1
|
Tkachenko A, Havranek O. Cell death signaling in human erythron: erythrocytes lose the complexity of cell death machinery upon maturation. Apoptosis 2025; 30:652-673. [PMID: 39924584 PMCID: PMC11947060 DOI: 10.1007/s10495-025-02081-5] [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] [Accepted: 01/20/2025] [Indexed: 02/11/2025]
Abstract
Over the recent years, our understanding of the cell death machinery of mature erythrocytes has been greatly expanded. It resulted in the discovery of several regulated cell death (RCD) pathways in red blood cells. Apoptosis (eryptosis) and necroptosis of erythrocytes share certain features with their counterparts in nucleated cells, but they are also critically different in particular details. In this review article, we summarize the cell death subroutines in the erythroid precursors (apoptosis, necroptosis, and ferroptosis) in comparison to mature erythrocytes (eryptosis and erythronecroptosis) to highlight the consequences of organelle clearance and associated loss of multiple components of the cell death machinery upon erythrocyte maturation. Recent advances in understanding the role of erythrocyte RCDs in health and disease have expanded potential clinical applications of these lethal subroutines, emphasizing their contribution to the development of anemia, microthrombosis, and endothelial dysfunction, as well as their role as diagnostic biomarkers and markers of erythrocyte storage-induced lesions. Fas signaling and the functional caspase-8/caspase-3 system are not indispensable for eryptosis, but might be retained in mature erythrocytes to mediate the crosstalk between both erythrocyte-associated RCDs. The ability of erythrocytes to switch between eryptosis and necroptosis suggests that their cell death is not a simple unregulated mechanical disintegration, but a tightly controlled process. This allows investigation of eventual pharmacological interventions aimed at individual cell death subroutines of erythrocytes.
Collapse
Affiliation(s)
- Anton Tkachenko
- First Faculty of Medicine, BIOCEV, Charles University, Průmyslová 595, 25250, Vestec, Czech Republic.
| | - Ondrej Havranek
- First Faculty of Medicine, BIOCEV, Charles University, Průmyslová 595, 25250, Vestec, Czech Republic
- First Department of Medicine - Hematology, General University Hospital and First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
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
|
Maggio A, Napolitano M, Taher AT, Bou-Fakhredin R, Ostuni MA. Reframing thalassaemia syndrome as a benign haematopoietic stem cell disorder. Br J Haematol 2025; 206:464-477. [PMID: 39676308 DOI: 10.1111/bjh.19919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 11/12/2024] [Indexed: 12/17/2024]
Abstract
Thalassaemia, caused by over 250 mutations in the beta globin gene, changes the haematopoietic stem cell (HSC) differentiation, leading to ineffective erythropoiesis. This Wider Perspective article overlooks its underlying nature as a benign HSC disorder with a significant impact on the erythroid cell lineage. The simplicity of managing symptoms through transfusions and iron chelation therapy has shifted the focus away from the development of cell-based treatments. The identification of the beta039 mutation by Chang and Kan in 1979 marked a turning point, suggesting as main approach the molecular level by correcting the beta globin chain imbalances through gene insertion and editing. However, challenges of technology have delayed the implementation of these strategies for over four decades. In contrast, the past two decades have witnessed significant advances in the treatment of HSC disorders of the myeloid clone which are driven by a 'target cell strategy'. Many current and innovative treatments for thalassaemia are now adopting this approach, highlighting the importance of identifying suitable candidates through risk stratification. This manuscript explores the evolving understanding of thalassaemia syndromes as congenital HSC disorders of the erythroid clone and examines the implications of this perspective for the development of future treatments.
Collapse
Affiliation(s)
- Aurelio Maggio
- Campus of Haematology Franco and Piera Cutino, AOOR Villa Sofia-V. Cervello, Palermo, Italy
| | - Mariasanta Napolitano
- Campus of Haematology Franco and Piera Cutino, AOOR Villa Sofia-V. Cervello, Palermo, Italy
- Dipartimento PROMISE, Università degli Studi di AOUP "P. Giaccone", Palermo, Italy
| | - Ali T Taher
- Division of Hematology and Oncology, Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Rayan Bou-Fakhredin
- Division of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | | |
Collapse
|
4
|
Jana S, Alayash AI. Exploring the Molecular Interplay Between Oxygen Transport, Cellular Oxygen Sensing, and Mitochondrial Respiration. Antioxid Redox Signal 2025. [PMID: 39846399 DOI: 10.1089/ars.2023.0428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Significance: The mitochondria play a key role in maintaining oxygen homeostasis under normal oxygen tension (normoxia) and during oxygen deprivation (hypoxia). This is a critical balancing act between the oxygen content of the blood, the tissue oxygen sensing mechanisms, and the mitochondria, which ultimately consume most oxygen for energy production. Recent Advances: We describe the well-defined role of the mitochondria in oxygen metabolism with a special focus on the impact on blood physiology and pathophysiology. Critical Issues: Fundamental questions remain regarding the impact of mitochondrial responses to changes in overall blood oxygen content under normoxic and hypoxic states and in the case of impaired oxygen sensing in various cardiovascular and pulmonary complications including blood disorders involving hemolysis and hemoglobin toxicity, ischemia reperfusion, and even in COVID-19 disease. Future Directions: Understanding the nature of the crosstalk among normal homeostatic pathways, oxygen carrying by hemoglobin, utilization of oxygen by the mitochondrial respiratory chain machinery, and oxygen sensing by hypoxia-inducible factor proteins, may provide a target for future therapeutic interventions. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Sirsendu Jana
- Laboratory of Biochemistry and Vascular Biology, Center for Biologics Evaluation and Research, Food and Drug Administration (FDA), Silver Spring, Maryland, USA
| | - Abdu I Alayash
- Laboratory of Biochemistry and Vascular Biology, Center for Biologics Evaluation and Research, Food and Drug Administration (FDA), Silver Spring, Maryland, USA
| |
Collapse
|
5
|
Joly A, Schott A, Phadke I, Gonzalez-Menendez P, Kinet S, Taylor N. Beyond ATP: Metabolite Networks as Regulators of Physiological and Pathological Erythroid Differentiation. Physiology (Bethesda) 2025; 40:0. [PMID: 39226028 DOI: 10.1152/physiol.00035.2024] [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: 07/15/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024] Open
Abstract
Hematopoietic stem cells (HSCs) possess the capacity for self-renewal and the sustained production of all mature blood cell lineages. It has been well established that a metabolic rewiring controls the switch of HSCs from a self-renewal state to a more differentiated state, but it is only recently that we have appreciated the importance of metabolic pathways in regulating the commitment of progenitors to distinct hematopoietic lineages. In the context of erythroid differentiation, an extensive network of metabolites, including amino acids, sugars, nucleotides, fatty acids, vitamins, and iron, is required for red blood cell (RBC) maturation. In this review, we highlight the multifaceted roles via which metabolites regulate physiological erythropoiesis as well as the effects of metabolic perturbations on erythroid lineage commitment and differentiation. Of note, the erythroid differentiation process is associated with an exceptional breadth of solute carrier (SLC) metabolite transporter upregulation. Finally, we discuss how recent research, revealing the critical impact of metabolic reprogramming in diseases of disordered and ineffective erythropoiesis, has created opportunities for the development of novel metabolic-centered therapeutic strategies.
Collapse
Affiliation(s)
- Axel Joly
- Université de Montpellier, CNRS, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
| | - Arthur Schott
- Université de Montpellier, CNRS, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
| | - Ira Phadke
- Université de Montpellier, CNRS, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
- Pediatric Oncology Branch, CCR, NCI, National Institutes of Health, Bethesda, Maryland, United States
| | - Pedro Gonzalez-Menendez
- Departamento de Morfologia y Biologia Celular, Instituto Universitario de Oncologia del Principado de Asturias (IUOPA), Universidad de Oviedo, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Sandrina Kinet
- Université de Montpellier, CNRS, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
| | - Naomi Taylor
- Université de Montpellier, CNRS, Institut de Génétique Moléculaire de Montpellier, Montpellier, France
- Pediatric Oncology Branch, CCR, NCI, National Institutes of Health, Bethesda, Maryland, United States
| |
Collapse
|
6
|
Kidoguchi S, Torii K, Okada T, Yamano T, Iwamura N, Miyagi K, Toyama T, Iwano M, Miyazaki R, Shigematsu Y, Kimura H. Fatty Acid β-Oxidation May Be Associated with the Erythropoietin Resistance Index in Stable Patients Undergoing Haemodialysis. Diagnostics (Basel) 2024; 14:2295. [PMID: 39451618 PMCID: PMC11506985 DOI: 10.3390/diagnostics14202295] [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: 09/13/2024] [Revised: 10/09/2024] [Accepted: 10/13/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND/OBJECTIVES Lipid metabolism and adiponectin modulate erythropoiesis in vitro and in general population studies and may also affect responsiveness to erythropoietin in patients undergoing haemodialysis (HD). However, little is known about the impact of lipid-associated biomarkers on reticulocyte production and erythropoietin resistance index (ERI) in patients undergoing HD. Therefore, we aimed to investigate their impacts in 167 stable patients undergoing HD. METHODS Pre-dialysis blood samples were collected and analysed for reticulocyte counts and serum lipid profiles by routine analyses and serum carnitine profiles (C0-C18) by LC-MS/MS. ERI was calculated as erythropoietin dose/kg/week normalized for haemoglobin levels. RESULTS The independent positive determinants of reticulocyte count were log [Triglyceride (TG)] and logC18:1. A large proportion of longer-chain acylcarnitines was positively correlated with reticulocyte counts, possibly resulting from the accumulation of acylcarnitines in mitochondria undergoing fateful exocytosis from reticulocytes. These results indicate a possible association between reticulocyte formation and reduced β-oxidation, which occurs during the peripheral phase of erythroblast enucleation. Total cholesterol (TC) and log [C2/(C16 + C18:1)] as a putative marker of β-oxidation efficiency were negative independent determinants of ERI. Moreover, acyl chain length had a significantly positive impact on the correlation coefficients of individual acylcarnitines with ERI, suggesting that enhanced β-oxidation may be associated with reduced ERI. Finally, adiponectin had no independent association with reticulocyte counts or ERI despite its negative association with HDL-C levels. CONCLUSIONS Enhanced fatty acid β-oxidation and higher TC levels may be associated with lower ERI, whereas higher TG levels and longer acylcarnitines may be related to the latest production of reticulocytes in stable patients undergoing HD.
Collapse
Affiliation(s)
- Shuhei Kidoguchi
- Department of Clinical Laboratory, University of Fukui Hospital, Fukui 910-1193, Japan; (S.K.); (K.T.); (T.O.); (T.Y.); (N.I.); (T.T.)
| | - Kunio Torii
- Department of Clinical Laboratory, University of Fukui Hospital, Fukui 910-1193, Japan; (S.K.); (K.T.); (T.O.); (T.Y.); (N.I.); (T.T.)
- Department of Clinical Laboratory, Japanese Red Cross Fukui Hospital, Fukui 918-8501, Japan
| | - Toshiharu Okada
- Department of Clinical Laboratory, University of Fukui Hospital, Fukui 910-1193, Japan; (S.K.); (K.T.); (T.O.); (T.Y.); (N.I.); (T.T.)
| | - Tomoko Yamano
- Department of Clinical Laboratory, University of Fukui Hospital, Fukui 910-1193, Japan; (S.K.); (K.T.); (T.O.); (T.Y.); (N.I.); (T.T.)
| | - Nanami Iwamura
- Department of Clinical Laboratory, University of Fukui Hospital, Fukui 910-1193, Japan; (S.K.); (K.T.); (T.O.); (T.Y.); (N.I.); (T.T.)
| | - Kyoko Miyagi
- Department of Internal Medicine, Fujita Memorial Hospital, Fukui 910-0004, Japan; (K.M.); (R.M.)
| | - Tadashi Toyama
- Department of Clinical Laboratory, University of Fukui Hospital, Fukui 910-1193, Japan; (S.K.); (K.T.); (T.O.); (T.Y.); (N.I.); (T.T.)
- Division of Nephrology, Department of General Medicine, School of Medicine, University of Fukui, Fukui 910-1193, Japan
| | - Masayuki Iwano
- Division of Nephrology, Department of General Medicine, School of Medicine, University of Fukui, Fukui 910-1193, Japan
| | - Ryoichi Miyazaki
- Department of Internal Medicine, Fujita Memorial Hospital, Fukui 910-0004, Japan; (K.M.); (R.M.)
| | - Yosuke Shigematsu
- Department of Pediatrics, Faculty of Medical Sciences, University of Fukui, Fukui 910-1193, Japan;
| | - Hideki Kimura
- Department of Clinical Laboratory, University of Fukui Hospital, Fukui 910-1193, Japan; (S.K.); (K.T.); (T.O.); (T.Y.); (N.I.); (T.T.)
- Division of Nephrology, Department of General Medicine, School of Medicine, University of Fukui, Fukui 910-1193, Japan
| |
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 X, Cui T, Yan H, Zhao L, Zang R, Li H, Wang H, Zhang B, Zhou J, Liu Y, Yue W, Xi J, Pei X. Enhancing terminal erythroid differentiation in human embryonic stem cells through TRIB3 overexpression. Heliyon 2024; 10:e37463. [PMID: 39309892 PMCID: PMC11415673 DOI: 10.1016/j.heliyon.2024.e37463] [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: 02/14/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024] Open
Abstract
Tribbles pseudokinase 3 (TRIB3) expression significantly increases during terminal erythropoiesis in vivo. However, we found that TRIB3 expression remained relatively low during human embryonic stem cell (hESC) erythropoiesis, particularly in the late stage, where it is typically active. TRIB3 was expressed in megakaryocyte-erythrocyte progenitor cells and its low expression was necessary for megakaryocyte differentiation. Thus, we proposed that the high expression during late stage of erythropoiesis could be the clue for promotion of maturation of hESC-derived erythroid cells. To our knowledge, the role of TRIB3 in the late stage of erythropoiesis remains ambiguous. To address this, we generated inducible TRIB3 overexpression hESCs, named TRIB3tet-on OE H9, based on a Tet-On system. Then, we analyzed hemoglobin expression, condensed chromosomes, organelle clearance, and enucleation with or without doxycycline treatment. TRIB3tet-on OE H9 cells generated erythrocytes with a high proportion of orthochromatic erythroblast in flow cytometry, enhanced hemoglobin and related protein expression in Western blot, decreased nuclear area size, promoted enucleation rate, decreased lysosome and mitochondria number, more colocalization of LC3 with LAMP1 (lysosome marker) and TOM20 (mitochondria marker) and up-regulated mitophagy-related protein expression after treatment with 2 μg/mL doxycycline. Our results showed that TRIB3 overexpression during terminal erythropoiesis may promote the maturation of erythroid cells. Therefore, our study delineates the role of TRIB3 in terminal erythropoiesis, and reveals TRIB3 as a key regulator of UPS and downstream mitophagy by ensuring appropriate mitochondrial clearance during the compaction of chromatin.
Collapse
Affiliation(s)
| | | | - Hao Yan
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Lingping Zhao
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Ruge Zang
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Hongyu Li
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Haiyang Wang
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Biao Zhang
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Junnian Zhou
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Yiming Liu
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Wen Yue
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Jiafei Xi
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Xuetao Pei
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| |
Collapse
|
9
|
Yan H, Zang R, Cui T, Liu Y, Zhang B, Zhao L, Li H, Zhou J, Wang H, Zeng Q, Xu L, Zhou Y, Pei X, Xi J, Yue W. PROTAC-mediated vimentin degradation promotes terminal erythroid differentiation of pluripotent stem cells. Stem Cell Res Ther 2024; 15:310. [PMID: 39294765 PMCID: PMC11412063 DOI: 10.1186/s13287-024-03910-1] [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: 06/10/2024] [Accepted: 08/28/2024] [Indexed: 09/21/2024] Open
Abstract
BACKGROUND Human pluripotent stem cells (hPSCs), including human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs), can undergo erythroid differentiation, offering a potentially invaluable resource for generating large quantities of erythroid cells. However, the majority of erythrocytes derived from hPSCs fail to enucleate compared with those derived from cord blood progenitors, with an unknown molecular basis for this difference. The expression of vimentin (VIM) is retained in erythroid cells differentiated from hPSCs but is absent in mature erythrocytes. Further exploration is required to ascertain whether VIM plays a critical role in enucleation and to elucidate the underlying mechanisms. METHODS In this study, we established a hESC line with reversible vimentin degradation (dTAG-VIM-H9) using the proteolysis-targeting chimera (PROTAC) platform. Various time-course studies, including erythropoiesis from CD34+ human umbilical cord blood and three-dimensional (3D) organoid culture from hESCs, morphological analysis, quantitative real-time PCR (qRT-PCR), western blotting, flow cytometry, karyotyping, cytospin, Benzidine-Giemsa staining, immunofluorescence assay, and high-speed cell imaging analysis, were conducted to examine and compare the characteristics of hESCs and those with vimentin degradation, as well as their differentiated erythroid cells. RESULTS Vimentin expression diminished during normal erythropoiesis in CD34+ cord blood cells, whereas it persisted in erythroid cells differentiated from hESC. Depletion of vimentin using the degradation tag (dTAG) system promotes erythroid enucleation in dTAG-VIM-H9 cells. Nuclear polarization of erythroblasts is elevated by elimination of vimentin. CONCLUSIONS VIM disappear during the normal maturation of erythroid cells, whereas they are retained in erythroid cells differentiated from hPSCs. We found that retention of vimentin during erythropoiesis impairs erythroid enucleation from hPSCs. Using the PROTAC platform, we validated that vimentin degradation by dTAG accelerates the enucleation rate in dTAG-VIM-H9 cells by enhancing nuclear polarization.
Collapse
Affiliation(s)
- Hao Yan
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Ruge Zang
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China.
| | - Tiantian Cui
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Yiming Liu
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Biao Zhang
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Lingpin Zhao
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Hongyu Li
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Juannian Zhou
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Haiyang Wang
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Quan Zeng
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Lei Xu
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Yuqi Zhou
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Xuetao Pei
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China
| | - Jiafei Xi
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China.
| | - Wen Yue
- Beijing Institute of Radiation Medicine, Beijing, 100850, P. R. China.
| |
Collapse
|
10
|
Fairley LH, Grimm A, Herff SA, Eckert A. Translocator protein (TSPO) ligands attenuate mitophagy deficits in the SH-SY5Y cellular model of Alzheimer's disease via the autophagy adaptor P62. Biochimie 2024; 224:132-138. [PMID: 38280505 DOI: 10.1016/j.biochi.2024.01.012] [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: 10/05/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 01/29/2024]
Abstract
Mitochondrial dysfunction has been widely implicated in the pathogenesis of Alzheimer's disease (AD), with accumulation of damaged and dysfunctional mitochondria occurring early in the disease. Mitophagy, which governs mitochondrial turnover and quality control, is impaired in the AD brain, and strategies aimed at enhancing mitophagy have been identified as promising therapeutic targets. The translocator protein (TSPO) is an outer mitochondrial membrane protein that is upregulated in AD, and ligands targeting TSPO have been shown to exert neuroprotective effects in mouse models of AD. However, whether TSPO ligands modulate mitophagy in AD has not been explored. Here, we provide evidence that the TSPO-specific ligands Ro5-4864 and XBD173 attenuate mitophagy deficits and mitochondrial fragmentation in a cellular model of AD overexpressing the human amyloid precursor protein (APP). Ro5-4864 and XBD173 appear to enhance mitophagy via modulation of the autophagic cargo receptor P62/SQSTM1, in the absence of an effect on PARK2, PINK1, or LC3 level. Taken together, these findings indicate that TSPO ligands may be promising therapeutic agents for ameliorating mitophagy deficits in AD.
Collapse
Affiliation(s)
- Lauren H Fairley
- Research Cluster, Molecular & Cognitive Neuroscience, Neurobiology Laboratory for Brain Aging and Mental Health, University of Basel, Basel, Switzerland.
| | - Amandine Grimm
- Research Cluster, Molecular & Cognitive Neuroscience, Neurobiology Laboratory for Brain Aging and Mental Health, University of Basel, Basel, Switzerland; Psychiatric University Clinics, Basel, Switzerland
| | - Steffen A Herff
- The MARCS Institute for Brain, Behaviour and Development, Western Sydney University, Australia
| | - Anne Eckert
- Research Cluster, Molecular & Cognitive Neuroscience, Neurobiology Laboratory for Brain Aging and Mental Health, University of Basel, Basel, Switzerland; Psychiatric University Clinics, Basel, Switzerland
| |
Collapse
|
11
|
Menon V, Slavinsky M, Hermine O, Ghaffari S. Mitochondrial regulation of erythropoiesis in homeostasis and disease. Br J Haematol 2024; 205:429-439. [PMID: 38946206 PMCID: PMC11619715 DOI: 10.1111/bjh.19600] [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: 03/12/2024] [Accepted: 06/06/2024] [Indexed: 07/02/2024]
Abstract
Erythroid cells undergo a highly complex maturation process, resulting in dynamic changes that generate red blood cells (RBCs) highly rich in haemoglobin. The end stages of the erythroid cell maturation process primarily include chromatin condensation and nuclear polarization, followed by nuclear expulsion called enucleation and clearance of mitochondria and other organelles to finally generate mature RBCs. While healthy RBCs are devoid of mitochondria, recent evidence suggests that mitochondria are actively implicated in the processes of erythroid cell maturation, erythroblast enucleation and RBC production. However, the extent of mitochondrial participation that occurs during these ultimate steps is not completely understood. This is specifically important since abnormal RBC retention of mitochondria or mitochondrial DNA contributes to the pathophysiology of sickle cell and other disorders. Here we review some of the key findings so far that elucidate the importance of this process in various aspects of erythroid maturation and RBC production under homeostasis and disease conditions.
Collapse
Affiliation(s)
- Vijay Menon
- Department of Cell, Developmental & Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Mary Slavinsky
- Department of Cell, Developmental & Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Olivier Hermine
- Department Hematology, Hôpital Necker, Assistance Publique Hôpitaux de Paris, University Paris Descartes
- INSERM U1163 and CNRS 8254, Imagine Institute, Université Sorbonne Paris Cité, Paris, France
| | - Saghi Ghaffari
- Department of Cell, Developmental & Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| |
Collapse
|
12
|
Ikeda M, Kato H, Shima H, Matsumoto M, Furukawa E, Yan Y, Liao R, Xu J, Muto A, Fujiwara T, Harigae H, Bresnick EH, Igarashi K. Heme-dependent induction of mitophagy program during differentiation of murine erythroid cells. Exp Hematol 2023; 118:21-30. [PMID: 36481429 PMCID: PMC10161131 DOI: 10.1016/j.exphem.2022.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 11/25/2022] [Accepted: 11/26/2022] [Indexed: 12/12/2022]
Abstract
Although establishment and maintenance of mitochondria are essential for the production of massive amounts of heme in erythroblasts, mitochondria must be degraded upon terminal differentiation to red blood cells (RBCs), thus creating a biphasic regulatory process. Previously, we reported that iron deficiency in mice promotes mitochondrial retention in RBCs, suggesting that a proper amount of iron and/or heme is necessary for the degradation of mitochondria during erythroblast maturation. Because the transcription factor GATA1 regulates autophagy in erythroid cells, which involves mitochondrial clearance (mitophagy), we investigated the relationship between iron or heme and mitophagy by analyzing the expression of genes related to GATA1 and autophagy and the impact of iron or heme restriction on the amount of mitochondria. We found that heme promotes the expression of GATA1-regulated mitophagy-related genes and the induction of mitophagy. GATA1 might induce the expression of the autophagy-related genes Atg4d and Stk11 for mitophagy through a heme-dependent mechanism in murine erythroleukemia (MEL) cells and a genetic rescue system with G1E-ER-GATA1 erythroblast cells derived from Gata1-null murine embryonic stem cells. These results provide evidence for a biphasic mechanism in which mitochondria are essential for heme generation, and the heme generated during differentiation promotes mitophagy and mitochondrial disposal. This mechanism provides a molecular framework for understanding this fundamentally important cell biological process.
Collapse
Affiliation(s)
- Masatoshi Ikeda
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Seiryo-machi, Sendai, Japan
| | - Hiroki Kato
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Seiryo-machi, Sendai, Japan; Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Seiryo-machi, Sendai, Japan
| | - Hiroki Shima
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Seiryo-machi, Sendai, Japan
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Seiryo-machi, Sendai, Japan; Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Seiryo-machi, Sendai, Japan
| | - Eijiro Furukawa
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Seiryo-machi, Sendai, Japan
| | - Yan Yan
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Seiryo-machi, Sendai, Japan
| | - Ruiqi Liao
- Department of Cell and Regenerative Biology, Wisconsin Blood Cancer Research Institute, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Jian Xu
- Children's Medical Center Research Institute, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX
| | - Akihiko Muto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Seiryo-machi, Sendai, Japan
| | - Tohru Fujiwara
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Seiryo-machi, Sendai, Japan
| | - Hideo Harigae
- Department of Hematology and Rheumatology, Tohoku University Graduate School of Medicine, Seiryo-machi, Sendai, Japan
| | - Emery H Bresnick
- Department of Cell and Regenerative Biology, Wisconsin Blood Cancer Research Institute, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Seiryo-machi, Sendai, Japan; Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Seiryo-machi, Sendai, Japan.
| |
Collapse
|
13
|
Mitochondria: Emerging Consequential in Sickle Cell Disease. J Clin Med 2023; 12:jcm12030765. [PMID: 36769414 PMCID: PMC9917941 DOI: 10.3390/jcm12030765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
Advanced mitochondrial multi-omics indicate a multi-facet involvement of mitochondria in the physiology of the cell, changing the perception of mitochondria from being just the energy-generating organelles to organelles that highly influence cell structure, function, signaling, and cell fate. This sets mitochondrial dysfunction in the centerstage of numerous acquired and genetic diseases. Sickle cell disease is also being increasingly associated with mitochondrial anomalies and the pathophysiology of sickle cell disease finds mitochondria at crucial intersections in the pathological cascade. Altered mitophagy, increased ROS, and mitochondrial DNA all contribute to the condition and its severity. Such mitochondrial aberrations lead to consequent mitochondrial retention in red blood cells in sickle cell diseases, increased oxidation in the cellular environment, inflammation, worsened vaso-occlusive crisis, etc. There are increasing studies indicating mitochondrial significance in sickle cell disease, consequently providing an opportunity to target it for improving the outcomes of treatment. Identification of the impaired mitochondrial attributes in sickle cell disease and their modulation by therapeutic interventions can impart a better management of the disease. This review aims to describe the mitochondria in the perspective of sicke cell disease so as to provide the reader an overview of the emerging mitochondrial stance in sickle cell disease.
Collapse
|
14
|
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
|
15
|
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
|
16
|
Dussouchaud A, Jacob J, Secq C, Verbavatz JM, Moras M, Larghero J, Fader CM, Ostuni MA, Lefevre SD. Transmission Electron Microscopy to Follow Ultrastructural Modifications of Erythroblasts Upon ex vivo Human Erythropoiesis. Front Physiol 2022; 12:791691. [PMID: 35222062 PMCID: PMC8864112 DOI: 10.3389/fphys.2021.791691] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 12/23/2021] [Indexed: 01/02/2023] Open
Abstract
Throughout mammal erythroid differentiation, erythroblasts undergo enucleation and organelle clearance becoming mature red blood cell. Organelles are cleared by autophagic pathways non-specifically targeting organelles and cytosolic content or by specific mitophagy targeting mitochondria. Mitochondrial functions are essential to coordinate metabolism reprogramming, cell death, and differentiation balance, and also synthesis of heme, the prosthetic group needed in hemoglobin assembly. In mammals, mitochondria subcellular localization and mitochondria interaction with other structures as endoplasmic reticulum and nucleus might be of importance for the removal of the nucleus, that is, the enucleation. Here, we aim to characterize by electron microscopy the changes in ultrastructure of cells over successive stages of human erythroblast differentiation. We focus on mitochondria to gain insights into intracellular localization, ultrastructure, and contact with other organelles. We found that mitochondria are progressively cleared with a significant switch between PolyE and OrthoE stages, acquiring a rounded shape and losing contact sites with both ER (MAM) and nucleus (NAM). We studied intracellular vesicle trafficking and found that endosomes and MVBs, known to be involved in iron traffic and heme synthesis, are increased during BasoE to PolyE transition; autophagic structures such as autophagosomes increase from ProE to OrthoE stages. Finally, consistent with metabolic switch, glycogen accumulation was observed in OrthoE stage.
Collapse
Affiliation(s)
- Alice Dussouchaud
- Université de Paris and Université des Antilles, INSERM, BIGR, Paris, France
| | - Julieta Jacob
- Laboratorio de Biología Celular y Molecular, Instituto de Histología y Embriología, CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Charles Secq
- Université de Paris and Université des Antilles, INSERM, BIGR, Paris, France
| | | | - Martina Moras
- Université de Paris and Université des Antilles, INSERM, BIGR, Paris, France
| | - Jérôme Larghero
- CNRS, UMR 7592, Institut Jacques Monod, Université de Paris, Paris, France
| | - Claudio M. Fader
- Laboratorio de Biología Celular y Molecular, Instituto de Histología y Embriología, CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Mariano A. Ostuni
- Université de Paris and Université des Antilles, INSERM, BIGR, Paris, France
| | - Sophie D. Lefevre
- Université de Paris and Université des Antilles, INSERM, BIGR, Paris, France
- *Correspondence: Sophie D. Lefevre,
| |
Collapse
|
17
|
Zhang Y, Xu Y, Zhang S, Lu Z, Li Y, Zhao B. The regulation roles of Ca 2+ in erythropoiesis: What have we learned? Exp Hematol 2021; 106:19-30. [PMID: 34879257 DOI: 10.1016/j.exphem.2021.12.192] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/15/2021] [Accepted: 12/03/2021] [Indexed: 01/09/2023]
Abstract
Calcium (Ca2+) is an important second messenger molecule in the body, regulating cell cycle and fate. There is growing evidence that intracellular Ca2+ levels play functional roles in the total physiological process of erythroid differentiation, including the proliferation and differentiation of erythroid progenitor cells, terminal enucleation, and mature red blood cell aging and clearance. Moreover, recent research on the pathology of erythroid disorders has made great progress in the past decades, indicating that calcium ion hemostasis is closely related to ineffective erythropoiesis and increased sensitivity to stress factors. In this review, we summarized what is known about the functional roles of intracellular Ca2+ in erythropoiesis and erythrocyte-related diseases, with an emphasis on the regulation of the intracellular Ca2+ homeostasis during erythroid differentiation. An understanding of the regulation roles of Ca2+ homeostasis in erythroid differentiation will facilitate further studies and eventually molecular identification of the pathways involved in the pathological process of erythroid disorders, providing new therapeutic opportunities in erythrocyte-related disease.
Collapse
Affiliation(s)
- Yuanzhen Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yan Xu
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shujing Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhiyuan Lu
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yuan Li
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Baobing Zhao
- Department of Pharmacology, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China; Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
18
|
Decreased PGC1β expression results in disrupted human erythroid differentiation, impaired hemoglobinization and cell cycle exit. Sci Rep 2021; 11:17129. [PMID: 34429458 PMCID: PMC8385110 DOI: 10.1038/s41598-021-96585-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/10/2021] [Indexed: 11/08/2022] Open
Abstract
Production of red blood cells relies on proper mitochondrial function, both for their increased energy demands during differentiation and for proper heme and iron homeostasis. Mutations in genes regulating mitochondrial function have been reported in patients with anemia, yet their pathophysiological role often remains unclear. PGC1β is a critical coactivator of mitochondrial biogenesis, with increased expression during terminal erythroid differentiation. The role of PGC1β has however mainly been studied in skeletal muscle, adipose and hepatic tissues, and its function in erythropoiesis remains largely unknown. Here we show that perturbed PGC1β expression in human hematopoietic stem/progenitor cells from both bone marrow and cord blood results in impaired formation of early erythroid progenitors and delayed terminal erythroid differentiation in vitro, with accumulations of polychromatic erythroblasts, similar to MDS-related refractory anemia. Reduced levels of PGC1β resulted in deregulated expression of iron, heme and globin related genes in polychromatic erythroblasts, and reduced hemoglobin content in the more mature bone marrow derived reticulocytes. Furthermore, PGC1β knock-down resulted in disturbed cell cycle exit with accumulation of erythroblasts in S-phase and enhanced expression of G1-S regulating genes, with smaller reticulocytes as a result. Taken together, we demonstrate that PGC1β is directly involved in production of hemoglobin and regulation of G1-S transition and is ultimately required for proper terminal erythroid differentiation.
Collapse
|