1
|
Chukwuemeka CG, Ndubueze CW, Kolawole AV, Joseph JN, Oladipo IH, Ofoezie EF, Annor-Yeboah SA, Bello ARE, Ganiyu SO. In vitro erythropoiesis: the emerging potential of induced pluripotent stem cells (iPSCs). BLOOD SCIENCE 2025; 7:e00215. [PMID: 39726795 PMCID: PMC11671056 DOI: 10.1097/bs9.0000000000000215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Due to global blood shortages and restricted donor blood storage, the focus has switched to the in vitro synthesis of red blood cells (RBCs) from induced pluripotent stem cells (iPSCs) as a potential solution. Many processes are required to synthesize RBCs from iPSCs, including the production of iPSCs from human or animal cells, differentiation of iPSCs into hematopoietic stem cells, culturing, and maturation of the hematopoietic stem cells (HSC) to make functional erythrocytes. Previous investigations on the in vitro production of erythrocytes have shown conflicting results. Some studies have demonstrated substantial yields of functional erythrocytes, whereas others have observed low yields of enucleated cells. Before large-scale in vitro RBC production can be achieved, several challenges which have limited its application in the clinic must be overcome. These issues include optimizing differentiation techniques to manufacture vast amounts of functional RBCs, upscaling the manufacturing process, cost-effectiveness, and assuring the production of RBCs with good manufacturing practices (GMP) before they can be used for therapeutic purposes.
Collapse
Affiliation(s)
| | - Chizaram W. Ndubueze
- Chester Medical School, University of Chester, Exton Park, Chester CH1 4BJ, England
| | - Adeola V. Kolawole
- Chester Medical School, University of Chester, Exton Park, Chester CH1 4BJ, England
| | - Joshua N. Joseph
- College of Science, University of Massey, Tennent Drive, Massey University, Palmerston North 4410, New Zealand
- Resilient Agriculture, AgResearch Limited, Grasslands Research Centre Tennent Drive, Fitzherbert Palmerston North 4410, New Zealand
| | - Ifeoluwa H. Oladipo
- Chester Medical School, University of Chester, Exton Park, Chester CH1 4BJ, England
| | - Ezichi F. Ofoezie
- Chester Medical School, University of Chester, Exton Park, Chester CH1 4BJ, England
| | | | - Abdur-Rahman Eneye Bello
- Chester Medical School, University of Chester, Exton Park, Chester CH1 4BJ, England
- Department of Biochemistry, Confluence University of Science and Technology, Osara, Kogi State, Nigeria
| | - Sodiq O. Ganiyu
- Chester Medical School, University of Chester, Exton Park, Chester CH1 4BJ, England
| |
Collapse
|
2
|
Wang X, Cui T, Yan H, Zhao L, Zang R, Li H, Wang H, Zhang B, Zhou J, Liu Y, Yue W, Xi J, Pei X. Enhancing terminal erythroid differentiation in human embryonic stem cells through TRIB3 overexpression. Heliyon 2024; 10:e37463. [PMID: 39309892 PMCID: PMC11415673 DOI: 10.1016/j.heliyon.2024.e37463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 09/02/2024] [Accepted: 09/04/2024] [Indexed: 09/25/2024] Open
Abstract
Tribbles pseudokinase 3 (TRIB3) expression significantly increases during terminal erythropoiesis in vivo. However, we found that TRIB3 expression remained relatively low during human embryonic stem cell (hESC) erythropoiesis, particularly in the late stage, where it is typically active. TRIB3 was expressed in megakaryocyte-erythrocyte progenitor cells and its low expression was necessary for megakaryocyte differentiation. Thus, we proposed that the high expression during late stage of erythropoiesis could be the clue for promotion of maturation of hESC-derived erythroid cells. To our knowledge, the role of TRIB3 in the late stage of erythropoiesis remains ambiguous. To address this, we generated inducible TRIB3 overexpression hESCs, named TRIB3tet-on OE H9, based on a Tet-On system. Then, we analyzed hemoglobin expression, condensed chromosomes, organelle clearance, and enucleation with or without doxycycline treatment. TRIB3tet-on OE H9 cells generated erythrocytes with a high proportion of orthochromatic erythroblast in flow cytometry, enhanced hemoglobin and related protein expression in Western blot, decreased nuclear area size, promoted enucleation rate, decreased lysosome and mitochondria number, more colocalization of LC3 with LAMP1 (lysosome marker) and TOM20 (mitochondria marker) and up-regulated mitophagy-related protein expression after treatment with 2 μg/mL doxycycline. Our results showed that TRIB3 overexpression during terminal erythropoiesis may promote the maturation of erythroid cells. Therefore, our study delineates the role of TRIB3 in terminal erythropoiesis, and reveals TRIB3 as a key regulator of UPS and downstream mitophagy by ensuring appropriate mitochondrial clearance during the compaction of chromatin.
Collapse
Affiliation(s)
| | | | - Hao Yan
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Lingping Zhao
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Ruge Zang
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Hongyu Li
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Haiyang Wang
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Biao Zhang
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Junnian Zhou
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Yiming Liu
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Wen Yue
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Jiafei Xi
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| | - Xuetao Pei
- Beijing Institute of Radiation Medicine, Beijing, 100850, PR China
| |
Collapse
|
3
|
Wang E, Liu S, Zhang X, Peng Q, Yu H, Gao L, Xie A, Ma D, Zhao G, Cheng L. An Optimized Human Erythroblast Differentiation System Reveals Cholesterol-Dependency of Robust Production of Cultured Red Blood Cells Ex Vivo. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303471. [PMID: 38481061 PMCID: PMC11165465 DOI: 10.1002/advs.202303471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 11/01/2023] [Indexed: 06/12/2024]
Abstract
The generation of cultured red blood cells (cRBCs) ex vivo represents a potentially unlimited source for RBC transfusion and other cell therapies. Human cRBCs can be generated from the terminal differentiation of proliferating erythroblasts derived from hematopoietic stem/progenitor cells or erythroid precursors in peripheral blood mononuclear cells. Efficient differentiation and maturation into cRBCs highly depend on replenishing human plasma, which exhibits variable potency across donors or batches and complicates the consistent cRBC production required for clinical translation. Hence, the role of human plasma in erythroblast terminal maturation is investigated and uncovered that 1) a newly developed cell culture basal medium mimicking the metabolic profile of human plasma enhances cell growth and increases cRBC yield upon erythroblast terminal differentiation and 2) LDL-carried cholesterol, as a substitute for human plasma, is sufficient to support erythroid survival and terminal differentiation ex vivo. Consequently, a chemically-defined optimized medium (COM) is developed, enabling robust generation of cRBCs from erythroblasts of multiple origins, with improved enucleation efficiency and higher reticulocyte yield, without the need for supplementing human plasma or serum. In addition, the results reveal the crucial role of lipid metabolism during human terminal erythropoiesis.
Collapse
Affiliation(s)
- Enyu Wang
- Department of HematologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
- Blood and Cell Therapy InstituteAnhui Provincial Key Laboratory of Blood Research and ApplicationsUniversity of Science and Technology of ChinaHefeiAnhui230027China
- Department of Electronic Engineering and Information ScienceUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Senquan Liu
- Department of HematologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
- Blood and Cell Therapy InstituteAnhui Provincial Key Laboratory of Blood Research and ApplicationsUniversity of Science and Technology of ChinaHefeiAnhui230027China
- School of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Xinye Zhang
- School of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Qingyou Peng
- School of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Huijuan Yu
- School of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Lei Gao
- Blood and Cell Therapy InstituteAnhui Provincial Key Laboratory of Blood Research and ApplicationsUniversity of Science and Technology of ChinaHefeiAnhui230027China
- School of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - An Xie
- Blood and Cell Therapy InstituteAnhui Provincial Key Laboratory of Blood Research and ApplicationsUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Ding Ma
- Department of HematologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
- Blood and Cell Therapy InstituteAnhui Provincial Key Laboratory of Blood Research and ApplicationsUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Gang Zhao
- Blood and Cell Therapy InstituteAnhui Provincial Key Laboratory of Blood Research and ApplicationsUniversity of Science and Technology of ChinaHefeiAnhui230027China
- Department of Electronic Engineering and Information ScienceUniversity of Science and Technology of ChinaHefeiAnhui230027China
| | - Linzhao Cheng
- Department of HematologyThe First Affiliated Hospital of USTCDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230001China
- Blood and Cell Therapy InstituteAnhui Provincial Key Laboratory of Blood Research and ApplicationsUniversity of Science and Technology of ChinaHefeiAnhui230027China
- School of Basic Medical SciencesDivision of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiAnhui230027China
- Division of HematologyJohns Hopkins University School of MedicineBaltimoreMD21205USA
| |
Collapse
|
4
|
Ju H, Sohn Y, Nam Y, Rim YA. Progresses in overcoming the limitations of in vitro erythropoiesis using human induced pluripotent stem cells. Stem Cell Res Ther 2024; 15:142. [PMID: 38750578 PMCID: PMC11094930 DOI: 10.1186/s13287-024-03754-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/04/2024] [Indexed: 05/19/2024] Open
Abstract
Researchers have attempted to generate transfusable oxygen carriers to mitigate RBC supply shortages. In vitro generation of RBCs using stem cells such as hematopoietic stem and progenitor cells (HSPCs), embryonic stem cells (ESCs), and induced pluripotent stem cells (iPSCs) has shown promise. Specifically, the limited supplies of HSPCs and ethical issues with ESCs make iPSCs the most promising candidate for in vitro RBC generation. However, researchers have encountered some major challenges when using iPSCs to produce transfusable RBC products, such as enucleation and RBC maturation. In addition, it has proven difficult to manufacture these products on a large scale. In this review, we provide a brief overview of erythropoiesis and examine endeavors to recapitulate erythropoiesis in vitro using various cell sources. Furthermore, we explore the current obstacles and potential solutions aimed at enabling the large-scale production of transfusable RBCs in vitro.
Collapse
Affiliation(s)
- Hyeonwoo Ju
- Department of Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Yeowon Sohn
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea
| | - Yoojun Nam
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, South Korea.
- YiPSCELL Inc., L2 Omnibus Park, Banpo-dearo 222, Seocho-gu, Seoul, 06591, Republic of Korea.
| | - Yeri Alice Rim
- YiPSCELL Inc., L2 Omnibus Park, Banpo-dearo 222, Seocho-gu, Seoul, 06591, Republic of Korea.
- CiSTEM laboratory, Convergent Research Consortium for Immunologic Disease, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| |
Collapse
|
5
|
Gunawardena N, Chou ST. Generation of red blood cells from induced pluripotent stem cells. Curr Opin Hematol 2024; 31:115-121. [PMID: 38362913 PMCID: PMC10959681 DOI: 10.1097/moh.0000000000000810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
PURPOSE OF REVIEW Human induced pluripotent stem cells (iPSCs) are an attractive source to generate in-vitro-derived blood for use as transfusable and reagent red cells. We review recent advancements in the field and the remaining limitations for clinical use. RECENT FINDINGS For iPSC-derived red blood cell (RBC) generation, recent work has optimized culture conditions to omit feeder cells, enhance red cell maturation, and produce cells that mimic fetal or adult-type RBCs. Genome editing provides novel strategies to improve cell yield and create designer RBCs with customized antigen phenotypes. SUMMARY Current protocols support red cell production that mimics embryonic and fetal hematopoiesis and cell yield sufficient for diagnostic RBC reagents. Ongoing challenges to generate RBCs for transfusion include recapitulating definitive erythropoiesis to produce functional adult-type cells, increasing scalability of culture conditions, and optimizing high-density manufacturing capacity.
Collapse
Affiliation(s)
| | - Stella T Chou
- Division of Hematology, Department of Pediatrics
- Division of Transfusion Medicine, Department of Pathology, Children's Hospital of Philadelphia, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
6
|
Jiang JH, Ren RT, Cheng YJ, Li XX, Zhang GR. Immune cells and RBCs derived from human induced pluripotent stem cells: method, progress, prospective challenges. Front Cell Dev Biol 2024; 11:1327466. [PMID: 38250324 PMCID: PMC10796611 DOI: 10.3389/fcell.2023.1327466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/14/2023] [Indexed: 01/23/2024] Open
Abstract
Blood has an important role in the healthcare system, particularly in blood transfusions and immunotherapy. However, the occurrence of outbreaks of infectious diseases worldwide and seasonal fluctuations, blood shortages are becoming a major challenge. Moreover, the narrow specificity of immune cells hinders the widespread application of immune cell therapy. To address this issue, researchers are actively developing strategies for differentiating induced pluripotent stem cells (iPSCs) into blood cells in vitro. The establishment of iPSCs from terminally differentiated cells such as fibroblasts and blood cells is a straightforward process. However, there is need for further refinement of the protocols for differentiating iPSCs into immune cells and red blood cells to ensure their clinical applicability. This review aims to provide a comprehensive overview of the strategies and challenges facing the generation of iPSC-derived immune cells and red blood cells.
Collapse
Affiliation(s)
- Jin-he Jiang
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
| | - Ru-tong Ren
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
| | - Yan-jie Cheng
- Institute of Biomedical and Health Science, School of Life and Health Science, Anhui Science and Technology University, Chuzhou, Anhui, China
| | - Xin-xin Li
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
| | - Gui-rong Zhang
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
| |
Collapse
|
7
|
Boccacci Y, Dumont N, Doyon Y, Laganière J. Accessory-cell-free differentiation of hematopoietic stem and progenitor cells into mature red blood cells. Cytotherapy 2023; 25:1242-1248. [PMID: 37598334 DOI: 10.1016/j.jcyt.2023.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 07/14/2023] [Accepted: 07/25/2023] [Indexed: 08/21/2023]
Abstract
BACKGROUND AIMS The culture and ex vivo engineering of red blood cells (RBCs) can help characterize genetic variants, model diseases, and may eventually spur the development of applications in transfusion medicine. In the last decade, improvements to the in vitro production of RBCs have enabled efficient erythroid progenitor proliferation and high enucleation levels from several sources of hematopoietic stem and progenitor cells (HSPCs). Despite these advances, there remains a need for refining the terminal step of in vitro human erythropoiesis, i.e., the terminal maturation of reticulocytes into erythrocytes, so that it can occur without feeder or accessory cells and animal-derived components. METHODS Here, we describe the near-complete erythroid differentiation of cultured RBCs (cRBCs) from adult HSPCs in accessory-cell-free and xeno-free conditions. RESULTS The approach improves post-enucleation cell integrity and cell survival, and it enables subsequent storage of cRBCs for up to 42 days in classical additive solution conditions without any specialized equipment. CONCLUSIONS We foresee that these improvements will facilitate the characterization of RBCs derived from gene-edited HSPCs.
Collapse
Affiliation(s)
- Yelena Boccacci
- Medical Affairs and Innovation, Héma-Qubec, Québec, Quebec, Canada; Centre Hospitalier Universitaire de Québec Research Center, Université Laval, Québec, Quebec, Canada
| | - Nellie Dumont
- Medical Affairs and Innovation, Héma-Qubec, Québec, Quebec, Canada
| | - Yannick Doyon
- Centre Hospitalier Universitaire de Québec Research Center, Université Laval, Québec, Quebec, Canada
| | - Josée Laganière
- Medical Affairs and Innovation, Héma-Qubec, Québec, Quebec, Canada.
| |
Collapse
|
8
|
Jeon SB, Koh H, Han AR, Kim J, Lee S, Lee JH, Im SS, Yoon YS, Lee JH, Lee JY. Ferric citrate and apo-transferrin enable erythroblast maturation with β-globin from hemogenic endothelium. NPJ Regen Med 2023; 8:46. [PMID: 37626061 PMCID: PMC10457393 DOI: 10.1038/s41536-023-00320-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Red blood cell (RBC) generation from human pluripotent stem cells (PSCs) offers potential for innovative cell therapy in regenerative medicine as well as developmental studies. Ex vivo erythropoiesis from PSCs is currently limited by the low efficiency of functional RBCs with β-globin expression in culture systems. During induction of β-globin expression, the absence of a physiological microenvironment, such as a bone marrow niche, may impair cell maturation and lineage specification. Here, we describe a simple and reproducible culture system that can be used to generate erythroblasts with β-globin expression. We prepared a two-dimensional defined culture with ferric citrate treatment based on definitive hemogenic endothelium (HE). Floating erythroblasts derived from HE cells were primarily CD45+CD71+CD235a+ cells, and their number increased remarkably upon Fe treatment. Upon maturation, the erythroblasts cultured in the presence of ferric citrate showed high transcriptional levels of β-globin and enrichment of genes associated with heme synthesis and cell cycle regulation, indicating functionality. The rapid maturation of these erythroblasts into RBCs was observed when injected in vivo, suggesting the development of RBCs that were ready to grow. Hence, induction of β-globin expression may be explained by the effects of ferric citrate that promote cell maturation by binding with soluble transferrin and entering the cells.Taken together, upon treatment with Fe, erythroblasts showed advanced maturity with a high transcription of β-globin. These findings can help devise a stable protocol for the generation of clinically applicable RBCs.
Collapse
Affiliation(s)
- Soo-Been Jeon
- CHA Advanced Research Institute, Bundang CHA Medical Center, CHA University, Seongnam, Kyunggi-do, 13488, South Korea
| | - Hyebin Koh
- Futuristic Animal Resource & Research Center (FARRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - A-Reum Han
- CHA Advanced Research Institute, Bundang CHA Medical Center, CHA University, Seongnam, Kyunggi-do, 13488, South Korea
| | - Jieun Kim
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Republic of Korea
| | - Sunghun Lee
- CHA Advanced Research Institute, Bundang CHA Medical Center, CHA University, Seongnam, Kyunggi-do, 13488, South Korea
| | - Jae-Ho Lee
- Department of Physiology, Keimyung University School of Medicine, Daegu, 42601, Korea
| | - Seung-Soon Im
- Department of Physiology, Keimyung University School of Medicine, Daegu, 42601, Korea
| | - Young-Sup Yoon
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
- Department of Medicine, Emory University, Atlanta, USA
| | - Jong-Hee Lee
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea.
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Republic of Korea.
| | - Ji Yoon Lee
- CHA Advanced Research Institute, Bundang CHA Medical Center, CHA University, Seongnam, Kyunggi-do, 13488, South Korea.
- Department of Biomedical Science, CHA University, Seongnam, Kyunggi-do, 13488, South Korea.
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
9
|
Lee SJ, Jung C, Oh JE, Kim S, Lee S, Lee JY, Yoon YS. Generation of Red Blood Cells from Human Pluripotent Stem Cells-An Update. Cells 2023; 12:1554. [PMID: 37296674 PMCID: PMC10253210 DOI: 10.3390/cells12111554] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
Red blood cell (RBC) transfusion is a lifesaving medical procedure that can treat patients with anemia and hemoglobin disorders. However, the shortage of blood supply and risks of transfusion-transmitted infection and immune incompatibility present a challenge for transfusion. The in vitro generation of RBCs or erythrocytes holds great promise for transfusion medicine and novel cell-based therapies. While hematopoietic stem cells and progenitors derived from peripheral blood, cord blood, and bone marrow can give rise to erythrocytes, the use of human pluripotent stem cells (hPSCs) has also provided an important opportunity to obtain erythrocytes. These hPSCs include both human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs). As hESCs carry ethical and political controversies, hiPSCs can be a more universal source for RBC generation. In this review, we first discuss the key concepts and mechanisms of erythropoiesis. Thereafter, we summarize different methodologies to differentiate hPSCs into erythrocytes with an emphasis on the key features of human definitive erythroid lineage cells. Finally, we address the current limitations and future directions of clinical applications using hiPSC-derived erythrocytes.
Collapse
Affiliation(s)
- Shin-Jeong Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.-J.L.); (C.J.); (J.E.O.); (S.K.)
- Research and Development Center, KarisBio Inc., 50-1 Yonsei-Ro, Avison Biomedical Research Center Room 525, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Cholomi Jung
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.-J.L.); (C.J.); (J.E.O.); (S.K.)
- Department of Internal Medicine, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Jee Eun Oh
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.-J.L.); (C.J.); (J.E.O.); (S.K.)
- Research and Development Center, KarisBio Inc., 50-1 Yonsei-Ro, Avison Biomedical Research Center Room 525, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Sangsung Kim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.-J.L.); (C.J.); (J.E.O.); (S.K.)
- Research and Development Center, KarisBio Inc., 50-1 Yonsei-Ro, Avison Biomedical Research Center Room 525, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Sangho Lee
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Ji Yoon Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.-J.L.); (C.J.); (J.E.O.); (S.K.)
| | - Young-sup Yoon
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.-J.L.); (C.J.); (J.E.O.); (S.K.)
- Research and Development Center, KarisBio Inc., 50-1 Yonsei-Ro, Avison Biomedical Research Center Room 525, Seodaemun-gu, Seoul 03722, Republic of Korea
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA;
| |
Collapse
|
10
|
Gallego-Murillo JS, Yağcı N, Pinho EM, Wahl SA, van den Akker E, von Lindern M. Iron-loaded deferiprone can support full hemoglobinization of cultured red blood cells. Sci Rep 2023; 13:6960. [PMID: 37117329 PMCID: PMC10147612 DOI: 10.1038/s41598-023-32706-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 03/31/2023] [Indexed: 04/30/2023] Open
Abstract
Iron, supplemented as iron-loaded transferrin (holotransferrin), is an essential nutrient in mammalian cell cultures, particularly for erythroid cultures. The high cost of human transferrin represents a challenge for large scale production of red blood cells (RBCs) and for cell therapies in general. We evaluated the use of deferiprone, a cell membrane-permeable drug for iron chelation therapy, as an iron carrier for erythroid cultures. Iron-loaded deferiprone (Def3·Fe3+, at 52 µmol/L) could eliminate the need for holotransferrin supplementation during in vitro expansion and differentiation of erythroblast cultures to produce large numbers of enucleated RBC. Only the first stage, when hematopoietic stem cells committed to erythroblasts, required holotransferrin supplementation. RBCs cultured in presence of Def3·Fe3+ or holotransferrin (1000 µg/mL) were similar with respect to differentiation kinetics, expression of cell-surface markers CD235a and CD49d, hemoglobin content, and oxygen association/dissociation. Replacement of holotransferrin supplementation by Def3·Fe3+ was also successful in cultures of myeloid cell lines (MOLM13, NB4, EOL1, K562, HL60, ML2). Thus, iron-loaded deferiprone can partially replace holotransferrin as a supplement in chemically defined cell culture medium. This holds promise for a significant decrease in medium cost and improved economic perspectives of the large scale production of red blood cells for transfusion purposes.
Collapse
Affiliation(s)
- Joan Sebastián Gallego-Murillo
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center (UMC), Amsterdam, The Netherlands
- Department of Biotechnology, Faculty of Applied Sciences, Delft University of Technology, Delft, The Netherlands
- Meatable, Alexander Fleminglaan 1, 2613AX, Delft, The Netherlands
| | - Nurcan Yağcı
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center (UMC), Amsterdam, The Netherlands
| | - Eduardo Machado Pinho
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center (UMC), Amsterdam, The Netherlands
- Department of Bioengineering, Faculty of Engineering, University of Porto, Porto, Portugal
| | - Sebastian Aljoscha Wahl
- Department of Biotechnology, Faculty of Applied Sciences, Delft University of Technology, Delft, The Netherlands
- Lehrstuhl Für Bioverfahrenstechnik, Friedrich-Alexander Universität Erlangen-Nürnberg, Paul-Gordan-Str. 3, 91052, Erlangen, Germany
| | - Emile van den Akker
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center (UMC), Amsterdam, The Netherlands
| | - Marieke von Lindern
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Center (UMC), Amsterdam, The Netherlands.
| |
Collapse
|
11
|
Cho YK, Kim HK, Kwon SS, Jeon SH, Cheong JW, Nam KT, Kim HS, Kim S, Kim HO. In vitro erythrocyte production using human-induced pluripotent stem cells: determining the best hematopoietic stem cell sources. Stem Cell Res Ther 2023; 14:106. [PMID: 37101221 PMCID: PMC10132444 DOI: 10.1186/s13287-023-03305-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 03/28/2023] [Indexed: 04/28/2023] Open
Abstract
BACKGROUND Blood transfusion is an essential part of medicine. However, many countries have been facing a national blood crisis. To address this ongoing blood shortage issue, there have been efforts to generate red blood cells (RBCs) in vitro, especially from human-induced pluripotent stem cells (hiPSCs). However, the best source of hiPSCs for this purpose is yet to be determined. METHODS In this study, hiPSCs were established from three different hematopoietic stem cell sources-peripheral blood (PB), cord blood (CB) and bone marrow (BM) aspirates (n = 3 for each source)-using episomal reprogramming vectors and differentiated into functional RBCs. Various time-course studies including immunofluorescence assay, quantitative real-time PCR, flow cytometry, karyotyping, morphological analysis, oxygen binding capacity analysis, and RNA sequencing were performed to examine and compare the characteristics of hiPSCs and hiPSC-differentiated erythroid cells. RESULTS hiPSC lines were established from each of the three sources and were found to be pluripotent and have comparable characteristics. All hiPSCs differentiated into erythroid cells, but there were discrepancies in differentiation and maturation efficiencies: CB-derived hiPSCs matured into erythroid cells the fastest while PB-derived hiPSCs required a longer time for maturation but showed the highest degree of reproducibility. BM-derived hiPSCs gave rise to diverse types of cells and exhibited poor differentiation efficiency. Nonetheless, erythroid cells differentiated from all hiPSC lines mainly expressed fetal and/or embryonic hemoglobin, indicating that primitive erythropoiesis occurred. Their oxygen equilibrium curves were all left-shifted. CONCLUSIONS Collectively, both PB- and CB-derived hiPSCs were favorably reliable sources for the clinical production of RBCs in vitro, despite several challenges that need to be overcome. However, owing to the limited availability and the large amount of CB required to produce hiPSCs, and the results of this study, the advantages of using PB-derived hiPSCs for RBC production in vitro may outweigh those of using CB-derived hiPSCs. We believe that our findings will facilitate the selection of optimal hiPSC lines for RBC production in vitro in the near future.
Collapse
Affiliation(s)
- Youn Keong Cho
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyun-Kyung Kim
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Soon Sung Kwon
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Su-Hee Jeon
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - June-Won Cheong
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ki Taek Nam
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Han-Soo Kim
- Department of Biomedical Sciences, Catholic Kwandong University College of Medical Convergence, Gangneung-si, Gangwon-do, Republic of Korea
| | - Sinyoung Kim
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | - Hyun Ok Kim
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
12
|
Cvekl A, Camerino MJ. Generation of Lens Progenitor Cells and Lentoid Bodies from Pluripotent Stem Cells: Novel Tools for Human Lens Development and Ocular Disease Etiology. Cells 2022; 11:3516. [PMID: 36359912 PMCID: PMC9658148 DOI: 10.3390/cells11213516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
In vitro differentiation of human pluripotent stem cells (hPSCs) into specialized tissues and organs represents a powerful approach to gain insight into those cellular and molecular mechanisms regulating human development. Although normal embryonic eye development is a complex process, generation of ocular organoids and specific ocular tissues from pluripotent stem cells has provided invaluable insights into the formation of lineage-committed progenitor cell populations, signal transduction pathways, and self-organization principles. This review provides a comprehensive summary of recent advances in generation of adenohypophyseal, olfactory, and lens placodes, lens progenitor cells and three-dimensional (3D) primitive lenses, "lentoid bodies", and "micro-lenses". These cells are produced alone or "community-grown" with other ocular tissues. Lentoid bodies/micro-lenses generated from human patients carrying mutations in crystallin genes demonstrate proof-of-principle that these cells are suitable for mechanistic studies of cataractogenesis. Taken together, current and emerging advanced in vitro differentiation methods pave the road to understand molecular mechanisms of cataract formation caused by the entire spectrum of mutations in DNA-binding regulatory genes, such as PAX6, SOX2, FOXE3, MAF, PITX3, and HSF4, individual crystallins, and other genes such as BFSP1, BFSP2, EPHA2, GJA3, GJA8, LIM2, MIP, and TDRD7 represented in human cataract patients.
Collapse
Affiliation(s)
- Aleš Cvekl
- Departments Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Michael John Camerino
- Departments Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
13
|
An HH, Gagne AL, Maguire JA, Pavani G, Abdulmalik O, Gadue P, French DL, Westhoff CM, Chou ST. The use of pluripotent stem cells to generate diagnostic tools for transfusion medicine. Blood 2022; 140:1723-1734. [PMID: 35977098 PMCID: PMC9707399 DOI: 10.1182/blood.2022015883] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 08/04/2022] [Indexed: 12/14/2022] Open
Abstract
Red blood cell (RBC) transfusion is one of the most common medical treatments, with more than 10 million units transfused per year in the United States alone. Alloimmunization to foreign Rh proteins (RhD and RhCE) on donor RBCs remains a challenge for transfusion effectiveness and safety. Alloantibody production disproportionately affects patients with sickle cell disease who frequently receive blood transfusions and exhibit high genetic diversity in the Rh blood group system. With hundreds of RH variants now known, precise identification of Rh antibody targets is hampered by the lack of appropriate reagent RBCs with uncommon Rh antigen phenotypes. Using a combination of human-induced pluripotent stem cell (iPSC) reprogramming and gene editing, we designed a renewable source of cells with unique Rh profiles to facilitate the identification of complex Rh antibodies. We engineered a very rare Rh null iPSC line lacking both RHD and RHCE. By targeting the AAVS1 safe harbor locus in this Rh null background, any combination of RHD or RHCE complementary DNAs could be reintroduced to generate RBCs that express specific Rh antigens such as RhD alone (designated D--), Goa+, or DAK+. The RBCs derived from these iPSCs (iRBCs) are compatible with standard laboratory assays used worldwide and can determine the precise specificity of Rh antibodies in patient plasma. Rh-engineered iRBCs can provide a readily accessible diagnostic tool and guide future efforts to produce an alternative source of rare RBCs for alloimmunized patients.
Collapse
Affiliation(s)
- Hyun Hyung An
- Division of Hematology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Alyssa L. Gagne
- Department of Pathology and Laboratory Medicine, Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Jean Ann Maguire
- Department of Pathology and Laboratory Medicine, Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Giulia Pavani
- Department of Pathology and Laboratory Medicine, Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Osheiza Abdulmalik
- Division of Hematology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Paul Gadue
- Department of Pathology and Laboratory Medicine, Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Deborah L. French
- Department of Pathology and Laboratory Medicine, Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | | | - Stella T. Chou
- Division of Hematology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Division of Transfusion Medicine, Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA
| |
Collapse
|
14
|
Bernecker C, Matzhold EM, Kolb D, Avdili A, Rohrhofer L, Lampl A, Trötzmüller M, Singer H, Oldenburg J, Schlenke P, Dorn I. Membrane Properties of Human Induced Pluripotent Stem Cell-Derived Cultured Red Blood Cells. Cells 2022; 11:cells11162473. [PMID: 36010549 PMCID: PMC9406338 DOI: 10.3390/cells11162473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 12/16/2022] Open
Abstract
Cultured red blood cells from human induced pluripotent stem cells (cRBC_iPSCs) are a promising source for future concepts in transfusion medicine. Before cRBC_iPSCs will have entrance into clinical or laboratory use, their functional properties and safety have to be carefully validated. Due to the limitations of established culture systems, such studies are still missing. Improved erythropoiesis in a recently established culture system, closer simulating the physiological niche, enabled us to conduct functional characterization of enucleated cRBC_iPSCs with a focus on membrane properties. Morphology and maturation stage of cRBC_iPSCs were closer to native reticulocytes (nRETs) than to native red blood cells (nRBCs). Whereas osmotic resistance of cRBC_iPSCs was similar to nRETs, their deformability was slightly impaired. Since no obvious alterations in membrane morphology, lipid composition, and major membrane associated protein patterns were observed, reduced deformability might be caused by a more primitive nature of cRBC_iPSCs comparable to human embryonic- or fetal liver erythropoiesis. Blood group phenotyping of cRBC_iPSCs further confirmed the potency of cRBC_iPSCs as a prospective device in pre-transfusional routine diagnostics. Therefore, RBC membrane analyses obtained in this study underscore the overall prospects of cRBC_iPSCs for their future application in the field of transfusion medicine.
Collapse
Affiliation(s)
- Claudia Bernecker
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Eva Maria Matzhold
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Dagmar Kolb
- Core Facility Ultrastructure Analysis, Medical University of Graz, 8010 Graz, Austria
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, 8010 Graz, Austria
| | - Afrim Avdili
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Lisa Rohrhofer
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Annika Lampl
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Martin Trötzmüller
- Core Facility Mass Spectrometry, Center for Medical Research, Medical University of Graz, 8010 Graz, Austria
| | - Heike Singer
- Institute of Experimental Haematology and Transfusion Medicine, University Clinic Bonn, 53127 Bonn, Germany
| | - Johannes Oldenburg
- Institute of Experimental Haematology and Transfusion Medicine, University Clinic Bonn, 53127 Bonn, Germany
| | - Peter Schlenke
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, 8036 Graz, Austria
| | - Isabel Dorn
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Graz, 8036 Graz, Austria
- Correspondence:
| |
Collapse
|
15
|
Yu S, Vassilev S, Lim ZR, Sivalingam J, Lam ATL, Ho V, Renia L, Malleret B, Reuveny S, Oh SKW. Selection of O-negative induced pluripotent stem cell clones for high-density red blood cell production in a scalable perfusion bioreactor system. Cell Prolif 2022; 55:e13218. [PMID: 35289971 PMCID: PMC9357363 DOI: 10.1111/cpr.13218] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 01/27/2022] [Accepted: 02/18/2022] [Indexed: 12/31/2022] Open
Abstract
Objectives Large‐scale generation of universal red blood cells (RBCs) from O‐negative (O‐ve) human induced pluripotent stem cells (hiPSCs) holds the potential to alleviate worldwide shortages of blood and provide a safe and secure year‐round supply. Mature RBCs and reticulocytes, the immature counterparts of RBCs generated during erythropoiesis, could also find important applications in research, for example in malaria parasite infection studies. However, one major challenge is the lack of a high‐density culture platform for large‐scale generation of RBCs in vitro. Materials and Methods We generated 10 O‐ve hiPSC clones and evaluated their potential for mesoderm formation and erythroid differentiation. We then used a perfusion bioreactor system to perform studies with high‐density cultures of erythroblasts in vitro. Results Based on their tri‐lineage (and specifically mesoderm) differentiation potential, we isolated six hiPSC clones capable of producing functional erythroblasts. Using the best performing clone, we demonstrated the small‐scale generation of high‐density cultures of erythroblasts in a perfusion bioreactor system. After process optimization, we were able to achieve a peak cell density of 34.7 million cells/ml with 92.2% viability in the stirred bioreactor. The cells expressed high levels of erythroblast markers, showed oxygen carrying capacity, and were able to undergo enucleation. Conclusions This study demonstrated a scalable platform for the production of functional RBCs from hiPSCs. The perfusion culture platform we describe here could pave the way for large volume‐controlled bioreactor culture for the industrial generation of high cell density erythroblasts and RBCs.
Collapse
Affiliation(s)
- SuE Yu
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Republic of Singapore
| | - Svetlan Vassilev
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Republic of Singapore
| | - Zhong Ri Lim
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Republic of Singapore
| | - Jaichandran Sivalingam
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Republic of Singapore
| | - Alan Tin Lun Lam
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Republic of Singapore
| | - Valerie Ho
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Republic of Singapore
| | - Laurent Renia
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Republic of Singapore.,A*STAR Infectious Diseases Labs, Agency for Science, Technology and Research, Singapore, Republic of Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Republic of Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore, Republic of Singapore
| | - Benoit Malleret
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Republic of Singapore.,Department of Microbiology and Immunology, Immunology Translational Research Programme, Yong Loo Lin School of Medicine, Immunology Programme, Life Sciences Institute, National University of Singapore, Singapore, Republic of Singapore
| | - Shaul Reuveny
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Republic of Singapore
| | - Steve Kah Weng Oh
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Republic of Singapore
| |
Collapse
|
16
|
Ferenz K, Karaman O, Shah SB. Artificial red blood cells. NANOTECHNOLOGY FOR HEMATOLOGY, BLOOD TRANSFUSION, AND ARTIFICIAL BLOOD 2022:397-427. [DOI: 10.1016/b978-0-12-823971-1.00018-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
17
|
Mukherjee K, Bieker JJ. Transcriptional Control of Gene Expression and the Heterogeneous Cellular Identity of Erythroblastic Island Macrophages. Front Genet 2021; 12:756028. [PMID: 34880902 PMCID: PMC8646026 DOI: 10.3389/fgene.2021.756028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/04/2021] [Indexed: 11/13/2022] Open
Abstract
During definitive erythropoiesis, maturation of erythroid progenitors into enucleated reticulocytes requires the erythroblastic island (EBI) niche comprising a central macrophage attached to differentiating erythroid progenitors. Normally, the macrophage provides a nurturing environment for maturation of erythroid cells. Its critical physiologic importance entails aiding in recovery from anemic insults, such as systemic stress or acquired disease. Considerable interest in characterizing the central macrophage of the island niche led to the identification of putative cell surface markers enriched in island macrophages, enabling isolation and characterization. Recent studies focus on bulk and single cell transcriptomics of the island macrophage during adult steady-state erythropoiesis and embryonic erythropoiesis. They reveal that the island macrophage is a distinct cell type but with widespread cellular heterogeneity, likely suggesting distinct developmental origins and biological function. These studies have also uncovered transcriptional programs that drive gene expression in the island macrophage. Strikingly, the master erythroid regulator EKLF/Klf1 seems to also play a major role in specifying gene expression in island macrophages, including a putative EKLF/Klf1-dependent transcription circuit. Our present review and analysis of mouse single cell genetic patterns suggest novel expression characteristics that will enable a clear enrichment of EBI subtypes and resolution of island macrophage heterogeneity. Specifically, the discovery of markers such as Epor, and specific features for EKLF/Klf1-expressing island macrophages such as Sptb and Add2, or for SpiC-expressing island macrophage such as Timd4, or for Maf/Nr1h3-expressing island macrophage such as Vcam1, opens exciting possibilities for further characterization of these unique macrophage cell types in the context of their critical developmental function.
Collapse
Affiliation(s)
- Kaustav Mukherjee
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai School of Medicine, New York, NY, United States.,Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY, United States
| | - James J Bieker
- Department of Cell, Developmental, and Regenerative Biology, Mount Sinai School of Medicine, New York, NY, United States.,Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY, United States.,Tisch Cancer Center, Mount Sinai School of Medicine, New York, NY, United States.,Mindich Child Health and Development Institute, Mount Sinai School of Medicine, New York, NY, United States
| |
Collapse
|
18
|
Industrially Compatible Transfusable iPSC-Derived RBCs: Progress, Challenges and Prospective Solutions. Int J Mol Sci 2021; 22:ijms22189808. [PMID: 34575977 PMCID: PMC8472628 DOI: 10.3390/ijms22189808] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 02/06/2023] Open
Abstract
Amidst the global shortfalls in blood supply, storage limitations of donor blood and the availability of potential blood substitutes for transfusion applications, society has pivoted towards in vitro generation of red blood cells (RBCs) as a means to solve these issues. Many conventional research studies over the past few decades have found success in differentiating hematopoietic stem and progenitor cells (HSPCs) from cord blood, adult bone marrow and peripheral blood sources. More recently, techniques that involve immortalization of erythroblast sources have also gained traction in tackling this problem. However, the RBCs generated from human induced pluripotent stem cells (hiPSCs) still remain as the most favorable solution due to many of its added advantages. In this review, we focus on the breakthroughs for high-density cultures of hiPSC-derived RBCs, and highlight the major challenges and prospective solutions throughout the whole process of erythropoiesis for hiPSC-derived RBCs. Furthermore, we elaborate on the recent advances and techniques used to achieve cost-effective, high-density cultures of GMP-compliant RBCs, and on their relevant novel applications after downstream processing and purification.
Collapse
|
19
|
Pellegrin S, Severn CE, Toye AM. Towards manufactured red blood cells for the treatment of inherited anemia. Haematologica 2021; 106:2304-2311. [PMID: 34042406 PMCID: PMC8409035 DOI: 10.3324/haematol.2020.268847] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Indexed: 11/21/2022] Open
Abstract
Patients with inherited anemia and hemoglobinopathies (such as sickle cell disease and β-thalassemia) are treated with red blood cell (RBC) transfusions to alleviate their symptoms. Some of these patients may have rare blood group types or go on to develop alloimmune reactions, which can make it difficult to source compatible blood in the donor population. Laboratory-grown RBC represent a particularly attractive alternative which could satisfy an unmet clinical need. The challenge, however, is to produce - from a limited number of stem cells - the 2x1012 RBC required for a standard adult therapeutic dose. Encouraging progress has been made in RBC production from adult stem cells under good manufacturing practice. In 2011, the Douay group conducted a successful proof-of-principle mini-transfusion of autologous manufactured RBC in a single volunteer. In the UK, a trial is planned to assess whether manufactured RBC are equivalent to RBC produced naturally in donors, by testing an allogeneic mini-dose of laboratory-grown manufactured RBC in multiple volunteers. This review discusses recent progress in the erythroid culture field as well as opportunities for further scaling up of manufactured RBC production for transfusion practice.
Collapse
Affiliation(s)
- Stephanie Pellegrin
- School of Biochemistry, Biomedical Sciences Building; National Institute for Health Research (NIHR) Blood and Transplant Research Unit in Red Blood Cell Products, University of Bristol.
| | - Charlotte E Severn
- School of Biochemistry, Biomedical Sciences Building; National Institute for Health Research (NIHR) Blood and Transplant Research Unit in Red Blood Cell Products, University of Bristol.
| | - Ashley M Toye
- School of Biochemistry, Biomedical Sciences Building; National Institute for Health Research (NIHR) Blood and Transplant Research Unit in Red Blood Cell Products, University of Bristol; Bristol Institute of Transfusion Sciences, NHSBT Filton. Bristol.
| |
Collapse
|
20
|
Di Buduo CA, Aguilar A, Soprano PM, Bocconi A, Miguel CP, Mantica G, Balduini A. Latest culture techniques: cracking the secrets of bone marrow to mass-produce erythrocytes and platelets ex vivo. Haematologica 2021; 106:947-957. [PMID: 33472355 PMCID: PMC8017859 DOI: 10.3324/haematol.2020.262485] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
Since the dawn of medicine, scientists have carefully observed, modeled and interpreted the human body to improve healthcare. At the beginning there were drawings and paintings, now there is three-dimensional modeling. Moving from two-dimensional cultures and towards complex and relevant biomaterials, tissue-engineering approaches have been developed in order to create three-dimensional functional mimics of native organs. The bone marrow represents a challenging organ to reproduce because of its structure and composition that confer it unique biochemical and mechanical features to control hematopoiesis. Reproducing the human bone marrow niche is instrumental to answer the growing demand for human erythrocytes and platelets for fundamental studies and clinical applications in transfusion medicine. In this review, we discuss the latest culture techniques and technological approaches to obtain functional platelets and erythrocytes ex vivo. This is a rapidly evolving field that will define the future of targeted therapies for thrombocytopenia and anemia, but also a long-term promise for new approaches to the understanding and cure of hematologic diseases.
Collapse
Affiliation(s)
| | - Alicia Aguilar
- Department of Molecular Medicine, University of Pavia, Pavia
| | - Paolo M Soprano
- Department of Molecular Medicine, University of Pavia, Pavia
| | - Alberto Bocconi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy; Department of Chemistry, Materials and Chemical Engineering G. Natta, Politecnico di Milano, Milano
| | | | | | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy; Department of Biomedical Engineering, Tufts University, Medford, MA
| |
Collapse
|
21
|
Liu S, Wu M, Lancelot M, Deng J, Gao Y, Roback JD, Chen T, Cheng L. BMI1 enables extensive expansion of functional erythroblasts from human peripheral blood mononuclear cells. Mol Ther 2021; 29:1918-1932. [PMID: 33484967 PMCID: PMC8116606 DOI: 10.1016/j.ymthe.2021.01.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/26/2020] [Accepted: 01/12/2021] [Indexed: 01/06/2023] Open
Abstract
Transfusion of red blood cells (RBCs) from ABO-matched but genetically unrelated donors is commonly used for treating anemia and acute blood loss. Increasing demand and insufficient supply for donor RBCs, especially those of universal blood types or free of known and unknown pathogens, has called for ex vivo generation of functional RBCs by large-scale cell culture. However, generating physiological numbers of transfusable cultured RBCs (cRBCs) ex vivo remains challenging, due to our inability to either extensively expand primary RBC precursors (erythroblasts) or achieve efficient enucleation once erythroblasts have been expanded and induced to differentiation and maturation. Here, we report that ectopic expression of the human BMI1 gene confers extensive expansion of human erythroblasts, which can be derived readily from adult peripheral blood mononuclear cells of either healthy donors or sickle cell patients. These extensively expanded erythroblasts (E3s) are able to proliferate exponentially (>1 trillion-fold in 2 months) in a defined culture medium. Expanded E3 cells are karyotypically normal and capable of terminal maturation with approximately 50% enucleation. Additionally, E3-derived cRBCs can circulate in a mouse model following transfusion similar to primary human RBCs. Therefore, we provide a facile approach of generating physiological numbers of human functional erythroblasts ex vivo.
Collapse
Affiliation(s)
- Senquan Liu
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China; Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Mengyao Wu
- Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Division of Hematology, Huashan Hospital of Fudan University, Shanghai 200040, China
| | - Moira Lancelot
- Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jiusheng Deng
- Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yongxing Gao
- Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - John D Roback
- Center for Transfusion and Cellular Therapies, Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Tong Chen
- Division of Hematology, Huashan Hospital of Fudan University, Shanghai 200040, China.
| | - Linzhao Cheng
- Blood and Cell Therapy Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, China; Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
22
|
Ferguson DCJ, Mokim JH, Meinders M, Moody ERR, Williams TA, Cooke S, Trakarnsanga K, Daniels DE, Ferrer-Vicens I, Shoemark D, Tipgomut C, Macinnes KA, Wilson MC, Singleton BK, Frayne J. Characterization and evolutionary origin of novel C 2H 2 zinc finger protein (ZNF648) required for both erythroid and megakaryocyte differentiation in humans. Haematologica 2020; 106:2859-2873. [PMID: 33054117 PMCID: PMC8561289 DOI: 10.3324/haematol.2020.256347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Indexed: 01/01/2023] Open
Abstract
Human ZNF648 is a novel poly C-terminal C2H2 zinc finger protein identified amongst the most dysregulated proteins in erythroid cells differentiated from iPSC. Its nuclear localisation and structure indicate it is likely a DNA-binding protein. Using a combination of ZNF648 overexpression in an iPSC line and primary adult erythroid cells, ZNF648 knockdown in primary adult erythroid cells and megakaryocytes, comparative proteomics and transcriptomics we show that ZNF648 is required for both erythroid and megakaryocyte differentiation. Orthologues of ZNF648 were detected across Mammals, Reptilia, Actinopterygii, in some Aves, Amphibia and Coelacanthiformes suggesting the gene originated in the common ancestor of Osteichthyes (Euteleostomi or bony fish). Conservation of the C-terminal zinc finger domain is higher, with some variation in zinc finger number but a core of at least six zinc fingers conserved across all groups, with the N-terminus recognisably similar within but not between major lineages. This suggests the N-terminus of ZNF648 evolves faster than the C-terminus, however this is not due to exon-shuffling as the entire coding region of ZNF648 is within a single exon. As for other such transcription factors, the N-terminus likely carries out regulatory functions, but showed no sequence similarity to any known domains. The greater functional constraint on the zinc finger domain suggests ZNF648 binds at least some similar regions of DNA in the different organisms. However, divergence of the N-terminal region may enable differential expression, allowing adaptation of function in the different organisms.
Collapse
Affiliation(s)
- Daniel C. J. Ferguson
- School of Biochemistry, University of Bristol, Bristol, UK,*DCJF and JHM contributed equally as co-first authors
| | - Juraidah Haji Mokim
- School of Biochemistry, University of Bristol, Bristol, UK,*DCJF and JHM contributed equally as co-first authors
| | | | | | - Tom A. Williams
- School of Biological Sciences, University of Bristol, Bristol, UK
| | - Sarah Cooke
- School of Biochemistry, University of Bristol, Bristol, UK
| | - Kongtana Trakarnsanga
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Deborah E. Daniels
- School of Biochemistry, University of Bristol, Bristol, UK,NIHR Blood and Transplant Research Unit in Red Blood Cell Products, University of Bristol, Bristol, UK
| | | | | | - Chartsiam Tipgomut
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Katherine A. Macinnes
- School of Biochemistry, University of Bristol, Bristol, UK,NIHR Blood and Transplant Research Unit in Red Blood Cell Products, University of Bristol, Bristol, UK
| | | | - Belinda K. Singleton
- NIHR Blood and Transplant Research Unit in Red Blood Cell Products, University of Bristol, Bristol, UK,Bristol Institute for Transfusion Sciences, National Health Service Blood and Transplant (NHSBT), Bristol, UK
| | - Jan Frayne
- School of Biochemistry, University of Bristol, BS8 1TD, UK.; NIHR Blood and Transplant Research Unit in Red blood cell products, University of Bristol, Bristol BS8 1TD, UK.
| |
Collapse
|
23
|
Tolu SS, Wang K, Yan Z, Zhang S, Roberts K, Crouch AS, Sebastian G, Chaitowitz M, Fornari ED, Schwechter EM, Uehlinger J, Manwani D, Minniti CP, Bouhassira EE. Characterization of Hematopoiesis in Sickle Cell Disease by Prospective Isolation of Stem and Progenitor Cells. Cells 2020; 9:cells9102159. [PMID: 32987729 PMCID: PMC7598721 DOI: 10.3390/cells9102159] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 12/24/2022] Open
Abstract
The consequences of sickle cell disease (SCD) include ongoing hematopoietic stress, hemolysis, vascular damage, and effect of chronic therapies, such as blood transfusions and hydroxyurea, on hematopoietic stem and progenitor cell (HSPC) have been poorly characterized. We have quantified the frequencies of nine HSPC populations by flow cytometry in the peripheral blood of pediatric and adult patients, stratified by treatment and control cohorts. We observed broad differences between SCD patients and healthy controls. SCD is associated with 10 to 20-fold increase in CD34dim cells, a two to five-fold increase in CD34bright cells, a depletion in Megakaryocyte-Erythroid Progenitors, and an increase in hematopoietic stem cells, when compared to controls. SCD is also associated with abnormal expression of CD235a as well as high levels CD49f antigen expression. These findings were present to varying degrees in all patients with SCD, including those on chronic therapy and those who were therapy naive. HU treatment appeared to normalize many of these parameters. Chronic stress erythropoiesis and inflammation incited by SCD and HU therapy have long been suspected of causing premature aging of the hematopoietic system, and potentially increasing the risk of hematological malignancies. An important finding of this study was that the observed concentration of CD34bright cells and of all the HSPCs decreased logarithmically with time of treatment with HU. This correlation was independent of age and specific to HU treatment. Although the number of circulating HSPCs is influenced by many parameters, our findings suggest that HU treatment may decrease premature aging and hematologic malignancy risk compared to the other therapeutic modalities in SCD.
Collapse
Affiliation(s)
- Seda S. Tolu
- Department of Medicine, Division of Hematology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (S.S.T.); (A.S.C.); (G.S.); (M.C.); (C.P.M.)
| | - Kai Wang
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (K.W.); (Z.Y.); (S.Z.); (K.R.)
| | - Zi Yan
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (K.W.); (Z.Y.); (S.Z.); (K.R.)
| | - Shouping Zhang
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (K.W.); (Z.Y.); (S.Z.); (K.R.)
| | - Karl Roberts
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (K.W.); (Z.Y.); (S.Z.); (K.R.)
| | - Andrew S. Crouch
- Department of Medicine, Division of Hematology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (S.S.T.); (A.S.C.); (G.S.); (M.C.); (C.P.M.)
| | - Gracy Sebastian
- Department of Medicine, Division of Hematology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (S.S.T.); (A.S.C.); (G.S.); (M.C.); (C.P.M.)
| | - Mark Chaitowitz
- Department of Medicine, Division of Hematology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (S.S.T.); (A.S.C.); (G.S.); (M.C.); (C.P.M.)
| | - Eric D. Fornari
- Department of Orthopedic Surgery, Montefiore Medical Center, Bronx, NY 10461, USA; (E.D.F.); (E.M.S.)
| | - Evan M. Schwechter
- Department of Orthopedic Surgery, Montefiore Medical Center, Bronx, NY 10461, USA; (E.D.F.); (E.M.S.)
| | - Joan Uehlinger
- Department of Pathology, Division of Transfusion Medicine, Montefiore Health System, Bronx, NY 10467, USA;
| | - Deepa Manwani
- Pediatric Hematology/Oncology/Marrow and Blood Cell Transplantation, Montefiore Health System, Bronx, NY 10467, USA;
| | - Caterina P. Minniti
- Department of Medicine, Division of Hematology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (S.S.T.); (A.S.C.); (G.S.); (M.C.); (C.P.M.)
| | - Eric E. Bouhassira
- Department of Medicine, Division of Hematology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (S.S.T.); (A.S.C.); (G.S.); (M.C.); (C.P.M.)
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (K.W.); (Z.Y.); (S.Z.); (K.R.)
- Correspondence: ; Tel.: +1-718-430-2000
| |
Collapse
|
24
|
Park YJ, Jeon SH, Kim HK, Suh EJ, Choi SJ, Kim S, Kim HO. Human induced pluripotent stem cell line banking for the production of rare blood type erythrocytes. J Transl Med 2020; 18:236. [PMID: 32532292 PMCID: PMC7291485 DOI: 10.1186/s12967-020-02403-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/04/2020] [Indexed: 02/07/2023] Open
Abstract
Background The in vitro production of mature human red blood cells (RBCs) from induced pluripotent stem cells (iPSCs) has been the focus of research to meet the high demand for blood transfusions. However, limitations like high costs and technological requirements restrict the use of RBCs produced by iPSC differentiation to specific circumstances, such as for patients with rare blood types or alloimmunized patients. In this study, we developed a detailed protocol for the generation of iPSC lines derived from peripheral blood of donors with O D-positive blood and rare blood types (D–and Jr(a-)) and subsequent erythroid differentiation. Methods Mononuclear cells separated from the peripheral blood of O D-positive and rare blood type donors were cultured to produce and expand erythroid progenitors and reprogrammed into iPSCs. A 31-day serum-free, xeno-free erythroid differentiation protocol was used to generate reticulocytes. The stability of iPSC lines was confirmed with chromosomal analysis and RT-PCR. Morphology and cell counts were determined by microscopy observations and flow cytometry. Results Cells from all donors were successfully used to generate iPSC lines, which were differentiated into erythroid precursors without any apparent chromosomal mutations. This differentiation protocol resulted in moderate erythrocyte yield per iPSC. Conclusions It has previously only been hypothesized that erythroid differentiation from iPSCs could be used to produce RBCs for transfusion to patients with rare blood types or who have been alloimmunized. Our results demonstrate the feasibility of producing autologous iPSC-differentiated RBCs for clinical transfusions in patients without alternative options.
Collapse
Affiliation(s)
- Yu Jin Park
- Department of Laboratory Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.,Department of Laboratory Medicine, Armed Forces Yangju Hospital, Yangju-si, Gyeonggi-do, Korea
| | - Su-Hee Jeon
- Department of Laboratory Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyun-Kyung Kim
- Department of Laboratory Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Eun Jung Suh
- Department of Laboratory Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Seung Jun Choi
- Department of Laboratory Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Sinyoung Kim
- Department of Laboratory Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyun Ok Kim
- Department of Laboratory Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
25
|
Bernecker C, Ackermann M, Lachmann N, Rohrhofer L, Zaehres H, Araúzo-Bravo MJ, van den Akker E, Schlenke P, Dorn I. Enhanced Ex Vivo Generation of Erythroid Cells from Human Induced Pluripotent Stem Cells in a Simplified Cell Culture System with Low Cytokine Support. Stem Cells Dev 2019; 28:1540-1551. [PMID: 31595840 PMCID: PMC6882453 DOI: 10.1089/scd.2019.0132] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Red blood cell (RBC) differentiation from human induced pluripotent stem cells (hiPSCs) offers great potential for developmental studies and innovative therapies. However, ex vivo erythropoiesis from hiPSCs is currently limited by low efficiency and unphysiological conditions of common culture systems. Especially, the absence of a physiological niche may impair cell growth and lineage-specific differentiation. We here describe a simplified, xeno- and feeder-free culture system for prolonged RBC generation that uses low numbers of supporting cytokines [stem cell factor (SCF), erythropoietin (EPO), and interleukin 3 (IL-3)] and is based on the intermediate development of a “hematopoietic cell forming complex (HCFC).” From this HCFC, CD43+ hematopoietic cells (purity >95%) were continuously released into the supernatant and could be collected repeatedly over a period of 6 weeks for further erythroid differentiation. The released cells were mainly CD34+/CD45+ progenitors with high erythroid colony-forming potential and CD36+ erythroid precursors. A total of 1.5 × 107 cells could be harvested from the supernatant of one six-well plate, showing 100- to 1000-fold amplification during subsequent homogeneous differentiation into GPA+ erythroid cells. Mean enucleation rates near 40% (up to 60%) further confirmed the potency of the system. These benefits may be explained by the generation of a niche within the HCFC that mimics the spatiotemporal signaling of the physiological microenvironment in which erythropoiesis occurs. Compared to other protocols, this method provides lower complexity, less cytokine and medium consumption, higher cellular output, and better enucleation. In addition, slight modifications in cytokine addition shift the system toward continuous generation of granulocytes and macrophages.
Collapse
Affiliation(s)
- Claudia Bernecker
- Department of Blood Group Serology and Transfusion Medicine, Medical University Graz, Graz, Austria
| | - Mania Ackermann
- RG Translational Hematology of Congenital Diseases, Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Nico Lachmann
- RG Translational Hematology of Congenital Diseases, Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Cluster of Excellence, Hannover Medical School, Hannover, Germany
| | - Lisa Rohrhofer
- Department of Blood Group Serology and Transfusion Medicine, Medical University Graz, Graz, Austria
| | - Holm Zaehres
- Department of Anatomy and Molecular Embryology, Ruhr-University Bochum, Bochum, Germany
| | - Marcos J Araúzo-Bravo
- Computational Biology and Systems Biomedicine Research Group, Biodonostia Health Research Institute, San Sebastián, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | | | - Peter Schlenke
- Department of Blood Group Serology and Transfusion Medicine, Medical University Graz, Graz, Austria
| | - Isabel Dorn
- Department of Blood Group Serology and Transfusion Medicine, Medical University Graz, Graz, Austria
| |
Collapse
|
26
|
Differentiation of Baboon ( Papio anubis) Induced-Pluripotent Stem Cells into Enucleated Red Blood Cells. Cells 2019; 8:cells8101282. [PMID: 31635069 PMCID: PMC6829891 DOI: 10.3390/cells8101282] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/04/2019] [Accepted: 10/16/2019] [Indexed: 01/14/2023] Open
Abstract
As cell culture methods and stem cell biology have progressed, the in vitro production of cultured RBCs (cRBCs) has emerged as a viable option to produce cells for transfusion or to carry therapeutic cargoes. RBCs produced in culture can be quality-tested either by xeno-transfusion of human cells into immuno-deficient animals, or by transfusion of autologous cells in immuno-competent models. Although murine xeno-transfusion methods have improved, they must be complemented by studies in immuno-competent models. Non-human primates (NHPs) are important pre-clinical, large animal models due to their high biological and developmental similarities with humans, including their comparable hematopoietic and immune systems. Among NHPs, baboons are particularly attractive to validate cRBCs because of the wealth of data available on the characteristics of RBCs in this species that have been generated by past blood transfusion studies. We report here that we have developed a method to produce enucleated cRBCs by differentiation of baboon induced pluripotent stem cells (iPSCs). This method will enable the use of baboons to evaluate therapeutic cRBCs and generate essential pre-clinical data in an immuno-competent, large animal model. Production of the enucleated baboon cRBCs was achieved by adapting the PSC-RED protocol that we previously developed for human cells. Baboon-PSC-RED is an efficient chemically-defined method to differentiate iPSCs into cRBCs that are about 40% to 50% enucleated. PSC-RED is relatively low cost because it requires no albumin and only small amounts of recombinant transferrin.
Collapse
|