1
|
Lu L, Liu C, Chen L, Zhang X, Su Y, Chou Z, Liang Y, Song Y. Understanding erythroid physiology and pathology in humanized mice: A closer look. Br J Haematol 2025; 206:1272-1284. [PMID: 40007143 PMCID: PMC12078861 DOI: 10.1111/bjh.20023] [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: 10/27/2024] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
Erythropoiesis, the process of red blood cell (RBC) development from haematopoietic stem cells, is crucial in haematology research due to its intricate regulation and implications in various pathologies such as anaemia and haemoglobinopathies. Humanized mice, created by introducing human cells or tissues into immunodeficient mice, offer a promising avenue in vivo approach. However, challenges persist in fully replicating human erythropoiesis in these models, particularly in generating mature human RBCs capable of sustained circulation. This review discusses the differences between human and mouse erythropoiesis, recent progress made using refined humanized mouse models for studying human erythropoiesis and erythropoietic disorders, the challenges that impede a faithful mimicking of human phenotypes in these mice and recommendations for future research improvements. Despite progress being made, enhancing the translational potential of humanized mouse models for human erythropoiesis research remains a priority.
Collapse
Affiliation(s)
- Lu Lu
- State Key Laboratory of Oncology in South China, Department of Hematologic Oncology, Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Chenfei Liu
- State Key Laboratory of Oncology in South China, Department of Hematologic Oncology, Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Lezong Chen
- State Key Laboratory of Oncology in South China, Department of Hematologic Oncology, Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Xumiao Zhang
- State Key Laboratory of Oncology in South China, Department of Hematologic Oncology, Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Yinglin Su
- State Key Laboratory of Oncology in South China, Department of Hematologic Oncology, Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Zhenzhen Chou
- State Key Laboratory of Oncology in South China, Department of Hematologic Oncology, Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Yang Liang
- State Key Laboratory of Oncology in South China, Department of Hematologic Oncology, Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Yuanbin Song
- State Key Laboratory of Oncology in South China, Department of Hematologic Oncology, Collaborative Innovation Center for Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| |
Collapse
|
2
|
Zhang S, Zhang Y, Li Y, Zhang Z, Li H, Xu M, Lu Z, Li Y, Zhao B. Fine-tuned calcium homeostasis is crucial for murine erythropoiesis. FEBS J 2025; 292:1934-1949. [PMID: 39838539 DOI: 10.1111/febs.17401] [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: 07/29/2024] [Revised: 10/25/2024] [Accepted: 11/21/2024] [Indexed: 01/23/2025]
Abstract
Intracellular calcium (Ca2+) is a crucial signaling molecule involved in multiple cellular processes. However, the functional role of Ca2+ in terminal erythropoiesis remains unclear. Here, we uncovered the dynamics of intracellular Ca2+ levels during mouse erythroid development. By using the calcium ionophore ionomycin, we found that low Ca2+ levels are required for the expansion of erythroid progenitors, whereas higher Ca2+ levels led to the differentiation and proliferation of early-stage erythroblasts. Intracellular Ca2+ levels were then gradually reduced, which is required for the nuclear condensation and polarisation at the late stage of erythroid differentiation. However, elevated Ca2+ levels in late-stage erythroblasts, achieved by using ionomycin, promoted erythroid enucleation via calmodulin (CaM)/calcium/calmodulin-dependent protein kinase kinase 1 (CaMKK1)/AMPK signaling. These data suggest that the reduction of intracellular Ca2+ plays a double-edged role at the late stage of erythroid differentiation, which is beneficial for nuclear condensation but compromises terminal enucleation. Our study highlighted the importance of the fine-tuned regulation of intracellular Ca2+ during terminal erythropoiesis, providing cues for the efficient generation of mature and enucleated erythrocytes in vitro.
Collapse
Affiliation(s)
- Shujing Zhang
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuanzhen Zhang
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yanxia Li
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhiyue Zhang
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hui Li
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Miaomiao Xu
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhiyuan Lu
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yuan Li
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Baobing Zhao
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
3
|
Canals C, Gonzalez-Santesteban C, Vinyets I, Gracia M, Boto N, Salgado M, Moreno I, Rodríguez-Aliberas M, Casanovas E, Muñiz-Diaz E, Nogués N. CD36 antibodies in isoimmunized African-origin pregnant women: three years experience in Spain. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2025; 23:123-127. [PMID: 39133626 PMCID: PMC11925251 DOI: 10.2450/bloodtransfus.812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/11/2024] [Indexed: 03/21/2025]
Affiliation(s)
- Carme Canals
- Immunohematology Laboratory, Banc de Sang i Teixits, Barcelona, Spain
| | | | | | - Mercedes Gracia
- Immunohematology Laboratory, Banc de Sang i Teixits, Barcelona, Spain
| | - Neus Boto
- Immunohematology Laboratory, Banc de Sang i Teixits, Barcelona, Spain
| | - Marta Salgado
- Immunohematology Laboratory, Banc de Sang i Teixits, Barcelona, Spain
| | - Immaculada Moreno
- Immunohematology Laboratory, Banc de Sang i Teixits, Barcelona, Spain
| | | | - Enric Casanovas
- Immunohematology Laboratory, Banc de Sang i Teixits, Barcelona, Spain
| | | | - Núria Nogués
- Immunohematology Laboratory, Banc de Sang i Teixits, Barcelona, Spain
| |
Collapse
|
4
|
Yang X, Cheng L, Xin Y, Zhang J, Chen X, Xu J, Zhang M, Feng R, Hyle J, Qi W, Rosikiewicz W, Xu B, Li C, Xu P. CTCF is selectively required for maintaining chromatin accessibility and gene expression in human erythropoiesis. Genome Biol 2025; 26:44. [PMID: 40022213 PMCID: PMC11869676 DOI: 10.1186/s13059-025-03510-z] [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: 05/15/2024] [Accepted: 02/19/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND CTCF is considered as the most essential transcription factor regulating chromatin architecture and gene expression. However, genome-wide impact of CTCF on erythropoiesis has not been extensively investigated. RESULTS Using a state-of-the-art human erythroid progenitor cell model (HUDEP-2 and HEL cell lines), we systematically investigate the effects of acute CTCF loss by an auxin-inducible degron system on transcriptional programs, chromatin accessibility, CTCF genome occupancy, and genome architecture. By integrating multi-omics datasets, we reveal that acute CTCF loss notably disrupts genome-wide chromatin accessibility and the transcription network. We detect over thousands of decreased chromatin accessibility regions but only a few hundred increased regions after CTCF depletion in HUDEP-2 and HEL lines, suggesting the role of CTCF in maintaining proper chromatin openness in the erythroid lineage. CTCF depletion in the erythroid context notably disrupts the boundary integrity of topologically associating domains and chromatin loops but does not affect nuclear compartmentalization. We find erythroid lineage-specific genes, including some metabolism-related genes, are suppressed at immature and mature stages. Notably, we find a subset of genes whose transcriptional levels increase upon CTCF depletion, accompanied by decreased chromatin accessibility regions enriched with the GATA motif. We further decipher the molecular mechanism underlying the CTCF/GATA2 repression axis through distal non-coding chromatin regions. These results suggest a suppressive role of CTCF in gene expression during erythroid lineage specification. CONCLUSIONS Our study reveals a novel role of CTCF in regulating erythroid differentiation by maintaining its proper chromatin openness and gene expression network, which extends our understanding of CTCF biology.
Collapse
Affiliation(s)
- Xue Yang
- Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Hematology, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Li Cheng
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Present address: GenAssist Therapeutics Incorporation, Suzhou, Jiangsu Province, 215000, China
| | - Ye Xin
- Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Hematology, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Jianxiang Zhang
- Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Hematology, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Xinfeng Chen
- Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Hematology, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Jinchao Xu
- Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Hematology, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Mengli Zhang
- Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Hematology, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, 215123, China
| | - Ruopeng Feng
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Judith Hyle
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Wenjie Qi
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Wojciech Rosikiewicz
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Beisi Xu
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| | - Chunliang Li
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| | - Peng Xu
- Cyrus Tang Medical Institute, National Clinical Research Center for Hematologic Diseases, State Key Laboratory of Radiation Medicine and Protection, Collaborative Innovation Center of Hematology, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, 215123, China.
| |
Collapse
|
5
|
Ling Y, Du H, Zhang Y, Fu Q, He C. Advances in the generation of erythrocytes from stem cells in vitro. Hematology 2024; 29:2427932. [PMID: 39658930 DOI: 10.1080/16078454.2024.2427932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 11/05/2024] [Indexed: 12/12/2024] Open
Abstract
Red blood cell transfusion is the main treatment to improve anemia caused by many reasons and has important clinical application value. However, the supply of red blood cells at home and abroad is currently very tight. Therefore, the utilization of stem cells to prepare erythrocytes for clinical use is expected to become a new mode of blood supply and security in the future. This review describes the process of erythropoiesis regulation in vivo, summarizes the latest research progress of in vitro erythropoiesis, and points out the current challenges of in vitro erythropoiesis, which is expected to provide a new idea for solving the problem of insufficient clinical erythropoiesis supply.
Collapse
Affiliation(s)
- Yating Ling
- Department of Laboratory Medicine, Nanjing Red Cross Blood Center, Nanjing, Jiangsu, People's Republic of China
| | - Hailin Du
- Department of Laboratory Medicine, Nanjing Red Cross Blood Center, Nanjing, Jiangsu, People's Republic of China
| | - Yu Zhang
- Department of Laboratory Medicine, Nanjing Red Cross Blood Center, Nanjing, Jiangsu, People's Republic of China
| | - Qiang Fu
- Department of Laboratory Medicine, Nanjing Red Cross Blood Center, Nanjing, Jiangsu, People's Republic of China
| | - Chengtao He
- Department of Laboratory Medicine, Nanjing Red Cross Blood Center, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
6
|
Metral S, Genetet S, Gamain B, Mouro-Chanteloup I. JK-1, a useful erythroleukemic cell line model to study a controlled erythroid differentiation from progenitors to terminal erythropoiesis. Sci Rep 2024; 14:25885. [PMID: 39468295 PMCID: PMC11519332 DOI: 10.1038/s41598-024-76897-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024] Open
Abstract
New hematopoietic cell models have recently emerged through immortalization of CD34 cells to study and understand various molecular mechanisms of erythropoiesis. Here, we characterize the JK-1 CML-derived cell line, previously shown to spontaneously differentiate without cytokines. Using an epigenetic differentiation inhibitor that keeps JK-1 in an early differentiation phase, we characterized 2 progenitor stages: BFU-E JK-1 and CFU-E JK-1 with CD34+/CD36- and CD34-/CD36 + phenotypes respectively. Then, using the PFI-1 inducer known to synchronously control the terminal differentiation of JK-1 cells, 5 precursor stages were obtained (ProE, Baso1-2, PolyC, OrthoC) and characterized via cell morphology, CD49a and Band3 markers. Enlarged phenotyping was carried out for the earlier phase, and expression kinetics of membrane proteins such as RhAG, RhD/CE, CD47, DARC and CD44 were performed on each stage of the terminal phase. Furthermore, since JK-1 offers the unique property of covering a broad spectrum of differentiation stages, we explored deeper the GATA2/GATA1 and the non-erythroid/erythroid spectrin 'switching'. The possibility of obtaining large quantities of JK-1 cells at each stage of differentiation, as shown in this study, as well as the potential to genetically modify these cells, via CrisprCas9, makes their use of considerable interest for studying pathologies occurring during erythropoiesis.
Collapse
Affiliation(s)
- Sylvain Metral
- Université Paris Cité and Université des Antilles, Inserm U1134, BIGR, Paris, France
| | - Sandrine Genetet
- Université Paris Cité and Université des Antilles, Inserm U1134, BIGR, Paris, France
| | - Benoît Gamain
- Sorbonne Université, Inserm U1135, CNRS ERL 8255, Paris, France
- Centre d'Immunologie et des Maladies Infectieuses, Faculté de médecine Sorbonne Université, 5e étage - Bureau 506 91 boulevard de l'Hôpital, 75013, Paris, France
| | - Isabelle Mouro-Chanteloup
- Sorbonne Université, Inserm U1135, CNRS ERL 8255, Paris, France.
- Centre d'Immunologie et des Maladies Infectieuses, Faculté de médecine Sorbonne Université, 5e étage - Bureau 506 91 boulevard de l'Hôpital, 75013, Paris, France.
| |
Collapse
|
7
|
Newton LM, Fowler VM, Humbert PO. Erythroblast enucleation at a glance. J Cell Sci 2024; 137:jcs261673. [PMID: 39397781 PMCID: PMC11529606 DOI: 10.1242/jcs.261673] [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] [Indexed: 10/15/2024] Open
Abstract
Erythroid enucleation, the penultimate step in mammalian erythroid terminal differentiation, is a unique cellular process by which red blood cells (erythrocytes) remove their nucleus and accompanying nuclear material. This complex, multi-stage event begins with chromatin compaction and cell cycle arrest and ends with generation of two daughter cells: a pyrenocyte, which contains the expelled nucleus, and an anucleate reticulocyte, which matures into an erythrocyte. Although enucleation has been compared to asymmetric cell division (ACD), many mechanistic hallmarks of ACD appear to be absent. Instead, enucleation appears to rely on mechanisms borrowed from cell migration, endosomal trafficking and apoptosis, as well as unique cellular interactions within the microenvironment. In this Cell Science at a Glance article and the accompanying poster, we summarise current insights into the morphological features and genetic drivers regulating the key intracellular events that culminate in erythroid enucleation and engulfment of pyrenocytes by macrophages within the bone marrow microenvironment.
Collapse
Affiliation(s)
- Lucas M. Newton
- Department of Biochemistry and Chemistry, La Trobe University, Melbourne, VIC 3073, Australia
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3073, Australia
| | - Velia M. Fowler
- Department of Biological Sciences, University of Delaware, Newark, DE 19711, USA
| | - Patrick O. Humbert
- Department of Biochemistry and Chemistry, La Trobe University, Melbourne, VIC 3073, Australia
- La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3073, Australia
- Department of Biochemistry and Pharmacology, University of Melbourne, Parkville, VIC 3010, Australia
- Department of Clinical Pathology, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
8
|
Wang J, Liang Y, Xu C, Gao J, Tong J, Shi L. The heterogeneity of erythroid cells: insight at the single-cell transcriptome level. Cell Tissue Res 2024; 397:179-192. [PMID: 38953986 DOI: 10.1007/s00441-024-03903-9] [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: 12/11/2023] [Accepted: 06/19/2024] [Indexed: 07/04/2024]
Abstract
Erythroid cells, the most prevalent cell type in blood, are one of the earliest products and permeate through the entire process of hematopoietic development in the human body, the oxygen-transporting function of which is crucial for maintaining overall health and life support. Previous investigations into erythrocyte differentiation and development have primarily focused on population-level analyses, lacking the single-cell perspective essential for comprehending the intricate pathways of erythroid maturation, differentiation, and the encompassing cellular heterogeneity. The continuous optimization of single-cell transcriptome sequencing technology, or single-cell RNA sequencing (scRNA-seq), provides a powerful tool for life sciences research, which has a particular superiority in the identification of unprecedented cell subgroups, the analyzing of cellular heterogeneity, and the transcriptomic characteristics of individual cells. Over the past decade, remarkable strides have been taken in the realm of single-cell RNA sequencing technology, profoundly enhancing our understanding of erythroid cells. In this review, we systematically summarize the recent developments in single-cell transcriptome sequencing technology and emphasize their substantial impact on the study of erythroid cells, highlighting their contributions, including the exploration of functional heterogeneity within erythroid populations, the identification of novel erythrocyte subgroups, the tracking of different erythroid lineages, and the unveiling of mechanisms governing erythroid fate decisions. These findings not only invigorate erythroid cell research but also offer new perspectives on the management of diseases related to erythroid cells.
Collapse
Affiliation(s)
- Jingwei Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Yipeng Liang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Changlu Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Jie Gao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Jingyuan Tong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
| | - Lihong Shi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
- CAMS Center for Stem Cell Medicine, PUMC Department of Stem Cell and Regenerative Medicine, Tianjin, 300020, China.
| |
Collapse
|
9
|
Pan X, Köberle M, Ghashghaeinia M. Vitamin C-Dependent Uptake of Non-Heme Iron by Enterocytes, Its Impact on Erythropoiesis and Redox Capacity of Human Erythrocytes. Antioxidants (Basel) 2024; 13:968. [PMID: 39199214 PMCID: PMC11352176 DOI: 10.3390/antiox13080968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/01/2024] Open
Abstract
In the small intestine, nutrients from ingested food are absorbed and broken down by enterocytes, which constitute over 95% of the intestinal epithelium. Enterocytes demonstrate diet- and segment-dependent metabolic flexibility, enabling them to take up large amounts of glutamine and glucose to meet their energy needs and transfer these nutrients into the bloodstream. During glycolysis, ATP, lactate, and H+ ions are produced within the enterocytes. Based on extensive but incomplete glutamine oxidation large amounts of alanine or lactate are produced. Lactate, in turn, promotes hypoxia-inducible factor-1α (Hif-1α) activation and Hif-1α-dependent transcription of various proton channels and exchangers, which extrude cytoplasmic H+-ions into the intestinal lumen. In parallel, the vitamin C-dependent and duodenal cytochrome b-mediated conversion of ferric iron into ferrous iron progresses. Finally, the generated electrochemical gradient is utilized by the divalent metal transporter 1 for H+-coupled uptake of non-heme Fe2+-ions. Iron efflux from enterocytes, subsequent binding to the plasma protein transferrin, and systemic distribution supply a wide range of cells with iron, including erythroid precursors essential for erythropoiesis. In this review, we discuss the impact of vitamin C on the redox capacity of human erythrocytes and connect enterocyte function with iron metabolism, highlighting its effects on erythropoiesis.
Collapse
Affiliation(s)
- Xia Pan
- Physiological Institute, Department of Vegetative and Clinical Physiology, Eberhard Karls University of Tübingen, 72074 Tübingen, Germany
| | - Martin Köberle
- Department of Dermatology and Allergology, School of Medicine and Health, Technical University of Munich, Biedersteinerstr. 29, 80802 München, Germany
| | - Mehrdad Ghashghaeinia
- Physiological Institute, Department of Vegetative and Clinical Physiology, Eberhard Karls University of Tübingen, 72074 Tübingen, Germany
| |
Collapse
|
10
|
Kumar S, Arora R, Gupta S, Ahuja N, Bhagyaraj E, Nanduri R, Kalra R, Khare AK, Kumawat S, Kaushal V, Sharma M, Gupta P. Nuclear receptor Rev-erbα role in fine-tuning erythropoietin gene expression. Blood Adv 2024; 8:3705-3717. [PMID: 38748870 PMCID: PMC11296239 DOI: 10.1182/bloodadvances.2023012228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 05/08/2024] [Indexed: 07/13/2024] Open
Abstract
ABSTRACT The regulation of red blood cell (RBC) homeostasis by erythropoietin (EPO) is critical for O2 transport and maintaining the adequate number of RBCs in vertebrates. Therefore, dysregulation in EPO synthesis results in disease conditions such as polycythemia in the case of excessive EPO production and anemia, which occurs when EPO is inadequately produced. EPO plays a crucial role in treating anemic patients; however, its overproduction can increase blood viscosity, potentially leading to fatal heart failure. Consequently, the identification of druggable transcription factors and their associated ligands capable of regulating EPO offers a promising therapeutic approach to address EPO-related disorders. This study unveils a novel regulatory mechanism involving 2 pivotal nuclear receptors (NRs), Rev-ERBA (Rev-erbα, is a truncation of reverse c-erbAa) and RAR-related orphan receptor A (RORα), in the control of EPO gene expression. Rev-erbα acts as a cell-intrinsic negative regulator, playing a vital role in maintaining erythropoiesis at the correct level. It accomplishes this by directly binding to newly identified response elements within the human and mouse EPO gene promoter, thereby repressing EPO production. These findings are further supported by the discovery that a Rev-erbα agonist (SR9011) effectively suppresses hypoxia-induced EPO expression in mice. In contrast, RORα functions as a positive regulator of EPO gene expression, also binding to the same response elements in the promoter to induce EPO production. Finally, the results of this study revealed that the 2 NRs, Rev-erbα and RORα, influence EPO synthesis in a negative and positive manner, respectively, suggesting that the modulating activity of these 2 NRs could provide a method to target disorders linked with EPO dysregulation.
Collapse
Affiliation(s)
- Sumit Kumar
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Rashmi Arora
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Shalini Gupta
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Nancy Ahuja
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Ella Bhagyaraj
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Ravikanth Nanduri
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Rashi Kalra
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Asheesh Kumar Khare
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Saumyata Kumawat
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Vipashu Kaushal
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Mahathi Sharma
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh, India
| | - Pawan Gupta
- Department of Molecular Biology, CSIR-Institute of Microbial Technology, Chandigarh, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
11
|
Liu G, Hou Y, Jin X, Zhang Y, Sun C, Huang C, Ren Y, Gao J, Wang X, Jiang X. PI3K/HSCB axis facilitates FOG1 nuclear translocation to promote erythropoiesis and megakaryopoiesis. eLife 2024; 13:RP95815. [PMID: 38757931 PMCID: PMC11101173 DOI: 10.7554/elife.95815] [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] [Indexed: 05/18/2024] Open
Abstract
Erythropoiesis and megakaryopoiesis are stringently regulated by signaling pathways. However, the precise molecular mechanisms through which signaling pathways regulate key transcription factors controlling erythropoiesis and megakaryopoiesis remain partially understood. Herein, we identified heat shock cognate B (HSCB), which is well known for its iron-sulfur cluster delivery function, as an indispensable protein for friend of GATA 1 (FOG1) nuclear translocation during erythropoiesis of K562 human erythroleukemia cells and cord-blood-derived human CD34+CD90+hematopoietic stem cells (HSCs), as well as during megakaryopoiesis of the CD34+CD90+HSCs. Mechanistically, HSCB could be phosphorylated by phosphoinositol-3-kinase (PI3K) to bind with and mediate the proteasomal degradation of transforming acidic coiled-coil containing protein 3 (TACC3), which otherwise detained FOG1 in the cytoplasm, thereby facilitating FOG1 nuclear translocation. Given that PI3K is activated during both erythropoiesis and megakaryopoiesis, and that FOG1 is a key transcription factor for these processes, our findings elucidate an important, previously unrecognized iron-sulfur cluster delivery independent function of HSCB in erythropoiesis and megakaryopoiesis.
Collapse
Affiliation(s)
- Gang Liu
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal UniversityChangchunChina
| | - Yunxuan Hou
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal UniversityChangchunChina
| | - Xin Jin
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal UniversityChangchunChina
| | - Yixue Zhang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal UniversityChangchunChina
| | - Chaoyue Sun
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal UniversityChangchunChina
| | - Chengquan Huang
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal UniversityChangchunChina
| | - Yujie Ren
- Key Laboratory of Molecular Epigenetics of the Ministry of Education, Northeast Normal UniversityChangchunChina
| | - Jianmin Gao
- School of Chemistry, Northeast Normal UniversityChangchunChina
| | - Xiuli Wang
- School of Life Science, Northeast Normal UniversityChangchunChina
| | - Xiumei Jiang
- School of Chemistry, Northeast Normal UniversityChangchunChina
| |
Collapse
|
12
|
Siraj L, Castro RI, Dewey H, Kales S, Nguyen TTL, Kanai M, Berenzy D, Mouri K, Wang QS, McCaw ZR, Gosai SJ, Aguet F, Cui R, Vockley CM, Lareau CA, Okada Y, Gusev A, Jones TR, Lander ES, Sabeti PC, Finucane HK, Reilly SK, Ulirsch JC, Tewhey R. Functional dissection of complex and molecular trait variants at single nucleotide resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.05.592437. [PMID: 38766054 PMCID: PMC11100724 DOI: 10.1101/2024.05.05.592437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Identifying the causal variants and mechanisms that drive complex traits and diseases remains a core problem in human genetics. The majority of these variants have individually weak effects and lie in non-coding gene-regulatory elements where we lack a complete understanding of how single nucleotide alterations modulate transcriptional processes to affect human phenotypes. To address this, we measured the activity of 221,412 trait-associated variants that had been statistically fine-mapped using a Massively Parallel Reporter Assay (MPRA) in 5 diverse cell-types. We show that MPRA is able to discriminate between likely causal variants and controls, identifying 12,025 regulatory variants with high precision. Although the effects of these variants largely agree with orthogonal measures of function, only 69% can plausibly be explained by the disruption of a known transcription factor (TF) binding motif. We dissect the mechanisms of 136 variants using saturation mutagenesis and assign impacted TFs for 91% of variants without a clear canonical mechanism. Finally, we provide evidence that epistasis is prevalent for variants in close proximity and identify multiple functional variants on the same haplotype at a small, but important, subset of trait-associated loci. Overall, our study provides a systematic functional characterization of likely causal common variants underlying complex and molecular human traits, enabling new insights into the regulatory grammar underlying disease risk.
Collapse
Affiliation(s)
- Layla Siraj
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Program in Biophysics, Harvard Graduate School of Arts and Sciences, Boston, MA, USA
- Harvard-Massachusetts Institute of Technology MD/PhD Program, Harvard Medical School, Boston, MA, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | | | | | | | | | - Masahiro Kanai
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, MA, USA
| | | | | | - Qingbo S. Wang
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA USA
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | | | - Sager J. Gosai
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, USA
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - François Aguet
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Illumina Artificial Intelligence Laboratory, Illumina, San Diego, CA, USA
| | - Ran Cui
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Caleb A. Lareau
- Program in Computational and Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Alexander Gusev
- Harvard Medical School and Dana-Farber Cancer Institute, Boston, MA, USA
| | - Thouis R. Jones
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Eric S. Lander
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Biology, MIT, Cambridge, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Pardis C. Sabeti
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Hilary K. Finucane
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA USA
| | - Steven K. Reilly
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
- Wu Tsai Institute, Yale University, New Haven, CT, USA
| | - Jacob C. Ulirsch
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA USA
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, USA
- Illumina Artificial Intelligence Laboratory, Illumina, San Diego, CA, USA
| | - Ryan Tewhey
- The Jackson Laboratory, Bar Harbor, ME, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
13
|
Xu Y, Guo R, Huang T, Guo C. miRNA-7145-cuedc2 axis controls hematopoiesis through JAK1/STAT3 signaling pathway. Cell Death Discov 2024; 10:209. [PMID: 38697957 PMCID: PMC11066045 DOI: 10.1038/s41420-024-01977-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024] Open
Abstract
Hematopoiesis ensures tissue oxygenation, and remodeling as well as immune protection in vertebrates. During embryogenesis, hemangioblasts are the source of all blood cells. Gata1a and pu.1 are co-expressed in hemangioblasts before hemangioblasts are differentiated into blood cells. However, the genes that determine the differentiation of hemangioblasts into myeloid or erythroid cell lineages have not been fully uncovered. Here we showed that miRNA-7145, a miRNA with previously unknown function, was enriched in erythrocytes at the definitive wave, but not expressed in myeloid cells. Overexpression and loss-of-function analysis of miRNA-7145 revealed that miRNA-7145 functions as a strong inhibitor for myeloid progenitor cell differentiation while driving erythropoiesis during the primitive wave. Furthermore, we confirmed that cuedc2 is one of miRNA-7145 targeted-genes. Overexpression or knock-down of cuedc2 partially rescues the phenotype caused by miRNA-7145 overexpression or loss-of-function. As well, overexpression and loss-of-function analysis of cuedc2 showed that cuedc2 is required for myelopoiesis at the expense of erythropoiesis. Finally, we found that overexpression of zebrafish cuedc2 in 293 T cell inhibits the JAK1/STAT3 signaling pathway. Collectively, our results uncover a previously unknown miRNA-7145-cuedc2 axis, which regulate hematopoiesis through inhibiting the JAK1/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Yongsheng Xu
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China.
| | - Rui Guo
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China
| | - Tao Huang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China
| | - Chunming Guo
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China.
| |
Collapse
|
14
|
Țichil I, Mitre I, Zdrenghea MT, Bojan AS, Tomuleasa CI, Cenariu D. A Review of Key Regulators of Steady-State and Ineffective Erythropoiesis. J Clin Med 2024; 13:2585. [PMID: 38731114 PMCID: PMC11084473 DOI: 10.3390/jcm13092585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Erythropoiesis is initiated with the transformation of multipotent hematopoietic stem cells into committed erythroid progenitor cells in the erythroblastic islands of the bone marrow in adults. These cells undergo several stages of differentiation, including erythroblast formation, normoblast formation, and finally, the expulsion of the nucleus to form mature red blood cells. The erythropoietin (EPO) pathway, which is activated by hypoxia, induces stimulation of the erythroid progenitor cells and the promotion of their proliferation and survival as well as maturation and hemoglobin synthesis. The regulation of erythropoiesis is a complex and dynamic interaction of a myriad of factors, such as transcription factors (GATA-1, STAT5), cytokines (IL-3, IL-6, IL-11), iron metabolism and cell cycle regulators. Multiple microRNAs are involved in erythropoiesis, mediating cell growth and development, regulating oxidative stress, erythrocyte maturation and differentiation, hemoglobin synthesis, transferrin function and iron homeostasis. This review aims to explore the physiology of steady-state erythropoiesis and to outline key mechanisms involved in ineffective erythropoiesis linked to anemia, chronic inflammation, stress, and hematological malignancies. Studying aberrations in erythropoiesis in various diseases allows a more in-depth understanding of the heterogeneity within erythroid populations and the development of gene therapies to treat hematological disorders.
Collapse
Affiliation(s)
- Ioana Țichil
- Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (I.M.); (M.T.Z.); (A.S.B.); (C.I.T.); (D.C.)
- Department of Haematology, “Ion Chiricuta” Institute of Oncology, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Ileana Mitre
- Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (I.M.); (M.T.Z.); (A.S.B.); (C.I.T.); (D.C.)
| | - Mihnea Tudor Zdrenghea
- Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (I.M.); (M.T.Z.); (A.S.B.); (C.I.T.); (D.C.)
- Department of Haematology, “Ion Chiricuta” Institute of Oncology, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Anca Simona Bojan
- Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (I.M.); (M.T.Z.); (A.S.B.); (C.I.T.); (D.C.)
- Department of Haematology, “Ion Chiricuta” Institute of Oncology, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
| | - Ciprian Ionuț Tomuleasa
- Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (I.M.); (M.T.Z.); (A.S.B.); (C.I.T.); (D.C.)
- Department of Haematology, “Ion Chiricuta” Institute of Oncology, 34–36 Republicii Street, 400015 Cluj-Napoca, Romania
- MEDFUTURE—Research Centre for Advanced Medicine, 8 Louis Pasteur Street, 400347 Cluj-Napoca, Romania
| | - Diana Cenariu
- Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania; (I.M.); (M.T.Z.); (A.S.B.); (C.I.T.); (D.C.)
- MEDFUTURE—Research Centre for Advanced Medicine, 8 Louis Pasteur Street, 400347 Cluj-Napoca, Romania
| |
Collapse
|
15
|
Melczer M, Jiménez Lamana J, Justo-Vega A, Hanser O, Ndaw S, Lobinski R. Multielement analysis of single red blood cells by single cell - inductively coupled plasma tandem mass spectrometry. Talanta 2024; 267:125226. [PMID: 37742394 DOI: 10.1016/j.talanta.2023.125226] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/23/2023] [Accepted: 09/19/2023] [Indexed: 09/26/2023]
Abstract
A method for the analysis of essential metals (Fe, Cu, Mg, and Zn) and non-metals (P, S) in single red blood cells was developed by single cell (SC)-ICP-MS. The use of a triple quadrupole configuration (MS/MS) enabled an effective elimination of polyatomic interferences, which affect the accuracy of ICP-MS analysis using a single quadrupole mass analyzer. Fixation with glutaraldehyde for at least 90 days was developed to improve the quantification of elements in a single red blood cell. The experimental conditions were optimized while special attention was paid to the residence time of analytes in the plasma. Addition of a surfactant (0.05% (v/v) Tween80®) improved quantification of elements in fixed red blood cells. The detection limits obtained by SC-ICP-MS/MS were lower than for ICP-MS, especially for S and P (3 fg and 1.7 fg. cell-1 instead of 163 and 6.3 fg. cell-1, respectively).
Collapse
Affiliation(s)
- Mathieu Melczer
- French Research and Safety Institute for the Prevention of Occupational Accidents and Diseases (INRS), Toxicology and Biomonitoring Division, 1 Rue Morvan, F-54519, Vandoeuvre les Nancy, France.
| | - Javier Jiménez Lamana
- Universite de Pau et des Pays de l'Adour, E2S UPPA, CNRS, Institute of Analytical and Physical Chemistry for Environment and Materials (IPREM-UMR5254), 64053, Pau, France
| | - Ana Justo-Vega
- Universite de Pau et des Pays de l'Adour, E2S UPPA, CNRS, Institute of Analytical and Physical Chemistry for Environment and Materials (IPREM-UMR5254), 64053, Pau, France; Group of Trace Elements, Spectroscopy and Speciation (GETEE), Materials Institute (iMATUS), Department of Analytical Chemistry, Nutrition and Bromatology, Faculty of Chemistry, University of Santiago de Compostela, Avda das Ciencias, s/n, 15782, Santiago de Compostela, Spain
| | - Ogier Hanser
- French Research and Safety Institute for the Prevention of Occupational Accidents and Diseases (INRS), Toxicology and Biomonitoring Division, 1 Rue Morvan, F-54519, Vandoeuvre les Nancy, France
| | - Sophie Ndaw
- French Research and Safety Institute for the Prevention of Occupational Accidents and Diseases (INRS), Toxicology and Biomonitoring Division, 1 Rue Morvan, F-54519, Vandoeuvre les Nancy, France
| | - Ryszard Lobinski
- Universite de Pau et des Pays de l'Adour, E2S UPPA, CNRS, Institute of Analytical and Physical Chemistry for Environment and Materials (IPREM-UMR5254), 64053, Pau, France; Chair of Analytical Chemistry, Faculty of Chemistry, Warsaw University of Technology, ul. Noakowskiego 3, 00-664, Warswawa, Poland
| |
Collapse
|
16
|
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
|
17
|
Walulik A, Łysak K, Błaszkiewicz M, Górecki I, Gomułka K. The Role of Neutrophils in ANCA-Associated Vasculitis: The Pathogenic Role and Diagnostic Utility of Autoantibodies. Int J Mol Sci 2023; 24:17217. [PMID: 38139045 PMCID: PMC10743134 DOI: 10.3390/ijms242417217] [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: 10/26/2023] [Revised: 12/01/2023] [Accepted: 12/03/2023] [Indexed: 12/24/2023] Open
Abstract
Recent years have brought progress in understanding the role of the neutrophil, dispelling the dogma of homogeneous cells mainly involved in the prime defence against pathogens, shedding light on their pathogenic role in inflammatory diseases and on the importance of antineutrophil-cytoplasmic antibodies' pathogenic role in ANCA-associated vasculitides vasculitis (AAV). Myeloperoxidase (MPO) and proteinase 3 (PR3) expressed in neutrophil granulocytes are the most common targets for ANCAs and contribute to the formation of MPO-ANCAs and PR3-ANCAs which, released to the bloodstream, become an excellent diagnostic tool for AAV. In this study, we focus on increasing the clinical and experimental evidence that supports the pathogenic role of ANCAs in AAV. Additionally, we discuss the diagnostic utility of ANCAs for disease activity and prognosis in AAV. Understanding the central role of ANCAs in AAV is crucial for advancing our knowledge of these complex disorders and developing targeted therapeutic strategies in the era of personalized medicine.
Collapse
Affiliation(s)
- Agata Walulik
- Student Scientific Group of Adult Allergology and Internal Medicine, Wroclaw Medical University, 50-369 Wrocław, Poland; (A.W.); (I.G.)
| | - Kinga Łysak
- Faculty of Medicine, Medical University of Gdansk, 80-210 Gdańsk, Poland;
| | - Michał Błaszkiewicz
- Student Scientific Group of Adult Allergology and Internal Medicine, Wroclaw Medical University, 50-369 Wrocław, Poland; (A.W.); (I.G.)
| | - Ignacy Górecki
- Student Scientific Group of Adult Allergology and Internal Medicine, Wroclaw Medical University, 50-369 Wrocław, Poland; (A.W.); (I.G.)
| | - Krzysztof Gomułka
- Department of Internal Medicine, Pneumology and Allergology, Wroclaw Medical University, 50-369 Wrocław, Poland
| |
Collapse
|
18
|
Tang P, Wang H. Regulation of erythropoiesis: emerging concepts and therapeutic implications. Hematology 2023; 28:2250645. [PMID: 37639548 DOI: 10.1080/16078454.2023.2250645] [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: 04/19/2023] [Accepted: 08/17/2023] [Indexed: 08/31/2023] Open
Abstract
The process of erythropoiesis is complex and involves the transfer of cells from the yolk sac to the fetal hepar and, ultimately, to the bone marrow during embryonic development. Within the bone marrow, erythroid progenitor cells undergo several stages to generate reticulocytes that enter the bloodstream. Erythropoiesis is regulated by various factors, with erythropoietin (EPO) synthesized by the kidney being the promoting factor and hepcidin synthesized by the hepar inhibiting iron mobilization. Transcription factors, such as GATA and KLF, also play a crucial role in erythropoiesis. Disruption of any of these factors can lead to abnormal erythropoiesis, resulting in red cell excess, red cell deficiency, or abnormal morphological function. This review provides a general description of erythropoiesis, as well as its regulation, highlighting the significance of understanding the process for the diagnosis and treatment of various hematological disorders.
Collapse
Affiliation(s)
- Pu Tang
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin, People's Republic of China
| | - Huaquan Wang
- Department of Hematology, General Hospital, Tianjin Medical University, Tianjin, People's Republic of China
| |
Collapse
|
19
|
Han Y, Wang S, Wang Y, Huang Y, Gao C, Guo X, Chen L, Zhao H, An X. Comprehensive Characterization and Global Transcriptome Analysis of Human Fetal Liver Terminal Erythropoiesis. GENOMICS, PROTEOMICS & BIOINFORMATICS 2023; 21:1117-1132. [PMID: 37657739 PMCID: PMC11082260 DOI: 10.1016/j.gpb.2023.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 05/19/2023] [Accepted: 08/26/2023] [Indexed: 09/03/2023]
Abstract
The fetal liver (FL) is the key erythropoietic organ during fetal development, but knowledge on human FL erythropoiesis is very limited. In this study, we sorted primary erythroblasts from FL cells and performed RNA sequencing (RNA-seq) analyses. We found that temporal gene expression patterns reflected changes in function during primary human FL terminal erythropoiesis. Notably, the expression of genes enriched in proteolysis and autophagy was up-regulated in orthochromatic erythroblasts (OrthoEs), suggesting the involvement of these pathways in enucleation. We also performed RNA-seq of in vitro cultured erythroblasts derived from FL CD34+ cells. Comparison of transcriptomes between the primary and cultured erythroblasts revealed significant differences, indicating impacts of the culture system on gene expression. Notably, the expression of lipid metabolism-related genes was increased in cultured erythroblasts. We further immortalized erythroid cell lines from FL and cord blood (CB) CD34+ cells (FL-iEry and CB-iEry, respectively). FL-iEry and CB-iEry were immortalized at the proerythroblast stage and can be induced to differentiate into OrthoEs, but their enucleation ability was very low. Comparison of the transcriptomes between OrthoEs with and without enucleation capability revealed the down-regulation of pathways involved in chromatin organization and mitophagy in OrthoEs without enucleation capacity, indicating that defects in chromatin organization and mitophagy contribute to the inability of OrthoEs to enucleate. Additionally, the expression of HBE1, HBZ, and HBG2 was up-regulated in FL-iEry compared with CB-iEry, and such up-regulation was accompanied by down-regulated expression of BCL11A and up-regulated expression of LIN28B and IGF2BP1. Our study provides new insights into human FL erythropoiesis and rich resources for future studies.
Collapse
Affiliation(s)
- Yongshuai Han
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065, USA
| | - Shihui Wang
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065, USA; Institute of Hematology, People's Hospital of Zhengzhou University, Zhengzhou 450003, China
| | - Yaomei Wang
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065, USA; Department of Hematology, the Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Yumin Huang
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065, USA; Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, China
| | - Chengjie Gao
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065, USA
| | - Xinhua Guo
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065, USA
| | - Lixiang Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Huizhi Zhao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Xiuli An
- Laboratory of Membrane Biology, New York Blood Center, New York, NY 10065, USA.
| |
Collapse
|
20
|
Fu Y, Li Z, Lin W, Yao J, Jiang X, Shu Q, Mao X, Tu J, Liang X, Li L. Extramedullary hematopoiesis contributes to enhanced erythropoiesis during pregnancy via TGF-β signaling. Front Immunol 2023; 14:1295717. [PMID: 38045690 PMCID: PMC10693449 DOI: 10.3389/fimmu.2023.1295717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 11/06/2023] [Indexed: 12/05/2023] Open
Abstract
Red blood cells are the predominant cellular component in human body, and their numbers increase significantly during pregnancy due to heightened erythropoiesis. CD71+ erythroid cells (CECs) are immature red blood cells, encompassing erythroblasts and reticulocytes, constitute a rare cell population primarily found in the bone marrow, although they are physiologically enriched in the neonatal mouse spleen and human cord blood. Presently, the mechanisms underlying the CECs expansion during pregnancy remain largely unexplored. Additionally, the mechanisms and roles associated with extramedullary hematopoiesis (EMH) of erythroid cells during pregnancy have yet to be fully elucidated. In this study, our objective was to examine the underlying mechanisms of erythroid-biased hematopoiesis during pregnancy. Our findings revealed heightened erythropoiesis and elevated CECs in both human and mouse pregnancies. The increased presence of transforming growth factor (TGF)-β during pregnancy facilitated the differentiation of CD34+ hematopoietic stem and progenitor cells (HSPCs) into CECs, without impacting HSPCs proliferation, ultimately leading to enhanced erythropoiesis. The observed increase in CECs during pregnancy was primarily attributed to EMH occurring in the spleen. During mouse pregnancy, splenic stromal cells were found to have a significant impact on splenic erythropoiesis through the activation of TGF-β signaling. Conversely, splenic macrophages were observed to contribute to extramedullary erythropoiesis in a TGF-β-independent manner. Our results suggest that splenic stromal cells play a crucial role in promoting extramedullary erythropoiesis and the production of CECs during pregnancy, primarily through TGF-β-dependent mechanisms.
Collapse
Affiliation(s)
- Yao Fu
- Department of Obstetrics, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China
- Post-doctoral Scientific Research Station of Clinical Medicine, Jinan University, Guangzhou, China
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhengjuan Li
- South China University of Technology School of Medicine, Guangzhou, China
| | - Wen Lin
- South China University of Technology School of Medicine, Guangzhou, China
| | - Jingxin Yao
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiang Jiang
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qun Shu
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoyuan Mao
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiaoqin Tu
- Department of Obstetrics and Gynecology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xinyuan Liang
- Department of Obstetrics, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, Shenzhen, China
- Post-doctoral Scientific Research Station of Clinical Medicine, Jinan University, Guangzhou, China
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liping Li
- Department of Obstetrics, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
21
|
Boccacci Y, Dumont N, Doyon Y, Laganière J. Accessory-cell-free differentiation of hematopoietic stem and progenitor cells into mature red blood cells. Cytotherapy 2023; 25:1242-1248. [PMID: 37598334 DOI: 10.1016/j.jcyt.2023.07.007] [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: 02/24/2023] [Revised: 07/14/2023] [Accepted: 07/25/2023] [Indexed: 08/21/2023]
Abstract
BACKGROUND AIMS The culture and ex vivo engineering of red blood cells (RBCs) can help characterize genetic variants, model diseases, and may eventually spur the development of applications in transfusion medicine. In the last decade, improvements to the in vitro production of RBCs have enabled efficient erythroid progenitor proliferation and high enucleation levels from several sources of hematopoietic stem and progenitor cells (HSPCs). Despite these advances, there remains a need for refining the terminal step of in vitro human erythropoiesis, i.e., the terminal maturation of reticulocytes into erythrocytes, so that it can occur without feeder or accessory cells and animal-derived components. METHODS Here, we describe the near-complete erythroid differentiation of cultured RBCs (cRBCs) from adult HSPCs in accessory-cell-free and xeno-free conditions. RESULTS The approach improves post-enucleation cell integrity and cell survival, and it enables subsequent storage of cRBCs for up to 42 days in classical additive solution conditions without any specialized equipment. CONCLUSIONS We foresee that these improvements will facilitate the characterization of RBCs derived from gene-edited HSPCs.
Collapse
Affiliation(s)
- Yelena Boccacci
- Medical Affairs and Innovation, Héma-Qubec, Québec, Quebec, Canada; Centre Hospitalier Universitaire de Québec Research Center, Université Laval, Québec, Quebec, Canada
| | - Nellie Dumont
- Medical Affairs and Innovation, Héma-Qubec, Québec, Quebec, Canada
| | - Yannick Doyon
- Centre Hospitalier Universitaire de Québec Research Center, Université Laval, Québec, Quebec, Canada
| | - Josée Laganière
- Medical Affairs and Innovation, Héma-Qubec, Québec, Quebec, Canada.
| |
Collapse
|
22
|
de Paula CP, de Oliveira da Silva JPM, Romanello KS, Bernardo VS, Torres FF, da Silva DGH, da Cunha AF. Peroxiredoxins in erythrocytes: far beyond the antioxidant role. J Mol Med (Berl) 2023; 101:1335-1353. [PMID: 37728644 DOI: 10.1007/s00109-023-02368-7] [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: 02/23/2023] [Revised: 08/17/2023] [Accepted: 08/31/2023] [Indexed: 09/21/2023]
Abstract
The red blood cells (RBCs) are essential to transport oxygen (O2) and nutrients throughout the human body. Changes in the structure or functioning of the erythrocytes can lead to several deficiencies, such as hemolytic anemias, in which an increase in reactive oxidative species generation is involved in the pathophysiological process, playing a significant role in the severity of several clinical manifestations. There are important lines of defense against the damage caused by oxidizing molecules. Among the antioxidant molecules, the enzyme peroxiredoxin (Prx) has the higher decomposition power of hydrogen peroxide, especially in RBCs, standing out because of its abundance. This review aimed to present the recent findings that broke some paradigms regarding the three isoforms of Prxs found in RBC (Prx1, Prx2, and Prx6), showing that in addition to their antioxidant activity, these enzymes may have supplementary roles in transducing peroxide signals, as molecular chaperones, protecting from membrane damage, and maintenance of iron homeostasis, thus contributing to the overall survival of human RBCs, roles that seen to be disrupted in hemolytic anemia conditions.
Collapse
Affiliation(s)
- Carla Peres de Paula
- Genetics and Evolution Department, Biological and Health Sciences Center, Federal University of São Carlos, São Carlos, Brazil.
- Biotechnology Graduate Program, Exact and Technology Sciences Center, Federal University of São Carlos, São Carlos, Brazil.
| | - João Pedro Maia de Oliveira da Silva
- Genetics and Evolution Department, Biological and Health Sciences Center, Federal University of São Carlos, São Carlos, Brazil
- Evolutionary Genetics and Molecular Biology Graduate Program, Biological and Health Sciences Center, Federal University of São Carlos, São Carlos, Brazil
| | - Karen Simone Romanello
- Genetics and Evolution Department, Biological and Health Sciences Center, Federal University of São Carlos, São Carlos, Brazil
- Evolutionary Genetics and Molecular Biology Graduate Program, Biological and Health Sciences Center, Federal University of São Carlos, São Carlos, Brazil
| | | | | | - Danilo Grünig Humberto da Silva
- Department of Biology, Paulista State University, São Paulo, Brazil
- Federal University of Mato Grosso do Sul, Campus de Três Lagoas, Três Lagoas, Mato Grosso do Sul, Brazil
| | - Anderson Ferreira da Cunha
- Genetics and Evolution Department, Biological and Health Sciences Center, Federal University of São Carlos, São Carlos, Brazil.
| |
Collapse
|
23
|
Lewis JE, Hergott CB. The Immunophenotypic Profile of Healthy Human Bone Marrow. Clin Lab Med 2023; 43:323-332. [PMID: 37481314 DOI: 10.1016/j.cll.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
Flow cytometry enables multiparametric characterization of hematopoietic cell immunophenotype. Deviations from normal immunophenotypic patterns comprise a cardinal feature of many hematopoietic neoplasms, underscoring the ongoing essentiality of flow cytometry as a diagnostic tool. However, understanding of aberrant hematopoiesis requires an equal understanding of normal hematopoiesis as a comparator. In this review, we outline key features of healthy adult hematopoiesis and lineage specification as illuminated by flow cytometry and provide diagrams illustrating what a diagnostician may observe in flow cytometric plots. These features provide a profile of baseline hematopoiesis, to which clinical samples with suspected neoplasia may be compared.
Collapse
Affiliation(s)
- Joshua E Lewis
- Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA; Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA
| | - Christopher B Hergott
- Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA; Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA.
| |
Collapse
|
24
|
Pikora K, Krętowska-Grunwald A, Krawczuk-Rybak M, Sawicka-Żukowska M. Diagnostic Value and Prognostic Significance of Nucleated Red Blood Cells (NRBCs) in Selected Medical Conditions. Cells 2023; 12:1817. [PMID: 37508482 PMCID: PMC10378384 DOI: 10.3390/cells12141817] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/01/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Nucleated red blood cells (NRBCs) are premature erythrocyte precursors that reside in the bone marrow of humans of all ages as an element of erythropoiesis. They rarely present in healthy adults' circulatory systems but can be found circulating in fetuses and neonates. An NRBC count is a cost-effective laboratory test that is currently rarely used in everyday clinical practice; it is mostly used in the diagnosis of hematological diseases/disorders relating to erythropoiesis, anemia, or hemolysis. However, according to several studies, it may be used as a biomarker in the diagnosis and clinical outcome prognosis of preterm infants or severely ill adult patients. This would allow for a quick diagnosis of life-threatening conditions and the prediction of a possible change in a patient's condition, especially in relation to patients in the intensive care unit. In this review, we sought to summarize the possible use of NRBCs as a prognostic marker in various disease entities. Research into the evaluation of the NRBCs in the pediatric population most often concerns neonatal hypoxia, the occurrence and consequences of asphyxia, and overall neonatal mortality. Among adults, NRBCs can be used to predict changes in clinical condition and mortality in critically ill patients, including those with sepsis, trauma, ARDS, acute pancreatitis, or severe cardiovascular disease.
Collapse
Affiliation(s)
- Katarzyna Pikora
- Department of Pediatric Oncology and Hematology, Medical University of Bialystok, Jerzego Waszyngtona 17, 15-274 Bialystok, Poland
| | - Anna Krętowska-Grunwald
- Department of Pediatric Oncology and Hematology, Medical University of Bialystok, Jerzego Waszyngtona 17, 15-274 Bialystok, Poland
| | - Maryna Krawczuk-Rybak
- Department of Pediatric Oncology and Hematology, Medical University of Bialystok, Jerzego Waszyngtona 17, 15-274 Bialystok, Poland
| | - Małgorzata Sawicka-Żukowska
- Department of Pediatric Oncology and Hematology, Medical University of Bialystok, Jerzego Waszyngtona 17, 15-274 Bialystok, Poland
| |
Collapse
|
25
|
Schippel N, Sharma S. Dynamics of human hematopoietic stem and progenitor cell differentiation to the erythroid lineage. Exp Hematol 2023; 123:1-17. [PMID: 37172755 PMCID: PMC10330572 DOI: 10.1016/j.exphem.2023.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/04/2023] [Accepted: 05/07/2023] [Indexed: 05/15/2023]
Abstract
Erythropoiesis, the development of erythrocytes from hematopoietic stem cells, occurs through four phases: erythroid progenitor (EP) development, early erythropoiesis, terminal erythroid differentiation (TED), and maturation. According to the classical model that is based on immunophenotypic profiles of cell populations, each of these phases comprises multiple differentiation states that arise in a hierarchical manner. After segregation of lymphoid potential, erythroid priming begins during progenitor development and progresses through progenitor cell types that have multilineage potential. Complete separation of the erythroid lineage is achieved during early erythropoiesis with the formation of unipotent EPs: burst-forming unit-erythroid and colony-forming unit-erythroid. These erythroid-committed progenitors undergo TED and maturation, which involves expulsion of the nucleus and remodeling to form functional biconcave, hemoglobin-filled erythrocytes. In the last decade or so, many studies employing advanced techniques such as single-cell RNA-sequencing (scRNA-seq) as well as the conventional methods, including colony-forming cell assays and immunophenotyping, have revealed heterogeneity within the stem, progenitor, and erythroblast stages, and uncovered alternate paths for segregation of erythroid lineage potential. In this review, we provide an in-depth account of immunophenotypic profiles of all cell types within erythropoiesis, highlight studies that demonstrate heterogeneous erythroid stages, and describe deviations to the classical model of erythropoiesis. Overall, although scRNA-seq approaches have provided new insights, flow cytometry remains relevant and is the primary method for validation of novel immunophenotypes.
Collapse
Affiliation(s)
- Natascha Schippel
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ
| | - Shalini Sharma
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ.
| |
Collapse
|
26
|
Ganesh K, Joshi MB. Neutrophil sub-types in maintaining immune homeostasis during steady state, infections and sterile inflammation. Inflamm Res 2023; 72:1175-1192. [PMID: 37212866 PMCID: PMC10201050 DOI: 10.1007/s00011-023-01737-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/20/2023] [Accepted: 04/28/2023] [Indexed: 05/23/2023] Open
Abstract
INTRODUCTION Neutrophils are component of innate immune system and a) eliminate pathogens b) maintain immune homeostasis by regulating other immune cells and c) contribute to the resolution of inflammation. Neutrophil mediated inflammation has been described in pathogenesis of various diseases. This indicates neutrophils do not represent homogeneous population but perform multiple functions through confined subsets. Hence, in the present review we summarize various studies describing the heterogeneous nature of neutrophils and associated functions during steady state and pathological conditions. METHODOLOGY We performed extensive literature review with key words 'Neutrophil subpopulations' 'Neutrophil subsets', Neutrophil and infections', 'Neutrophil and metabolic disorders', 'Neutrophil heterogeneity' in PUBMED. RESULTS Neutrophil subtypes are characterized based on buoyancy, cell surface markers, localization and maturity. Recent advances in high throughput technologies indicate the existence of functionally diverse subsets of neutrophils in bone marrow, blood and tissues in both steady state and pathological conditions. Further, we found proportions of these subsets significantly vary in pathological conditions. Interestingly, stimulus specific activation of signalling pathways in neutrophils have been demonstrated. CONCLUSION Neutrophil sub-populations differ among diseases and hence, mechanisms regulating formation, sustenance, proportions and functions of these sub-types vary between physiological and pathological conditions. Hence, mechanistic insights of neutrophil subsets in disease specific manner may facilitate development of neutrophil-targeted therapies.
Collapse
Affiliation(s)
- Kailash Ganesh
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Planetarium Complex, Manipal, 576104, India
| | - Manjunath B Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Planetarium Complex, Manipal, 576104, India.
| |
Collapse
|
27
|
Li W, Acker JP. CD71 + RBCs: A potential immune mediator in transfusion. Transfus Apher Sci 2023:103721. [PMID: 37173208 DOI: 10.1016/j.transci.2023.103721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Donor - recipient sex - mismatched transfusion is associated with increased mortality. The mechanisms for this are not clear, but it may relate to transfusion-related immunomodulation. Recently, CD71+ erythroid cells (CECs), including reticulocytes (CD71+ RBCs) and erythroblasts, have been identified as potent immunoregulatory cells. The proportion of CD71+ RBCs in the peripheral blood is sufficient to play a potential immunomodulatory role. Differences in the quantity of CD71+ RBCs are dependent on blood donor sex. The total number of CD71+ RBCs in red cell concentrates is also affected by blood manufacturing methods, and storage duration. As a component of the total CECs, CD71+ RBCs can affect innate and adaptive immune cells. Phagocytosed CECs directly reduce TNF-α production from macrophages. CECs can also suppress the production of TNF-α production from antigen presenting cells. Moreover, CECs can suppress T cell proliferation thorough immune mediation and / or direct cell-to-cell interactions. Different in their biophysical features compared to mature RBCs, blood donor CD71+ RBCs may be preferential targets for the macrophages. This report summarizes the currently literature supporting an important role for CD71+ RBCs in adverse transfusion reactions including immune mediation and sepsis.
Collapse
Affiliation(s)
- Wenhui Li
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada
| | - Jason P Acker
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Canada; Innovation and Portfolio Management, Canadian Blood Services, Edmonton.
| |
Collapse
|
28
|
Bou-Fakhredin R, Rivella S, Cappellini MD, Taher AT. Pathogenic Mechanisms in Thalassemia I: Ineffective Erythropoiesis and Hypercoagulability. Hematol Oncol Clin North Am 2023; 37:341-351. [PMID: 36907607 DOI: 10.1016/j.hoc.2022.12.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Erythropoiesis is the physiological process that results in the production of red blood cells (RBCs). In conditions of pathologically altered erythropoiesis or ineffective erythropoiesis, as in the case of β-thalassemia, the reduced ability of erythrocytes to differentiate, survive and deliver oxygen stimulates a state of stress that leads to the ineffective production of RBCs. We herein describe the main features of erythropoiesis and its regulation in addition to the mechanisms behind ineffective erythropoiesis development in β-thalassemia. Finally, we review the pathophysiology of hypercoagulability and vascular disease development in β-thalassemia and the currently available prevention and treatment modalities.
Collapse
Affiliation(s)
- Rayan Bou-Fakhredin
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Stefano Rivella
- Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Maria Domenica Cappellini
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy; UOC General Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Ali T Taher
- Division of Hematology-Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon.
| |
Collapse
|
29
|
Yusoff NA, Abd Hamid Z, Budin SB, Taib IS. Linking Benzene, in Utero Carcinogenicity and Fetal Hematopoietic Stem Cell Niches: A Mechanistic Review. Int J Mol Sci 2023; 24:ijms24076335. [PMID: 37047305 PMCID: PMC10094243 DOI: 10.3390/ijms24076335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/19/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
Previous research reported that prolonged benzene exposure during in utero fetal development causes greater fetal abnormalities than in adult-stage exposure. This phenomenon increases the risk for disease development at the fetal stage, particularly carcinogenesis, which is mainly associated with hematological malignancies. Benzene has been reported to potentially act via multiple modes of action that target the hematopoietic stem cell (HSCs) niche, a complex microenvironment in which HSCs and multilineage hematopoietic stem and progenitor cells (HSPCs) reside. Oxidative stress, chromosomal aberration and epigenetic modification are among the known mechanisms mediating benzene-induced genetic and epigenetic modification in fetal stem cells leading to in utero carcinogenesis. Hence, it is crucial to monitor exposure to carcinogenic benzene via environmental, occupational or lifestyle factors among pregnant women. Benzene is a well-known cause of adult leukemia. However, proof of benzene involvement with childhood leukemia remains scarce despite previously reported research linking incidences of hematological disorders and maternal benzene exposure. Furthermore, accumulating evidence has shown that maternal benzene exposure is able to alter the developmental and functional properties of HSPCs, leading to hematological disorders in fetus and children. Since HSPCs are parental blood cells that regulate hematopoiesis during the fetal and adult stages, benzene exposure that targets HSPCs may induce damage to the population and trigger the development of hematological diseases. Therefore, the mechanism of in utero carcinogenicity by benzene in targeting fetal HSPCs is the primary focus of this review.
Collapse
|
30
|
Gallivan A, Alejandro M, Kanu A, Zekaryas N, Horneman H, Hong LK, Vinchinsky E, Lavelle D, Diamond AM, Molokie RE, Ramasamy J, Rivers A. Reticulocyte mitochondrial retention increases reactive oxygen species and oxygen consumption in mouse models of sickle cell disease and phlebotomy-induced anemia. Exp Hematol 2023:S0301-472X(23)00033-4. [PMID: 36934777 DOI: 10.1016/j.exphem.2023.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 03/18/2023]
Abstract
Sickle cell disease (SCD) is caused by a mutation of the β-globin gene that results in the production of hemoglobin S (HbS). People with SCD experience anemia, severe acute pain episodes, persistent chronic pain, multiorgan damage, and a reduced life span. The pathophysiology of SCD caused by the polymerization of HbS on deoxygenation results in red cell deformability and the generation of reactive oxygen species (ROS). These 2 factors lead to red cell fragility and hemolysis. Reticulocytosis is an independent predictor of disease morbidity and mortality in SCD. We previously established that humans and mice with SCD exhibit abnormal mitochondrial retention in erythrocytes increasing ROS-associated hemolysis. Here, we investigated the hypothesis that mitochondrial retention and increased ROS are a consequence of stress erythropoiesis. Our results show clearly that stress erythropoiesis in phlebotomized, anemic AA mice results in mitochondrial retention and increased ROS in reticulocytes. We observed that elevated mitochondrial retention in reticulocytes also alters oxygen consumption and potentially contributes to increased HbS polymerization and red blood cell hemolysis. Therefore, these events occurring due to stress erythropoiesis contribute significantly to the pathology of SCD and suggest new therapeutic targets.
Collapse
Affiliation(s)
- Anne Gallivan
- UCSF Benioff Children's Hospital Oakland, Oakland, CA
| | | | - Amarachi Kanu
- UCSF Benioff Children's Hospital Oakland, Oakland, CA
| | | | - Hart Horneman
- UCSF Benioff Children's Hospital Oakland, Oakland, CA
| | | | | | - Don Lavelle
- University of Illinois at Chicago, Chicago, IL; Jesse Brown VA Medical Center, Chicago, IL
| | | | - Robert E Molokie
- University of Illinois at Chicago, Chicago, IL; Jesse Brown VA Medical Center, Chicago, IL
| | | | - Angela Rivers
- UCSF Benioff Children's Hospital Oakland, Oakland, CA.
| |
Collapse
|
31
|
Maag AH, Swanton H, Kull M, Vegi NM, Feuring M. Immunophenotypical profiling of myeloid neoplasms with erythroid predominance using mass cytometry (CyTOF). Cytometry A 2023. [PMID: 36647792 DOI: 10.1002/cyto.a.24716] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 12/10/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023]
Abstract
Acute erythroid leukemia (AEL) is a disease continuum between Myelodysplastic syndrome (MDS) and Acute myeloid leukemia (AML) with the cellular hallmark of uncontrolled proliferation and impaired differentiation of erythroid progenitor cells. First described by Giovanni di Guglielmo in 1917 AEL accounts for less than 5% of all de novo AML cases. There have been efforts to characterize AEL at a molecular level, describing recurrent alterations in TP53, NPM1 and FLT3 genes. A genomic analysis of AEL cases confirmed its complexity. Despite these advances, the biology underlying erythroid proliferations remains unclear and the prognosis is dismal with a median survival of only 3 months for pure erythroid leukemia (PEL). Marker combinations suitable for the identification and characterization of leukemic stem cell (LSC) candidates, monitoring measurable residual disease (MRD) during chemotherapy treatment and the development of innovative targeted therapies are missing. Here, we developed a mass cytometry panel for an in-depth characterization of erythroid and myeloid blast cell populations from human AEL bone marrow samples in comparison to other AML subtypes and healthy counterparts. A total of 8 AEL samples were analyzed and compared to 28 AML samples from different molecular subtypes, healthy bone marrow counterparts (n = 5) and umbilical cord blood (n = 6) using high-dimensional mass cytometry. Identification of erythroid and myeloid blast populations in high-dimensional mass cytometry data enabled a refined view on erythroblast differentiation stages present in AEL erythroid blasts and revealed immunophenotypical profiles specific to AEL. Profiling of phenotypic LSCs revealed aberrant erythroid marker expression in the CD34+ CD38- stem cell compartment. In addition, the identification of novel candidate surface marker combinations and aberrancies might enhance clinical diagnostics of AEL. We present a high-parameter mass cytometry approach feasible for immunophenotypical analysis of blast and stem cell populations in myeloid neoplasms with erythroid predominance laying the foundation for more precise experimental approaches in the future.
Collapse
Affiliation(s)
- Abdul-Habib Maag
- Comprehensive Cancer Center, Institute of Experimental Cancer Research, University Hospital of Ulm, Ulm, Germany
| | - Helen Swanton
- Comprehensive Cancer Center, Institute of Experimental Cancer Research, University Hospital of Ulm, Ulm, Germany
| | - Miriam Kull
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Naidu M Vegi
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Michaela Feuring
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| |
Collapse
|
32
|
Song P, Cai YC, Chen MX, Chen SH, Chen JX. Enhanced phosphatidylserine exposure and erythropoiesis in Babesia microti-infected mice. Front Microbiol 2023; 13:1083467. [PMID: 36687590 PMCID: PMC9846230 DOI: 10.3389/fmicb.2022.1083467] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 12/08/2022] [Indexed: 12/02/2023] Open
Abstract
INTRODUCTION Babesia microti (B. microti) is the dominant species responsible for human babesiosis, which is associated with severe hemolytic anemia and splenomegaly because it infects mammalian erythrocytes. The actual prevalence of B. microti is thought to have been substantially underestimated. METHODS In this study, Bagg's albino/c (BALB/c) mice were intraperitoneally injected with B. microti-infected erythrocytes, and parasitemia was subsequently measured by calculating the proportion of infected erythrocytes. The ultrastructure of infected erythrocytes was observed using scanning and transmission electron microscopes. Quantifying phosphatidylserine (PS) exposure, oxidative stress, intracellular Ca2+, and erythropoiesis of erythrocytes were done using flow cytometry. The physiological indicators were analyzed using a Mindray BC-5000 Vet automatic hematology analyzer. RESULTS Of note, 40.7 ± 5.9% of erythrocytes changed their structure and shrunk in the B. microti-infected group. The percentage of annexin V-positive erythrocytes and the levels of reactive oxygen species (ROS) in the erythrocytes were higher in the B. microti-infected group than in the control group at 10 dpi. Significant splenomegaly and severe anemia were also observed following B. microti infection. The parasitemia level in the B. microti-infected splenectomized group was higher than that of the B. microti-infected sham group. The population of early erythroblasts increased, and the late erythroblasts decreased in both the bone marrow and spleen tissues of the B. microti-infected group at 10 dpi. DISCUSSION PS exposure and elevated ROS activities were hallmarks of eryptosis in the B. microti-infected group. This study revealed for the first time that B. microti could also induce eryptosis. At the higher parasitemia phase, the occurrence of severe anemia and significant changes in the abundance of erythroblasts in B. microti-infected mice group were established. The spleen plays a critical protective role in controlling B. microti infection and preventing anemia. B. microti infection could cause a massive loss of late erythroblasts and induce erythropoiesis.
Collapse
Affiliation(s)
- Peng Song
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
- NHC Key Laboratory of Parasite and Vector Biology, Ministry of Public Health, Shanghai, China
- WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, China
- Hainan Tropical Diseases Research Center (Chinese Center for Tropical Diseases Research, Hainan), Haikou, Hainan, China
| | - Yu-Chun Cai
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
- NHC Key Laboratory of Parasite and Vector Biology, Ministry of Public Health, Shanghai, China
- WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, China
| | - Mu-Xin Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
- NHC Key Laboratory of Parasite and Vector Biology, Ministry of Public Health, Shanghai, China
- WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, China
- Hainan Tropical Diseases Research Center (Chinese Center for Tropical Diseases Research, Hainan), Haikou, Hainan, China
| | - Shao-Hong Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
- NHC Key Laboratory of Parasite and Vector Biology, Ministry of Public Health, Shanghai, China
- WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, China
| | - Jia-Xu Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
- NHC Key Laboratory of Parasite and Vector Biology, Ministry of Public Health, Shanghai, China
- WHO Collaborating Centre for Tropical Diseases, National Center for International Research on Tropical Diseases, Ministry of Science and Technology, Shanghai, China
- Hainan Tropical Diseases Research Center (Chinese Center for Tropical Diseases Research, Hainan), Haikou, Hainan, China
| |
Collapse
|
33
|
Cappellini MD, Taher AT, Verma A, Shah F, Hermine O. Erythropoiesis in lower-risk myelodysplastic syndromes and beta-thalassemia. Blood Rev 2022; 59:101039. [PMID: 36577601 DOI: 10.1016/j.blre.2022.101039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
The hematologic disorders myelodysplastic syndromes and beta-thalassemia are characterized by ineffective erythropoiesis and anemia, often managed with regular blood transfusions. Erythropoiesis, the process by which sufficient numbers of functional erythrocytes are produced from hematopoietic stem cells, is highly regulated, and defects can negatively affect the proliferation, differentiation, and survival of erythroid precursors. Treatments that directly target the underlying mechanisms of ineffective erythropoiesis are limited, and management of anemia with regular blood transfusions imposes a significant burden on patients, caregivers, and health care systems. There is therefore a strong unmet need for treatments that can restore effective erythropoiesis. Novel therapies are beginning to address this need by targeting a variety of mechanisms underlying erythropoiesis. Herein, we provide an overview of the role of ineffective erythropoiesis in myelodysplastic syndromes and beta-thalassemia, discuss unmet needs in targeting ineffective erythropoiesis, and describe current management strategies and emerging treatments for these disorders.
Collapse
Affiliation(s)
| | - Ali T Taher
- Department of Internal Medicine, American University of Beirut Medical Center, Halim and Aida Daniel Academic and Clinical Center, Beirut, Lebanon.
| | - Amit Verma
- Albert Einstein College of Medicine, New York, NY, USA.
| | - Farrukh Shah
- Department of Haematology, Whittington Health NHS Trust, London, UK.
| | - Olivier Hermine
- Department of Hematology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, University Paris Cité, Paris, France; INSERM U1163 and CNRS 8254, Imagine Institute, Université Sorbonne Paris Cité, Paris, France.
| |
Collapse
|
34
|
Bouthelier A, Fernández-Arroyo L, Mesa-Ciller C, Cibrian D, Martín-Cófreces NB, Castillo-González R, Calero M, Herráez-Aguilar D, Guajardo-Grence A, Pacheco AM, Marcos-Jiménez A, Quiroga B, Morado M, Monroy F, Muñoz-Calleja C, Sánchez-Madrid F, Urrutia AA, Aragonés J. Erythroid SLC7A5/SLC3A2 amino acid carrier controls red blood cell size and maturation. iScience 2022; 26:105739. [PMID: 36582828 PMCID: PMC9792907 DOI: 10.1016/j.isci.2022.105739] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 10/31/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Inhibition of the heterodimeric amino acid carrier SLC7A5/SLC3A2 (LAT1/CD98) has been widely studied in tumor biology but its role in physiological conditions remains largely unknown. Here we show that the SLC7A5/SLC3A2 heterodimer is constitutively present at different stages of erythroid differentiation but absent in mature erythrocytes. Administration of erythropoietin (EPO) further induces SLC7A5/SLC3A2 expression in circulating reticulocytes, as it also occurs in anemic conditions. Although Slc7a5 gene inactivation in the erythrocyte lineage does not compromise the total number of circulating red blood cells (RBCs), their size and hemoglobin content are significantly reduced accompanied by a diminished erythroblast mTORC1 activity. Furthermore circulating Slc7a5-deficient reticulocytes are characterized by lower transferrin receptor (CD71) expression as well as mitochondrial activity, suggesting a premature transition to mature RBCs. These data reveal that SLC7A5/SLC3A2 ensures adequate maturation of reticulocytes as well as the proper size and hemoglobin content of circulating RBCs.
Collapse
Affiliation(s)
- Antonio Bouthelier
- Research Unit, Hospital of Santa Cristina, Research Institute Princesa (IP), Autonomous University of Madrid, 28009 Madrid, Spain
| | - Lucía Fernández-Arroyo
- Research Unit, Hospital of Santa Cristina, Research Institute Princesa (IP), Autonomous University of Madrid, 28009 Madrid, Spain
| | - Claudia Mesa-Ciller
- Research Unit, Hospital of Santa Cristina, Research Institute Princesa (IP), Autonomous University of Madrid, 28009 Madrid, Spain
| | - Danay Cibrian
- Immunology Department, Hospital de la Princesa, Instituto Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, Madrid, Spain,Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Noa Beatriz Martín-Cófreces
- Immunology Department, Hospital de la Princesa, Instituto Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, Madrid, Spain,Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Raquel Castillo-González
- Immunology Department, Hospital de la Princesa, Instituto Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, Madrid, Spain,Department of Vascular Biology and Inflammation, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain,Pathology Anatomy Department, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Macarena Calero
- Departamento de Química Física, Universidad Complutense de Madrid, Ciudad Universitaria s/n, Madrid, Spain,Translational Biophysics. Instituto de Investigación Sanitaria Hospital Doce de Octubre (Imas12), Madrid, Spain
| | - Diego Herráez-Aguilar
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Ctra. Pozuelo-Majadahonda Km 1,800, 28223, Pozuelo de Alarcón, Madrid, Spain
| | - Andrea Guajardo-Grence
- Research Unit, Hospital of Santa Cristina, Research Institute Princesa (IP), Autonomous University of Madrid, 28009 Madrid, Spain
| | - Ana María Pacheco
- Research Unit, Hospital of Santa Cristina, Research Institute Princesa (IP), Autonomous University of Madrid, 28009 Madrid, Spain
| | - Ana Marcos-Jiménez
- Immunology Department, Hospital de la Princesa, Instituto Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Borja Quiroga
- Nephrology Department, Hospital de la Princesa, Instituto Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Marta Morado
- Hematology Department, Hospital Universitario La Paz, Madrid, Spain
| | - Francisco Monroy
- Departamento de Química Física, Universidad Complutense de Madrid, Ciudad Universitaria s/n, Madrid, Spain,Translational Biophysics. Instituto de Investigación Sanitaria Hospital Doce de Octubre (Imas12), Madrid, Spain
| | - Cecilia Muñoz-Calleja
- Immunology Department, Hospital de la Princesa, Instituto Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, Madrid, Spain
| | - Francisco Sánchez-Madrid
- Immunology Department, Hospital de la Princesa, Instituto Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, Madrid, Spain,Nephrology Department, Hospital de la Princesa, Instituto Investigación Sanitaria Princesa, Universidad Autónoma de Madrid, Madrid, Spain,CIBER de Enfermedades Cardiovasculares, Carlos III Health Institute, Madrid, Spain
| | - Andrés A. Urrutia
- Research Unit, Hospital of Santa Cristina, Research Institute Princesa (IP), Autonomous University of Madrid, 28009 Madrid, Spain
| | - Julián Aragonés
- Research Unit, Hospital of Santa Cristina, Research Institute Princesa (IP), Autonomous University of Madrid, 28009 Madrid, Spain,CIBER de Enfermedades Cardiovasculares, Carlos III Health Institute, Madrid, Spain,Corresponding author
| |
Collapse
|
35
|
Yoshinaga M, Han K, Morgens DW, Horii T, Kobayashi R, Tsuruyama T, Hia F, Yasukura S, Kajiya A, Cai T, Cruz PHC, Vandenbon A, Suzuki Y, Kawahara Y, Hatada I, Bassik MC, Takeuchi O. The N 6-methyladenosine methyltransferase METTL16 enables erythropoiesis through safeguarding genome integrity. Nat Commun 2022; 13:6435. [PMID: 36307435 PMCID: PMC9616860 DOI: 10.1038/s41467-022-34078-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 10/12/2022] [Indexed: 02/07/2023] Open
Abstract
During erythroid differentiation, the maintenance of genome integrity is key for the success of multiple rounds of cell division. However, molecular mechanisms coordinating the expression of DNA repair machinery in erythroid progenitors are poorly understood. Here, we discover that an RNA N6-methyladenosine (m6A) methyltransferase, METTL16, plays an essential role in proper erythropoiesis by safeguarding genome integrity via the control of DNA-repair-related genes. METTL16-deficient erythroblasts exhibit defective differentiation capacity, DNA damage and activation of the apoptotic program. Mechanistically, METTL16 controls m6A deposition at the structured motifs in DNA-repair-related transcripts including Brca2 and Fancm mRNAs, thereby upregulating their expression. Furthermore, a pairwise CRISPRi screen revealed that the MTR4-nuclear RNA exosome complex is involved in the regulation of METTL16 substrate mRNAs in erythroblasts. Collectively, our study uncovers that METTL16 and the MTR4-nuclear RNA exosome act as essential regulatory machinery to maintain genome integrity and erythropoiesis.
Collapse
Affiliation(s)
- Masanori Yoshinaga
- grid.258799.80000 0004 0372 2033Department of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501 Japan
| | - Kyuho Han
- grid.168010.e0000000419368956Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - David W. Morgens
- grid.168010.e0000000419368956Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Takuro Horii
- grid.256642.10000 0000 9269 4097Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, 371-8512 Japan
| | - Ryosuke Kobayashi
- grid.256642.10000 0000 9269 4097Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, 371-8512 Japan
| | - Tatsuaki Tsuruyama
- grid.258799.80000 0004 0372 2033Department of Drug and Discovery Medicine, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501 Japan
| | - Fabian Hia
- grid.258799.80000 0004 0372 2033Department of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501 Japan
| | - Shota Yasukura
- grid.258799.80000 0004 0372 2033Department of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501 Japan
| | - Asako Kajiya
- grid.258799.80000 0004 0372 2033Department of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501 Japan
| | - Ting Cai
- grid.258799.80000 0004 0372 2033Department of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501 Japan
| | - Pedro H. C. Cruz
- grid.136593.b0000 0004 0373 3971Department of RNA Biology and Neuroscience, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Alexis Vandenbon
- grid.258799.80000 0004 0372 2033Laboratory of Tissue Homeostasis, Institute for Life and Medical Sciences, Kyoto University, Kyoto, 606-8507 Japan
| | - Yutaka Suzuki
- grid.26999.3d0000 0001 2151 536XLaboratory of Functional Genomics, Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, 277-8562 Japan
| | - Yukio Kawahara
- grid.136593.b0000 0004 0373 3971Department of RNA Biology and Neuroscience, Graduate School of Medicine, Osaka University, Osaka, 565-0871 Japan
| | - Izuho Hatada
- grid.256642.10000 0000 9269 4097Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, 371-8512 Japan ,grid.256642.10000 0000 9269 4097Viral Vector Core, Gunma University Initiative for Advanced Research (GIAR), Gunma, 371-8512 Japan
| | - Michael C. Bassik
- grid.168010.e0000000419368956Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305 USA
| | - Osamu Takeuchi
- grid.258799.80000 0004 0372 2033Department of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501 Japan
| |
Collapse
|
36
|
Dumarchey A, Lavazec C, Verdier F. Erythropoiesis and Malaria, a Multifaceted Interplay. Int J Mol Sci 2022; 23:ijms232112762. [PMID: 36361552 PMCID: PMC9657351 DOI: 10.3390/ijms232112762] [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: 09/30/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 01/24/2023] Open
Abstract
One of the major pathophysiologies of malaria is the development of anemia. Although hemolysis and splenic clearance are well described as causes of malarial anemia, abnormal erythropoiesis has been observed in malaria patients and may contribute significantly to anemia. The interaction between inadequate erythropoiesis and Plasmodium parasite infection, which partly occurs in the bone marrow, has been poorly investigated to date. However, recent findings may provide new insights. This review outlines clinical and experimental studies describing different aspects of ineffective erythropoiesis and dyserythropoiesis observed in malaria patients and in animal or in vitro models. We also highlight the various human and parasite factors leading to erythropoiesis disorders and discuss the impact that Plasmodium parasites may have on the suppression of erythropoiesis.
Collapse
Affiliation(s)
- Aurélie Dumarchey
- Inserm U1016, CNRS UMR8104, Université Paris Cité, Institut Cochin, 75014 Paris, France
- Laboratoire d’Excellence GR-Ex, 75015 Paris, France
| | - Catherine Lavazec
- Inserm U1016, CNRS UMR8104, Université Paris Cité, Institut Cochin, 75014 Paris, France
- Laboratoire d’Excellence GR-Ex, 75015 Paris, France
| | - Frédérique Verdier
- Inserm U1016, CNRS UMR8104, Université Paris Cité, Institut Cochin, 75014 Paris, France
- Laboratoire d’Excellence GR-Ex, 75015 Paris, France
- Correspondence:
| |
Collapse
|
37
|
Chen Y, Huang J, Guo Z, Zhu Z, Shao Y, Li L, Yang Y, Yu Y, Liu L, Sun B. Primitive genotypic characteristics in umbilical cord neutrophils identified by single-cell transcriptome profiling and functional prediction. Front Immunol 2022; 13:970909. [PMID: 36105817 PMCID: PMC9464600 DOI: 10.3389/fimmu.2022.970909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/15/2022] [Indexed: 11/17/2022] Open
Abstract
The function and heterogeneity of neutrophils in neonatal umbilical cord blood (UCB) have not been characterized. In this study, we analyzed the neutrophils in UCB and healthy adults using single-cell RNA sequencing analysis for the first time. We found that neutrophils divided into six subpopulations (G2, G3, G4, G5a, G5b, and G5c) with different marker genes and different functions under homeostasis. Compared with healthy adults, neutrophils of UCB were more naïve and have more obvious degranulation and activation functions. Moreover, we found significant differences in the amount and function of G5b cells between healthy adults and UCB. The amount of G5b group in UCB was lower, but it has more degranulation, secretion and activation functions. In addition, we noted a new subset of G5c labeled by CD52, which almost did not exist in UCB. Besides, its differential genes were enriched in terms such as protein synthesis and mRNA transcription. Furthermore, uncharacteristic transcription factors ZNF-276, ZNF-319 and ZNF-354A were identified in our study. In summary, we first examined the heterogeneity and functional diversity of neutrophils in UCB, and these data provided new insights into the mechanism of neutrophil-mediated diseases of neonates and the wider use of neutrophils in UCB.
Collapse
Affiliation(s)
- Yi Chen
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Department of Burns and Plastic Surgery, Affiliated Huaian No.1 People’s Hospital, Nanjing Medical University, Huai’an, China
| | - Jiamin Huang
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Zaiwen Guo
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Zhechen Zhu
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Yiming Shao
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Linbin Li
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Yunxi Yang
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Yanzhen Yu
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Lu Liu
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Bingwei Sun
- Department of Burns and Plastic Surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- *Correspondence: Bingwei Sun,
| |
Collapse
|
38
|
Lu Z, Xu G, Li Y, Lu C, Shen Y, Zhao B. Discovery of N-arylcinnamamides as novel erythroblast enucleation inducers. Bioorg Chem 2022; 128:106105. [PMID: 36031698 DOI: 10.1016/j.bioorg.2022.106105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/12/2022] [Accepted: 08/17/2022] [Indexed: 01/28/2023]
Abstract
Derivation of mature red blood cells (RBCs) from stem cells in vitro is a promising solution to the current shortage of blood supply, in which terminal enucleation is the rate-limiting step. Here we discovered two cinnamamides B8 and B16 showed potential activities of enhancing the enucleation of erythroblasts through the screening of "in-house" compound library. Subsequently, twenty-four N-arylcinnamamides were rationally designed and synthesized on the basis of the structure of B8 and B16, in which N-(9H-carbazol-2-yl)cinnamamide (KS-2) significantly elevated the percentage of reticulocytes in the cultured mouse fetal liver cells in vitro (relative enucleation = 2.43). The underlying mechanism of KS-2 in promoting mouse erythroid enucleation is accelerating the process of cell cycle exit via p53 activation in late stage erythrocytes. These results strongly suggest that compound KS-2 is worthy of further study as a potential erythrocyte enucleation inducer.
Collapse
Affiliation(s)
- Zhiyuan Lu
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Guangsen Xu
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yanxia Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Chunhua Lu
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yuemao Shen
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Baobing Zhao
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| |
Collapse
|
39
|
Peng Y, Tang L, Li Y, Song J, Liu H, Wang P, Zhong Z, Yang Y, Wang S, Chen L, Zhang J, Zhang S, Wang Z, Li M, Liang L, Liu J. Comprehensive proteomic analysis reveals dynamic phospho-profiling in human early erythropoiesis. Br J Haematol 2022; 199:427-442. [PMID: 35974424 DOI: 10.1111/bjh.18407] [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: 02/16/2022] [Revised: 07/25/2022] [Accepted: 08/03/2022] [Indexed: 12/01/2022]
Abstract
Normal early erythropoiesis depends on the precise regulation of protein expression and phosphorylation modification. Dysregulation of protein levels or modification contributes to erythroid disorders. To date, the dynamics of protein phosphorylation profiling across human erythroid development is not fully understood. Here, we characterized quantitative proteomic and phosphoproteomic profiling by tandem mass-tagging technology. We systemically built phospho-expression profiling and expression clusters of 11 414 phosphopeptides for human early erythropoiesis. The standardization methods for multitier integrative analyses revealed multiple functional modules of phosphoproteins (e.g., regulation of the G2/M transition) and active phosphorylated signalling (e.g., cell cycle-related pathways). Our further analysis revealed that CDK family members were the main kinases that phosphorylate substrates in erythroid progenitors and identified that CDK9 played an important role in the proliferation of erythroid progenitors. Collectively, our phosphoproteomic profiling, integrative network analysis and functional studies define landscapes of the phosphoproteome and reveal signalling pathways that are involved in human early erythropoiesis. This study will serve as a valuable resource for further investigations of phosphatase and kinase functions in human erythropoiesis and erythroid-related diseases.
Collapse
Affiliation(s)
- Yuanliang Peng
- Department of Hematology, The Second Xiangya Hospital of Central South University, Molecular Biology Research Center, Center for Medical Genetics, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
| | - Li Tang
- School of Information Science and Engineering, Central South University, Changsha, China
| | - Yanan Li
- Department of Hematology, The Second Xiangya Hospital of Central South University, Molecular Biology Research Center, Center for Medical Genetics, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
| | - Jianhui Song
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Hong Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Pan Wang
- Department of Hematology, The Second Xiangya Hospital of Central South University, Molecular Biology Research Center, Center for Medical Genetics, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
| | - Zhizhou Zhong
- School of Information Science and Engineering, Central South University, Changsha, China
| | - Yifei Yang
- School of Information Science and Engineering, Central South University, Changsha, China
| | - Shihui Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Lixiang Chen
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Ji Zhang
- Department of Clinical Laboratory, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Shijie Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Zi Wang
- Department of Hematology, The Second Xiangya Hospital of Central South University, Molecular Biology Research Center, Center for Medical Genetics, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
| | - Min Li
- School of Information Science and Engineering, Central South University, Changsha, China
| | - Long Liang
- Department of Hematology, The Second Xiangya Hospital of Central South University, Molecular Biology Research Center, Center for Medical Genetics, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
| | - Jing Liu
- Department of Hematology, The Second Xiangya Hospital of Central South University, Molecular Biology Research Center, Center for Medical Genetics, School of Life Sciences, Hunan Province Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
| |
Collapse
|
40
|
Stolla MC, Reilly A, Bergantinos R, Stewart S, Thom N, Clough CA, Wellington R, Stolitenko R, Abkowitz JL, Doulatov S. ATG4A regulates human erythroid maturation and mitochondrial clearance. Blood Adv 2022; 6:3579-3589. [PMID: 35443024 PMCID: PMC9631553 DOI: 10.1182/bloodadvances.2021005910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 03/15/2022] [Indexed: 01/09/2023] Open
Abstract
Autophagy is a self-degradation pathway that is essential for erythropoiesis. During erythroid differentiation, autophagy facilitates the degradation of macromolecules and the programmed clearance of mitochondria. Impaired mitochondrial clearance results in anemia and alters the lifespan of red blood cells in vivo. While several essential autophagy genes contribute to autophagy in erythropoiesis, little is known about erythroid-specific mediators of this pathway. Genetic analysis of primary human erythroid and nonerythroid cells revealed the selective upregulation of the core autophagy gene ATG4A in maturing human erythroid cells. Because the function of ATG4A in erythropoiesis is unknown, we evaluated its role using an ex vivo model of human erythropoiesis. Depletion of ATG4A in primary human hematopoietic stem and progenitor cells selectively impaired erythroid but not myeloid lineage differentiation, resulting in reduced red cell production, delayed terminal differentiation, and impaired enucleation. Loss of ATG4A impaired autophagy and mitochondrial clearance, giving rise to reticulocytes with retained mitochondria and autophagic vesicles. In summary, our study identifies ATG4A as a cell type-specific regulator of autophagy in erythroid development.
Collapse
Affiliation(s)
| | | | | | | | - Neele Thom
- Division of Hematology, Department of Medicine
| | - Courtnee A. Clough
- Division of Hematology, Department of Medicine
- Molecular and Cellular Biology Program
| | - Rachel C. Wellington
- Division of Hematology, Department of Medicine
- Molecular and Cellular Biology Program
| | | | - Janis L. Abkowitz
- Division of Hematology, Department of Medicine
- Institute for Stem Cell and Regenerative Medicine, and
- Department of Genome Sciences, University of Washington, Seattle, WA
| | - Sergei Doulatov
- Division of Hematology, Department of Medicine
- Molecular and Cellular Biology Program
- Institute for Stem Cell and Regenerative Medicine, and
- Department of Genome Sciences, University of Washington, Seattle, WA
| |
Collapse
|
41
|
The Effect of Low-Energy Laser-Driven Ultrashort Pulsed Electron Beam Irradiation on Erythropoiesis and Oxidative Stress in Rats. Int J Mol Sci 2022; 23:ijms23126692. [PMID: 35743135 PMCID: PMC9223873 DOI: 10.3390/ijms23126692] [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: 05/18/2022] [Revised: 06/06/2022] [Accepted: 06/10/2022] [Indexed: 12/04/2022] Open
Abstract
Anemia is a commonly observed consequence of whole-body exposure to a dose of X-ray or gamma irradiation of the order of the mean lethal dose in mammals, and it is an important factor for the determination of the survival of animals. The aim of this study was to unravel the effect of laser-driven ultrashort pulsed electron beam (UPEB) irradiation on the process of erythropoiesis and the redox state in the organism. Wistar rats were exposed to laser-driven UPEB irradiation, after which the level of oxidative stress and the activities of different antioxidant enzymes, as well as blood smears, bone marrow imprints and sections, erythroblastic islets, hemoglobin and hematocrit, hepatic iron, DNA, and erythropoietin levels, were assessed on the 1st, 3rd, 7th, 14th, and 28th days after irradiation. Despite the fact that laser-driven UPEB irradiation requires quite low doses and repetition rates to achieve the LD50 in rats, our findings suggest that whole-body exposure with this new type of irradiation causes relatively mild anemia in rats, with subsequent fast recovery up to the 28th day. Moreover, this novel type of irradiation causes highly intense processes of oxidative stress, which, despite being relatively extinguished, did not reach the physiologically stable level even at the 28th day after irradiation due to the violations in the antioxidant system of the organism.
Collapse
|
42
|
Malczewska-Lenczowska J, Orysiak J, Majorczyk E, Sitkowski D, Starczewski M, Żmijewski P. HIF-1α and NFIA-AS2 Polymorphisms as Potential Determinants of Total Hemoglobin Mass in Endurance Athletes. J Strength Cond Res 2022; 36:1596-1604. [PMID: 35622109 DOI: 10.1519/jsc.0000000000003686] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
ABSTRACT Malczewska-Lenczowska, J, Orysiak, J, Majorczyk, E, Sitkowski, D, Starczewski, M, and Żmijewski, P. HIF-1α and NFIA-AS2 polymorphisms as potential determinants of total hemoglobin mass in endurance athletes. J Strength Cond Res 36(6): 1596-1604, 2022-The aims of this study were to examine (1) the genotype distribution of rs11549465:C>T of the HIF-1α gene and rs1572312:C>A of the NFIA-AS2 gene; (2) the association between the genes and hematological status in endurance-oriented athletes; and (3) the association between the NFIA-AS2 gene and aerobic capacity in cyclists. Two hundred thirty-eight well-trained athletes (female n = 90, male n = 148) participated in the study. Total hemoglobin mass (tHbmass), blood morphology, intravascular volumes, i.e., erythrocyte volume (EV), blood volume (BV) and plasma volume (PV), and aerobic capacity indices, e.g., peak oxygen uptake (V̇o2peak), and power at anaerobic threshold (PAT) were determined. In both studied genes, the CC genotype was predominant. In the HIF-1α gene, there were no differences in genotype and allele distribution among athletes from different disciplines and between sexes. The distribution of genotypes and alleles of the NFIA-AS2 gene differed significantly in male athletes; the frequency of A allele carriers (CA + AA) was significantly higher in cyclists than in rowers and middle- and long-distance runners. The athletes with CC genotype of NF1A-AS2 had significantly higher relative values of: tHbmass (total female athletes, cyclists), PV, BV (cyclists), and EV (total male athletes, cyclists) and PAT (cyclists) than A allele carriers (CA + AA genotypes). In conclusion, our study indicates that NFIA-AS2 rs1572312:C>A polymorphism was associated with hematological status in endurance athletes, as well as aerobic capacity indices in male cyclists. It suggests that this polymorphism may be a determinant of quantity of hemoglobin and intrtavascular volumes, which in turn can have an impact on aerobic performance.
Collapse
Affiliation(s)
| | - Joanna Orysiak
- Department of Ergonomics, Central Institute for Labour Protection, National Research Institute, Warsaw, Poland
| | - Edyta Majorczyk
- Faculty of Physical Education and Physiotherapy, Institute of Physiotherapy, University of Technology, Opole, Poland
| | - Dariusz Sitkowski
- Department of Physiology, Institute of Sport-National Research Institute, Warsaw, Poland; and
| | - Michał Starczewski
- Department of Physiology, Institute of Sport-National Research Institute, Warsaw, Poland; and
| | - Piotr Żmijewski
- Department of Physiology, Institute of Sport-National Research Institute, Warsaw, Poland; and
| |
Collapse
|
43
|
Development and Validation of a Sensitive Flow Cytometric Method for Determining CECs in RBC Products. Clin Chim Acta 2022; 530:119-125. [DOI: 10.1016/j.cca.2022.03.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 11/23/2022]
|
44
|
Russ BS. Anemia: Considerations for the Athletic Population. Strength Cond J 2022. [DOI: 10.1519/ssc.0000000000000712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
45
|
Stevens-Hernandez CJ, Bruce LJ. Reticulocyte Maturation. MEMBRANES 2022; 12:311. [PMID: 35323786 PMCID: PMC8953437 DOI: 10.3390/membranes12030311] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/03/2022] [Accepted: 03/08/2022] [Indexed: 02/04/2023]
Abstract
Changes to the membrane proteins and rearrangement of the cytoskeleton must occur for a reticulocyte to mature into a red blood cell (RBC). Different mechanisms of reticulocyte maturation have been proposed to reduce the size and volume of the reticulocyte plasma membrane and to eliminate residual organelles. Lysosomal protein degradation, exosome release, autophagy and the extrusion of large autophagic-endocytic hybrid vesicles have been shown to contribute to reticulocyte maturation. These processes may occur simultaneously or perhaps sequentially. Reticulocyte maturation is incompletely understood and requires further investigation. RBCs with membrane defects or cation leak disorders caused by genetic variants offer an insight into reticulocyte maturation as they present characteristics of incomplete maturation. In this review, we compare the structure of the mature RBC membrane with that of the reticulocyte. We discuss the mechanisms of reticulocyte maturation with a focus on incomplete reticulocyte maturation in red cell variants.
Collapse
Affiliation(s)
- Christian J Stevens-Hernandez
- Bristol Institute for Transfusion Sciences, National Health Service (NHS) Blood and Transplant, Bristol BS34 7QH, UK
- School of Biochemistry, University of Bristol, Bristol BS8 ITD, UK
| | - Lesley J Bruce
- Bristol Institute for Transfusion Sciences, National Health Service (NHS) Blood and Transplant, Bristol BS34 7QH, UK
| |
Collapse
|
46
|
Multi-Omics Analysis in β-Thalassemia Using an HBB Gene-Knockout Human Erythroid Progenitor Cell Model. Int J Mol Sci 2022; 23:ijms23052807. [PMID: 35269949 PMCID: PMC8911073 DOI: 10.3390/ijms23052807] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/21/2021] [Accepted: 12/27/2021] [Indexed: 12/21/2022] Open
Abstract
β-thalassemia is a hematologic disease that may be associated with significant morbidity and mortality. Increased expression of HBG1/2 can ameliorate the severity of β-thalassemia. Compared to the unaffected population, some β-thalassemia patients display elevated HBG1/2 expression levels in their red blood cells. However, the magnitude of up-regulation does not reach the threshold of self-healing, and thus, the molecular mechanism underlying HBG1/2 expression in the context of HBB-deficiency requires further elucidation. Here, we performed a multi-omics study examining chromatin accessibility, transcriptome, proteome, and phosphorylation patterns in the HBB homozygous knockout of the HUDEP2 cell line (HBB-KO). We found that up-regulation of HBG1/2 in HBB-KO cells was not induced by the H3K4me3-mediated genetic compensation response. Deletion of HBB in human erythroid progenitor cells resulted in increased ROS levels and production of oxidative stress, which led to an increased rate of apoptosis. Furthermore, in response to oxidative stress, slower cell cycle progression and proliferation were observed. In addition, stress erythropoiesis was initiated leading to increased intracellular HBG1/2 expression. This molecular model was also validated in the single-cell transcriptome of hematopoietic stem cells from β-hemoglobinopathy patients. These findings further the understanding of HBG1/2 gene regulatory networks and provide novel clinical insights into β-thalassemia phenotypic diversity.
Collapse
|
47
|
Ulyanova T, Cherone JM, Sova P, Papayannopoulou T. α4-Integrin deficiency in human CD34+ cells engenders precocious erythroid differentiation but inhibits enucleation. Exp Hematol 2022; 108:16-25. [DOI: 10.1016/j.exphem.2022.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/21/2022] [Accepted: 01/25/2022] [Indexed: 11/26/2022]
|
48
|
Soboleva S, Åkerstrand H, Miharada K. Transcriptomic analysis of functional diversity of human umbilical cord blood hematopoietic stem/progenitor cells in erythroid differentiation. Int J Hematol 2022; 115:481-488. [PMID: 35088351 DOI: 10.1007/s12185-022-03292-1] [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: 11/24/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 11/26/2022]
Abstract
Hematopoietic stem cells (HSC) give rise to all types of blood lineages, including red blood cells (RBC). Hematopoietic stem/progenitor cells (HSPC) are known to be functionally diverse in terms of their self-renewal potential and lineage output. Consequently, investigation of molecular heterogeneity in the differentiation potential of HSPC is vital to identify novel regulators that affect generation of specific cell types, especially RBC. Here, we compared the erythroid potential of CD34+ hematopoietic stem and progenitor cells from 50 different umbilical cord blood (UCB) donors and discovered that those donors gave rise to diverse frequencies of Glycophorin-A+ erythroid cells after in vitro differentiation, despite having similar frequencies of phenotypic HSC initially. RNA sequencing revealed that genes involved in G protein-coupled receptor (GPCR) signaling were significantly up-regulated in the high-erythroid output donors. When we chemically modified two main signaling elements in this pathway, adenylyl cyclase (AC) and phosphodiesterase (PDE), we observed that inhibition of PDE led to 10 times higher yield of Glycophorin-A+ cells than activation of AC. Our findings suggest that GPCR signaling, and particularly the cAMP-related pathway, contributes to the diversity of erythroid potential among UCB donors.
Collapse
Affiliation(s)
- Svetlana Soboleva
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Hugo Åkerstrand
- Division of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Kenichi Miharada
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden.
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
49
|
Zhao H, Wei Z, Shen G, Chen Y, Hao X, Li S, Wang R. Poly(rC)-binding proteins as pleiotropic regulators in hematopoiesis and hematological malignancy. Front Oncol 2022; 12:1045797. [PMID: 36452487 PMCID: PMC9701828 DOI: 10.3389/fonc.2022.1045797] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/26/2022] [Indexed: 11/16/2022] Open
Abstract
Poly(rC)-binding proteins (PCBPs), a defined subfamily of RNA binding proteins, are characterized by their high affinity and sequence-specific interaction with poly-cytosine (poly-C). The PCBP family comprises five members, including hnRNP K and PCBP1-4. These proteins share a relatively similar structure motif, with triple hnRNP K homology (KH) domains responsible for recognizing and combining C-rich regions of mRNA and single- and double-stranded DNA. Numerous studies have indicated that PCBPs play a prominent role in hematopoietic cell growth, differentiation, and tumorigenesis at multiple levels of regulation. Herein, we summarized the currently available literature regarding the structural and functional divergence of various PCBP family members. Furthermore, we focused on their roles in normal hematopoiesis, particularly in erythropoiesis. More importantly, we also discussed and highlighted their involvement in carcinogenesis, including leukemia and lymphoma, aiming to clarify the pleiotropic roles and molecular mechanisms in the hematopoietic compartment.
Collapse
Affiliation(s)
- Huijuan Zhao
- Henan International Joint Laboratory of Thrombosis and Hemostasis, Henan University of Science and Technology, Luoyang, China.,Basic Medical College, Henan University of Science and Technology, Luoyang, China
| | - Ziqing Wei
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Guomin Shen
- Henan International Joint Laboratory of Thrombosis and Hemostasis, Henan University of Science and Technology, Luoyang, China.,Basic Medical College, Henan University of Science and Technology, Luoyang, China
| | - Yixiang Chen
- Henan International Joint Laboratory of Thrombosis and Hemostasis, Henan University of Science and Technology, Luoyang, China.,Basic Medical College, Henan University of Science and Technology, Luoyang, China
| | - Xueqin Hao
- Basic Medical College, Henan University of Science and Technology, Luoyang, China
| | - Sanqiang Li
- Basic Medical College, Henan University of Science and Technology, Luoyang, China
| | - Rong Wang
- Department of Clinical Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
50
|
Lu Z, Huang L, Li Y, Xu Y, Zhang R, Zhou Q, Sun Q, Lu Y, Chen J, Shen Y, Li J, Zhao B. Fine-Tuning of Cholesterol Homeostasis Controls Erythroid Differentiation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2102669. [PMID: 34739188 PMCID: PMC8805577 DOI: 10.1002/advs.202102669] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 10/01/2021] [Indexed: 05/12/2023]
Abstract
Lipid metabolism is essential for stemness maintenance, self-renewal, and differentiation of stem cells, however, the regulatory function of cholesterol metabolism in erythroid differentiation is poorly studied. In the present study, a critical role for cholesterol homeostasis in terminal erythropoiesis is uncovered. The master transcriptional factor GATA1 binds to Sterol-regulatory element binding protein 2 (SREBP2) to downregulate cholesterol biosynthesis, leading to a gradual reduction in intracellular cholesterol levels. It is further shown that reduced cholesterol functions to block erythroid proliferation via the cholesterol/mTORC1/ribosome biogenesis axis, which coordinates cell cycle exit in the late stages of erythroid differentiation. The interaction of GATA1 and SREBP2 also provides a feedback loop for regulating globin expression through the transcriptional control of NFE2 by SREBP2. Importantly, it is shown that disrupting intracellular cholesterol hemostasis resulted in defect of terminal erythroid differentiation in vivo. These findings demonstrate that fine-tuning of cholesterol homeostasis emerges as a key mechanism for regulating erythropoiesis.
Collapse
Affiliation(s)
- Zhiyuan Lu
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Lixia Huang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yanxia Li
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yan Xu
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Ruihao Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Qian Zhou
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Qi Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yi Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Junjie Chen
- Analysis and Measurement Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen, Fujian, 361001, China
| | - Yuemao Shen
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jian Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Baobing Zhao
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| |
Collapse
|