1
|
Deleuze V, Soler E, Andrieu-Soler C. Protocol for efficient CRISPR-Cas9-mediated fluorescent tag knockin in hard-to-transfect erythroid cell lines. STAR Protoc 2024; 5:103016. [PMID: 38640065 PMCID: PMC11044133 DOI: 10.1016/j.xpro.2024.103016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/01/2024] [Accepted: 03/27/2024] [Indexed: 04/21/2024] Open
Abstract
Precise insertion of fluorescent tags by CRISPR-Cas9-mediated homologous recombination (HR) in mammalian genes is a powerful tool allowing to study gene function and protein gene products. Here, we present a protocol for efficient HR-mediated targeted insertion of fluorescent markers in the genome of hard-to-transfect erythroid cell lines MEL (mouse erythroleukemic) and MEDEP (mouse ES cell-derived erythroid progenitor line). We describe steps for plasmid construction, electroporation, amplification, and verification of genome editing. We then detail procedures for isolating positive clones and validating knockin clones. For complete details on the use and execution of this protocol, please refer to Deleuze et al.1.
Collapse
Affiliation(s)
- Virginie Deleuze
- IGMM University Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Université' de Paris, Paris, France
| | - Eric Soler
- IGMM University Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Université' de Paris, Paris, France.
| | - Charlotte Andrieu-Soler
- IGMM University Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Université' de Paris, Paris, France.
| |
Collapse
|
2
|
Nai A, Cordero-Sanchez C, Tanzi E, Pagani A, Silvestri L, Di Modica SM. Cellular and animal models for the investigation of β-thalassemia. Blood Cells Mol Dis 2024; 104:102761. [PMID: 37271682 DOI: 10.1016/j.bcmd.2023.102761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/24/2023] [Accepted: 05/26/2023] [Indexed: 06/06/2023]
Abstract
β-Thalassemia is a genetic form of anemia due to mutations in the β-globin gene, that leads to ineffective and extramedullary erythropoiesis, abnormal red blood cells and secondary iron-overload. The severity of the disease ranges from mild to lethal anemia based on the residual levels of globins production. Despite being a monogenic disorder, the pathophysiology of β-thalassemia is multifactorial, with different players contributing to the severity of anemia and secondary complications. As a result, the identification of effective therapeutic strategies is complex, and the treatment of patients is still suboptimal. For these reasons, several models have been developed in the last decades to provide experimental tools for the study of the disease, including erythroid cell lines, cultures of primary erythroid cells and transgenic animals. Years of research enabled the optimization of these models and led to decipher the mechanisms responsible for globins deregulation and ineffective erythropoiesis in thalassemia, to unravel the role of iron homeostasis in the disease and to identify and validate novel therapeutic targets and agents. Examples of successful outcomes of these analyses include iron restricting agents, currently tested in the clinics, several gene therapy vectors, one of which was recently approved for the treatment of most severe patients, and a promising gene editing strategy, that has been shown to be effective in a clinical trial. This review provides an overview of the available models, discusses pros and cons, and the key findings obtained from their study.
Collapse
Affiliation(s)
- Antonella Nai
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, via Olgettina 60, Milan, Italy; Vita-Salute San Raffaele University, via Olgettina 58, Milan, Italy.
| | - Celia Cordero-Sanchez
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, via Olgettina 60, Milan, Italy
| | - Emanuele Tanzi
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, via Olgettina 60, Milan, Italy
| | - Alessia Pagani
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, via Olgettina 60, Milan, Italy
| | - Laura Silvestri
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, via Olgettina 60, Milan, Italy; Vita-Salute San Raffaele University, via Olgettina 58, Milan, Italy
| | - Simona Maria Di Modica
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, via Olgettina 60, Milan, Italy
| |
Collapse
|
3
|
Deleuze V, Garcia L, Rouaisnel B, Salma M, Kinoo A, Andrieu-Soler C, Soler E. Efficient genome editing in erythroid cells unveils novel MYB target genes and regulatory functions. iScience 2023; 26:107641. [PMID: 37670779 PMCID: PMC10475484 DOI: 10.1016/j.isci.2023.107641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/09/2023] [Accepted: 08/11/2023] [Indexed: 09/07/2023] Open
Abstract
Targeted genome editing holds great promise in biology. However, efficient genome modification, including gene knock-in (KI), remains an unattained goal in multiple cell types and loci due to poor transfection efficiencies and low target genes expression, impeding the positive selection of recombined cells. Here, we describe a genome editing approach to achieve efficient gene targeting using hard to transfect erythroid cell lines. We demonstrate robust fluorescent protein KI efficiency in low expressed transcription factor (TF) genes (e.g., Myb or Zeb1). We further show the ability to target two independent loci in individual cells, exemplified by MYB-GFP and NuMA-Cherry double KI, allowing multicolor labeling of regulatory factors at physiological endogenous levels. Our KI tagging approach allowed us to perform genome-wide TF analysis at increased signal-to-noise ratios, and highlighted previously unidentified MYB target genes and pathways. Overall, we establish a versatile CRISPR-Cas9-based platform, offering attractive opportunities for the dissection of the erythroid differentiation process.
Collapse
Affiliation(s)
| | - Leonor Garcia
- IGMM, University Montpellier, CNRS, Montpellier, France
| | | | - Mohammad Salma
- IGMM, University Montpellier, CNRS, Montpellier, France
- Laboratory of Excellence GR-Ex, Université de Paris, Paris, France
| | - Alexia Kinoo
- IGMM, University Montpellier, CNRS, Montpellier, France
| | - Charlotte Andrieu-Soler
- IGMM, University Montpellier, CNRS, Montpellier, France
- Laboratory of Excellence GR-Ex, Université de Paris, Paris, France
| | - Eric Soler
- IGMM, University Montpellier, CNRS, Montpellier, France
- Laboratory of Excellence GR-Ex, Université de Paris, Paris, France
| |
Collapse
|
4
|
Wu J, Moriwaki K, Asuka T, Nakai R, Kanda S, Taniguchi M, Sugiyama T, Yoshimura SI, Kunii M, Nagasawa T, Hosen N, Miyoshi E, Harada A. EHBP1L1, an apicobasal polarity regulator, is critical for nuclear polarization during enucleation of erythroblasts. Blood Adv 2023; 7:3382-3394. [PMID: 37042948 PMCID: PMC10345855 DOI: 10.1182/bloodadvances.2022008930] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 03/13/2023] [Accepted: 03/31/2023] [Indexed: 04/13/2023] Open
Abstract
Cell polarity, the asymmetric distribution of proteins and organelles, is permanently or transiently established in various cell types and plays an important role in many physiological events. epidermal growth factor receptor substrate 15 homology domain-binding protein 1-like 1 (EHBP1L1) is an adapter protein that is localized on recycling endosomes and regulates apical-directed transport in polarized epithelial cells. However, the role of EHBP1L1 in nonepithelial cells, remains unknown. Here, Ehbp1l1-/- mice showed impaired erythroblast enucleation. Further analyses showed that nuclear polarization before enucleation was impaired in Ehbp1l1-/- erythroblasts. It was also revealed that EHBP1L1 interactors Rab10, Bin1, and dynamin were involved in erythroblast enucleation. In addition, Ehbp1l1-/- erythrocytes exhibited stomatocytic morphology and dehydration. These defects in erythroid cells culminated in early postnatal anemic lethality in Ehbp1l1-/- mice. Moreover, we found the mislocalization of nuclei and mitochondria in the skeletal muscle cells of Ehbp1l1-/- mice, as observed in patients with centronuclear myopathy with genetic mutations in Bin1 or dynamin 2. Taken together, our findings indicate that the Rab8/10-EHBP1L1-Bin1-dynamin axis plays an important role in multiple cell polarity systems in epithelial and nonepithelial cells.
Collapse
Affiliation(s)
- Ji Wu
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Kenta Moriwaki
- Department of Biochemistry, Toho University School of Medicine, Tokyo, Japan
| | - Tatsuya Asuka
- Department of Molecular Biochemistry and Clinical Investigation, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Ritsuko Nakai
- Department of Hematology and Oncology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Satoshi Kanda
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Manabu Taniguchi
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Tatsuki Sugiyama
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Shin-ichiro Yoshimura
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Masataka Kunii
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Takashi Nagasawa
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Naoki Hosen
- Department of Hematology and Oncology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Eiji Miyoshi
- Department of Molecular Biochemistry and Clinical Investigation, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Akihiro Harada
- Department of Cell Biology, Graduate School of Medicine, Osaka University, Suita, Japan
| |
Collapse
|
5
|
Cervellera CF, Mazziotta C, Di Mauro G, Iaquinta MR, Mazzoni E, Torreggiani E, Tognon M, Martini F, Rotondo JC. Immortalized erythroid cells as a novel frontier for in vitro blood production: current approaches and potential clinical application. Stem Cell Res Ther 2023; 14:139. [PMID: 37226267 PMCID: PMC10210309 DOI: 10.1186/s13287-023-03367-8] [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: 01/17/2023] [Accepted: 05/05/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Blood transfusions represent common medical procedures, which provide essential supportive therapy. However, these procedures are notoriously expensive for healthcare services and not without risk. The potential threat of transfusion-related complications, such as the development of pathogenic infections and the occurring of alloimmunization events, alongside the donor's dependence, strongly limits the availability of transfusion units and represents significant concerns in transfusion medicine. Moreover, a further increase in the demand for donated blood and blood transfusion, combined with a reduction in blood donors, is expected as a consequence of the decrease in birth rates and increase in life expectancy in industrialized countries. MAIN BODY An emerging and alternative strategy preferred over blood transfusion is the in vitro production of blood cells from immortalized erythroid cells. The high survival capacity alongside the stable and longest proliferation time of immortalized erythroid cells could allow the generation of a large number of cells over time, which are able to differentiate into blood cells. However, a large-scale, cost-effective production of blood cells is not yet a routine clinical procedure, as being dependent on the optimization of culture conditions of immortalized erythroid cells. CONCLUSION In our review, we provide an overview of the most recent erythroid cell immortalization approaches, while also describing and discussing related advancements of establishing immortalized erythroid cell lines.
Collapse
Affiliation(s)
- Christian Felice Cervellera
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
| | - Chiara Mazziotta
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
- Department of Medical Sciences, Center for Studies on Gender Medicine, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
| | - Giulia Di Mauro
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
| | - Maria Rosa Iaquinta
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
- Department of Medical Sciences, Center for Studies on Gender Medicine, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
| | - Elisa Mazzoni
- Department of Chemical, Pharmaceutical and Agricultural Sciences-DOCPAS, University of Ferrara, 44121, Ferrara, Italy
| | - Elena Torreggiani
- Department of Chemical, Pharmaceutical and Agricultural Sciences-DOCPAS, University of Ferrara, 44121, Ferrara, Italy
| | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy.
- Department of Medical Sciences, Center for Studies on Gender Medicine, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy.
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121, Ferrara, Italy.
| | - John Charles Rotondo
- Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy.
- Department of Medical Sciences, Center for Studies on Gender Medicine, University of Ferrara, 64/b, Fossato di Mortara Street, 44121, Ferrara, Italy.
| |
Collapse
|
6
|
Ohmori S, Takai J, Uemura S, Otsuki A, Mori T, Ohneda K, Moriguchi T. The Il6 -39 kb enhancer containing clustered GATA2- and PU.1-binding sites is essential for Il6 expression in murine mast cells. iScience 2022; 25:104942. [PMID: 36072552 PMCID: PMC9442365 DOI: 10.1016/j.isci.2022.104942] [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: 02/17/2022] [Revised: 06/17/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Mast cells serve as a first-line defense of innate immunity. Interleukin-6 (IL-6) induced by bacterial lipopolysaccharide (LPS) in mast cells plays a crucial role in antibacterial protection. The zinc finger transcription factor GATA2 cooperatively functions with the ETS family transcription factor PU.1 in multiple mast cell activities. However, the regulatory landscape directed by GATA2 and PU.1 under inflammation remains elusive. We herein showed that a large proportion of GATA2-binding peaks were closely located with PU.1-binding peaks in distal cis-regulatory regions of inflammatory cytokine genes in mast cells. Notably, GATA2 and PU.1 played crucial roles in promoting LPS-mediated inflammatory cytokine production. Genetic ablation of GATA2-PU.1-clustered binding sites at the Il6 -39 kb region revealed its central role in LPS-induced Il6 expression in mast cells. We demonstrate a novel collaborative activity of GATA2 and PU.1 in cytokine induction upon inflammatory stimuli via the GATA2-PU.1 overlapping sites in the distal cis-regulatory regions. GATA2- and PU.1-binding peaks are closely located in distal enhancers of cytokine genes GATA2 and PU.1 play crucial roles in promoting LPS-mediated cytokine induction The Il6 -39 kb enhancer containing GATA2 and PU.1 motifs are crucial for Il6 induction GATA2 inhibitor exerts anti-inflammatory effects via reducing cytokine induction
Collapse
|
7
|
Mennen RH, Oldenburger MM, Piersma AH. Endoderm and mesoderm derivatives in embryonic stem cell differentiation and their use in developmental toxicity testing. Reprod Toxicol 2021; 107:44-59. [PMID: 34861400 DOI: 10.1016/j.reprotox.2021.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/25/2021] [Accepted: 11/29/2021] [Indexed: 02/06/2023]
Abstract
Embryonic stem cell differentiation models have increasingly been applied in non-animal test systems for developmental toxicity. After the initial focus on cardiac differentiation, attention has also included an array of neuro-ectodermal differentiation routes. Alternative differentiation routes in the mesodermal and endodermal germ lines have received less attention. This review provides an inventory of achievements in the latter areas of embryonic stem cell differentiation, with a view to possibilities for their use in non-animal test systems in developmental toxicology. This includes murine and human stem cell differentiation models, and also gains information from the field of stem cell use in regenerative medicine. Endodermal stem cell derivatives produced in vitro include hepatocytes, pancreatic cells, lung epithelium, and intestinal epithelium, and mesodermal derivatives include cardiac muscle, osteogenic, vascular and hemopoietic cells. This inventory provides an overview of studies on the different cell types together with biomarkers and culture conditions that stimulate these differentiation routes from embryonic stem cells. These models may be used to expand the spectrum of embryonic stem cell based new approach methodologies in non-animal developmental toxicity testing.
Collapse
Affiliation(s)
- R H Mennen
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands.
| | | | - A H Piersma
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands; Institute for Risk Assessment Sciences (IRAS), Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
8
|
Soboleva S, Kurita R, Kajitani N, Åkerstrand H, Miharada K. Establishment of an immortalized human erythroid cell line sustaining differentiation potential without inducible gene expression system. Hum Cell 2021; 35:408-417. [PMID: 34817797 DOI: 10.1007/s13577-021-00652-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/17/2021] [Indexed: 11/30/2022]
Abstract
Ex vivo manufactured red blood cells (RBC) generated from immortalized erythroid cell lines which can continuously grow are expected to become a significant alternative in future transfusion therapies. The ectopic expression of human papilloma virus (HPV) E6/E7 gene has successfully been employed to establish these cell lines. To induce differentiation and maturation of the immortalized cell lines, terminating the HPV-E6/E7 expression through a gene induction system has been believed to be essential. Here, we report that erythroid cell lines established from human bone marrow using simple expression of HPV-E6/E7 are capable of normal erythroid differentiation, without turning gene expression off. Through simply changing cell culture conditions, a newly established cell line, Erythroid Line from Lund University (ELLU), is able to differentiate toward mature cells, including enucleated reticulocytes. ELLU is heterogeneous and, unexpectedly, clones expressing adult hemoglobin rapidly differentiate and produce fragile cells. Upon differentiation, other ELLU clones shift from fetal to adult hemoglobin expression, giving rise to more mature cells. Our findings propose that it is not necessary to employ gene induction systems to establish immortalized erythroid cell lines sustaining differentiation potential and describe novel cellular characteristics for desired functionally competent clones.
Collapse
Affiliation(s)
- Svetlana Soboleva
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Ryo Kurita
- Department of Research and Development, Central Blood Institute, Japanese Red Cross Society, Tokyo, Japan
| | - Naoko Kajitani
- Division of Medical Microbiology, Lund University, Lund, Sweden
| | - Hugo Åkerstrand
- Division of Molecular Hematology, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Kenichi Miharada
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden. .,International Research Center for Medical Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
9
|
Comprehensive proteomic analysis of murine terminal erythroid differentiation. Blood Adv 2021; 4:1464-1477. [PMID: 32282884 DOI: 10.1182/bloodadvances.2020001652] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 03/04/2020] [Indexed: 12/12/2022] Open
Abstract
Murine-based cellular models have provided and continue to provide many useful insights into the fundamental mechanisms of erythropoiesis, as well as insights into the pathophysiology of inherited and acquired red cell disorders. Although detailed information on many aspects of these cell models is available, comprehensive proteomic data are lacking. This is a critical knowledge gap, as proteins are effectors of most biologic processes. To address this critical unmet need, proteomes of the murine cell lines Friend erythroleukemia (MEL), GATA1 erythroid (G1ER), and embryonic stem cell-derived erythroid progenitor (MEDEP) and proteomes of cultured murine marrow-derived erythroblasts at different stages of terminal erythroid differentiation were analyzed. The proteomes of MEDEP cells and primary murine erythroid cells were most similar, whereas those of MEL and G1ER cells were more distantly related. We demonstrated that the overall cellular content of histones does not decrease during terminal differentiation, despite strong chromatin condensation. Comparison of murine and human proteomes throughout terminal erythroid differentiation revealed that many noted transcriptomic changes were significantly dampened at the proteome level, especially at the end of the terminal differentiation process. Analysis of the early events associated with induction of terminal differentiation in MEDEP cells revealed divergent alterations in associated transcriptomes and proteomes. These proteomic data are powerful and valuable tools for the study of fundamental mechanisms of normal and disordered erythropoiesis and will be of broad interest to a wide range of investigators for making the appropriate choice of various cell lines to study inherited and acquired diseases of the erythrocyte.
Collapse
|
10
|
Richard C, Viret S, Cantero Aguilar L, Lefevre C, Leduc M, Faouzi EH, Azar N, Lavazec C, Mayeux P, Verdier F. Myotonic dystrophy kinase-related CDC42-binding kinase α, a new transferrin receptor type 2-binding partner, is a regulator of erythropoiesis. Am J Hematol 2021; 96:480-492. [PMID: 33476437 DOI: 10.1002/ajh.26104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 01/01/2023]
Abstract
Efficient erythropoiesis relies on the expression of the transferrin receptor type 2 (TFR2). In erythroid precursors, TFR2 facilitates the export of the erythropoietin receptor (EPOR) to cell surface, which ensures the survival and proliferation of erythroblasts. Although TFR2 has a crucial role in erythropoiesis regulation, its mechanism of action remains to be clarified. To understand its role better, we aimed at identifying its protein partners by mass-spectrometry after immunoprecipitation in erythroid cells. Here we report the kinase MRCKα (myotonic dystrophy kinase-related CDC42-binding kinase α) as a new partner of both TFR2 and EPOR in erythroblasts. We show that MRCKα is co-expressed with TFR2, and TFR1 during terminal differentiation and regulates the internalization of the two types of transferrin receptors. The knockdown of MRCKα by shRNA in human primary erythroblasts leads to a decreased cell surface expression of both TFR1 and TFR2, an increased cell-surface expression of EPOR, and a delayed differentiation. Additionally, knockout of Mrckα in the murine MEDEP cells also leads to a striking delay in erythropoiesis, showcasing the importance of this kinase in both species. Our data highlight the importance of MRCKα in the regulation of erythropoiesis.
Collapse
Affiliation(s)
- Cyrielle Richard
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
| | - Sophie Viret
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
| | - Lilia Cantero Aguilar
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
| | - Carine Lefevre
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
| | - Marjorie Leduc
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
- Plateforme Protéomique 3P5‐Proteom'IC, Université de Paris, Institut Cochin, INSERM, U1016, CNRS UMR8104 Paris France
| | - El Hassan Faouzi
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
| | - Nabih Azar
- Unité d'Hémobiothérapie, Hôpital La Pitié Salpêtrière Paris France
| | - Catherine Lavazec
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
| | - Patrick Mayeux
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
- Plateforme Protéomique 3P5‐Proteom'IC, Université de Paris, Institut Cochin, INSERM, U1016, CNRS UMR8104 Paris France
| | - Frédérique Verdier
- Université de Paris, Institut Cochin, INSERM U1016‐CNRS UMR8104 Paris France
- Laboratory of Excellence GR‐Ex Paris France
| |
Collapse
|
11
|
Goto H, Miyamoto M, Kihara A. Direct uptake of sphingosine-1-phosphate independent of phospholipid phosphatases. J Biol Chem 2021; 296:100605. [PMID: 33785361 PMCID: PMC8093947 DOI: 10.1016/j.jbc.2021.100605] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 12/20/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a lipid mediator that is relatively abundant in plasma and plays an important role in the vascular and immune systems. To date, the only known mechanism for removing S1P from plasma has been dephosphorylation by phospholipid phosphatases (PLPPs) on the surface of cells in contact with the plasma. However, there remains a possibility that PLPP-independent dephosphorylation or direct S1P uptake into cells could occur. To examine these possibilities, here we generated triple KO (TKO) HAP1 cells that lacked all PLPPs (PLPP1–3) present in mammals. In the TKO cells, the intracellular metabolism of externally added deuterium-labeled S1P to ceramide was reduced to 17% compared with the WT cells, indicating that most extracellular S1P is dephosphorylated by PLPPs and then taken up into cells. However, this result also reveals the existence of a PLPP-independent S1P uptake pathway. Tracer experiments using [32P]S1P showed the existence of a direct S1P uptake pathway that functions without prior dephosphorylation. Overexpression of sphingolipid transporter 2 (SPNS2) or of major facilitator superfamily domain containing 2B (MFSD2B), both known S1P efflux transporters, in TKO cells increased the direct uptake of S1P, whereas KO of MFSD2B in TKO cells reduced this uptake. These results suggest that these are channel-type transporters and capable of not only exporting but also importing S1P. Furthermore, we observed that erythroid cells expressing MFSD2B, exhibited high S1P uptake activity. Our findings describing direct S1P uptake may contribute to the elucidation of the molecular mechanisms that regulate plasma S1P concentration.
Collapse
Affiliation(s)
- Hirotaka Goto
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | | | - Akio Kihara
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
12
|
Dysregulation of erythropoiesis and altered erythroblastic NMDA receptor-mediated calcium influx in Lrfn2-deficient mice. PLoS One 2021; 16:e0245624. [PMID: 33481887 PMCID: PMC7822338 DOI: 10.1371/journal.pone.0245624] [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: 10/14/2020] [Accepted: 01/04/2021] [Indexed: 01/12/2023] Open
Abstract
LRFN2 encodes a synaptic adhesion-like molecule that physically interacts with N-methyl-D-aspartate (NMDA) receptor 1 and its scaffold proteins. Previous studies in humans and mice have demonstrated its genetic association with neurodevelopmental disorders such as learning deficiency and autism. In this study, we showed that Lrfn2-deficient (KO) mice exhibit abnormalities of erythropoietic systems due to altered NMDA receptor function. In mature Lrfn2 KO male mice, peripheral blood tests showed multilineage abnormalities, including normocytic erythrocythemia, and reduced platelet volume. Colony forming unit assay using bone marrow cells revealed decreases in the counts of erythrocyte progenitors (CFU-E) as well as granulocytes and monocyte progenitors (CFU-GM). Whole bone marrow cell staining showed that serum erythropoietin (EPO) level was decreased and EPO receptor-like immunoreactivity was increased. Flow cytometry analysis of bone marrow cells revealed increased early erythroblast count and increased transferrin receptor expression in late erythroblasts. Further, we found that late erythroblasts in Lrfn2 KO exhibited defective NMDA receptor-mediated calcium influx, which was inhibited by the NMDA receptor antagonist MK801. These results indicate that Lrfn2 has biphasic roles in hematopoiesis and is associated with the functional integrity of NMDA receptors in hematopoietic cells. Furthermore, taken together with previous studies that showed the involvement of NMDA receptors in hematopoiesis, the results of this study indicate that Lrfn2 may regulate erythropoiesis through its regulatory activity on NMDA receptors.
Collapse
|
13
|
Kiatpakdee B, Sato K, Otsuka Y, Arashiki N, Chen Y, Tsumita T, Otsu W, Yamamoto A, Kawata R, Yamazaki J, Sugimoto Y, Takada K, Mohandas N, Inaba M. Cholesterol-binding protein TSPO2 coordinates maturation and proliferation of terminally differentiating erythroblasts. J Biol Chem 2020; 295:8048-8063. [PMID: 32358067 PMCID: PMC7278357 DOI: 10.1074/jbc.ra119.011679] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 04/20/2020] [Indexed: 02/02/2023] Open
Abstract
TSPO2 (translocator protein 2) is a transmembrane protein specifically expressed in late erythroblasts and has been postulated to mediate intracellular redistribution of cholesterol. We identified TSPO2 as the causative gene for the HK (high-K+) trait with immature red cell phenotypes in dogs and investigated the effects of the TSPO2 defects on erythropoiesis in HK dogs with the TSPO2 mutation and Tspo2 knockout (Tspo2−/−) mouse models. Bone marrow–derived erythroblasts from HK dogs showed increased binucleated and apoptotic cells at various stages of maturation and shed large nuclei with incomplete condensation when cultured in the presence of erythropoietin, indicating impaired maturation and cytokinesis. The canine TSPO2 induces cholesterol accumulation in the endoplasmic reticulum and could thereby regulate cholesterol availability by changing intracellular cholesterol distribution in erythroblasts. Tspo2−/− mice consistently showed impaired cytokinesis with increased binucleated erythroblasts, resulting in compensated anemia, and their red cell membranes had increased Na,K-ATPase, resembling the HK phenotype in dogs. Tspo2-deficient mouse embryonic stem cell–derived erythroid progenitor (MEDEP) cells exhibited similar morphological defects associated with a cell-cycle arrest at the G2/M phase, resulting in decreased cell proliferation and had a depletion in intracellular unesterified and esterified cholesterol. When the terminal maturation was induced, Tspo2−/− MEDEP cells showed delays in hemoglobinization; maturation-associated phenotypic changes in CD44, CD71, and TER119 expression; and cell-cycle progression. Taken together, these findings imply that TSPO2 is essential for coordination of maturation and proliferation of erythroblasts during normal erythropoiesis.
Collapse
Affiliation(s)
- Benjaporn Kiatpakdee
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Kota Sato
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Yayoi Otsuka
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Nobuto Arashiki
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Yuqi Chen
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Takuya Tsumita
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Wataru Otsu
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Akito Yamamoto
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Reo Kawata
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Jumpei Yamazaki
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | | | - Kensuke Takada
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Narla Mohandas
- Red Cell Physiology Laboratory, New York Blood Center, New York, New York, USA
| | - Mutsumi Inaba
- Laboratory of Molecular Medicine, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
14
|
Fibach E. Erythropoiesis In Vitro-A Research and Therapeutic Tool in Thalassemia. J Clin Med 2019; 8:jcm8122124. [PMID: 31810354 PMCID: PMC6947291 DOI: 10.3390/jcm8122124] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 11/26/2019] [Accepted: 11/29/2019] [Indexed: 12/13/2022] Open
Abstract
Thalassemia (thal) is a hereditary chronic hemolytic anemia due to a partial or complete deficiency in the production of globin chains, in most cases, α or β, which compose, together with the iron-containing porphyrins (hemes), the hemoglobin molecules in red blood cells (RBC). The major clinical symptom of β-thal is severe chronic anemia—a decrease in RBC number and their hemoglobin content. In spite of the improvement in therapy, thal still severely affects the quality of life of the patients and their families and imposes a substantial financial burden on the community. These considerations position β-thal, among other hemoglobinopathies, as a major health and social problem that deserves increased efforts in research and its clinical application. These efforts are based on clinical studies, experiments in animal models and the use of erythroid cells grown in culture. The latter include immortal cell lines and cultures initiated by erythroid progenitor and stem cells derived from the blood and RBC producing (erythropoietic) sites of normal and thal donors, embryonic stem cells, and recently, "induced pluripotent stem cells" generated by manipulation of differentiated somatic cells. The present review summarizes the use of erythroid cultures, their technological aspects and their contribution to the research and its clinical application in thal. The former includes deciphering of the normal and pathological biology of the erythroid cell development, and the latter—their role in developing innovative therapeutics—drugs and methods of gene therapy, as well as providing an alternative source of RBC that may complement or substitute blood transfusions.
Collapse
Affiliation(s)
- Eitan Fibach
- The Hematology Department, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| |
Collapse
|
15
|
GATA2 and PU.1 Collaborate To Activate the Expression of the Mouse Ms4a2 Gene, Encoding FcεRIβ, through Distinct Mechanisms. Mol Cell Biol 2019; 39:MCB.00314-19. [PMID: 31501274 DOI: 10.1128/mcb.00314-19] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/02/2019] [Indexed: 12/20/2022] Open
Abstract
GATA factors GATA1 and GATA2 and ETS factor PU.1 are known to function antagonistically during hematopoietic development. In mouse mast cells, however, these factors are coexpressed and activate the expression of the Ms4a2 gene encoding the β chain of the high-affinity IgE receptor (FcεRI). The present study showed that these factors cooperatively regulate Ms4a2 gene expression through distinct mechanisms. Although GATA2 and PU.1 contributed almost equally to Ms4a2 gene expression, gene ablation experiments revealed that simultaneous knockdown of both factors showed neither a synergistic nor an additive effect. A chromatin immunoprecipitation analysis showed that they shared DNA binding to the +10.4-kbp region downstream of the Ms4a2 gene with chromatin looping factor LDB1, whereas the proximal -60-bp region was exclusively bound by GATA2 in a mast cell-specific manner. Ablation of PU.1 significantly reduced the level of GATA2 binding to both the +10.4-kbp and -60-bp regions. Surprisingly, the deletion of the +10.4-kbp region by genome editing completely abolished the Ms4a2 gene expression as well as the cell surface expression of FcεRI. These results suggest that PU.1 and LDB1 play central roles in the formation of active chromatin structure whereas GATA2 directly activates the Ms4a2 promoter.
Collapse
|
16
|
Ohneda K, Ohmori S, Yamamoto M. Mouse Tryptase Gene Expression is Coordinately Regulated by GATA1 and GATA2 in Bone Marrow-Derived Mast Cells. Int J Mol Sci 2019; 20:ijms20184603. [PMID: 31533351 PMCID: PMC6770354 DOI: 10.3390/ijms20184603] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 09/09/2019] [Accepted: 09/11/2019] [Indexed: 12/12/2022] Open
Abstract
Mast cell tryptases have crucial roles in allergic and inflammatory diseases. The mouse tryptase genes represent a cluster of loci on chromosome 16p3.3. While their functional studies have been extensively performed, transcriptional regulation of tryptase genes is poorly understood. In this study, we examined the molecular basis of the tryptase gene expression in bone marrow-derived mast cells (BMMCs) of C57BL/6 mice and in MEDMC-BRC6 mast cells. The expression of the Tpsb2 and Tpsg1 genes, which reside at the 3′-end of the tryptase locus, is significantly decreased by the reduction of the GATA transcription factors GATA1 or GATA2. Chromatin immunoprecipitation assays have shown that the GATA factors bind at multiple regions within the locus, including 1.0 and 72.8 kb upstream of the Tpsb2 gene, and that GATA1 and GATA2 facilitate each other’s DNA binding activity to these regions. Deletion of the −72.8 kb region by genome editing significantly reduced the Tpsb2 and Tpsg1 mRNA levels in MEDMC-BRC6 cells. Furthermore, binding of CTCF and the cohesin subunit Rad21 was found upstream of the −72.8 kb region and was significantly reduced in the absence of GATA1. These results suggest that mouse tryptase gene expression is coordinately regulated by GATA1 and GATA2 in BMMCs.
Collapse
Affiliation(s)
- Kinuko Ohneda
- Department of Pharmacy, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki 370-0033, Japan;
- Correspondence: ; Tel.: +81-22-274-5990
| | - Shin’ya Ohmori
- Department of Pharmacy, Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki 370-0033, Japan;
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai 980-8573, Japan;
| |
Collapse
|
17
|
Zingariello M, Bardelli C, Sancillo L, Ciaffoni F, Genova ML, Girelli G, Migliaccio AR. Dexamethasone Predisposes Human Erythroblasts Toward Impaired Lipid Metabolism and Renders Their ex vivo Expansion Highly Dependent on Plasma Lipoproteins. Front Physiol 2019; 10:281. [PMID: 31019464 PMCID: PMC6458278 DOI: 10.3389/fphys.2019.00281] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 03/04/2019] [Indexed: 02/04/2023] Open
Abstract
Cultures of stem cells from discarded sources supplemented with dexamethasone, a synthetic glucocorticoid receptor agonist, generate cultured red blood cells (cRBCs) in numbers sufficient for transfusion. According to the literature, however, erythroblasts generated with dexamethasone exhibit low enucleation rates giving rise to cRBCs that survive poorly in vivo. The knowledge that the glucocorticoid receptor regulates lipid metabolism and that lipid composition dictates the fragility of the plasma membrane suggests that insufficient lipid bioavailability restrains generation of cRBCs. To test this hypothesis, we first compared the expression profiling of erythroblasts generated with or without dexamethasone. This analysis revealed differences in expression of 55 genes, 6 of which encoding proteins involved in lipid metabolism. These were represented by genes encoding the mitochondrial proteins 3-Hydroxymethyl-3-Methylglutaryl-CoA lyase, upregulated, and 3-Oxoacid CoA-Transferase1 and glycerol-3-phosphate acyltransferase1, both downregulated, and the proteins ATP-binding cassette transporter1 and Hydroxysteroid-17-Beta-Dehydrogenase7, upregulated, and cAMP-dependent protein kinase catalytic subunit beta, downregulated. This profiling predicts that dexamethasone, possibly by interfering with mitochondrial functions, impairs the intrinsic lipid metabolism making the synthesis of the plasma membrane of erythroid cells depend on lipid-uptake from external sources. Optical and electron microscopy analyses confirmed that the mitochondria of erythroblasts generated with dexamethasone are abnormal and that their plasma membranes present pebbles associated with membrane ruptures releasing exosomes and micro-vesicles. These results indicate that the lipid supplements of media currently available are not adequate for cRBCs. To identify better lipid supplements, we determined the number of erythroblasts generated in synthetic media supplemented with either currently used liposomes or with lipoproteins purified from human plasma [the total lipoprotein fraction (TL) or its high (HDL), low (LDL) and very low (VLDL) density lipoprotein components]. Both LDL and VLDL generated numbers of erythroid cells 3-2-fold greater than that observed in controls. These greater numbers were associated with 2-3-fold greater amplification of erythroid cells due both to increased proliferation and to resistance to stress-induced death. In conclusion, dexamethasone impairs lipid metabolism making ex vivo expansion of erythroid cells highly dependent on lipid absorbed from external sources and the use of LDL and VLDL as lipid supplements improves the generation of cRBCs.
Collapse
Affiliation(s)
- Maria Zingariello
- Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Claudio Bardelli
- Department of Biomedical and NeuroMotor Sciences, Alma Mater Studiorum University, Bologna, Italy
| | - Laura Sancillo
- Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | | | - Maria Luisa Genova
- Department of Biomedical and NeuroMotor Sciences, Alma Mater Studiorum University, Bologna, Italy
| | | | - Anna Rita Migliaccio
- Department of Biomedical and NeuroMotor Sciences, Alma Mater Studiorum University, Bologna, Italy
| |
Collapse
|
18
|
Yang C, Hashimoto M, Lin QXX, Tan DQ, Suda T. Sphingosine-1-phosphate signaling modulates terminal erythroid differentiation through the regulation of mitophagy. Exp Hematol 2019; 72:47-59.e1. [DOI: 10.1016/j.exphem.2019.01.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/10/2019] [Accepted: 01/11/2019] [Indexed: 01/17/2023]
|
19
|
Diepstraten ST, Hart AH. Modelling human haemoglobin switching. Blood Rev 2018; 33:11-23. [PMID: 30616747 DOI: 10.1016/j.blre.2018.06.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/11/2018] [Accepted: 06/14/2018] [Indexed: 12/22/2022]
Abstract
Genetic lesions of the β-globin gene result in haemoglobinopathies such as β-thalassemia and sickle cell disease. To discover and test new molecular medicines for β-haemoglobinopathies, cell-based and animal models are now being widely utilised. However, multiple in vitro and in vivo models are required due to the complex structure and regulatory mechanisms of the human globin gene locus, subtle species-specific differences in blood cell development, and the influence of epigenetic factors. Advances in genome sequencing, gene editing, and precision medicine have enabled the first generation of molecular therapies aimed at reactivating, repairing, or replacing silenced or damaged globin genes. Here we compare and contrast current animal and cell-based models, highlighting their complementary strengths, reflecting on how they have informed the scope and direction of the field, and describing some of the novel molecular and precision medicines currently under development or in clinical trial.
Collapse
Affiliation(s)
- Sarah T Diepstraten
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia.
| | - Adam H Hart
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia.
| |
Collapse
|
20
|
Minetti G, Achilli C, Perotti C, Ciana A. Continuous Change in Membrane and Membrane-Skeleton Organization During Development From Proerythroblast to Senescent Red Blood Cell. Front Physiol 2018; 9:286. [PMID: 29632498 PMCID: PMC5879444 DOI: 10.3389/fphys.2018.00286] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 03/12/2018] [Indexed: 12/11/2022] Open
Abstract
Within the context of erythropoiesis and the possibility of producing artificial red blood cells (RBCs) in vitro, a most critical step is the final differentiation of enucleated erythroblasts, or reticulocytes, to a fully mature biconcave discocyte, the RBC. Reviewed here is the current knowledge about this fundamental maturational process. By combining literature data with our own experimental evidence we propose that the early phase in the maturation of reticulocytes to RBCs is driven by a membrane raft-based mechanism for the sorting of disposable membrane proteins, mostly the no longer needed transferrin receptor (TfR), to the multivesicular endosome (MVE) as cargo of intraluminal vesicles that are subsequently exocytosed as exosomes, consistently with the seminal and original observation of Johnstone and collaborators of more than 30 years ago (Pan BT, Johnstone RM. Cell. 1983;33:967-978). According to a strikingly selective sorting process, the TfR becomes cargo destined to exocytosis while other molecules, including the most abundant RBC transmembrane protein, band 3, are completely retained in the cell membrane. It is also proposed that while this process could be operating in the early maturational steps in the bone marrow, additional mechanism(s) must be at play for the final removal of the excess reticulocyte membrane that is observed to occur in the circulation. This processing will most likely require the intervention of the spleen, whose function is also necessary for the continuous remodeling of the RBC membrane all along this cell's circulatory life.
Collapse
Affiliation(s)
- Giampaolo Minetti
- Laboratori di Biochimica, Dipartimento di Biologia e Biotecnologie, Università degli Studi di Pavia, Pavia, Italy
| | - Cesare Achilli
- Laboratori di Biochimica, Dipartimento di Biologia e Biotecnologie, Università degli Studi di Pavia, Pavia, Italy
| | - Cesare Perotti
- Servizio Immunoematologia e Medicina Trasfusionale, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Annarita Ciana
- Laboratori di Biochimica, Dipartimento di Biologia e Biotecnologie, Università degli Studi di Pavia, Pavia, Italy
| |
Collapse
|
21
|
MFSD2B is a sphingosine 1-phosphate transporter in erythroid cells. Sci Rep 2018; 8:4969. [PMID: 29563527 PMCID: PMC5862976 DOI: 10.1038/s41598-018-23300-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 03/02/2018] [Indexed: 12/22/2022] Open
Abstract
Sphingosine 1-phosphate (S1P) is an intercellular signaling molecule present in blood. Erythrocytes have a central role in maintaining the S1P concentration in the blood stream. We previously demonstrated that S1P is exported from erythrocytes by a glyburide-sensitive S1P transporter. However, the gene encoding the S1P transporter in erythrocytes is unknown. In this study, we found that the mouse erythroid cell line, MEDEP-E14, has S1P export activity and exhibits properties that are consistent with those of erythrocytes. Using microarray analysis of MEDEP-E14 cells and its parental cell line, E14TG2a, we identified several candidate genes for S1P export activity. Of those genes, only one gene, Mfsd2b, showed S1P transport activity. The properties of S1P release by MFSD2B were similar to those in erythrocytes. Moreover, knockout of MFSD2B in MEDEP-E14 cells decreased S1P export from the cells. These results strongly suggest that MFSD2B is a novel S1P transporter in erythroid cells.
Collapse
|
22
|
|
23
|
Zhang Y, Wang C, Wang L, Shen B, Guan X, Tian J, Ren Z, Ding X, Ma Y, Dai W, Jiang Y. Large-Scale Ex Vivo Generation of Human Red Blood Cells from Cord Blood CD34 + Cells. Stem Cells Transl Med 2017; 6:1698-1709. [PMID: 28618139 PMCID: PMC5689780 DOI: 10.1002/sctm.17-0057] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 05/08/2017] [Indexed: 12/17/2022] Open
Abstract
The ex vivo generation of human red blood cells on a large scale from hematopoietic stem and progenitor cells has been considered as a potential method to overcome blood supply shortages. Here, we report that functional human erythrocytes can be efficiently produced from cord blood (CB) CD34+ cells using a bottle turning device culture system. Safety and efficiency studies were performed in murine and nonhuman primate (NHP) models. With the selected optimized culture conditions, one human CB CD34+ cell could be induced ex vivo to produce up to 200 million erythrocytes with a purity of 90.1% ± 6.2% and 50% ± 5.7% (mean ± SD) for CD235a+ cells and enucleated cells, respectively. The yield of erythrocytes from one CB unit (5 million CD34+ cells) could be, in theory, equivalent to 500 blood transfusion units in clinical application. Moreover, induced human erythrocytes had normal hemoglobin content and could continue to undergo terminal maturation in the murine xenotransplantation model. In NHP model, xenotransplantation of induced human erythrocytes enhanced hematological recovery and ameliorated the hypoxia situation in the primates with hemorrhagic anemia. These findings suggested that the ex vivo-generated erythrocytes could be an alternative blood source for traditional transfusion products in the clinic. Stem Cells Translational Medicine 2017;6:1698-1709.
Collapse
Affiliation(s)
- Yu Zhang
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| | - Chen Wang
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.,Biopharmagen Corp, Suzhou, China
| | - Lan Wang
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| | - Bin Shen
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| | - Xin Guan
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| | - Jing Tian
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| | - Zhihua Ren
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.,Biopharmagen Corp, Suzhou, China
| | - Xinxin Ding
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.,College of Nanoscale Science, SUNY Polytechnic Institute, Albany, New York, USA
| | - Yupo Ma
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.,Department of Pathology, School of Medicine, The State University of New York at Stony Brook, Stony Brook, New York, USA
| | - Wei Dai
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.,Department of Environment Medicine, New York University Langone Medical center, Tuxedo, New York, USA
| | - Yongping Jiang
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.,Biopharmagen Corp, Suzhou, China
| |
Collapse
|
24
|
Shibagaki S, Tahara-Hanaoka S, Hiroyama T, Nakamura Y, Shibuya A. Long-term survival of the mouse ES cell-derived mast cell, MEDMC-BRC6, in mast cell-deficient KitW-sh/W-sh mice. Int Immunol 2017; 29:235-242. [PMID: 28431112 DOI: 10.1093/intimm/dxx022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 04/12/2017] [Indexed: 01/04/2023] Open
Abstract
Mast cells (MCs) play pivotal roles in allergic reactions and the host defense against microbial infection through the IgE-dependent and IgE-independent signaling pathways. MC lines that can be analyzed both in vitro and in vivo would be useful for the study of MC-dependent immune responses. Here, we investigated the functional characteristics of a mouse embryonic stem cell-derived MC-like cell line, MEDMC-BRC6. The cell line expressed FcεRI and c-Kit and showed degranulation and production of inflammatory cytokines and chemokines, including TNF-α, IL-6 and MCP-1, upon cross-linking FcεRI with IgE. These cytokines and chemokines were also produced by the cell line by stimulation of TLR2 and TLR4. MEDMC-BRC6 survived in the peritoneal cavity and the ear skin for at least 6 months after the transfer into genetically compatible MC-deficient KitW-sh/W-sh mice, in which systemic anaphylaxis was successfully induced. Thus, MEDMC-BRC6 cells represent a potent tool for investigating the functions of MCs in vitro and in vivo.
Collapse
Affiliation(s)
- Shohei Shibagaki
- Department of Immunology, Faculty of Medicine.,Doctoral Program in Graduate School of Comprehensive Human Sciences
| | - Satoko Tahara-Hanaoka
- Department of Immunology, Faculty of Medicine.,Life Science Center of Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, 1-1-1, Ten-nodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Takashi Hiroyama
- Cell Engineering Division, RIKEN BioResource Center, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Center, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | - Akira Shibuya
- Department of Immunology, Faculty of Medicine.,Life Science Center of Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, 1-1-1, Ten-nodai, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
25
|
Caminal M, Labrozzi JP, Oliver-Vila I, Alzaga-Gragera M, Marín-Gallén S, Pla A, García J, Vives J. Ex vivo production of red blood cells from human cord blood. BMC Proc 2015. [PMCID: PMC4685347 DOI: 10.1186/1753-6561-9-s9-p67] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
|
26
|
Kanemaru K, Noguchi E, Tokunaga T, Nagai K, Hiroyama T, Nakamura Y, Tahara-Hanaoka S, Shibuya A. Tie2 Signaling Enhances Mast Cell Progenitor Adhesion to Vascular Cell Adhesion Molecule-1 (VCAM-1) through α4β1 Integrin. PLoS One 2015; 10:e0144436. [PMID: 26659448 PMCID: PMC4687632 DOI: 10.1371/journal.pone.0144436] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 11/18/2015] [Indexed: 12/21/2022] Open
Abstract
Mast cell (MC) activation contributes considerably to immune responses, such as host protection and allergy. Cell surface immunoreceptors expressed on MCs play an important role in MC activation. Although various immunoreceptors on MCs have been identified, the regulatory mechanism of MC activation is not fully understood. To understand the regulatory mechanisms of MC activation, we used gene expression analyses of human and mouse MCs to identify a novel immunoreceptor expressed on MCs. We found that Tek, which encodes Tie2, was preferentially expressed in the MCs of both humans and mice. However, Tie2 was not detected on the cell surface of the mouse MCs of the peritoneal cavity, ear skin, or colon lamina propria. In contrast, it was expressed on mouse bone marrow–derived MCs and bone marrow MC progenitors (BM-MCps). Stimulation of Tie2 by its ligand angiopoietin-1 induced tyrosine phosphorylation of Tie2 in MEDMC-BRC6, a mouse embryonic stem cell-derived mast cell line, and enhanced MEDMC-BRC6 and mouse BM-MCp adhesion to vascular cell adhesion molecule-1 (VCAM-1) through α4β1 integrin. These results suggest that Tie2 signaling induces α4β1 integrin activation on BM-MCps for adhesion to VCAM-1.
Collapse
Affiliation(s)
- Kazumasa Kanemaru
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Emiko Noguchi
- Department of Medical Genetics, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Takahiro Tokunaga
- Department of Medical Genetics, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Otorhinolaryngology Head and Neck Surgery, University of Fukui, Fukui, Japan
| | - Kei Nagai
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Takashi Hiroyama
- Cell Engineering Division, RIKEN BioResource Center, Kounodai, Tsukuba, Ibaraki, Japan
| | - Yukio Nakamura
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Cell Engineering Division, RIKEN BioResource Center, Kounodai, Tsukuba, Ibaraki, Japan
| | - Satoko Tahara-Hanaoka
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Life Science Center of Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
- Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Akira Shibuya
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Life Science Center of Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
- Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
- * E-mail:
| |
Collapse
|
27
|
Fauzi I, Panoskaltsis N, Mantalaris A. In Vitro Differentiation of Embryonic Stem Cells into Hematopoietic Lineage: Towards Erythroid Progenitor's Production. Methods Mol Biol 2015; 1341:217-34. [PMID: 26160454 DOI: 10.1007/7651_2015_218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Embryonic stem cells (ESCs) differentiation via embryoid body (EB) formation is an established method that generates the three germ layers. However, EB differentiation poses several problems including formation of heterogeneous cell populations. Herein, we described a differentiation protocol on enhancing mesoderm derivation from murine ESCs (mESCs) using conditioned medium (CM) from HepG2 cells. We used this technique to direct hematopoiesis by generating "embryoid-like" colonies (ELCs) from murine (m) ESCs without following standard formation of EBs. Our CM-mESCs group yielded an almost fivefold increase in ELC formation (p ≤ 0.05) and higher expression of mesoderm genes;-Brachyury-T, Goosecoid, and Flk-1 compared with control mESCs group. Hematopoietic colony formation from CM-mESCs was also enhanced by twofold at days 7 and 14 with earlier colony commitment compared to control mESCs (p ≤ 0.05). This early clonogenic capacity was confirmed morphologically by the presence of nucleated erythrocytes and macrophages as early as day 7 in culture using standard 14-day colony-forming assay. Early expression of hematopoietic primitive (ζ-globin) and definitive (β-globin) erythroid genes and proteins was also observed by day 7 in the CM-treated culture. These data indicate that hematopoietic cells more quickly differentiate from CM-treated, compared with those using standard EB approaches, and provide an efficient bioprocess platform for erythroid-specific differentiation of ESCs.
Collapse
Affiliation(s)
- Iliana Fauzi
- Biological Systems Engineering Laboratory, Department of Chemical Engineering and Chemical Technology, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Nicki Panoskaltsis
- Biological Systems Engineering Laboratory, Department of Chemical Engineering and Chemical Technology, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK.,Department of Hematology, Imperial College London, Northwick Park & St. Mark's campus, London, HA1 3UJ, UK
| | - Athanasios Mantalaris
- Biological Systems Engineering Laboratory, Department of Chemical Engineering and Chemical Technology, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK.
| |
Collapse
|
28
|
De novo generation of HSCs from somatic and pluripotent stem cell sources. Blood 2015; 125:2641-8. [PMID: 25762177 DOI: 10.1182/blood-2014-10-570234] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 02/13/2015] [Indexed: 01/19/2023] Open
Abstract
Generating human hematopoietic stem cells (HSCs) from autologous tissues, when coupled with genome editing technologies, is a promising approach for cellular transplantation therapy and for in vitro disease modeling, drug discovery, and toxicology studies. Human pluripotent stem cells (hPSCs) represent a potentially inexhaustible supply of autologous tissue; however, to date, directed differentiation from hPSCs has yielded hematopoietic cells that lack robust and sustained multilineage potential. Cellular reprogramming technologies represent an alternative platform for the de novo generation of HSCs via direct conversion from heterologous cell types. In this review, we discuss the latest advancements in HSC generation by directed differentiation from hPSCs or direct conversion from somatic cells, and highlight their applications in research and prospects for therapy.
Collapse
|
29
|
Varricchio L, Migliaccio AR. The role of glucocorticoid receptor (GR) polymorphisms in human erythropoiesis. AMERICAN JOURNAL OF BLOOD RESEARCH 2014; 4:53-72. [PMID: 25755906 PMCID: PMC4348794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 11/21/2014] [Indexed: 06/04/2023]
Abstract
Glucocorticoids are endogenous steroid hormones that regulate several biological functions including proliferation, differentiation and apoptosis in numerous cell types in response to stress. Synthetic glucocorticoids, such as dexamethasone (Dex) are used to treat a variety of diseases ranging from allergy to depression. Glucocorticoids exert their effects by passively entering into cells and binding to a specific Glucocorticoid Receptor (GR) present in the cytoplasm. Once activated by its ligand, GR may elicit cytoplasmic (mainly suppression of p53), and nuclear (regulation of transcription of GR responsive genes), responses. Human GR is highly polymorphic and may encode > 260 different isoforms. This polymorphism is emerging as the leading cause for the variability of phenotype and response to glucocorticoid therapy observed in human populations. Studies in mice and clinical observations indicate that GR controls also the response to erythroid stress. This knowledge has been exploited in-vivo by using synthetic GR agonists for treatment of the erythropoietin-refractory congenic Diamond Blackfan Anemia and in-vitro to develop culture conditions that may theoretically generate red cells in numbers sufficient for transfusion. However, the effect exerted by GR polymorphism on the variability of the phenotype of genetic and acquired erythroid disorders observed in the human population is still poorly appreciated. This review will summarize current knowledge on the biological activity of GR and of its polymorphism in non-hematopoietic diseases and discuss the implications of these observations for erythropoiesis.
Collapse
Affiliation(s)
- Lilian Varricchio
- Tisch Cancer Institute, Mount Sinai School of MedicineNew York, NY 10029, USA
| | - Anna Rita Migliaccio
- Tisch Cancer Institute, Mount Sinai School of MedicineNew York, NY 10029, USA
- Istituto Superiore di Sanita’ Viale Regina Elena 299Italy
| |
Collapse
|
30
|
Kang SJ, Park YI, So B, Kang HG. Sodium butyrate efficiently converts fully reprogrammed induced pluripotent stem cells from mouse partially reprogrammed cells. Cell Reprogram 2014; 16:345-54. [PMID: 25093667 DOI: 10.1089/cell.2013.0087] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Partially reprogrammed cells [preinduced pluripotent stem cells (pre-iPSCs)] commonly stall at epigenetic barriers, and this is one of the major failures in the reprogramming process. These cells can be converted to the fully reprogrammed state by reducing epigenetic blocks. In this study, we established three iPSC lines and two pre-iPSC lines induced by the doxycycline (dox)-inducible lentiviral system. In the pre-iPSC lines maintained under dox treatment (dox(+)), a small portion of embryonic stem cell (ESC)-like colonies spontaneously emerged after dox withdrawal (dox(-)), and major differentiation into fibroblast-like cells occurred. The spontaneous conversions based on the number of stage-specific embryonic antigen-1-positive (SSEA-1(+)) colonies were 0.006 ± 0.004% [mean ± standard deviation (SD)] for the #89-7D line and 0.016 ± 0.004% for the #102-2D line. The SSEA-1(+) colonies did not express the Nanog protein. However, the colonies showed characteristics typical of fully reprogrammed iPSCs after further expansion. To determine whether spontaneous conversion could be improved by epigenetic modification, we applied four small molecules-valproic acid (VPA), sodium butyrate (SB), trichostatin (TSA), and 5-aza-2'-deoxycytidine (5-Aza)-in both pre-iPSC lines. SB was the most effective molecule in enhancing the number of SSEA-1(+) colonies (32- to 39-fold) at day 5 of dox(-) treatment. In addition, the expression of pluripotent genes (sox2 and nanog) was increased by SB. After exposure to SB, we found that the expression of four reprogramming factors and cell cycle-related genes was relatively increased compared to their expression following dox(+) of pre-iPSCs. These changes caused by SB might play an important role in the spontaneous conversion of partially reprogrammed cells.
Collapse
Affiliation(s)
- Seok-Jin Kang
- Veterinary Drugs & Biologics , Animal and Plant Quarantine Agency, 430-824, Anyang, Republic of Korea
| | | | | | | |
Collapse
|
31
|
Fauzi I, Panoskaltsis N, Mantalaris A. Early exposure of murine embryonic stem cells to hematopoietic cytokines differentially directs definitive erythropoiesis and cardiomyogenesis in alginate hydrogel three-dimensional cultures. Stem Cells Dev 2014; 23:2720-9. [PMID: 24926614 DOI: 10.1089/scd.2014.0105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
HepG2-conditioned medium (CM) facilitates early differentiation of murine embryonic stem cells (mESCs) into hematopoietic cells in two-dimensional cultures through formation of embryoid-like colonies (ELCs), bypassing embryoid body (EB) formation. We now demonstrate that three-dimensional (3D) cultures of alginate-encapsulated mESCs cultured in a rotating wall vessel bioreactor can be differentially driven toward definitive erythropoiesis and cardiomyogenesis in the absence of ELC formation. Three groups were evaluated: mESCs in maintenance medium with leukemia inhibitory factor (LIF, control) and mESCs cultured with HepG2 CM (CM1 and CM2). Control and CM1 groups were cultivated for 8 days in early differentiation medium with murine stem cell factor (mSCF) followed by 10 days in hematopoietic differentiation medium (HDM) containing human erythropoietin, m-interleukin (mIL)-3, and mSCF. CM2 cells were cultured for 18 days in HDM, bypassing early differentiation. In CM1, a fivefold expansion of hematopoietic colonies was observed at day 14, with enhancement of erythroid progenitors, hematopoietic genes (Gata-2 and SCL), erythroid genes (EKLF and β-major globin), and proteins (Gata-1 and β-globin), although ζ-globin was not expressed. In contrast, CM2 primarily produced beating colonies in standard hematopoietic colony assay and expressed early cardiomyogenic markers, anti-sarcomeric α-actinin and Gata-4. In conclusion, a scalable, automatable, integrated, 3D bioprocess for the differentiation of mESC toward definitive erythroblasts has been established. Interestingly, cardiomyogenesis was also directed in a specific protocol with HepG2 CM and hematopoietic cytokines making this platform a useful tool for the study of erythroid and cardiomyogenic development.
Collapse
Affiliation(s)
- Iliana Fauzi
- 1 Biological Systems Engineering Laboratory , Department of Chemical Engineering and Chemical Technology, Imperial College London, London, United Kingdom
| | | | | |
Collapse
|
32
|
Singh VK, Saini A, Tsuji K, Sharma PB, Chandra R. Manufacturing blood ex vivo: a futuristic approach to deal with the supply and safety concerns. Front Cell Dev Biol 2014; 2:26. [PMID: 25364733 PMCID: PMC4206981 DOI: 10.3389/fcell.2014.00026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 05/26/2014] [Indexed: 12/13/2022] Open
Abstract
Blood transfusions are routinely done in every medical regimen and a worldwide established collection, processing/storage centers provide their services for the same. There have been extreme global demands for both raising the current collections and supply of safe/adequate blood due to increasingly demanding population. With, various risks remain associated with the donor derived blood, and a number of post collection blood screening and processing methods put extreme constraints on supply system especially in the underdeveloped countries. A logistic approach to manufacture erythrocytes ex-vivo by using modern tissue culture techniques have surfaced in the past few years. There are several reports showing the possibilities of RBCs (and even platelets/neutrophils) expansion under tightly regulated conditions. In fact, ex vivo synthesis of the few units of clinical grade RBCs from a single dose of starting material such as umbilical cord blood (CB) has been well established. Similarly, many different sources are also being explored for the same purpose, such as embryonic stem cells, induced pluripotent stem cells. However, the major concerns remain elusive before the manufacture and clinical use of different blood components may be used to successfully replace the present system of donor derived blood transfusion. The most important factor shall include the large scale of RBCs production from each donated unit within a limited time period and cost of their production, both of these issues need to be handled carefully since many of the recipients among developing countries are unable to pay even for the freely available donor derived blood. Anyways, keeping these issues in mind, present article shall be focused on the possibilities of blood production and their use in the near future.
Collapse
Affiliation(s)
- Vimal K Singh
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Abhishek Saini
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Kohichiro Tsuji
- Departments of Pediatric Hematology/Oncology, Research Hospital, The Institute of Medical Science, The University of Tokyo Hospital Tokyo, Japan
| | - P B Sharma
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Ramesh Chandra
- Dr B. R. Ambedkar Center for Biomedical Research, University of Delhi Delhi, India
| |
Collapse
|
33
|
|
34
|
Shah S, Huang X, Cheng L. Concise review: stem cell-based approaches to red blood cell production for transfusion. Stem Cells Transl Med 2013; 3:346-55. [PMID: 24361925 DOI: 10.5966/sctm.2013-0054] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Blood transfusion is a common procedure in modern medicine, and it is practiced throughout the world; however, many countries report a less than sufficient blood supply. Even in developed countries where the supply is currently adequate, projected demographics predict an insufficient supply as early as 2050. The blood supply is also strained during occasional widespread disasters and crises. Transfusion of blood components such as red blood cells (RBCs), platelets, or neutrophils is increasingly used from the same blood unit for multiple purposes and to reduce alloimmune responses. Even for RBCs and platelets lacking nuclei and many antigenic cell-surface molecules, alloimmunity could occur, especially in patients with chronic transfusion requirements. Once alloimmunization occurs, such patients require RBCs from donors with a different blood group antigen combination, making it a challenge to find donors after every successive episode of alloimmunization. Alternative blood substitutes such as synthetic oxygen carriers have so far proven unsuccessful. In this review, we focus on current research and technologies that permit RBC production ex vivo from hematopoietic stem cells, pluripotent stem cells, and immortalized erythroid precursors.
Collapse
Affiliation(s)
- Siddharth Shah
- Division of Hematology, Department of Medicine, and Stem Cell Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | |
Collapse
|
35
|
Rousseau GF, Giarratana MC, Douay L. Large-scale production of red blood cells from stem cells: what are the technical challenges ahead? Biotechnol J 2013; 9:28-38. [PMID: 24408610 DOI: 10.1002/biot.201200368] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 08/05/2013] [Accepted: 09/12/2013] [Indexed: 12/11/2022]
Abstract
Blood-transfusion centers regularly face the challenge of donor blood shortages, especially for rare blood groups. The possibility of producing universal red blood cells from stem cells industrially has become a possible alternative since the successful injection of blood generated in vitro into a human being in 2011. Although there remains many biological and regulatory issues concerning the efficacy and safety of this new product, the major challenge today for future clinical applications is switching from the current limited 2-dimensional production techniques to large-scale 3-dimensional bioreactors. In addition to requiring technological breakthroughs, the whole process also has to become at least five-fold more cost-efficient to match the current prices of high-quality blood products. The current review sums up the main biological advances of the past decade, outlines the key biotechnological challenges for the large-scale cost-effective production of red blood cells, proposes solutions based on strategies used in the bioindustry and presents the state-of-the-art of large-scale blood production.
Collapse
Affiliation(s)
- Guillaume F Rousseau
- UPMC University Paris 6, UMR_S938, Proliferation and Differentiation of Stem Cells, Paris, France; INSERM, UMR_S938, Proliferation and Differentiation of Stem Cells, Paris, France; Université Paris Diderot, Paris, France
| | | | | |
Collapse
|
36
|
Huang X, Shah S, Wang J, Ye Z, Dowey SN, Tsang KM, Mendelsohn LG, Kato GJ, Kickler TS, Cheng L. Extensive ex vivo expansion of functional human erythroid precursors established from umbilical cord blood cells by defined factors. Mol Ther 2013; 22:451-463. [PMID: 24002691 DOI: 10.1038/mt.2013.201] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 08/21/2013] [Indexed: 12/15/2022] Open
Abstract
There is a constant shortage of red blood cells (RBCs) from sufficiently matched donors for patients who need chronic transfusion. Ex vivo expansion and maturation of human erythroid precursors (erythroblasts) from the patients or optimally matched donors could represent a potential solution. Proliferating erythroblasts can be expanded from umbilical cord blood mononuclear cells (CB MNCs) ex vivo for 10(6)-10(7)-fold (in ~50 days) before proliferation arrest and reaching sufficient number for broad application. Here, we report that ectopic expression of three genetic factors (Sox2, c-Myc, and an shRNA against TP53 gene) associated with iPSC derivation enables CB-derived erythroblasts to undergo extended expansion (~10(68)-fold in ~12 months) in a serum-free culture condition without change of cell identity or function. These expanding erythroblasts maintain immature erythroblast phenotypes and morphology, a normal diploid karyotype and dependence on a specific combination of growth factors for proliferation throughout expansion period. When being switched to a terminal differentiation condition, these immortalized erythroblasts gradually exit cell cycle, decrease cell size, accumulate hemoglobin, condense nuclei and eventually give rise to enucleated hemoglobin-containing erythrocytes that can bind and release oxygen. Our result may ultimately lead to an alternative approach to generate unlimited numbers of RBCs for personalized transfusion medicine.
Collapse
Affiliation(s)
- Xiaosong Huang
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Stem Cell Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Siddharth Shah
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Stem Cell Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jing Wang
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Stem Cell Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zhaohui Ye
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Stem Cell Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sarah N Dowey
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Stem Cell Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kit Man Tsang
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Stem Cell Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Laurel G Mendelsohn
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Gregory J Kato
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Thomas S Kickler
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Linzhao Cheng
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Stem Cell Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
37
|
Li X, Wu Z, Fu X, Han W. How Far Are Stem-Cell-Derived Erythrocytes from the Clinical Arena? Bioscience 2013. [DOI: 10.1525/bio.2013.63.8.6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
38
|
Migliaccio AR. Stem cell-derived erythrocytes as upcoming players in blood transfusion. ISBT SCIENCE SERIES 2013; 8:165-171. [PMID: 26229549 PMCID: PMC4517842 DOI: 10.1111/voxs.12048] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Blood transfusion is current standard-of-care for genetic forms of anemia that would be otherwise lethal and allows implementation of aggressive cytotoxic/surgical therapies developed for numerous types of cancer. In developed countries the blood supply is adequate and sporadically even in excess. However, difficulties exist in finding blood with rare phenotypes to treat alloimmunized patients and the progressive ageing of the human population predicts that blood will become scarce by 2050. These considerations establish the need for the development of techniques to generate cultured red blood cell (cRBCs) as transfusion products. MATERIALS AND METHODS Recent progress in cell culture techniques is revolutionizing organ replacement therapies. Two new disciplines, cell therapy and tissue engineering, have been developed to generate in vitro therapeutic products for a variety of applications ranging from skin grafts to organ-function repairs. It is currently believed that these advances will eventually allow ex-vivo production of various cell types in numbers so great that, in the case of red cells, would be clinically adequate for transfusion. RESULTS Proof-of-principle in animal models indicate that cRBCs generated from murine embryonic stem cells protect mice from lethal anemia. Conditions to generate small amounts of clinical grade cRBCs have been established and the first-in-man administration of autologous cRBCs perfomed. The results of this trial indicate that cRBCs survive in vivo at least as long as their natural counterpart. DISCUSSION These ground-breaking reports have raised great excitement for clinical evaluation of cRBCs for transfusion. However, skepticism still persist that production of cRBCs in numbers sufficient for transfusion will ever be possible. This paper will discuss diagnostic and clinical goals pursuable with numbers of cRBCs that may be generated with current technology. CONCLUSION We are confident that development of relevant clinical goals achievable with current technologies will not only improve clinical care in transfusion medicine but will also foster studies to overcome scientific and technical barriers that render transfusion with cRBCs of the general population impractical today.
Collapse
Affiliation(s)
- A R Migliaccio
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore Sanità, Rome, Italy ; Tisch Cancer Institute, Mount Sinai School of Medicine, New York, NY, USA
| |
Collapse
|
39
|
Gao L, Thilakavathy K, Nordin N. A plethora of human pluripotent stem cells. Cell Biol Int 2013; 37:875-87. [DOI: 10.1002/cbin.10120] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 04/15/2013] [Indexed: 12/28/2022]
Affiliation(s)
- Liyang Gao
- Clinical Genetics Unit; Department of Obstetrics & Gynaecology; Faculty of Medicine & Health Sciences; Universiti Putra Malaysia; 43400; UPM Serdang; Selangor; Malaysia
| | | | | |
Collapse
|
40
|
Kurita R, Suda N, Sudo K, Miharada K, Hiroyama T, Miyoshi H, Tani K, Nakamura Y. Establishment of immortalized human erythroid progenitor cell lines able to produce enucleated red blood cells. PLoS One 2013; 8:e59890. [PMID: 23533656 PMCID: PMC3606290 DOI: 10.1371/journal.pone.0059890] [Citation(s) in RCA: 304] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 02/19/2013] [Indexed: 12/27/2022] Open
Abstract
Transfusion of red blood cells (RBCs) is a standard and indispensable therapy in current clinical practice. In vitro production of RBCs offers a potential means to overcome a shortage of transfusable RBCs in some clinical situations and also to provide a source of cells free from possible infection or contamination by microorganisms. Thus, in vitro production of RBCs may become a standard procedure in the future. We previously reported the successful establishment of immortalized mouse erythroid progenitor cell lines that were able to produce mature RBCs very efficiently. Here, we have developed a reliable protocol for establishing immortalized human erythroid progenitor cell lines that are able to produce enucleated RBCs. These immortalized cell lines produce functional hemoglobin and express erythroid-specific markers, and these markers are upregulated following induction of differentiation in vitro. Most importantly, these immortalized cell lines all produce enucleated RBCs after induction of differentiation in vitro, although the efficiency of producing enucleated RBCs remains to be improved further. To the best of our knowledge, this is the first demonstration of the feasibility of using immortalized human erythroid progenitor cell lines as an ex vivo source for production of enucleated RBCs.
Collapse
Affiliation(s)
- Ryo Kurita
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
| | - Noriko Suda
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
| | - Kazuhiro Sudo
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
| | - Kenichi Miharada
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
| | - Takashi Hiroyama
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
| | - Hiroyuki Miyoshi
- Subteam for Manipulation of Cell Fate, RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
| | - Kenzaburo Tani
- Department of Molecular Genetics, Division of Molecular and Clinical Genetics, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Fukuoka, Japan
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
- * E-mail:
| |
Collapse
|
41
|
Zeuner A, Martelli F, Vaglio S, Federici G, Whitsett C, Migliaccio AR. Concise review: stem cell-derived erythrocytes as upcoming players in blood transfusion. Stem Cells 2013; 30:1587-96. [PMID: 22644674 DOI: 10.1002/stem.1136] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Blood transfusions have become indispensable to treat the anemia associated with a variety of medical conditions ranging from genetic disorders and cancer to extensive surgical procedures. In developed countries, the blood supply is generally adequate. However, the projected decline in blood donor availability due to population ageing and the difficulty in finding rare blood types for alloimmunized patients indicate a need for alternative red blood cell (RBC) transfusion products. Increasing knowledge of processes that govern erythropoiesis has been translated into efficient procedures to produce RBC ex vivo using primary hematopoietic stem cells, embryonic stem cells, or induced pluripotent stem cells. Although in vitro-generated RBCs have recently entered clinical evaluation, several issues related to ex vivo RBC production are still under intense scrutiny: among those are the identification of stem cell sources more suitable for ex vivo RBC generation, the translation of RBC culture methods into clinical grade production processes, and the development of protocols to achieve maximal RBC quality, quantity, and maturation. Data on size, hemoglobin, and blood group antigen expression and phosphoproteomic profiling obtained on erythroid cells expanded ex vivo from a limited number of donors are presented as examples of the type of measurements that should be performed as part of the quality control to assess the suitability of these cells for transfusion. New technologies for ex vivo erythroid cell generation will hopefully provide alternative transfusion products to meet present and future clinical requirements.
Collapse
Affiliation(s)
- Ann Zeuner
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | | | | |
Collapse
|
42
|
Tashiro K, Omori M, Kawabata K, Hirata N, Yamaguchi T, Sakurai F, Takaki S, Mizuguchi H. Inhibition of Lnk in Mouse Induced Pluripotent Stem Cells Promotes Hematopoietic Cell Generation. Stem Cells Dev 2012; 21:3381-90. [DOI: 10.1089/scd.2012.0100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Katsuhisa Tashiro
- Laboratory of Stem Cell Regulation, National Institute of Biomedical Innovation, Osaka, Japan
| | - Miyuki Omori
- Laboratory of Stem Cell Regulation, National Institute of Biomedical Innovation, Osaka, Japan
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Kenji Kawabata
- Laboratory of Stem Cell Regulation, National Institute of Biomedical Innovation, Osaka, Japan
- Laboratory of Biomedical Innovation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Nobue Hirata
- Laboratory of Stem Cell Regulation, National Institute of Biomedical Innovation, Osaka, Japan
| | - Tomoko Yamaguchi
- Laboratory of Stem Cell Regulation, National Institute of Biomedical Innovation, Osaka, Japan
| | - Fuminori Sakurai
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Satoshi Takaki
- Department of Immune Regulation, National Center for Global Health and Medicine, Research Institute, Tokyo, Japan
| | - Hiroyuki Mizuguchi
- Laboratory of Stem Cell Regulation, National Institute of Biomedical Innovation, Osaka, Japan
- Laboratory of Biochemistry and Molecular Biology, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- The Center for Advanced Medical Engineering and Informatics, Osaka University, Osaka, Japan
| |
Collapse
|
43
|
Bouhassira EE. Concise review: production of cultured red blood cells from stem cells. Stem Cells Transl Med 2012; 1:927-33. [PMID: 23283554 PMCID: PMC3659674 DOI: 10.5966/sctm.2012-0097] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 10/04/2012] [Indexed: 01/11/2023] Open
Abstract
In the Western world, the volunteer-based collection system covers most transfusion needs, but transient shortages regularly develop and blood supplies are vulnerable to potentially major disruptions. The production of cultured red blood cells from stem cells is slowly emerging as a potential alternative. The various cell sources, the niche applications most likely to reach the clinic first, and some of the remaining technical issues are reviewed here.
Collapse
Affiliation(s)
- Eric E Bouhassira
- Departments of Cell Biology and Medicine, Albert Einstein College of Medicine, New York, NY, USA.
| |
Collapse
|
44
|
Fauzi I, Panoskaltsis N, Mantalaris A. Enhanced hematopoietic differentiation toward erythrocytes from murine embryonic stem cells with HepG2-conditioned medium. Stem Cells Dev 2012; 21:3152-61. [PMID: 22587789 DOI: 10.1089/scd.2012.0030] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Embryonic stem cell (ESC) differentiation via embryoid body (EB) formation is an established method that generates the 3 germ layers. However, EB differentiation poses several problems including formation of heterogeneous cell populations. Previously, we have enhanced mesoderm derivation from murine ESCs (mESCs) using conditioned medium (CM) from HepG2 cells. We used this technique to direct hematopoiesis by generating "embryoid-like" colonies (ELCs) from mESCs without standard formation of EBs. Two predifferentiation conditions were tested: (1) mESCs cultured 3 days in standard predifferentiation medium (control) and (2) mESCs cultured 3 days in HepG2 CM (CM-mESCs). Both groups were then exposed to primary differentiation for 8 days (ELC-formation period) and 14 days of hematopoietic differentiation. Enhanced mesoderm formation was observed in the CM-mESC group with an almost 5-fold increase in ELC formation (P ≤ 0.05) and higher expression of mesoderm genes-Brachyury-T, Goosecoid, and Flk-1-compared with those of control mESCs. Hematopoietic colony formation by CM-mESCs was also enhanced by 2-fold at days 7 and 14 with earlier colony commitment compared with those of control mESCs (P ≤ 0.05). This early clonogenic capacity was confirmed morphologically by the presence of nucleated erythrocytes and macrophages as early as day 7 in CM-mESC culture using standard 14-day colony-forming assay. Early expression of hematopoietic primitive (ζ-globin) and definitive (β-globin) erythroid genes and proteins was also observed by day 7 in CM-mESC cultures. These data indicate that hematopoietic cells more quickly differentiate from CM-mESCs, compared with those using standard EB approaches, and provide an efficient bioprocess platform for erythroid-specific differentiation of ESCs.
Collapse
Affiliation(s)
- Iliana Fauzi
- Biological Systems Engineering Laboratory, Department of Chemical Engineering and Chemical Technology, Imperial College London, London, United Kingdom
| | | | | |
Collapse
|
45
|
Migliaccio AR, Whitsett C, Papayannopoulou T, Sadelain M. The potential of stem cells as an in vitro source of red blood cells for transfusion. Cell Stem Cell 2012; 10:115-9. [PMID: 22305561 PMCID: PMC3676433 DOI: 10.1016/j.stem.2012.01.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Recent advances have increased excitement about the potential for therapeutic production of red blood cells (RBCs) in vitro. However, generation of RBCs in the large numbers required for transfusion remains a significant challenge. In this article, we summarize recent progress in producing RBCs from various cell sources, and discuss the hurdles that remain for translation into the clinical arena.
Collapse
|
46
|
Migliaccio AR, Masselli E, Varricchio L, Whitsett C. Ex-vivo expansion of red blood cells: how real for transfusion in humans? Blood Rev 2011; 26:81-95. [PMID: 22177597 DOI: 10.1016/j.blre.2011.11.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Blood transfusion is indispensable for modern medicine. In developed countries, the blood supply is adequate and safe but blood for alloimmunized patients is often unavailable. Concerns are increasing that donations may become inadequate in the future as the population ages prompting a search for alternative transfusion products. Improvements in culture conditions and proof-of-principle studies in animal models have suggested that ex-vivo expanded red cells may represent such a product. Compared to other cell therapies transfusion poses the unique challenge of requiring great cell doses (2.5×10(12) cells vs 10(7) cells). Although production of such cell numbers is theoretically possible, current technologies generate red cells in numbers sufficient only for safety studies. It is conceived that by the time these studies will be completed, technical barriers to mass cell production will have been eliminated making transfusion with ex-vivo generated red cells a reality.
Collapse
Affiliation(s)
- Anna Rita Migliaccio
- The Tisch Cancer Institute and Myeloproliferative Disease Research Consortium (MPD-RC), Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | | | | | |
Collapse
|
47
|
Abstract
In vitro RBC production from stem cells could represent an alternative to classic transfusion products. Until now the clinical feasibility of this concept has not been demonstrated. We addressed the question of the capacity of cultured RBCs (cRBCs) to survive in humans. By using a culture protocol permitting erythroid differentiation from peripheral CD34(+) HSC, we generated a homogeneous population of cRBC functional in terms of their deformability, enzyme content, capacity of their hemoglobin to fix/release oxygen, and expression of blood group antigens. We then demonstrated in the nonobese diabetes/severe combined immunodeficiency mouse that cRBC encountered in vivo the conditions necessary for their complete maturation. These data provided the rationale for injecting into one human a homogeneous sample of 10(10) cRBCs generated under good manufacturing practice conditions and labeled with (51)Cr. The level of these cells in the circulation 26 days after injection was between 41% and 63%, which compares favorably with the reported half-life of 28 ± 2 days for native RBCs. Their survival in vivo testifies globally to their quality and functionality. These data establish the proof of principle for transfusion of in vitro-generated RBCs and path the way toward new developments in transfusion medicine. This study is registered at http://www.clinicaltrials.gov as NCT0929266.
Collapse
|
48
|
Under HEMA conditions, self-replication of human erythroblasts is limited by autophagic death. Blood Cells Mol Dis 2011; 47:182-97. [PMID: 21775174 DOI: 10.1016/j.bcmd.2011.06.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 05/26/2011] [Accepted: 05/26/2011] [Indexed: 11/23/2022]
Abstract
The number of erythroblasts generated ex-vivo under human-erythroid massive-amplification conditions by mononuclear cells from one unit of adult blood (~10(10)) are insufficient for transfusion (~10(12) red cells), emphasizing the need for studies to characterize cellular interactions during culture to increase erythroblast production. To identify the cell populations which generate erythroblasts under human-erythroid-massive-amplification conditions and the factors that limit proliferation, day 10 non-erythroblasts and immature- and mature-erythroblasts were separated by sorting, labelled with carboxyfluorescein-diacetate-succinimidyl-ester and re-cultured either under these conditions (for proliferation, maturation and/or apoptosis/autophagy determinations) or in semisolid media (for progenitor cell determination). Non-erythroblasts contained 54% of the progenitor cells but did not grow under human-erythroid-massive-amplification conditions. Immature-erythroblasts contained 25% of the progenitor cells and generated erythroblasts under human-erythroid-massive-amplification conditions (FI at 48 h=2.57±1.15). Mature-erythroblasts did not generate colonies and died in human-erythroid-massive-amplification conditions. In sequential sorting/re-culture experiments, immature-erythroblasts retained the ability to generate erythroblasts for 6 days and generated 2-5-fold more cells than the corresponding unfractionated population, suggesting that mature-erythroblasts may limit erythroblast expansion. In co-cultures of carboxyfluorescein-diacetate-succinimidyl-ester-labelled-immature-erythroblasts with mature-erythroblasts at increasing ratios, cell numbers did not increase and proliferation, maturation and apoptotic rates were unchanged. However, Acridine Orange staining (a marker for autophagic death) increased from ~3.2% in cultures with immature-erythroblasts alone to 14-22% in cultures of mature-erythroblasts with and without immature-erythroblasts. In conclusion, these data identify immature-erythroblasts as the cells that generate additional erythroblasts in human-erythroid-massive-amplification cultures and autophagy as the leading cause of death limiting the final cellular output of these cultures.
Collapse
|
49
|
Establishment of novel embryonic stem (ES) cell lines from OG2/rtTA blastocysts. J Genet Genomics 2011; 38:289-95. [PMID: 21777853 DOI: 10.1016/j.jgg.2011.05.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2011] [Revised: 05/25/2011] [Accepted: 05/26/2011] [Indexed: 02/08/2023]
Abstract
Embryonic stem (ES) cells derived from the pre-implantation blastocyst-stage embryos have been widely used to investigate the molecular events determining pluripotency and cell lineage differentiation. As the first discovered ES-specific transcription factor, Oct4 has been considered as the core pluripotency factor of ES cells. In the present study, we successfully established seven ES lines from the blastocysts collected from female OG2 (Oct4-GFP transgenic) mice, which have been crossed with male rtTA transgenic mice. The pluripotency of the ES cell lines can be visualized by the expression of Oct4-GFP under fluorescent microscopy and germ-line transmission capability has been further confirmed. More importantly, the presence of rtTA could induce transgene's expression with the help of doxycycline. Therefore, these ES cell lines provide an excellent tool to further discover novel factors affecting pluripotency and to investigate the molecular mechanism of reprogramming in defined transcription factors mediated nuclear reprogramming.
Collapse
|
50
|
Hiroyama T, Miharada K, Kurita R, Nakamura Y. Plasticity of cells and ex vivo production of red blood cells. Stem Cells Int 2011; 2011:195780. [PMID: 21785608 PMCID: PMC3137953 DOI: 10.4061/2011/195780] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2011] [Accepted: 05/13/2011] [Indexed: 11/21/2022] Open
Abstract
The supply of transfusable red blood cells (RBCs) is not sufficient in many countries. If transfusable RBCs could be produced abundantly from certain resources, it would be very useful. Our group has developed a method to produce enucleated RBCs efficiently from hematopoietic stem/progenitor cells present in umbilical cord blood. More recently, it was reported that enucleated RBCs could be abundantly produced from human embryonic stem (ES) cells. The common obstacle for application of these methods is that they require very high cost to produce sufficient number of RBCs that are applicable in the clinic. If erythroid cell lines (immortalized cell lines) able to produce transfusable RBCs ex vivo were established, they would be valuable resources. Our group developed a robust method to obtain immortalized erythroid cell lines able to produce mature RBCs. To the best of our knowledge, this was the first paper to show the feasibility of establishing immortalized erythroid progenitor cell lines able to produce enucleated RBCs ex vivo. This result strongly suggests that immortalized human erythroid progenitor cell lines able to produce mature RBCs ex vivo can also be established.
Collapse
Affiliation(s)
- Takashi Hiroyama
- Cell Engineering Division, RIKEN BioResource Center, Koyadai 3-1-1, Tsukuba, Ibaraki 305-0074, Japan
| | | | | | | |
Collapse
|