1
|
Di Buduo CA, Careddu F, Metti S, Lunghi M, Diprima S, Camilotto V, Bruni G, Gianelli U, Tosi D, Perotti C, Del Fante C, Cazzola M, Braghetta P, Kaplan DL, Minetti G, Malcovati L, Balduini A. In vitro studies of human erythropoiesis using a 3D silk-based bone marrow model that generates erythroblastic islands. Blood Adv 2025; 9:2192-2206. [PMID: 39951616 DOI: 10.1182/bloodadvances.2024014905] [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: 09/23/2024] [Revised: 11/01/2024] [Accepted: 01/05/2025] [Indexed: 02/16/2025] Open
Abstract
ABSTRACT The pursuit of ex vivo erythrocyte generation has led to the development of various culture systems that simulate the bone marrow microenvironment. However, these models often fail to fully replicate the hematopoietic niche's complex dynamics. In our research, we use a comprehensive strategy that emphasizes physiological red blood cell (RBC) differentiation using a minimal cytokine regimen. A key innovation in our approach is the integration of a 3-dimensional (3D) silk-based scaffold engineered to mimic both the physical and chemical properties of human bone marrow. This scaffold facilitates critical macrophage-RBC interactions and incorporates fibronectin functionalization to support the formation of erythroblastic island (EBI)-like niches. We observed diverse stages of erythroblast maturation within these niches, driven by the activation of autophagy, which promotes organelle clearance and membrane remodeling. This process leads to reduced surface integrin expression and significantly enhances RBC enucleation. Using a specialized bioreactor chamber, millions of RBCs can be detached from the EBIs and collected in transfusion bags via dynamic perfusion. Inhibition of autophagy through pharmacological agents or α4 integrin blockade disrupted EBI formation, preventing cells from completing their final morphological transformations, having them trapped in the erythroblast stage. Our findings underscore the importance of the bone marrow niche in maintaining the structural integrity of EBIs and highlight the critical role of autophagy in facilitating organelle clearance during RBC maturation. RNA sequencing analysis further confirmed that these processes are uniquely supported by the 3D silk scaffold, which is essential for enhancing RBC production ex vivo.
Collapse
Affiliation(s)
| | | | - Samuele Metti
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Marco Lunghi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Santo Diprima
- Bioinformatic Division, Center for Omics Sciences, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Virginia Camilotto
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Department of Hematology Oncology, Hematology Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Giovanna Bruni
- Center for Colloid and Surface Science-Department of Chemistry, Physical-Chemistry Section, University of Pavia, Pavia, Italy
| | - Umberto Gianelli
- Department of Health Sciences, University of Milan, Struttura Complessa di Anatomia Patologica, Azienda Socio-Sanitaria Territoriale-Santi Paolo e Carlo, Milan, Italy
| | - Delfina Tosi
- Department of Health Sciences, University of Milan, Struttura Complessa di Anatomia Patologica, Azienda Socio-Sanitaria Territoriale-Santi Paolo e Carlo, Milan, Italy
| | - Cesare Perotti
- Division of Immunohaematology and Transfusion Service, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Claudia Del Fante
- Division of Immunohaematology and Transfusion Service, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Mario Cazzola
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Department of Hematology Oncology, Hematology Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Paola Braghetta
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA
| | - Giampaolo Minetti
- Department of Biology and Biotechnology "Lazzaro Spallanzani," University of Pavia, Pavia, Italy
| | - Luca Malcovati
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Department of Hematology Oncology, Hematology Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
| | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- Department of Biomedical Engineering, Tufts University, Medford, MA
| |
Collapse
|
2
|
Liu Q, Hao T, Lin Z, Fang Y, Li L, Huang D, Wu J, Zhao Y, Zhang X. AZD8055 Is More Effective Than Rapamycin in Inhibiting Proliferation and Promoting Mitochondrial Clearance in Erythroid Differentiation. Anal Cell Pathol (Amst) 2024; 2024:2639464. [PMID: 39411209 PMCID: PMC11479778 DOI: 10.1155/2024/2639464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 07/24/2024] [Accepted: 08/22/2024] [Indexed: 10/19/2024] Open
Abstract
Background: As an important downstream effector of various signaling pathways, mTOR plays critical roles in regulating many physiological processes including erythropoiesis. It is composed of two distinct complexes, mTORC1 and mTORC2, which differ in their components and downstream signaling effects. Our previous study revealed that the inhibition of mTORC1 by rapamycin significantly repressed the erythroid progenitor expansion in the early stage but promoted enucleation and mitochondria clearance in the late stage of erythroid differentiation. However, the particular roles and differences of mTORC1 and mTORC2 in the regulation of erythropoiesis still remain largely unknown. In the present study, we investigated the comparative effects of dual mTORC1/mTORC2 mTOR kinase inhibitor AZD8055 and mTORC1 inhibitor rapamycin on erythroid differentiation in K562 cells induced by hemin and erythropoiesis in β-thalassemia mouse model. Materials and Methods: In vitro erythroid differentiation model of hemin-induced K562 cells and β-thalassemia mouse model were treated with AZD8055 and rapamycin. Cell Counting Kit-8 was used to detect cell viability. The cell proliferation, cell cycle, erythroid surface marker expression, mitochondrial content, and membrane potential were determined and analyzed by flow cytometry and laser scanning confocal microscopy. Globin gene expression during erythroid differentiation was measured by RT-qPCR. The mTORC2/mTORC1 and autophagy pathway was evaluated using western blotting. Results: Both AZD8055 and rapamycin treatments increased the expression levels of the erythroid differentiation-specific markers, CD235a, α-globin, γ-globin, and ε-globin. Notably, AZD8055 suppressed the cell proliferation and promoted the mitochondrial clearance of hemin-induced K562 cells more effectively than rapamycin. In a mouse model of β-thalassemia, both rapamycin and AZD8055 remarkably improve erythroid cell maturation and anemia. Moreover, AZD8055 and rapamycin treatment inhibited the mTORC1 pathway and enhanced autophagy, whereas AZD8055 enhanced autophagy more effectively than rapamycin. Indeed, AZD8055 treatment inhibited both mTORC2 and mTORC1 pathway in hemin-induced K562 cells. Conclusion: AZD8055 is more effective than rapamycin in inhibiting proliferation and promoting mitochondrial clearance in erythroid differentiation, which might provide us one more therapeutic option other than rapamycin for ineffective erythropoiesis treatment in the future. These findings also provide some preliminary information indicating the roles of mTORC1 and mTORC2 in erythropoiesis, and further studies are necessary to dissect the underlying mechanisms.
Collapse
Affiliation(s)
- Qian Liu
- Department of Cardiology, The Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Tao Hao
- Department of Colorectal and Anal Surgery, The Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Ze Lin
- Shantou University Medical College, Shantou, Guangdong Province, China
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China
| | - Yipeng Fang
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Lei Li
- Department of Cardiology, The Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Daqi Huang
- Department of Cardiology, The Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Jianbo Wu
- Department of Cardiology, The Binzhou Medical University Hospital, Binzhou, Shandong Province, China
| | - Yanchao Zhao
- Binzhou Health Commission, Binzhou, Shandong Province, China
| | - Xin Zhang
- Shantou University Medical College, Shantou, Guangdong Province, China
- Department of Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China
- Laboratory of Medical Molecular Imaging, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong Province, China
| |
Collapse
|
3
|
Wang YY, Zou LP, Xu KF, Xu WS, Zhang MN, Lu Q, Tian XL, Pang LY, He W, Wang QH, Gao Y, Liu LY, Chen XQ, Ma SF, Chen HM, Dun S, Yang XY, Luo XM, Huang LL, Li YF. Long-term safety and influence on growth in patients receiving sirolimus: a pooled analysis. Orphanet J Rare Dis 2024; 19:299. [PMID: 39148107 PMCID: PMC11325711 DOI: 10.1186/s13023-024-03243-5] [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: 12/11/2022] [Accepted: 06/05/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND Sirolimus is increasingly utilized in treating diseases associated with mTOR pathway overactivation. Despite its potential, the lack of evidence regarding its long-term safety across all age groups, particularly in pediatric patients, has limited its further application. This study aims to assess the long-term safety of sirolimus, with a specific focus on its impact on growth patterns in pediatric patients. METHODS This pooled analysis inlcudes two prospective cohort studies spanning 10 years, including 1,738 participants (aged 5 days to 69 years) diagnosed with tuberous sclerosis and/or lymphangioleiomyomatosis. All participants were mTOR inhibitor-naive and received 1 mg/m²/day of sirolimus, with dose adjustments during a two-week titration period to maintain trough blood concentrations between 5 and 10 ng/ml (maximum dose 2 mg). Indicators of physical growth, hematopoietic, liver, renal function, and blood lipid levels were all primary outcomes and were analyzed. The adverse events and related management were also recorded. RESULTS Sirolimus administration did not lead to deviations from normal growth ranges, but higher doses exhibited a positive association with Z-scores exceeding 2 SD in height, weight, and BMI. Transient elevations in red blood cell and white blood cell counts, along with hyperlipidemia, were primarily observed within the first year of treatment. Other measured parameters remained largely unchanged, displaying only weak correlations with drug use. Stomatitis is the most common adverse event (920/1738, 52.9%). In adult females, menstrual disorders were observed in 48.5% (112/217). CONCLUSIONS Sirolimus's long-term administration is not associated with adverse effects on children's physical growth pattern, nor significant alterations in hematopoietic, liver, renal function, or lipid levels. A potential dose-dependent influence on growth merits further exploration. TRIAL REGISTRATION Pediatric patients: Chinese clinical trial registry, No. ChiCTR-OOB-15,006,535. Adult patients: ClinicalTrials, No. NCT03193892.
Collapse
Affiliation(s)
- Yang-Yang Wang
- Department of Pediatrics, The First Medical Center of PLA General Hospital, Chinese PLA General Hospital, Beijing, 100853, China
| | - Li-Ping Zou
- Department of Pediatrics, The First Medical Center of PLA General Hospital, Chinese PLA General Hospital, Beijing, 100853, China.
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, 100069, China.
| | - Kai-Feng Xu
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100730, China.
| | - Wen-Shuai Xu
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100730, China
| | - Meng-Na Zhang
- Department of Pediatrics, The First Medical Center of PLA General Hospital, Chinese PLA General Hospital, Beijing, 100853, China
| | - Qian Lu
- Department of Pediatrics, The First Medical Center of PLA General Hospital, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xin-Lun Tian
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100730, China
| | - Ling-Yu Pang
- Department of Neurology, Hebei Children's Hospital, Hebei, China
| | - Wen He
- Department of Pediatrics, The First Medical Center of PLA General Hospital, Chinese PLA General Hospital, Beijing, 100853, China
| | - Qiu-Hong Wang
- Department of Pediatrics, The First Medical Center of PLA General Hospital, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yang Gao
- Deyang People's Hospital, Sichuan, China
| | - Li-Ying Liu
- Department of Pediatrics, The First Medical Center of PLA General Hospital, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xiao-Qiao Chen
- Department of Pediatrics, The 904th Hospital of Joint Logistic Support Force, Jiangsu, China
| | - Shu-Fang Ma
- Department of Pediatrics, Binzhou Medical University Hospital, Shandong, China
| | - Hui-Min Chen
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Shuo Dun
- Department of Pediatrics, The First Medical Center of PLA General Hospital, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xiao-Yan Yang
- Department of Pediatrics, The First Medical Center of PLA General Hospital, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xiao-Mei Luo
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, 100069, China
| | - Lu-Lu Huang
- Department of Pediatrics, Zhujiang Hospital of Southern Medical University, Guangdong, China
| | - Yu-Fen Li
- Department of Pediatrics, Shandong Linyi People's Hospital, Shandong, China
| |
Collapse
|
4
|
Steiner L. Lost in translation. Blood 2024; 144:595-597. [PMID: 39115827 DOI: 10.1182/blood.2024024947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024] Open
|
5
|
Liu Q, Lin Z, Yue M, Wu J, Li L, Huang D, Fang Y, Zhang X, Hao T. Identification and validation of ferroptosis related markers in erythrocyte differentiation of umbilical cord blood-derived CD34 + cell by bioinformatic analysis. Front Genet 2024; 15:1365232. [PMID: 39139819 PMCID: PMC11319168 DOI: 10.3389/fgene.2024.1365232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 07/09/2024] [Indexed: 08/15/2024] Open
Abstract
Ferroptosis has been observed to play an important role during erythrocyte differentiation (ED). However, the biological gene markers and ferroptosis mechanisms in ED remain unknown. We downloaded the datasets of ED in human umbilical cord blood-derived CD34+ cells from the Gene Expression Omnibus database. Using median differentiation time, the sample was categorized into long and short groups. The differentially expressed ferroptosis-related genes (DE-FRGs) were screened using differential expression analysis. The enrichment analyses and a protein-protein interaction (PPI) network were conducted. To predict the ED stage, a logistic regression model was constructed using the least absolute shrinkage and selection operator (LASSO). Overall, 22 DE-FRGs were identified. Ferroptosis-related pathways were enriched using Gene Ontology and the Kyoto Encyclopedia of Genes and Genomes. Gene Set Enrichment Analysis and Gene Set Variation Analysis revealed the primary involvement of DE-FRGs in JAK-STAT, MAPK, PI3K-AKT-mTORC1, WNT, and NOTCH signaling pathways. Ten-hub DE-FRGs were obtained using PPI analysis. Furthermore, we constructed mRNA-microRNA (miRNA) and mRNA-transcription factor networks. Immune cell infiltration levels differed significantly during ED. LASSO regression analysis established a signature using six DE-FRGs (ATF3, CDH2, CHAC1, DDR2, DPP4, and GDF15) related to the ED stage. Bioinformatic analyses identified ferroptosis-associated genes during ED, which were further validated. Overall, we identified ferroptosis-related genes to predict their correlations in ED. Exploring the underlying mechanisms of ferroptosis may help us better understand pathophysiological changes in ED and provide new evidence for clinical transformation.
Collapse
Affiliation(s)
- Qian Liu
- Department of Cardiology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Ze Lin
- Shantou University Medical College, Shantou, Guangdong, China
| | - Minghui Yue
- Shantou University Medical College, Shantou, Guangdong, China
| | - Jianbo Wu
- Department of Cardiology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Lei Li
- Department of Cardiology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Daqi Huang
- Department of Cardiology, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Yipeng Fang
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Zhang
- Shantou University Medical College, Shantou, Guangdong, China
- Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
- Laboratory of Medical Molecular Imaging, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Tao Hao
- Department of Colorectal Surgery, Binzhou Medical University Hospital, Binzhou, Shandong, China
| |
Collapse
|
6
|
Wang Z, Jiang B, Song L, Sun M, Li C, Li X, Zheng W, Tao Y, Sun Q, Qi J. Patients with acquired pure red cell aplasia respond to PI3Kδ inhibitor rapidly. Am J Hematol 2024; 99:1431-1433. [PMID: 38607227 DOI: 10.1002/ajh.27325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 03/22/2024] [Accepted: 04/02/2024] [Indexed: 04/13/2024]
Affiliation(s)
- Zhenzhen Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Bo Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Lin Song
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Mingyuan Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Chunhong Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Xiaoxia Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Weiwei Zheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yuan Tao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Qi Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Junyuan Qi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| |
Collapse
|
7
|
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
|
8
|
Zhang H, Xue F, Zhao H, Chen L, Wang T, Wu X. DNA methylation status of DNAJA4 is essential for human erythropoiesis. Epigenomics 2022; 14:1249-1267. [DOI: 10.2217/epi-2022-0341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Aims: To investigate DNA methylation patterns in early and terminal stages of erythropoiesis, and to explore the function of differentially methylated genes in erythropoiesis and erythroid disorders. Materials & methods: Differential analysis of DNA methylation and gene expression during erythropoiesis, as well as weighted gene coexpression network analysis of acute myeloid leukemia was performed. Results: We identified four candidate genes that possessed differential methylation in the promoter regions. DNAJA4 affected proliferation, apoptosis and enucleation during terminal erythropoiesis and was associated with the prognosis of acute myeloid leukemia. DNAJA4 was specifically highly expressed in erythroleukemia and is associated with DNA methylation. Conclusion: DNAJA4 plays a crucial role for erythropoiesis and is regulated via DNA methylation. Dysregulation of DNAJA4 expression is associated with erythroid disorders.
Collapse
Affiliation(s)
- Hengchao Zhang
- School of Life Sciences, Zhengzhou University, Science Road 100, Zhengzhou, 450001, China
| | - Fumin Xue
- Department of Gastroenterology, Children’s Hospital affiliated of Zhengzhou University, Zhengzhou, 450000, China
| | - Huizhi Zhao
- School of Life Sciences, Zhengzhou University, Science Road 100, Zhengzhou, 450001, China
| | - Lixiang Chen
- School of Life Sciences, Zhengzhou University, Science Road 100, Zhengzhou, 450001, China
| | - Ting Wang
- School of Life Sciences, Zhengzhou University, Science Road 100, Zhengzhou, 450001, China
| | - Xiuyun Wu
- School of Life Sciences, Zhengzhou University, Science Road 100, Zhengzhou, 450001, China
| |
Collapse
|
9
|
Li J, Quan C, He YL, Cao Y, Chen Y, Wang YF, Wu LY. Autophagy regulated by the HIF/REDD1/mTORC1 signaling is progressively increased during erythroid differentiation under hypoxia. Front Cell Dev Biol 2022; 10:896893. [PMID: 36092719 PMCID: PMC9448881 DOI: 10.3389/fcell.2022.896893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
For hematopoietic stem and progenitor cells (HSPCs), hypoxia is a specific microenvironment known as the hypoxic niche. How hypoxia regulates erythroid differentiation of HSPCs remains unclear. In this study, we show that hypoxia evidently accelerates erythroid differentiation, and autophagy plays a pivotal role in this process. We further determine that mTORC1 signaling is suppressed by hypoxia to relieve its inhibition of autophagy, and with the process of erythroid differentiation, mTORC1 activity gradually decreases and autophagy activity increases accordingly. Moreover, we provide evidence that the HIF-1 target gene REDD1 is upregulated to suppress mTORC1 signaling and enhance autophagy, thereby promoting erythroid differentiation under hypoxia. Together, our study identifies that the enhanced autophagy by hypoxia favors erythroid maturation and elucidates a new regulatory pattern whereby autophagy is progressively increased during erythroid differentiation, which is driven by the HIF-1/REDD1/mTORC1 signaling in a hypoxic niche.
Collapse
|
10
|
Effect of Glucocorticosteroids in Diamond-Blackfan Anaemia: Maybe Not as Elusive as It Seems. Int J Mol Sci 2022; 23:ijms23031886. [PMID: 35163808 PMCID: PMC8837118 DOI: 10.3390/ijms23031886] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 02/01/2022] [Accepted: 02/05/2022] [Indexed: 12/22/2022] Open
Abstract
Diamond-Blackfan anaemia (DBA) is a red blood cell aplasia that in the majority of cases is associated with ribosomal protein (RP) aberrations. However, the mechanism by which this disorder leads to such a specific phenotype remains unclear. Even more elusive is the reason why non-specific agents such as glucocorticosteroids (GCs), also known as glucocorticoids, are an effective therapy for DBA. In this review, we (1) explore why GCs are successful in DBA treatment, (2) discuss the effect of GCs on erythropoiesis, and (3) summarise the GC impact on crucial pathways deregulated in DBA. Furthermore, we show that GCs do not regulate DBA erythropoiesis via a single mechanism but more likely via several interdependent pathways.
Collapse
|
11
|
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] [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 SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Lixia Huang
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Yanxia Li
- Key Laboratory of Chemical Biology (Ministry of Education)School of Pharmaceutical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Yan Xu
- Key Laboratory of Chemical Biology (Ministry of Education)School of Pharmaceutical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Ruihao Zhang
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Qian Zhou
- Key Laboratory of Chemical Biology (Ministry of Education)School of Pharmaceutical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Qi Sun
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Yi Lu
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Junjie Chen
- Analysis and Measurement CenterSchool of Pharmaceutical SciencesXiamen UniversityXiamenFujian361001China
| | - Yuemao Shen
- Key Laboratory of Chemical Biology (Ministry of Education)School of Pharmaceutical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Jian Li
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| | - Baobing Zhao
- Key Laboratory of Chemical Biology (Ministry of Education)School of Pharmaceutical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
- Department of PharmacologySchool of Pharmaceutical SciencesCheeloo College of MedicineShandong UniversityJinanShandong250012China
| |
Collapse
|
12
|
Li H, Jiang X, Shen X, Sun Y, Jiang N, Zeng J, Lin J, Yue L, Lai J, Li Y, Wu A, Wang L, Qin D, Huang F, Mei Q, Yang J, Wu J. TMEA, a Polyphenol in Sanguisorba officinalis, Promotes Thrombocytopoiesis by Upregulating PI3K/Akt Signaling. Front Cell Dev Biol 2021; 9:708331. [PMID: 34485295 PMCID: PMC8416095 DOI: 10.3389/fcell.2021.708331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/28/2021] [Indexed: 01/14/2023] Open
Abstract
Thrombocytopenia is closely linked with hemorrhagic diseases, for which induction of thrombopoiesis shows promise as an effective treatment. Polyphenols widely exist in plants and manifest antioxidation and antitumour activities. In this study, we investigated the thrombopoietic effect and mechanism of 3,3′,4′-trimethylellagic acid (TMEA, a polyphenol in Sanguisorba officinalis L.) using in silico prediction and experimental validation. A KEGG analysis indicated that PI3K/Akt signalling functioned as a crucial pathway. Furthermore, the virtual molecular docking results showed high-affinity binding (a docking score of 6.65) between TMEA and mTOR, suggesting that TMEA might target the mTOR protein to modulate signalling activity. After isolation of TMEA, in vitro and in vivo validation revealed that this compound could promote megakaryocyte differentiation/maturation and platelet formation. In addition, it enhanced the phosphorylation of PI3K, Akt, mTOR, and P70S6K and increased the expression of GATA-1 and NF-E2, which confirmed the mechanism prediction. In conclusion, our findings are the first to demonstrate that TMEA may provide a novel therapeutic strategy that relies on the PI3K/Akt/mTOR pathway to facilitate megakaryocyte differentiation and platelet production.
Collapse
Affiliation(s)
- Hong Li
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xueqin Jiang
- School of Pharmacy, Southwest Medical University, Luzhou, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Medical School, Sichuan University, Chengdu, China
| | - Xin Shen
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yueshan Sun
- School of Pharmacy, Southwest Medical University, Luzhou, China.,Medical Research Center, The Third People's Hospital of Chengdu, Chengdu, China
| | - Nan Jiang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jing Zeng
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jing Lin
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Liang Yue
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jia Lai
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yan Li
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Anguo Wu
- School of Pharmacy, Southwest Medical University, Luzhou, China.,The Key Laboratory of Medical Electrophysiology, Medical Key Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Ministry of Education of China, Institute of Cardiovascular Research, Luzhou, China
| | - Long Wang
- School of Pharmacy, Southwest Medical University, Luzhou, China.,The Key Laboratory of Medical Electrophysiology, Medical Key Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Ministry of Education of China, Institute of Cardiovascular Research, Luzhou, China
| | - Dalian Qin
- School of Pharmacy, Southwest Medical University, Luzhou, China.,The Key Laboratory of Medical Electrophysiology, Medical Key Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Ministry of Education of China, Institute of Cardiovascular Research, Luzhou, China
| | - Feihong Huang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Qibing Mei
- The Key Laboratory of Medical Electrophysiology, Medical Key Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Ministry of Education of China, Institute of Cardiovascular Research, Luzhou, China
| | - Jing Yang
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jianming Wu
- School of Pharmacy, Southwest Medical University, Luzhou, China.,The Key Laboratory of Medical Electrophysiology, Medical Key Laboratory for Drug Discovery and Druggability Evaluation of Sichuan Province, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, Ministry of Education of China, Institute of Cardiovascular Research, Luzhou, China
| |
Collapse
|
13
|
Jakubowska J, Pawlik B, Wyka K, Stolarska M, Kotulska K, Jóźwiak S, Młynarski W, Trelińska J. New Insights into Red Blood Cell Microcytosis upon mTOR Inhibitor Administration. Int J Mol Sci 2021; 22:6802. [PMID: 34202704 PMCID: PMC8268656 DOI: 10.3390/ijms22136802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/12/2021] [Accepted: 06/22/2021] [Indexed: 11/16/2022] Open
Abstract
The aim of this study was to evaluate the effect of everolimus, a mammalian target of rapamycin (mTOR) inhibitor, on red blood cell parameters in the context of iron homeostasis in patients with tuberous sclerosis complex (TSC) and evaluate its effect on cell size in vitro. Everolimus has a significant impact on red blood cell parameters in patients with TSC. The most common alteration was microcytosis. The mean MCV value decreased by 9.2%, 12%, and 11.8% after 3, 6, and 12 months of everolimus treatment. The iron level declined during the first 3 months, and human soluble transferrin receptor concentration increased during 6 months of therapy. The size of K562 cells decreased when cultured in the presence of 5 μM everolimus by approximately 8%. The addition of hemin to the cell culture with 5 μM everolimus did not prevent any decrease in cell size. The stage of erythroid maturation did not affect the response to everolimus. Our results showed that the mTOR inhibitor everolimus caused red blood cell microcytosis in vivo and in vitro. This effect is not clearly related to a deficit of iron and erythroid maturation. This observation confirms that mTOR signaling plays a complex role in the control of cell size.
Collapse
Affiliation(s)
- Justyna Jakubowska
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, ul. Sporna 36/50, 91-738 Lodz, Poland; (J.J.); (B.P.); (K.W.); (M.S.); (W.M.)
| | - Bartłomiej Pawlik
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, ul. Sporna 36/50, 91-738 Lodz, Poland; (J.J.); (B.P.); (K.W.); (M.S.); (W.M.)
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Krystyna Wyka
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, ul. Sporna 36/50, 91-738 Lodz, Poland; (J.J.); (B.P.); (K.W.); (M.S.); (W.M.)
| | - Małgorzata Stolarska
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, ul. Sporna 36/50, 91-738 Lodz, Poland; (J.J.); (B.P.); (K.W.); (M.S.); (W.M.)
| | - Katarzyna Kotulska
- Department of Neurology & Epileptology and Pediatric Rehabilitation, The Children’s Memorial Health Institute, ul. Dzieci Polskich 20, 00-999 Warsaw, Poland;
| | - Sergiusz Jóźwiak
- Department of Child Neurology, Medical University of Warsaw, ul. Banacha 1A, 02-097 Warsaw, Poland;
| | - Wojciech Młynarski
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, ul. Sporna 36/50, 91-738 Lodz, Poland; (J.J.); (B.P.); (K.W.); (M.S.); (W.M.)
| | - Joanna Trelińska
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, ul. Sporna 36/50, 91-738 Lodz, Poland; (J.J.); (B.P.); (K.W.); (M.S.); (W.M.)
| |
Collapse
|
14
|
Wang X, Song W, Zhang F, Huang R. Dihydroartemisinin Inhibits TGF-β-Induced Fibrosis in Human Tenon Fibroblasts via Inducing Autophagy. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:973-981. [PMID: 33688170 PMCID: PMC7937381 DOI: 10.2147/dddt.s280322] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 01/13/2021] [Indexed: 12/29/2022]
Abstract
Background The formation of hypertrophic scars (HS) can result in the failure of glaucoma surgery, and fibrosis is known to be closely associated with the progression of HS. Dihydroartemisinin (DHA) has been reported to inhibit the progression of fibrosis; however, whether DHA can alleviate the formation of HS remains unclear. Methods In the present study, in order to examine the effects of DHA on the progression of HS, human Tenon's capsule fibroblasts (HTFs) were isolated from patients who underwent glaucoma surgery. In addition, Western blot analysis, microtubule associated protein 1 light chain 3 α staining and reverse transcription-quantitative PCR were performed to detect protein and mRNA expression levels in the HTFs, respectively. Cell proliferation was detected by Ki67 staining. Flow cytometry was used to examine apoptosis and reactive oxygen species (ROS) levels in the HTFs. Results The results revealed that TGF-β promoted the proliferation and fibrosis of HTFs; however, DHA significantly reversed the effects of TGF-β by increasing cell autophagy. In addition, DHA notably induced the apoptosis of TGF-β-stimulated HTFs by increasing the ROS levels, while these increases were partially reversed by 3-methyladenine. Furthermore, DHA notably increased the expression of microRNA (miR)-145-5p in HTFs in a dose-dependent manner. Conclusion The present study demonstrated that DHA inhibits the TGF-β-induced fibrosis of HTFs by inducing autophagy. These findings may aid in the development of novel agents for the prevention of the formation of HS following glaucoma surgery.
Collapse
Affiliation(s)
- Xiaoyuan Wang
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Wuqi Song
- Department of Microbiology, Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Fengmin Zhang
- Department of Microbiology, Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| | - Renping Huang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150001, People's Republic of China
| |
Collapse
|
15
|
Liu Q, Luo L, Ren C, Zou M, Yang S, Cai B, Wu L, Wang Y, Fu S, Hua X, Tang N, Huang S, Huang X, Xin W, Chen F, Zhang X. The opposing roles of the mTOR signaling pathway in different phases of human umbilical cord blood-derived CD34 + cell erythropoiesis. Stem Cells 2020; 38:1492-1505. [PMID: 32871057 PMCID: PMC7693065 DOI: 10.1002/stem.3268] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 06/25/2020] [Accepted: 07/10/2020] [Indexed: 02/05/2023]
Abstract
As an indispensable, even lifesaving practice, red blood cell (RBC) transfusion is challenging due to several issues, including supply shortage, immune incompatibility, and blood-borne infections since donated blood is the only source of RBCs. Although large-scale in vitro production of functional RBCs from human stem cells is a promising alternative, so far, no such system has been reported to produce clinically transfusable RBCs due to the poor understanding of mechanisms of human erythropoiesis, which is essential for the optimization of in vitro erythrocyte generation system. We previously reported that inhibition of mammalian target of rapamycin (mTOR) signaling significantly decreased the percentage of erythroid progenitor cells in the bone marrow of wild-type mice. In contrast, rapamycin treatment remarkably improved terminal maturation of erythroblasts and anemia in a mouse model of β-thalassemia. In the present study, we investigated the effect of mTOR inhibition with rapamycin from different time points on human umbilical cord blood-derived CD34+ cell erythropoiesis in vitro and the underlying mechanisms. Our data showed that rapamycin treatment significantly suppressed erythroid colony formation in the commitment/proliferation phase of erythropoiesis through inhibition of cell-cycle progression and proliferation. In contrast, during the maturation phase of erythropoiesis, mTOR inhibition dramatically promoted enucleation and mitochondrial clearance by enhancing autophagy. Collectively, our results suggest contrasting roles for mTOR in regulating different phases of human erythropoiesis.
Collapse
Affiliation(s)
- Qian Liu
- Laboratory of Molecular CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
- Shantou University Medical CollegeShantouPeople's Republic of China
| | - Linhong Luo
- Department of Obstetrics and GynecologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
| | - Chunhong Ren
- Department of International Medical ServiceThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
| | - Muping Zou
- Laboratory of Molecular CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
| | - Siqin Yang
- Laboratory of Molecular CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
| | - Bozhi Cai
- Laboratory of Molecular CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
| | - Libiao Wu
- Laboratory of Molecular CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
| | - Yunsheng Wang
- Laboratory of Molecular CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
| | - Shan Fu
- Shantou University Medical CollegeShantouPeople's Republic of China
| | - Xu Hua
- Shantou University Medical CollegeShantouPeople's Republic of China
| | - Nianping Tang
- Shantou University Medical CollegeShantouPeople's Republic of China
| | - Shiping Huang
- Shantou University Medical CollegeShantouPeople's Republic of China
| | - Xianxi Huang
- Intensive Care Unit, The First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
| | - Wen Xin
- Beijing TransGen Biotech Co., Ltd.BeijingPeople's Republic of China
| | - Feiheng Chen
- Department of HematologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
| | - Xin Zhang
- Laboratory of Molecular CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
- Shantou University Medical CollegeShantouPeople's Republic of China
- Department of CardiologyThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
- Laboratory of Medical Molecular ImagingThe First Affiliated Hospital of Shantou University Medical CollegeShantouPeople's Republic of China
| |
Collapse
|
16
|
Atkinson SP. A preview of selected articles. Stem Cells 2020. [DOI: 10.1002/stem.3285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|