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
|
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
|
3
|
Xu L, Zeng Q, Liang L, Yang Z, Qu M, Li H, Zhang B, Zhang J, Yuan X, Chen L, Fan Z, He L, Nan X, Yue W, Xie X, Pei X. Generation of Rh D-negative blood using CRISPR/Cas9. Cell Prolif 2023; 56:e13486. [PMID: 37096780 PMCID: PMC10623963 DOI: 10.1111/cpr.13486] [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] [Received: 01/04/2023] [Revised: 03/27/2023] [Accepted: 04/12/2023] [Indexed: 04/26/2023] Open
Abstract
Blood supply shortages, especially the shortage of rare blood types, threaten the current medical system. Research on stem cells has shed light on in vitro blood cell manufacturing. The in vitro production of universal red blood cells (RBCs) from induced pluripotent stem cells (iPSCs) has become the focus of transfusion medicine. To obtain O-type Rh D-negative blood, we developed O-type Rh D-negative human (h)iPSCs using homology-directed repair (HDR)-based CRISPR/Cas9. HuAiPSCs derived from human umbilical arterial endothelial cells and showing haematopoietic differentiation preferences were selected for gene modification. Guide RNAs (gRNAs) were selected, and a donor template flanked by gRNA-directed homologous arms was set to introduce a premature stop code to RHD exon 2. CRISPR/Cas9 gene editing has resulted in the successful generation of an RHD knockout cell line. The HuAiPSC-A1-RHD-/- cell line was differentiated into haematopoietic stem/progenitor cells and subsequently into erythrocytes in the oxygen concentration-optimized differentiation scheme. HuAiPSC-A1-RHD-/- derived erythrocytes remained positive for the RBC markers CD71 and CD235a. These erythrocytes did not express D antigen and did not agglutinate in the presence of anti-Rh D reagents. In conclusion, taking the priority of haematopoietic preference hiPSCs, the HDR-based CRISPR/Cas9 system and optimizing the erythroid-lineage differentiation protocol, we first generated O-type Rh D-negative universal erythrocytes from RHD knockout HuAiPSCs. Its production is highly efficient and shows great potential for clinical applications.
Collapse
Affiliation(s)
- Lei Xu
- Stem Cell and Regenerative Medicine LabBeijing Institute of Radiation MedicineBeijingChina
| | - Quan Zeng
- Stem Cell and Regenerative Medicine LabBeijing Institute of Radiation MedicineBeijingChina
- South China Research Center for Stem Cell & Regenerative MedicineGuangzhouChina
| | - Liqing Liang
- Stem Cell and Regenerative Medicine LabBeijing Institute of Radiation MedicineBeijingChina
| | - Zhou Yang
- Stem Cell and Regenerative Medicine LabBeijing Institute of Radiation MedicineBeijingChina
| | - Mingyi Qu
- Stem Cell and Regenerative Medicine LabBeijing Institute of Radiation MedicineBeijingChina
- South China Research Center for Stem Cell & Regenerative MedicineGuangzhouChina
| | - Huilin Li
- Stem Cell and Regenerative Medicine LabBeijing Institute of Radiation MedicineBeijingChina
| | - Bowen Zhang
- Stem Cell and Regenerative Medicine LabBeijing Institute of Radiation MedicineBeijingChina
- South China Research Center for Stem Cell & Regenerative MedicineGuangzhouChina
| | - Jing Zhang
- Stem Cell and Regenerative Medicine LabBeijing Institute of Radiation MedicineBeijingChina
- South China Research Center for Stem Cell & Regenerative MedicineGuangzhouChina
| | - Xin Yuan
- Stem Cell and Regenerative Medicine LabBeijing Institute of Radiation MedicineBeijingChina
| | - Lin Chen
- Stem Cell and Regenerative Medicine LabBeijing Institute of Radiation MedicineBeijingChina
- South China Research Center for Stem Cell & Regenerative MedicineGuangzhouChina
| | - Zeng Fan
- Stem Cell and Regenerative Medicine LabBeijing Institute of Radiation MedicineBeijingChina
- South China Research Center for Stem Cell & Regenerative MedicineGuangzhouChina
| | - Lijuan He
- South China Research Center for Stem Cell & Regenerative MedicineGuangzhouChina
- Institute of Health Service and Transfusion MedicineBeijingChina
| | - Xue Nan
- South China Research Center for Stem Cell & Regenerative MedicineGuangzhouChina
| | - Wen Yue
- Stem Cell and Regenerative Medicine LabBeijing Institute of Radiation MedicineBeijingChina
- South China Research Center for Stem Cell & Regenerative MedicineGuangzhouChina
| | - Xiaoyan Xie
- Stem Cell and Regenerative Medicine LabBeijing Institute of Radiation MedicineBeijingChina
- South China Research Center for Stem Cell & Regenerative MedicineGuangzhouChina
| | - Xuetao Pei
- Stem Cell and Regenerative Medicine LabBeijing Institute of Radiation MedicineBeijingChina
- South China Research Center for Stem Cell & Regenerative MedicineGuangzhouChina
| |
Collapse
|
4
|
Kweon S, Kim S, Choi HS, Jo K, Park JM, Baek EJ. Current status of platelet manufacturing in 3D or bioreactors. Biotechnol Prog 2023; 39:e3364. [PMID: 37294031 DOI: 10.1002/btpr.3364] [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: 01/12/2023] [Revised: 05/09/2023] [Accepted: 05/09/2023] [Indexed: 06/10/2023]
Abstract
Blood shortages for transfusion are global issues of grave concern. As in vitro manufactured platelets are promising substitutes for blood donation, recent research has shown progresses including different cell sources, different bioreactors, and three-dimensional materials. The first-in-human clinical trial of cultured platelets using induced pluripotent stem cell-derived platelets began in Japan and demonstrated its quality, safety, and efficacy. A novel bioreactor with fluid motion for platelet production has been reported. Herein, we discuss various cell sources for blood cell production, recent advances in manufacturing processes, and clinical applications of cultured blood.
Collapse
Affiliation(s)
- Soonho Kweon
- Department of Research and Development, ArtBlood Inc, Seoul, Republic of Korea
| | - Suyeon Kim
- Department of Research and Development, ArtBlood Inc, Seoul, Republic of Korea
| | - Hye Sook Choi
- Department of Research and Development, ArtBlood Inc, Seoul, Republic of Korea
| | - Kyeongwon Jo
- Department of Research and Development, ArtBlood Inc, Seoul, Republic of Korea
| | - Ju Mi Park
- Department of Research and Development, ArtBlood Inc, Seoul, Republic of Korea
| | - Eun Jung Baek
- Department of Research and Development, ArtBlood Inc, Seoul, Republic of Korea
- Department of Translational Medicine, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
- Department of Laboratory Medicine, Hanyang University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
5
|
Jiamvoraphong N, Septham P, Klaihmon P, Laowtammathron C, U-Pratya Y, Lorthongpanich C, Issaragrisil S. Derivation of MUSIi016-A iPSCs from peripheral blood with blood type O Rh positive. Stem Cell Res 2023; 66:103014. [PMID: 36603353 DOI: 10.1016/j.scr.2022.103014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 12/20/2022] [Accepted: 12/30/2022] [Indexed: 01/01/2023] Open
Abstract
MUSIi016-A, a human induced pluripotent stem cell (iPSC), generated from peripheral blood mononuclear cells of a healthy blood group O Rh positive donor was reprogrammed using Sendai viral vectors containing Yamanaka's factors. MUSIi016-A iPSC showed pluripotent stem cell characteristics, highly expressed pluripotent markers, and a capacity to differentiate into all three embryonic cell lineages. This iPSC can be used as a model for the generation of blood cells in vitro.
Collapse
Affiliation(s)
- Nittaya Jiamvoraphong
- Siriraj Center of Excellence for Stem Cell Research, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Prapasri Septham
- Siriraj Center of Excellence for Stem Cell Research, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Phatchanat Klaihmon
- Siriraj Center of Excellence for Stem Cell Research, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chuti Laowtammathron
- Siriraj Center of Excellence for Stem Cell Research, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Yaowalak U-Pratya
- Siriraj Center of Excellence for Stem Cell Research, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Chanchao Lorthongpanich
- Siriraj Center of Excellence for Stem Cell Research, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem Cell Research, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand; Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Bangkok Hematology Center, Wattanosoth Hospital, BDMS Center of Excellence for Cancer, Bangkok 10310, Thailand
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Luanpitpong S, Kang X, Janan M, Thumanu K, Li J, Kheolamai P, Issaragrisil S. Metabolic sensor O-GlcNAcylation regulates erythroid differentiation and globin production via BCL11A. Stem Cell Res Ther 2022; 13:274. [PMID: 35739577 PMCID: PMC9219246 DOI: 10.1186/s13287-022-02954-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/24/2022] [Indexed: 12/25/2022] Open
Abstract
Background Human erythropoiesis is a tightly regulated, multistep process encompassing the differentiation of hematopoietic stem cells (HSCs) toward mature erythrocytes. Cellular metabolism is an important regulator of cell fate determination during the differentiation of HSCs. However, how O-GlcNAcylation, a posttranslational modification of proteins that is an ideal metabolic sensor, contributes to the commitment of HSCs to the erythroid lineage and to the terminal erythroid differentiation has not been addressed. Methods Cellular O-GlcNAcylation was manipulated using small molecule inhibition or CRISPR/Cas9 manipulation of catalyzing enzyme O-GlcNAc transferase (OGT) and removing enzyme O-GlcNAcase (OGA) in two cell models of erythroid differentiation, starting from: (i) human umbilical cord blood-derived CD34+ hematopoietic stem/progenitor cells (HSPCs) to investigate the erythroid lineage specification and differentiation; and (ii) human-derived erythroblastic leukemia K562 cells to investigate the terminal differentiation. The functional and regulatory roles of O-GlcNAcylation in erythroid differentiation, maturation, and globin production were investigated, and downstream signaling was delineated. Results First, we observed that two-step inhibition of OGT and OGA, which were established from the observed dynamics of O-GlcNAc level along the course of differentiation, promotes HSPCs toward erythroid differentiation and enucleation, in agreement with an upregulation of a multitude of erythroid-associated genes. Further studies in the efficient K562 model of erythroid differentiation confirmed that OGA inhibition and subsequent hyper-O-GlcNAcylation enhance terminal erythroid differentiation and affect globin production. Mechanistically, we found that BCL11A is a key mediator of O-GlcNAc-driven erythroid differentiation and β- and α-globin production herein. Additionally, analysis of biochemical contents using synchrotron-based Fourier transform infrared (FTIR) spectroscopy showed unique metabolic fingerprints upon OGA inhibition during erythroid differentiation, supporting that metabolic reprogramming plays a part in this process. Conclusions The evidence presented here demonstrated the novel regulatory role of O-GlcNAc/BCL11A axis in erythroid differentiation, maturation, and globin production that could be important in understanding erythropoiesis and hematologic disorders whose etiology is related to impaired erythroid differentiation and hemoglobinopathies. Our findings may lay the groundwork for future clinical applications toward an ex vivo production of functional human reticulocytes for transfusion from renewable cell sources, i.e., HSPCs and pluripotent stem cells. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02954-5.
Collapse
Affiliation(s)
- Sudjit Luanpitpong
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Siriraj Hospital, Bangkoknoi, Bangkok, 10700, Thailand.
| | - Xing Kang
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Siriraj Hospital, Bangkoknoi, Bangkok, 10700, Thailand
| | - Montira Janan
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Siriraj Hospital, Bangkoknoi, Bangkok, 10700, Thailand
| | - Kanjana Thumanu
- Synchrotron Light Research Institute (Public Organization), Nakhon Ratchasima, Thailand
| | - Jingting Li
- Institute of Precision Medicine, Department of Burns, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Pakpoom Kheolamai
- Center of Excellence in Stem Cell Research and Innovation, Faculty of Medicine, Thammasat University, Pathum Thani, 12120, Thailand.
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Siriraj Hospital, Bangkoknoi, Bangkok, 10700, Thailand.,Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
9
|
Nilsri N, Jangprasert P, Pawinwongchai J, Israsena N, Rojnuckarin P. Distinct effects of V617F and exon12-mutated JAK2 expressions on erythropoiesis in a human induced pluripotent stem cell (iPSC)-based model. Sci Rep 2021; 11:5255. [PMID: 33664283 PMCID: PMC7933160 DOI: 10.1038/s41598-021-83895-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 02/09/2021] [Indexed: 12/15/2022] Open
Abstract
Activating mutations affecting the JAK-STAT signal transduction is the genetic driver of myeloproliferative neoplasms (MPNs) which comprise polycythemia vera (PV), essential thrombocythemia (ET) and myelofibrosis. The JAK2p.V617F mutation can produce both erythrocytosis in PV and thrombocytosis in ET, while JAK2 exon 12 mutations cause only erythrocytosis. We hypothesized that these two mutations activated different intracellular signals. In this study, the induced pluripotent stem cells (iPSCs) were used to model JAK2-mutated MPNs. Normal iPSCs underwent lentiviral transduction to overexpress JAK2p.V617F or JAK2p.N542_E543del (JAK2exon12) under a doxycycline-inducible system. The modified iPSCs were differentiated into erythroid cells. Compared with JAK2V617F-iPSCs, JAK2exon12-iPSCs yielded more total CD71+GlycophorinA+ erythroid cells, displayed more mature morphology and expressed more adult hemoglobin after doxycycline induction. Capillary Western immunoassay revealed significantly higher phospho-STAT1 but lower phospho-STAT3 and lower Phospho-AKT in JAK2exon12-iPSCs compared with those of JAK2V617F-iPSCs in response to erythropoietin. Furthermore, interferon alpha and arsenic trioxide were tested on these modified iPSCs to explore their potentials for MPN therapy. Both agents preferentially inhibited proliferation and promoted apoptosis of the iPSCs expressing mutant JAK2 compared with those without doxycycline induction. In conclusion, the modified iPSC model can be used to investigate the mechanisms and search for new therapy of MPNs.
Collapse
Affiliation(s)
- Nungruthai Nilsri
- Doctor of Philosophy Program in Medical Sciences, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| | - Panchalee Jangprasert
- Interdisciplinary Program of Biomedical Sciences, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Nipan Israsena
- Stem Cell and Cell Therapy Research Unit, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Ponlapat Rojnuckarin
- Research Unit in Translational Hematology, Division of Hematology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, 10330, Thailand.
| |
Collapse
|
10
|
Ebrahimi M, Forouzesh M, Raoufi S, Ramazii M, Ghaedrahmati F, Farzaneh M. Differentiation of human induced pluripotent stem cells into erythroid cells. Stem Cell Res Ther 2020; 11:483. [PMID: 33198819 PMCID: PMC7667818 DOI: 10.1186/s13287-020-01998-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 10/25/2020] [Indexed: 02/07/2023] Open
Abstract
During the last years, several strategies have been made to obtain mature erythrocytes or red blood cells (RBC) from the bone marrow or umbilical cord blood (UCB). However, UCB-derived hematopoietic stem cells (HSC) are a limited source and in vitro large-scale expansion of RBC from HSC remains problematic. One promising alternative can be human pluripotent stem cells (PSCs) that provide an unlimited source of cells. Human PSCs, including embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), are self-renewing progenitors that can be differentiated to lineages of ectoderm, mesoderm, and endoderm. Several previous studies have revealed that human ESCs can differentiate into functional oxygen-carrying erythrocytes; however, the ex vivo expansion of human ESC-derived RBC is subjected to ethical concerns. Human iPSCs can be a suitable therapeutic choice for the in vitro/ex vivo manufacture of RBCs. Reprogramming of human somatic cells through the ectopic expression of the transcription factors (OCT4, SOX2, KLF4, c-MYC, LIN28, and NANOG) has provided a new avenue for disease modeling and regenerative medicine. Various techniques have been developed to generate enucleated RBCs from human iPSCs. The in vitro production of human iPSC-derived RBCs can be an alternative treatment option for patients with blood disorders. In this review, we focused on the generation of human iPSC-derived erythrocytes to present an overview of the current status and applications of this field.
Collapse
Affiliation(s)
- Mohsen Ebrahimi
- Neonatal and Children's Health Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mehdi Forouzesh
- Legal Medicine Organization of Iran, Legal Medicine Research Center, Legal Medicine organization, Tehran, Iran
| | - Setareh Raoufi
- Faculty of Medical Sciences and Technologies, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Ramazii
- Kerman University of Medical Sciences, University of Kerman, Kerman, Iran
| | - Farhoodeh Ghaedrahmati
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Farzaneh
- Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
11
|
Abonyo C, Shaviya N, Budambula V, Were T. Anemia Burden, Types and Associated Risk Factors among Kenyan Human Immunodeficiency Virus-1 and Mycobacterium Tuberculosis Co-infected Injection Substance Users. Ethiop J Health Sci 2020; 30:661-670. [PMID: 33911826 PMCID: PMC8047275 DOI: 10.4314/ejhs.v30i5.4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 05/19/2020] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Although injection substance users and individuals co-infected with Human Immunodeficiency Virus-1 and Mycobacterium tuberculosis suffer marked hematologic derangements, the rates, levels, morphologic types and associated risk factors of anemia among Human immunodeficiency virus and Mycobacterium tuberculosis coinfected injection substance users has not been reported in Kenya. METHODS This cross-sectional study determined anemia rates, levels and morphologic types. Anemia was associated with clinical markers of disease- underweight, immunosuppression and viral load. Complete blood count, CD4 T-cell enumeration and viral load were determined via standard laboratory methods. RESULTS All injection substance users had higher rates of anaemia (HIV+TB+ ISUs, 79.3%; HIV-TB+ISUs, 70.0%; HIV+TB- ISUs, 56.6% and HIV-TB- ISUs, 56.2%) relative to non-ISUs (16.6%; P<0.05). A significant proportion of HIV+TB+ISUs (47.8%) developed severe anemia than other clinical groups. The commonest morphologic type of anemia in HIV+TB+ISUs was microcytic hypochromic (43.5%) followed by normocytic hypochromic (17.4%) relative to the other clinical groups. HIV+TB+ ISUs with CD4 T-cells <200/uL (OR: 2.94, 95% CI: 1.41-6.13, P=0.004) and CD4 Tcells of 200-349/uL (OR: 3.24, 95% CI: 1.66-6.31, P=0.001) associated with higher odds of developing anemia. CONCLUSION This study revealed that severe anemia and microcytic hypochromic anemia are the most common erythrocytic sequelae among Human Immunodeficiency Virus-1 and Mycobacterium tuberculosis co-infected ISUs. Those with CD4 T-cells < 350/uL are utmost expected to develop anemia.
Collapse
Affiliation(s)
- Collins Abonyo
- School of Public Health Biomedical Sciences and Technology, Department of Medical Laboratory Sciences, Masinde Muliro University of Science and Technology, Kanya
| | - Nathan Shaviya
- School of Public Health Biomedical Sciences and Technology, Department of Medical Laboratory Sciences, Masinde Muliro University of Science and Technology, Kanya
| | - Valentine Budambula
- School of Applied and Health Sciences, Department of Environment and Health Sciences, Technical University of Mombasa, Kenya
| | - Tom Were
- School of Public Health Biomedical Sciences and Technology, Department of Medical Laboratory Sciences, Masinde Muliro University of Science and Technology, Kanya
| |
Collapse
|
12
|
Thompson C, Otero P, Srinageshwar B, Petersen RB, Dunbar GL, Rossignol J. Possible roles of epigenetics in stem cell therapy for Parkinson's disease. Epigenomics 2020; 12:647-656. [PMID: 32396465 DOI: 10.2217/epi-2019-0347] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disease with loss of dopaminergic neurons. PD has genetic and epigenetic influences that determine specific changes in the brain. Epigenetic changes result in defective methylation of genes leading to differential gene-expression causing PD. This review provides an overview of stem cell transplantations as potential therapies for PD, with a focus on the epigenetic changes, prior or following transplantation. To date, no reports have addressed epigenetic alterations following stem cell transplantation into the PD brain. Given the potential for affecting the efficacy of stem cell therapy, increased attention needs to be given to the epigenetic processes that occur during stem cell culture and transplantation to maximize the therapeutic potential of stem cells to PD.
Collapse
Affiliation(s)
- Cassandra Thompson
- Field Neurosciences Institute laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI 48859, USA.,Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI 48859, USA
| | - Paulina Otero
- Field Neurosciences Institute laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI 48859, USA.,Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI 48859, USA
| | - Bhairavi Srinageshwar
- Field Neurosciences Institute laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI 48859, USA.,Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI 48859, USA.,College of Medicine, Central Michigan University, Mt. Pleasant, MI 48859, USA
| | - Robert B Petersen
- College of Medicine, Central Michigan University, Mt. Pleasant, MI 48859, USA
| | - Gary L Dunbar
- Field Neurosciences Institute laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI 48859, USA.,Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI 48859, USA.,College of Medicine, Central Michigan University, Mt. Pleasant, MI 48859, USA.,Department of Psychology, Central Michigan University, Mt. Pleasant, MI 48859, USA.,Field Neurosciences Institute, St. Mary's of Michigan, Saginaw, MI 48604, USA
| | - Julien Rossignol
- Field Neurosciences Institute laboratory for Restorative Neurology, Central Michigan University, Mt. Pleasant, MI 48859, USA.,Program in Neuroscience, Central Michigan University, Mt. Pleasant, MI 48859, USA.,College of Medicine, Central Michigan University, Mt. Pleasant, MI 48859, USA
| |
Collapse
|
13
|
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
|
14
|
A β1-tubulin-based megakaryocyte maturation reporter system identifies novel drugs that promote platelet production. Blood Adv 2019; 2:2262-2272. [PMID: 30206099 DOI: 10.1182/bloodadvances.2018019547] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 08/11/2018] [Indexed: 12/17/2022] Open
Abstract
During maturation, megakaryocytes (MKs) express β1-tubulin (TUBB1) and rearrange their microtubule components to enlarge, form proplatelets, and eventually release platelets. The development of a platform to identify in vitro conditions that would efficiently promote MK development could potentially enable large-scale platelet production. Here, we show that an immortalized MK cell line (imMKCL) genetically modified to express the β1-tubulin-Venus reporter provides a practical system to efficiently monitor the in vitro production of platelet-like particles (PLPs). The Venus transgene was inserted downstream of the TUBB1 locus in imMKCLs using CRISPR/Cas9, and the expression was visualized by Venus fluorescence intensity. This imMKCL reporter line was then used for high-throughput drug screening. We identified several compounds that significantly improved the efficiency of PLP production in vitro under feeder-free conditions and showed a significant tendency to recover platelets in vivo in a mouse thrombocytopenia model induced by anti-GPIbα antibody administration. Interestingly, most of these compounds, including a WNT signaling pathway inhibitor, Wnt-C59, antagonized the aryl hydrocarbon receptor (AhR) to increase PLP production, confirming the crucial role of AhR inhibition in MK maturation. Consistently, small interfering RNA treatment against AhR increased the Venus intensity and PLP production. TCS 359, an FLT3 inhibitor, significantly increased PLP production independently of FLT3 or AhR. This study highlights the usefulness of the β1-tubulin reporter MK line as a useful tool to study the mechanisms underlying thrombopoiesis and to identify novel inducers of ex vivo platelet production.
Collapse
|
15
|
Recent Updates on Induced Pluripotent Stem Cells in Hematological Disorders. Stem Cells Int 2019; 2019:5171032. [PMID: 31191673 PMCID: PMC6525795 DOI: 10.1155/2019/5171032] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 03/31/2019] [Indexed: 02/07/2023] Open
Abstract
Over the past decade, enormous progress has been made in the field of induced pluripotent stem cells (iPSCs). Patients' somatic cells such as skin fibroblasts or blood cells can be used to generate disease-specific pluripotent stem cells, which have unlimited proliferation and can differentiate into all cell types of the body. Human iPSCs offer great promises and opportunities for treatments of degenerative diseases and studying disease pathology and drug screening. So far, many iPSC-derived disease models have led to the discovery of novel pathological mechanisms as well as new drugs in the pipeline that have been tested in the iPSC-derived cells for efficacy and potential toxicities. Furthermore, recent advances in genome editing technology in combination with the iPSC technology have provided a versatile platform for studying stem cell biology and regenerative medicine. In this review, an overview of iPSCs, patient-specific iPSCs for disease modeling and drug screening, applications of iPSCs and genome editing technology in hematological disorders, remaining challenges, and future perspectives of iPSCs in hematological diseases will be discussed.
Collapse
|
16
|
Gaggi G, Izzicupo P, Di Credico A, Sancilio S, Di Baldassarre A, Ghinassi B. Spare Parts from Discarded Materials: Fetal Annexes in Regenerative Medicine. Int J Mol Sci 2019; 20:ijms20071573. [PMID: 30934825 PMCID: PMC6479500 DOI: 10.3390/ijms20071573] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/24/2019] [Accepted: 03/26/2019] [Indexed: 12/13/2022] Open
Abstract
One of the main aims in regenerative medicine is to find stem cells that are easy to obtain and are safe and efficient in either an autologous or allogenic host when transplanted. This review provides an overview of the potential use of the fetal annexes in regenerative medicine: we described the formation of the annexes, their immunological features, the new advances in the phenotypical characterization of fetal annexes-derived stem cells, the progressions obtained in the analysis of both their differentiative potential and their secretoma, and finally, the potential use of decellularized fetal membranes. Normally discarded as medical waste, the umbilical cord and perinatal tissue not only represent a rich source of stem cells but can also be used as a scaffold for regenerative medicine, providing a suitable environment for the growth and differentiation of stem cells.
Collapse
Affiliation(s)
- Giulia Gaggi
- Department of Medicine and Aging Sciences, University "G. D'Annunzio" of Chieti-Pescara, 66100 Chieti, Italy.
| | - Pascal Izzicupo
- Department of Medicine and Aging Sciences, University "G. D'Annunzio" of Chieti-Pescara, 66100 Chieti, Italy.
| | - Andrea Di Credico
- Department of Medicine and Aging Sciences, University "G. D'Annunzio" of Chieti-Pescara, 66100 Chieti, Italy.
| | - Silvia Sancilio
- Department of Medicine and Aging Sciences, University "G. D'Annunzio" of Chieti-Pescara, 66100 Chieti, Italy.
| | - Angela Di Baldassarre
- Department of Medicine and Aging Sciences, University "G. D'Annunzio" of Chieti-Pescara, 66100 Chieti, Italy.
| | - Barbara Ghinassi
- Department of Medicine and Aging Sciences, University "G. D'Annunzio" of Chieti-Pescara, 66100 Chieti, Italy.
| |
Collapse
|
17
|
Chandrashekar S. Transfusion Medicine has Emerged: Redefining the Role of the Transfusion Specialists in the Changing Scenario. GLOBAL JOURNAL OF TRANSFUSION MEDICINE 2019. [DOI: 10.4103/gjtm.gjtm_59_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
18
|
Ko SF, Chen YT, Wallace CG, Chen KH, Sung PH, Cheng BC, Huang TH, Chen YL, Li YC, Chang HW, Lee MS, Yang CC, Yip HK. Inducible pluripotent stem cell-derived mesenchymal stem cell therapy effectively protected kidney from acute ischemia-reperfusion injury. Am J Transl Res 2018; 10:3053-3067. [PMID: 30416650 PMCID: PMC6220224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 09/05/2018] [Indexed: 06/09/2023]
Abstract
This study tested whether inducible pluripotent stem cell (iPSC)-derived mesenchymal stem cell (MSC) therapy could effectively protect kidney from acute ischemia (1 h) - reperfusion (5 day) injury (IRI). Male-adult SD-rats (n = 24) were equally categorized into groups 1 (sham-control), 2 [sham-control + iPSC-MSC (1.2 × 106 cells/rat)], 3 (IR only) and 4 (IR + iPSC-MSC). Blood urine nitrogen/creatinine levels and ratio of urine protein to creatinine, kidney weight and expressions of inflammation (TNF-α/NF-κB), oxidative-stress (NOX-1/NOX-2/oxidized protein) and apoptosis (mitochondrial-Bax/cleaved caspase-3/PARP) were significantly higher in group 3 than in groups 1, 2 and 4 and significantly higher in group 4 than in groups 1 and 2 (all P<0.0001), but showed no differences between groups 1 and 2, whereas the protein expressions of anti-inflammation (IL-4/IL-10) and endothelial (CD31/vWF) markers exhibited an opposite pattern to inflammation among the four groups (all P<0.0001). Protein expressions of angiogenesis (VEGF/CXCR4/SDF-1α) markers progressively increased from groups 1 to 4 (all P<0.0001). Cellular expressions of kidney injury score/DNA-damage (γ-H2AX)/apoptotic nuclei and glomerulus-tubular-damage (KIM/FSP-1) displayed an identical pattern to inflammation, whereas the cellular expressions of glomerulus-tubular-integrity (dystroglycan/podocin/p-cadherin/synaptopodin/ZO-1/fibronectin) revealed an opposite pattern to inflammation among the four groups (all P<0.0001). In conclusion, iPSC-derived MSC therapy effectively protected kidney against IRI.
Collapse
Affiliation(s)
- Sheung-Fat Ko
- Department of Radiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung 83301, Taiwan
| | - Yen-Ta Chen
- Division of Urology, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung 83301, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial HospitalKaohsiung 83301, Taiwan
| | | | - Kuan-Hung Chen
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung 83301, Taiwan
| | - Pei-Hsun Sung
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung 83301, Taiwan
| | - Ben-Chung Cheng
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung 83301, Taiwan
| | - Tien-Hung Huang
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung 83301, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial HospitalKaohsiung 83301, Taiwan
| | - Yi-Ling Chen
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung 83301, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial HospitalKaohsiung 83301, Taiwan
| | - Yi-Chen Li
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung 83301, Taiwan
| | - Hsueh-Wen Chang
- Department of Biological Sciences, National Sun Yat-sen UniversityKaohsiung 80424, Taiwan
| | - Mel S Lee
- Department of Orthopedics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung 83301, Taiwan
| | - Chih-Chao Yang
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung 83301, Taiwan
| | - Hon-Kan Yip
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung 83301, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial HospitalKaohsiung 83301, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial HospitalKaohsiung 83301, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical UniversityTaichung 40402, Taiwan
- Department of Nursing, Asia UniversityTaichung 41354, Taiwan
| |
Collapse
|
19
|
Ovchynnikova E, Aglialoro F, von Lindern M, van den Akker E. The Shape Shifting Story of Reticulocyte Maturation. Front Physiol 2018; 9:829. [PMID: 30050448 PMCID: PMC6050374 DOI: 10.3389/fphys.2018.00829] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 06/12/2018] [Indexed: 12/11/2022] Open
Abstract
The final steps of erythropoiesis involve unique cellular processes including enucleation and reorganization of membrane proteins and the cytoskeleton to produce biconcave erythrocytes. Surprisingly this process is still poorly understood. In vitro erythropoiesis protocols currently produce reticulocytes rather than biconcave erythrocytes. In addition, immortalized lines and iPSC-derived erythroid cell suffer from low enucleation and suboptimal final maturation potential. In light of the increasing prospect to use in vitro produced erythrocytes as (personalized) transfusion products or as therapeutic delivery agents, the mechanisms driving this last step of erythropoiesis are in dire need of resolving. Here we review the elusive last steps of reticulocyte maturation with an emphasis on protein sorting during the defining steps of reticulocyte formation during enucleation and maturation.
Collapse
Affiliation(s)
- Elina Ovchynnikova
- Department of Hematopoiesis, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Francesca Aglialoro
- Department of Hematopoiesis, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Marieke von Lindern
- Department of Hematopoiesis, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Emile van den Akker
- Department of Hematopoiesis, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|