythropoiesis, impeding the access to erythroid lineage and driving the coming cells toward the differentiation endpoint. This model may place an innovative implication for IFNγ-IRF1 axis to understand its in-depth mechanism on normal hematopoiesis and abnormal blood disorders, especially aplastic anemia.
Collapse
Affiliation(s)
- Wentian Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Laboratory of Blood Disease Gene Therapy, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Huijuan Zhao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Laboratory of Blood Disease Gene Therapy, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China; Medical College, Henan University of Science and Technology, Luoyang, 471023, Henan, China
| | - Yang Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Laboratory of Blood Disease Gene Therapy, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Ying Chi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Laboratory of Blood Disease Gene Therapy, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Xiang Lv
- State Key Laboratory of Medical Molecular Biology, Department of Pathophysiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.
| | - Lei Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Tianjin Laboratory of Blood Disease Gene Therapy, CAMS Key Laboratory of Gene Therapy for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
| |
Collapse
|
35
|
Long noncoding RNA PCED1B-AS1 promotes erythroid differentiation coordinating with GATA1 and chromatin remodeling. BLOOD SCIENCE 2019; 1:161-167. [PMID: 35402806 PMCID: PMC8975080 DOI: 10.1097/bs9.0000000000000031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 08/23/2019] [Indexed: 12/24/2022] Open
Abstract
Erythropoiesis is a complex and sophisticated multi-stage process regulated by a variety of factors, including the transcription factor GATA1 and non-coding RNA. GATA1 is regarded as an essential transcriptional regulator promoting transcription of erythroid-specific genes—such as long non-coding RNAs (lncRNA). Here, we comprehensively screened lncRNAs that were potentially regulated by GATA1 in erythroid cells. We identified a novel lncRNA—PCED1B-AS1—and verified its role in promoting erythroid differentiation of K562 erythroid cells. We also predicted a model in which PCED1B-AS1 participates in erythroid differentiation via dynamic chromatin remodeling involving GATA1. The relationship between lncRNA and chromatin in the process of erythroid differentiation remains to be revealed, and in our study we have carried out preliminary explorations.
Collapse
|
36
|
Nimker S, Sharma K, Saraswathy R, Chandna S. Delineating the Effects of Ionizing Radiation on Erythropoietic Lineage-Implications for Radiation Biodosimetry. HEALTH PHYSICS 2019; 116:677-693. [PMID: 30720544 DOI: 10.1097/hp.0000000000000975] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The overall lethality/morbidity of ionizing radiation exposure involves multiple forms of inhibitory or cytotoxic effects that may manifest in different tissues with a varying dose and time response. One of the major systemic effects leading to lethality of radiation includes its suppressive effect on hematopoiesis, which could be observed even at doses as low as 1-2 Gy, whereas effects on gastrointestinal and nervous systems appear at relatively higher doses in the same order. This article reviews the effects of radiation on the three distinct stages of erythropoiesis-formation of erythroid progenitor cells, differentiation of erythroid precursor cells, and terminal maturation. During these stepwise developmental processes, erythroid progenitor cells undergo rapid expansion to form terminally differentiated red blood cells that are continuously replenished from bone marrow into the circulating peripheral blood stream. Cellular radiation response depends upon many factors such as cell lineage, rate of proliferation, and differentiation status. Therefore, we discuss radiation-induced alterations during the progenitor, precursor, and terminal maturation stages and the implications thereof. Since biomarkers of ionizing radiation exposure in human populations are of great interest for assessing normal tissue injury as well as for biodosimetry in the event of accidental or incidental radiation exposures, we also highlight blood-based biomarkers that have potential utility for medical management.
Collapse
Affiliation(s)
- Shwetanjali Nimker
- Division of Natural Radiation Response Mechanisms, Institute of Nuclear Medicine and Allied Sciences, Timarpur, Delhi, India
- School of Biosciences and Technology, Vellore Institiute of Technology, Vellore, Tamil Nadu, India
| | - Kanupriya Sharma
- Division of Natural Radiation Response Mechanisms, Institute of Nuclear Medicine and Allied Sciences, Timarpur, Delhi, India
| | - Radha Saraswathy
- School of Biosciences and Technology, Vellore Institiute of Technology, Vellore, Tamil Nadu, India
| | - Sudhir Chandna
- Division of Natural Radiation Response Mechanisms, Institute of Nuclear Medicine and Allied Sciences, Timarpur, Delhi, India
| |
Collapse
|
37
|
Thermal Reduction of Graphene Oxide Mitigates Its In Vivo Genotoxicity Toward Xenopus laevis Tadpoles. NANOMATERIALS 2019; 9:nano9040584. [PMID: 30970633 PMCID: PMC6523888 DOI: 10.3390/nano9040584] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 03/28/2019] [Accepted: 04/08/2019] [Indexed: 12/12/2022]
Abstract
The worldwide increase of graphene family materials raises the question of the potential consequences resulting from their release in the environment and future consequences on ecosystem health, especially in the aquatic environment in which they are likely to accumulate. Thus, there is a need to evaluate the biological and ecological risk but also to find innovative solutions leading to the production of safer materials. This work focuses on the evaluation of functional group-safety relationships regarding to graphene oxide (GO) in vivo genotoxic potential toward X. laevis tadpoles. For this purpose, thermal treatments in H2 atmosphere were applied to produce reduced graphene oxide (rGOs) with different surface group compositions. Analysis performed indicated that GO induced disturbances in erythrocyte cell cycle leading to accumulation of cells in G0/G1 phase. Significant genotoxicity due to oxidative stress was observed in larvae exposed to low GO concentration (0.1 mg·L−1). Reduction of GO at 200 °C and 1000 °C produced a material that was no longer genotoxic at low concentrations. X-ray photoelectron spectroscopy (XPS) analysis indicated that epoxide groups may constitute a good candidate to explain the genotoxic potential of the most oxidized form of the material. Thermal reduction of GO may constitute an appropriate “safer-by-design” strategy for the development of a safer material for environment.
Collapse
|
38
|
Yang C, Hashimoto M, Lin QXX, Tan DQ, Suda T. Sphingosine-1-phosphate signaling modulates terminal erythroid differentiation through the regulation of mitophagy. Exp Hematol 2019; 72:47-59.e1. [DOI: 10.1016/j.exphem.2019.01.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/10/2019] [Accepted: 01/11/2019] [Indexed: 01/17/2023]
|
39
|
Hirota K. An intimate crosstalk between iron homeostasis and oxygen metabolism regulated by the hypoxia-inducible factors (HIFs). Free Radic Biol Med 2019; 133:118-129. [PMID: 30053508 DOI: 10.1016/j.freeradbiomed.2018.07.018] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/21/2018] [Accepted: 07/23/2018] [Indexed: 12/24/2022]
Abstract
Oxygen and iron are among the most abundant elements and have significant roles in human biology. Iron is essential for oxygen transport and is a component of molecular O2-carrying proteins, such as hemoglobin and myoglobin. Iron is also a constituent of redox enzymes and can occupy multiple oxidation states. An elaborate system has evolved to stringently regulate the concentrations of both, free iron and oxygen, in various sites of the body. The final destination for iron and oxygen in the cells is the mitochondria. The mitochondria require substantial amounts of iron for heme synthesis and maturation of iron-sulfur clusters, and oxygen, as the electron acceptor in oxidative phosphorylation. Therefore, the balance between the control of iron availability and the physiology of hypoxic responses is critical for maintaining cell homeostasis. Several lines of study have clearly demonstrated that the transcription factors, hypoxia-inducible factors (HIFs), play a central role in cellular adaptation to critically low oxygen levels in both normal and compromised tissues. It has also been shown that several target genes of HIFs are involved in iron homeostasis, reflecting the molecular links between oxygen homeostasis and iron metabolism. Furthermore, HIF activation is modulated by intracellular iron, through regulation of hydroxylase activity, which requires iron as a cofactor. In addition, HIF-2α translation is controlled by iron regulatory protein (IRP) activity, providing another level of interdependence between iron and oxygen homeostasis.
Collapse
Affiliation(s)
- Kiichi Hirota
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Japan.
| |
Collapse
|
40
|
Barbarani G, Fugazza C, Strouboulis J, Ronchi AE. The Pleiotropic Effects of GATA1 and KLF1 in Physiological Erythropoiesis and in Dyserythropoietic Disorders. Front Physiol 2019; 10:91. [PMID: 30809156 PMCID: PMC6379452 DOI: 10.3389/fphys.2019.00091] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 01/25/2019] [Indexed: 01/19/2023] Open
Abstract
In the last few years, the advent of new technological approaches has led to a better knowledge of the ontogeny of erythropoiesis during development and of the journey leading from hematopoietic stem cells (HSCs) to mature red blood cells (RBCs). Our view of a well-defined hierarchical model of hematopoiesis with a near-homogeneous HSC population residing at the apex has been progressively challenged in favor of a landscape where HSCs themselves are highly heterogeneous and lineages separate earlier than previously thought. The coordination of these events is orchestrated by transcription factors (TFs) that work in a combinatorial manner to activate and/or repress their target genes. The development of next generation sequencing (NGS) has facilitated the identification of pathological mutations involving TFs underlying hematological defects. The examples of GATA1 and KLF1 presented in this review suggest that in the next few years the number of TF mutations associated with dyserythropoietic disorders will further increase.
Collapse
Affiliation(s)
- Gloria Barbarani
- Dipartimento di Biotecnologie e Bioscienze, Università degli Studi Milano-Bicocca, Milan, Italy
| | - Cristina Fugazza
- Dipartimento di Biotecnologie e Bioscienze, Università degli Studi Milano-Bicocca, Milan, Italy
| | - John Strouboulis
- School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| | - Antonella E Ronchi
- Dipartimento di Biotecnologie e Bioscienze, Università degli Studi Milano-Bicocca, Milan, Italy
| |
Collapse
|
41
|
Steiner M, Schneider L, Yillah J, Gerlach K, Kuvardina ON, Meyer A, Maring A, Bonig H, Seifried E, Zörnig M, Lausen J. FUSE binding protein 1 (FUBP1) expression is upregulated by T-cell acute lymphocytic leukemia protein 1 (TAL1) and required for efficient erythroid differentiation. PLoS One 2019; 14:e0210515. [PMID: 30653565 PMCID: PMC6336336 DOI: 10.1371/journal.pone.0210515] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 12/23/2018] [Indexed: 11/18/2022] Open
Abstract
During erythropoiesis, haematopoietic stem cells (HSCs) differentiate in successive steps of commitment and specification to mature erythrocytes. This differentiation process is controlled by transcription factors that establish stage- and cell type-specific gene expression. In this study, we demonstrate that FUSE binding protein 1 (FUBP1), a transcriptional regulator important for HSC self-renewal and survival, is regulated by T-cell acute lymphocytic leukaemia 1 (TAL1) in erythroid progenitor cells. TAL1 directly activates the FUBP1 promoter, leading to increased FUBP1 expression during erythroid differentiation. The binding of TAL1 to the FUBP1 promoter is highly dependent on an intact GATA sequence in a combined E-box/GATA motif. We found that FUBP1 expression is required for efficient erythropoiesis, as FUBP1-deficient progenitor cells were limited in their potential of erythroid differentiation. Thus, the finding of an interconnection between GATA1/TAL1 and FUBP1 reveals a molecular mechanism that is part of the switch from progenitor- to erythrocyte-specific gene expression. In summary, we identified a TAL1/FUBP1 transcriptional relationship, whose physiological function in haematopoiesis is connected to proper erythropoiesis.
Collapse
Affiliation(s)
- Marlene Steiner
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt/Main, Germany
| | - Lucas Schneider
- Institute for Transfusion Medicine and Immunohematology, Goethe-University and German Red Cross Blood Service, Frankfurt am Main, Germany
| | - Jasmin Yillah
- Institute for Transfusion Medicine and Immunohematology, Goethe-University and German Red Cross Blood Service, Frankfurt am Main, Germany
| | - Katharina Gerlach
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt/Main, Germany
| | - Olga N. Kuvardina
- Institute for Transfusion Medicine and Immunohematology, Goethe-University and German Red Cross Blood Service, Frankfurt am Main, Germany
| | - Annekarin Meyer
- Institute for Transfusion Medicine and Immunohematology, Goethe-University and German Red Cross Blood Service, Frankfurt am Main, Germany
| | - Alisa Maring
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt/Main, Germany
| | - Halvard Bonig
- Institute for Transfusion Medicine and Immunohematology, Goethe-University and German Red Cross Blood Service, Frankfurt am Main, Germany
| | - Erhard Seifried
- Institute for Transfusion Medicine and Immunohematology, Goethe-University and German Red Cross Blood Service, Frankfurt am Main, Germany
| | - Martin Zörnig
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt/Main, Germany
- * E-mail: (MZ); (JL)
| | - Jörn Lausen
- Institute for Transfusion Medicine and Immunohematology, Goethe-University and German Red Cross Blood Service, Frankfurt am Main, Germany
- * E-mail: (MZ); (JL)
| |
Collapse
|
42
|
Liao C, Carlson BA, Paulson RF, Prabhu KS. The intricate role of selenium and selenoproteins in erythropoiesis. Free Radic Biol Med 2018; 127:165-171. [PMID: 29719207 PMCID: PMC6168382 DOI: 10.1016/j.freeradbiomed.2018.04.578] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 04/26/2018] [Indexed: 01/18/2023]
Abstract
Selenium (Se) is incorporated as the 21st amino acid selenocysteine (Sec) into the growing polypeptide chain of proteins involved in redox gatekeeper functions. Erythropoiesis presents a particular problem to redox regulation as the presence of iron, heme, and unpaired globin chains lead to high levels of free radical-mediated oxidative stress, which are detrimental to erythroid development and can lead to anemia. Under homeostatic conditions, bone marrow erythropoiesis produces sufficient erythrocytes to maintain homeostasis. In contrast, anemic stress induces an alternative pathway, stress erythropoiesis, which rapidly produces new erythrocytes at extramedullary sites, such as spleen, to alleviate anemia. Previous studies suggest that dietary Se protects erythrocytes from such oxidative damage and the absence of selenoproteins causes hemolysis of erythrocytes due to oxidative stress. Furthermore, Se deficiency or lack of selenoproteins severely impairs stress erythropoiesis exacerbating the anemia in rodent models and human patients. Interestingly, erythroid progenitors develop in close proximity with macrophages in structures referred to as erythroblastic islands (EBIs), where macrophage expression of selenoproteins appears to be critical for the expression of heme transporters to facilitate export of heme from macrophage stores to the developing erythroid cells. Here we review the role of Se and selenoproteins in the intrinsic development of erythroid cells in addition to their role in the development of the erythropoietic niche that supports the functional role of EBIs in erythroid expansion and maturation in the spleen during recovery from anemia.
Collapse
Affiliation(s)
- Chang Liao
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Bradley A Carlson
- Molecular Biology of Selenium Section, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert F Paulson
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA.
| | - K Sandeep Prabhu
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
43
|
Singh N, Tripathi AK, Sahu DK, Mishra A, Linan M, Argente B, Varkey J, Parida N, Chowdhry R, Shyam H, Alam N, Dixit S, Shankar P, Mishra A, Agarwal A, Yoo C, Bhatt MLB, Kant R. Differential genomics and transcriptomics between tyrosine kinase inhibitor-sensitive and -resistant BCR-ABL-dependent chronic myeloid leukemia. Oncotarget 2018; 9:30385-30418. [PMID: 30100996 PMCID: PMC6084383 DOI: 10.18632/oncotarget.25752] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/28/2018] [Indexed: 01/11/2023] Open
Abstract
Previously, it has been stated that the BCR-ABL fusion-protein is sufficient to induce Chronic Myeloid Leukemia (CML), but additional genomic-changes are required for disease progression. Hence, we profiled control and tyrosine kinase inhibitors (TKI) alone or in combination with other drug-treated CML-samples in different phases, categorized as drug-sensitive and drug-resistant on the basis of BCR-ABL transcripts, the marker of major molecular-response. Molecular-profiling was done using the molecular-inversion probe-based-array, Human Transcriptomics-Array2.0, and Axiom-Biobank genotyping-arrays. At the transcript-level, clusters of control, TKI-resistant and TKI-sensitive cases were correlated with BCR-ABL transcript-levels. Both at the gene- and exon-levels, up-regulation of MPO, TPX2, and TYMS and down-regulation of STAT6, FOS, TGFBR2, and ITK lead up-regulation of the cell-cycle, DNA-replication, DNA-repair pathways and down-regulation of the immune-system, chemokine- and interleukin-signaling, TCR, TGF beta and MAPK signaling pathways. A comparison between TKI-sensitive and TKI-resistant cases revealed up-regulation of LAPTM4B, HLTF, PIEZO2, CFH, CD109, ANGPT1 in CML-resistant cases, leading to up-regulation of autophagy-, protein-ubiquitination-, stem-cell-, complement-, TGFβ- and homeostasis-pathways with specific involvement of the Tie2 and Basigin signaling-pathway. Dysregulated pathways were accompanied with low CNVs in CP-new and CP-UT-TKI-sensitive-cases with undetectable BCR-ABL-copies. High CNVs (previously reported gain of 9q34) were observed in BCR-ABL-independent and -dependent TKI, non-sensitive-CP-UT/AP-UT/B-UT and B-new samples. Further, genotyping CML-CP-UT cases with BCR-ABL 0-to-77.02%-copies, the identified, rsID239798 and rsID9475077, were associated with FAM83B, a candidate for therapeutic resistance. The presence of BCR-ABL, additional genetic-events, dysregulated-signaling-pathways and rsIDs associated with FAM83B in TKI-resistant-cases can be used to develop a signature-profile that may help in monitoring therapy.
Collapse
Affiliation(s)
- Neetu Singh
- Molecular Biology Unit, Center for Advance Research, King George's Medical University, Lucknow, India
| | - Anil Kumar Tripathi
- Department of Clinical Hematology, King George's Medical University, Lucknow, India
| | - Dinesh Kumar Sahu
- Molecular Biology Unit, Center for Advance Research, King George's Medical University, Lucknow, India
| | - Archana Mishra
- Department of Cardio Thoracic and Vascular Surgery, King George's Medical University, Lucknow, India
| | | | | | | | - Niranjan Parida
- Molecular Biology Unit, Center for Advance Research, King George's Medical University, Lucknow, India
| | - Rebecca Chowdhry
- Department of Periodontics, King George's Medical University, Lucknow, India
| | - Hari Shyam
- Molecular Biology Unit, Center for Advance Research, King George's Medical University, Lucknow, India
| | - Nawazish Alam
- Molecular Biology Unit, Center for Advance Research, King George's Medical University, Lucknow, India
| | - Shivani Dixit
- Molecular Biology Unit, Center for Advance Research, King George's Medical University, Lucknow, India
| | - Pratap Shankar
- Molecular Biology Unit, Center for Advance Research, King George's Medical University, Lucknow, India
| | - Abhishek Mishra
- Molecular Biology Unit, Center for Advance Research, King George's Medical University, Lucknow, India
| | - Avinash Agarwal
- Department of Medicine, King George's Medical University, Lucknow, India
| | - Chris Yoo
- Systems Imagination, Scottsdale, Arizona, USA
| | | | - Ravi Kant
- All India Institute of Medical Sciences, Rishikesh, India
| |
Collapse
|
44
|
Oikonomidou PR, Rivella S. What can we learn from ineffective erythropoiesis in thalassemia? Blood Rev 2018; 32:130-143. [PMID: 29054350 PMCID: PMC5882559 DOI: 10.1016/j.blre.2017.10.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 09/30/2017] [Accepted: 10/02/2017] [Indexed: 02/07/2023]
Abstract
Erythropoiesis is a dynamic process regulated at multiple levels to balance proliferation, differentiation and survival of erythroid progenitors. Ineffective erythropoiesis is a key feature of various diseases, including β-thalassemia. The pathogenic mechanisms leading to ineffective erythropoiesis are complex and still not fully understood. Altered survival and decreased differentiation of erythroid progenitors are both critical processes contributing to reduced production of mature red blood cells. Recent studies have identified novel important players and provided major advances in the development of targeted therapeutic approaches. In this review, β-thalassemia is used as a paradigmatic example to describe our current knowledge on the mechanisms leading to ineffective erythropoiesis and novel treatments that may have the potential to improve the clinical phenotype of associated diseases in the future.
Collapse
Affiliation(s)
- Paraskevi Rea Oikonomidou
- Department of Pediatrics, Division of Hematology, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA.
| | - Stefano Rivella
- Department of Pediatrics, Division of Hematology, Children's Hospital of Philadelphia (CHOP), Philadelphia, PA, USA; Cell and Molecular Biology Graduate Group (CAMB), University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
45
|
Lee Y, Park C, Lee S, Lee D, Kim J. Isolation and Functional Examination of the Long Non-Coding RNA Redrum. Mol Cells 2018; 41:134-139. [PMID: 29237115 PMCID: PMC5824023 DOI: 10.14348/molcells.2018.2246] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 11/04/2017] [Indexed: 11/27/2022] Open
Abstract
Here, we report isolation of multiple long non-coding RNAs (lncRNAs) expressed tissue-specifically during murine embryogenesis. One of these, subsequently came to be known as Redrum, is expressed in erythropoietic cells in fetal liver and adult bone marrow. Redrum transcription is also detected during pregnancy in the spleen where extramedullary hematopoiesis takes place. In order to examine the function of Redrum in vivo, we generated a gene-targeted murine model and analyzed its embryonic and adult erythropoiesis. The homozygous mutant embryo showed no apparent deficiency or defect in erythropoiesis. Adult erythropoiesis in bone marrow and in the spleen during pregnancy likewise showed no detectable phenotype as red blood cells matured in normal fashion. The phenotype is in contrast to the reported function of Redrum in vitro, and our observation implies that Redrum plays in vivo an accessory or supplementary role whose loss is compatible with normal erythropoiesis.
Collapse
Affiliation(s)
- Yerim Lee
- Department of Life Science, Ewha Womans University, Seoul 03760,
Korea
| | - Charny Park
- Department of Life Science, Ewha Womans University, Seoul 03760,
Korea
- Ewha Research Center for Systems Biology, Seoul 03760,
Korea
| | - Sanghyuk Lee
- Department of Life Science, Ewha Womans University, Seoul 03760,
Korea
- Ewha Research Center for Systems Biology, Seoul 03760,
Korea
| | - Daekee Lee
- Department of Life Science, Ewha Womans University, Seoul 03760,
Korea
- Ewha Research Center for Systems Biology, Seoul 03760,
Korea
| | - Jaesang Kim
- Department of Life Science, Ewha Womans University, Seoul 03760,
Korea
- Ewha Research Center for Systems Biology, Seoul 03760,
Korea
| |
Collapse
|
46
|
Ferreira AF, Calin GA, Picanço-Castro V, Kashima S, Covas DT, de Castro FA. Hematopoietic stem cells from induced pluripotent stem cells - considering the role of microRNA as a cell differentiation regulator. J Cell Sci 2018; 131:131/4/jcs203018. [PMID: 29467236 DOI: 10.1242/jcs.203018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Although hematopoietic stem cell (HSC) therapy for hematological diseases can lead to a good outcome from the clinical point of view, the limited number of ideal donors, the comorbidity of patients and the increasing number of elderly patients may limit the application of this therapy. HSCs can be generated from induced pluripotent stem cells (iPSCs), which requires the understanding of the bone marrow and liver niches components and function in vivo iPSCs have been extensively applied in several studies involving disease models, drug screening and cellular replacement therapies. However, the somatic reprogramming by transcription factors is a low-efficiency process. Moreover, the reprogramming process is also regulated by microRNAs (miRNAs), which modulate the expression of the transcription factors OCT-4 (also known as POU5F1), SOX-2, KLF-4 and MYC, leading somatic cells to a pluripotent state. In this Review, we present an overview of the challenges of cell reprogramming protocols with regard to HSC generation from iPSCs, and highlight the potential role of miRNAs in cell reprogramming and in the differentiation of induced pluripotent stem cells.
Collapse
Affiliation(s)
- Aline F Ferreira
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences, University of São Paulo (USP), Ribeirão Preto, São Paulo 14040-903, Brazil
| | - George A Calin
- Department of Experimental Therapeutics, MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Virgínia Picanço-Castro
- Center of Cell Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto, São Paulo 14051-140, Brazil
| | - Simone Kashima
- Center of Cell Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto, São Paulo 14051-140, Brazil
| | - Dimas T Covas
- Center of Cell Therapy, Regional Blood Center of Ribeirão Preto, Ribeirão Preto, São Paulo 14051-140, Brazil.,Department of Internal Medicine, School of Medicine of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo 14049-900, Brazil
| | - Fabiola A de Castro
- Department of Clinical Analysis, Toxicology and Food Science, School of Pharmaceutical Sciences, University of São Paulo (USP), Ribeirão Preto, São Paulo 14040-903, Brazil
| |
Collapse
|
47
|
Gore AV, Pillay LM, Venero Galanternik M, Weinstein BM. The zebrafish: A fintastic model for hematopoietic development and disease. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2018; 7:e312. [PMID: 29436122 DOI: 10.1002/wdev.312] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 11/30/2017] [Accepted: 12/03/2017] [Indexed: 12/19/2022]
Abstract
Hematopoiesis is a complex process with a variety of different signaling pathways influencing every step of blood cell formation from the earliest precursors to final differentiated blood cell types. Formation of blood cells is crucial for survival. Blood cells carry oxygen, promote organ development and protect organs in different pathological conditions. Hematopoietic stem and progenitor cells (HSPCs) are responsible for generating all adult differentiated blood cells. Defects in HSPCs or their downstream lineages can lead to anemia and other hematological disorders including leukemia. The zebrafish has recently emerged as a powerful vertebrate model system to study hematopoiesis. The developmental processes and molecular mechanisms involved in zebrafish hematopoiesis are conserved with higher vertebrates, and the genetic and experimental accessibility of the fish and the optical transparency of its embryos and larvae make it ideal for in vivo analysis of hematopoietic development. Defects in zebrafish hematopoiesis reliably phenocopy human blood disorders, making it a highly attractive model system to screen small molecules to design therapeutic strategies. In this review, we summarize the key developmental processes and molecular mechanisms of zebrafish hematopoiesis. We also discuss recent findings highlighting the strengths of zebrafish as a model system for drug discovery against hematopoietic disorders. This article is categorized under: Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cell Differentiation and Reversion Vertebrate Organogenesis > Musculoskeletal and Vascular Nervous System Development > Vertebrates: Regional Development Comparative Development and Evolution > Organ System Comparisons Between Species.
Collapse
Affiliation(s)
- Aniket V Gore
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland
| | - Laura M Pillay
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland
| | - Marina Venero Galanternik
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland
| | - Brant M Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland
| |
Collapse
|
48
|
Vinjamur DS, Bauer DE. Growing and Genetically Manipulating Human Umbilical Cord Blood-Derived Erythroid Progenitor (HUDEP) Cell Lines. Methods Mol Biol 2018; 1698:275-284. [PMID: 29076097 DOI: 10.1007/978-1-4939-7428-3_17] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
The recently established human umbilical cord blood-derived erythroid progenitor (HUDEP) cell lines have equipped red blood cell researchers with valuable in vitro models of erythroid development. Of the three established HUDEP cell lines, HUDEP-2 cells express predominantly adult β-globin and most closely resemble adult erythroid cells. This chapter describes culture protocols for the maintenance and erythroid differentiation of HUDEP-2 cells. Methods to genetically manipulate HUDEP-2 cells using a CRISPR/Cas9 nuclease-based approach are also discussed.
Collapse
Affiliation(s)
- Divya S Vinjamur
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Daniel E Bauer
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA. .,Dana-Farber Cancer Institute, Harvard Medical School, Harvard Stem Cell Institute, Karp 8211, 1 Blackfan Circle, Boston, MA, 02115, USA.
| |
Collapse
|
49
|
Huang SC, Rehman MU, Lan YF, Qiu G, Zhang H, Iqbal MK, Luo HQ, Mehmood K, Zhang LH, Li JK. Tibial dyschondroplasia is highly associated with suppression of tibial angiogenesis through regulating the HIF-1α/VEGF/VEGFR signaling pathway in chickens. Sci Rep 2017; 7:9089. [PMID: 28831181 PMCID: PMC5567304 DOI: 10.1038/s41598-017-09664-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 07/20/2017] [Indexed: 12/28/2022] Open
Abstract
Tibial dyschondroplasia (TD) is an intractable poultry problem that is characterized by the appearance of non-vascularized and non-mineralized cartilage masses in tibial growth plates (TGPs). However, the role of angiogenesis inhibition in the occurrence of TD remains unknown. In this study, we found that, compared to low-altitude Arbor Acres chickens (AACs), high-altitude Tibetan chickens showed higher tibial vascular distributions that were accompanied by up-regulation of hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor A (VEGFA) and VEGF receptors. These observations provide insights into hypoxia-induced angiogenesis, which may be related to the absence of TD in high-altitude native Tibetan chickens. Importantly, hypoxia experiments also showed that during hypoxia, tibial angiogenesis was enhanced, which was due to pro-angiogenic factor up-regulation (including VEGFA, VEGFR1, VEGFR2, and IL-8), in AACs. Moreover, we observed that thiram-induced TD could strongly inhibit tibial angiogenesis in the hypertrophic zone through coordinated down-regulation of HIF-1α and pro-angiogenic factors, leading to a disruption in the blood supply to the TGP. Taken together, these findings reveal that the occurrence of TD is highly associated with inhibition of tibial angiogenesis through down-regulated expression of HIF-1α, VEGFA and VEGF receptors, which results in suppression of TGP development.
Collapse
Affiliation(s)
- Shu-Cheng Huang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Mujeeb Ur Rehman
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Yan-Fang Lan
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Gang Qiu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.,Laboratory of Detection and Monitoring of Highland Animal Disease, Tibet Agriculture and Animal Husbandry College, Linzhi, 860000, Tibet, People's Republic of China
| | - Hui Zhang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Muhammad Kashif Iqbal
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Hou-Qiang Luo
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.,Animal Science Department, Wenzhou Vocational College of Science and Technology, Wenzhou, 325006, People's Republic of China
| | - Khalid Mehmood
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.,University College of Veterinary & Animal Sciences, The Islamia University of Bahawalpur, Bahawalpur, 631000, Pakistan
| | - Li-Hong Zhang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Jia-Kui Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China. .,Laboratory of Detection and Monitoring of Highland Animal Disease, Tibet Agriculture and Animal Husbandry College, Linzhi, 860000, Tibet, People's Republic of China.
| |
Collapse
|
50
|
Chang HC, Huang DY, Wu MS, Chu CL, Tzeng SJ, Lin WW. Spleen tyrosine kinase mediates the actions of EPO and GM-CSF and coordinates with TGF-β in erythropoiesis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:687-696. [DOI: 10.1016/j.bbamcr.2017.01.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 01/20/2017] [Accepted: 01/23/2017] [Indexed: 12/12/2022]
|