1
|
Claessen MJAG, Yagci N, Fu K, Brandsma E, Kersten MJ, von Lindern M, van den Akker E. Production and stability of cultured red blood cells depends on the concentration of cholesterol in culture medium. Sci Rep 2024; 14:15592. [PMID: 38971841 PMCID: PMC11227516 DOI: 10.1038/s41598-024-66440-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024] Open
Abstract
The production of cultured red blood cells (cRBC) for transfusion purposes requires large scale cultures and downstream processes to purify enucleated cRBC. The membrane composition, and cholesterol content in particular, are important during proliferation of (pro)erythroblasts and for cRBC quality. Therefore, we tested the requirement for cholesterol in the culture medium during expansion and differentiation of erythroid cultures with respect to proliferation, enucleation and purification by filtration. The low cholesterol level (22 µg/dl) in serum free medium was sufficient to expand (pro)erythroblast cultures. Addition of 2.0 or 5.0 mg/dL of free cholesterol at the start of differentiation induction inhibited enucleation compared to the default condition containing 3.3 mg/dl total cholesterol derived from the addition of Omniplasma to serum free medium. Addition of 5.0 mg/dl cholesterol at day 5 of differentiation did not affect the enucleation process but significantly increased recovery of enucleated cRBC following filtration over leukodepletion filters. The addition of cholesterol at day 5 increased the osmotic resistance of cRBC. In conclusion, cholesterol supplementation after the onset of enucleation improved the robustness of cRBC and increased the yield of enucleated cRBC in the purification process.
Collapse
Affiliation(s)
- M J A G Claessen
- Department Research, Sanquin Blood Supply, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
- Department of Hematology, Amsterdam University Medical Centers, Cancer Center Amsterdam, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
- Landsteiner Laboratory, Amsterdam University Medical Center, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
| | - N Yagci
- Department Research, Sanquin Blood Supply, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
- Landsteiner Laboratory, Amsterdam University Medical Center, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
| | - K Fu
- Department Research, Sanquin Blood Supply, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
- Landsteiner Laboratory, Amsterdam University Medical Center, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
| | - E Brandsma
- Department Research, Sanquin Blood Supply, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
- Department of Life Sciences, Saxion University of Applied Sciences, M.H. Tromplaan 28, 7513AB, Enschede, The Netherlands
| | - M J Kersten
- Department of Hematology, Amsterdam University Medical Centers, Cancer Center Amsterdam, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - M von Lindern
- Department Research, Sanquin Blood Supply, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
- Landsteiner Laboratory, Amsterdam University Medical Center, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands
| | - E van den Akker
- Department Research, Sanquin Blood Supply, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands.
- Landsteiner Laboratory, Amsterdam University Medical Center, Plesmanlaan 125, 1066 CX, Amsterdam, The Netherlands.
| |
Collapse
|
2
|
Wang Y, Peng X. Bioinformatics analysis characterizes immune infiltration landscape and identifies potential blood biomarkers for heart transplantation. Transpl Immunol 2024; 84:102036. [PMID: 38499050 DOI: 10.1016/j.trim.2024.102036] [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: 12/04/2023] [Revised: 03/15/2024] [Accepted: 03/15/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND Cardiac allograft rejection (AR) remains a significant complication following heart transplantation. The primary objective of our study is to gain a comprehensive understanding of the fundamental mechanisms involved in AR and identify possible therapeutic targets. METHODS We acquired the GSE87301 dataset from the Gene Expression Omnibus database. In GSE87301, a comparison was conducted on blood samples from patients with and without cardiac allograft rejection (AR and NAR) to detect differentially expressed genes (DEGs). Enrichment analysis was conducted to identify the pathways that show significant enrichment during AR. Machine learning techniques, including the least absolute shrinkage and selection operator regression (LASSO) and random forest (RF) algorithms, were employed to identify potential genes for the diagnosis of AR. The diagnostic value was evaluated using a nomogram and receiver operating characteristic (ROC) curve. Additionally, immune cell infiltration was analyzed to explore any dysregulation of immune cells in AR. RESULTS A total of 114 DEGs were identified from the GSE87301 dataset. These DEGs were mainly found to be enriched in pathways related to the immune system. To identify the signature genes, the LASSO and RF algorithms were used, and four genes, namely ALAS2, HBD, EPB42, and FECH, were identified. The performance of these signature genes was evaluated using the receiver operating characteristic curve (ROC) analysis, which showed that the area under the curve (AUC) values for ALAS2, HBD, EPB42, and FECH were 0.906, 0.881, 0.900, and 0.856, respectively. These findings were further confirmed in the independent datasets and clinical samples. The selection of these specific genes was made to construct a nomogram, which demonstrated excellent diagnostic ability. Additionally, the results of the single-sample gene set enrichment analysis (ssGSEA) revealed that these genes may be involved in immune cell infiltration. CONCLUSION We identified four signature genes (ALAS2, HBD, EPB42, and FECH) as potential peripheral blood diagnostic candidates for AR diagnosis. Additionally, a nomogram was constructed to aid in the diagnosis of heart transplantation. This study offers valuable insights into the identification of candidate genes for heart transplantation using peripheral blood samples.
Collapse
Affiliation(s)
- Yujia Wang
- Queen Mary College of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xiaoping Peng
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, 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
|
Barnhoorn S, Milanese C, Li T, Dons L, Ghazvini M, Sette M, Farina S, Sproviero D, Payan-Gomez C, Mastroberardino PG. Orthogonal analysis of mitochondrial function in Parkinson's disease patients. Cell Death Dis 2024; 15:243. [PMID: 38570521 PMCID: PMC10991487 DOI: 10.1038/s41419-024-06617-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 03/08/2024] [Accepted: 03/18/2024] [Indexed: 04/05/2024]
Abstract
The etiopathology of Parkinson's disease has been associated with mitochondrial defects at genetic, laboratory, epidemiological, and clinical levels. These converging lines of evidence suggest that mitochondrial defects are systemic and causative factors in the pathophysiology of PD, rather than being mere correlates. Understanding mitochondrial biology in PD at a granular level is therefore crucial from both basic science and translational perspectives. In a recent study, we investigated mitochondrial alterations in fibroblasts obtained from PD patients assessing mitochondrial function in relation to clinical measures. Our findings demonstrated that the magnitude of mitochondrial alterations parallels disease severity. In this study, we extend these investigations to blood cells and dopamine neurons derived from induced pluripotent stem cells reprogrammed from PD patients. To overcome the inherent metabolic heterogeneity of blood cells, we focused our analyses on metabolically homogeneous, accessible, and expandable erythroblasts. Our results confirm the presence of mitochondrial anomalies in erythroblasts and induced dopamine neurons. Consistent with our previous findings in fibroblasts, we observed that mitochondrial alterations are reversible, as evidenced by enhanced mitochondrial respiration when PD erythroblasts were cultured in a galactose medium that restricts glycolysis. This observation indicates that suppression of mitochondrial respiration may constitute a protective, adaptive response in PD pathogenesis. Notably, this effect was not observed in induced dopamine neurons, suggesting their distinct bioenergetic behavior. In summary, we provide additional evidence for the involvement of mitochondria in the disease process by demonstrating mitochondrial abnormalities in additional cell types relevant to PD. These findings contribute to our understanding of PD pathophysiology and may have implications for the development of novel biomarkers and therapeutic strategies.
Collapse
Affiliation(s)
- Sander Barnhoorn
- Department of Molecular Genetics, Erasmus MC, Rotterdam, Netherlands
| | - Chiara Milanese
- IFOM-ETS, the AIRC Institute for molecular Oncology, Milan, Italy
| | - Tracy Li
- Erasmus MC iPS Facility, Erasmus MC, Rotterdam, Netherlands
| | - Lieke Dons
- Erasmus MC iPS Facility, Erasmus MC, Rotterdam, Netherlands
| | | | | | - Stefania Farina
- Department of Molecular Genetics, Erasmus MC, Rotterdam, Netherlands
| | - Daisy Sproviero
- IFOM-ETS, the AIRC Institute for molecular Oncology, Milan, Italy
| | | | - Pier G Mastroberardino
- Department of Molecular Genetics, Erasmus MC, Rotterdam, Netherlands.
- IFOM-ETS, the AIRC Institute for molecular Oncology, Milan, Italy.
- Università degli Studi dell'Aquila, L'Aquila, Italy.
| |
Collapse
|
5
|
Godard A, Seute R, Grimaldi A, Granier T, Chiaroni J, El Nemer W, De Grandis M. A comparative study of two routinely used protocols for ex vivo erythroid differentiation. Blood Cells Mol Dis 2024; 106:102829. [PMID: 38278056 DOI: 10.1016/j.bcmd.2024.102829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 01/18/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024]
Abstract
BACKGROUND Erythropoiesis is a complex developmental process in which a hematopoietic stem cell undergoes serial divisions and differentiates through well-defined stages to give rise to red blood cells. Over the last decades, several protocols have been developed to perform ex vivo erythroid differentiation, allowing investigation into erythropoiesis and red cell production in health and disease. RESULTS In the current study, we compared the two commonly used protocols by assessing the differentiation kinetics, synchronisation, and cellular yield, using molecular and cellular approaches. Peripheral blood CD34+ cells were cultured in a two-phase (2P) or a four-phase (4P) liquid culture (LC) and monitored for 20 days. Both protocols could recapitulate all stages of erythropoiesis and generate reticulocytes, although to different extents. Higher proliferation and viability rates were achieved in the 4P-LC, with a higher degree of terminal differentiation and enucleation, associated with higher levels of the erythroid-specific transcription factors GATA-1, KLF-1, and TAL-1. Although the 2P-LC protocol was less efficient regarding terminal erythroid differentiation and maturation, it showed a higher yield of erythroid progenitors in the erythropoietin (EPO)-free expansion phase. CONCLUSIONS We provide data supporting the use of one protocol or the other to study the biological processes occurring in the early or late stages of erythroid differentiation, depending on the physiological process or pathological defect under investigation in a given study.
Collapse
Affiliation(s)
- Auria Godard
- Etablissement Français du Sang PACA-Corse, Aix Marseille University, CNRS, ADES UMR 7268, 13005 Marseille, France; Laboratoire d'Excellence GR-Ex, 75000 Paris, France
| | - Robert Seute
- Etablissement Français du Sang PACA-Corse, Aix Marseille University, CNRS, ADES UMR 7268, 13005 Marseille, France; Laboratoire d'Excellence GR-Ex, 75000 Paris, France
| | - Alexandra Grimaldi
- Etablissement Français du Sang PACA-Corse, Aix Marseille University, CNRS, ADES UMR 7268, 13005 Marseille, France; Laboratoire d'Excellence GR-Ex, 75000 Paris, France
| | - Thomas Granier
- Etablissement Français du Sang PACA-Corse, Aix Marseille University, CNRS, ADES UMR 7268, 13005 Marseille, France; Laboratoire d'Excellence GR-Ex, 75000 Paris, France
| | - Jacques Chiaroni
- Etablissement Français du Sang PACA-Corse, Aix Marseille University, CNRS, ADES UMR 7268, 13005 Marseille, France; Laboratoire d'Excellence GR-Ex, 75000 Paris, France
| | - Wassim El Nemer
- Etablissement Français du Sang PACA-Corse, Aix Marseille University, CNRS, ADES UMR 7268, 13005 Marseille, France; Laboratoire d'Excellence GR-Ex, 75000 Paris, France
| | - Maria De Grandis
- Etablissement Français du Sang PACA-Corse, Aix Marseille University, CNRS, ADES UMR 7268, 13005 Marseille, France; Laboratoire d'Excellence GR-Ex, 75000 Paris, France.
| |
Collapse
|
6
|
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
|
7
|
Grochowska MM, Ferraro F, Mascaro AC, Natale D, Winkelaar A, Boumeester V, Breedveld GJ, Bonifati V, Mandemakers W. deCLUTTER2+ - a pipeline to analyze calcium traces in a stem cell model for ventral midbrain patterned astrocytes. Dis Model Mech 2023; 16:dmm049980. [PMID: 37260295 PMCID: PMC10309582 DOI: 10.1242/dmm.049980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 05/19/2023] [Indexed: 06/02/2023] Open
Abstract
Astrocytes are the most populous cell type of the human central nervous system and are essential for physiological brain function. Increasing evidence suggests multiple roles for astrocytes in Parkinson's disease, nudging a shift in the research focus, which historically pivoted around ventral midbrain dopaminergic neurons (vmDANs). Studying human astrocytes and other cell types in vivo remains challenging. However, in vitro-reprogrammed human stem cell-based models provide a promising alternative. Here, we describe a novel protocol for astrocyte differentiation from human stem cell-derived vmDAN-generating progenitors. This protocol simulates the regionalization, gliogenic switch, radial migration and final differentiation that occur in the developing human brain. We characterized the morphological, molecular and functional features of these ventral midbrain patterned astrocytes with a broad palette of techniques and identified novel candidate midbrain-astrocyte specific markers. In addition, we developed a new pipeline for calcium imaging data analysis called deCLUTTER2+ (deconvolution of Ca2+ fluorescent patterns) that can be used to discover spontaneous or cue-dependent patterns of Ca2+ transients. Altogether, our protocol enables the characterization of the functional properties of human ventral midbrain patterned astrocytes under physiological conditions and in disease.
Collapse
Affiliation(s)
- Martyna M. Grochowska
- Erasmus MC, University Medical Center Rotterdam, Department of Clinical Genetics, P.O. Box 2040, 3000 CA Rotterdam, Netherlands
| | - Federico Ferraro
- Erasmus MC, University Medical Center Rotterdam, Department of Clinical Genetics, P.O. Box 2040, 3000 CA Rotterdam, Netherlands
| | - Ana Carreras Mascaro
- Erasmus MC, University Medical Center Rotterdam, Department of Clinical Genetics, P.O. Box 2040, 3000 CA Rotterdam, Netherlands
| | - Domenico Natale
- Erasmus MC, University Medical Center Rotterdam, Department of Clinical Genetics, P.O. Box 2040, 3000 CA Rotterdam, Netherlands
| | - Amber Winkelaar
- Erasmus MC, University Medical Center Rotterdam, Department of Clinical Genetics, P.O. Box 2040, 3000 CA Rotterdam, Netherlands
| | - Valerie Boumeester
- Erasmus MC, University Medical Center Rotterdam, Department of Clinical Genetics, P.O. Box 2040, 3000 CA Rotterdam, Netherlands
| | - Guido J. Breedveld
- Erasmus MC, University Medical Center Rotterdam, Department of Clinical Genetics, P.O. Box 2040, 3000 CA Rotterdam, Netherlands
| | - Vincenzo Bonifati
- Erasmus MC, University Medical Center Rotterdam, Department of Clinical Genetics, P.O. Box 2040, 3000 CA Rotterdam, Netherlands
| | - Wim Mandemakers
- Erasmus MC, University Medical Center Rotterdam, Department of Clinical Genetics, P.O. Box 2040, 3000 CA Rotterdam, Netherlands
| |
Collapse
|
8
|
Gallego‐Murillo JS, Iacono G, van der Wielen LAM, van den Akker E, von Lindern M, Wahl SA. Expansion and differentiation of ex vivo cultured erythroblasts in scalable stirred bioreactors. Biotechnol Bioeng 2022; 119:3096-3116. [PMID: 35879812 PMCID: PMC9804173 DOI: 10.1002/bit.28193] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 07/14/2022] [Accepted: 07/23/2022] [Indexed: 01/05/2023]
Abstract
Transfusion of donor-derived red blood cells (RBCs) is the most common form of cell therapy. Production of transfusion-ready cultured RBCs (cRBCs) is a promising replacement for the current, fully donor-dependent therapy. A single transfusion unit, however, contains 2 × 1012 RBC, which requires large scale production. Here, we report on the scale-up of cRBC production from static cultures of erythroblasts to 3 L stirred tank bioreactors, and identify the effect of operating conditions on the efficiency of the process. Oxygen requirement of proliferating erythroblasts (0.55-2.01 pg/cell/h) required sparging of air to maintain the dissolved oxygen concentration at the tested setpoint (2.88 mg O2 /L). Erythroblasts could be cultured at dissolved oxygen concentrations as low as 0.7 O2 mg/ml without negative impact on proliferation, viability or differentiation dynamics. Stirring speeds of up to 600 rpm supported erythroblast proliferation, while 1800 rpm led to a transient halt in growth and accelerated differentiation followed by a recovery after 5 days of culture. Erythroblasts differentiated in bioreactors, with final enucleation levels and hemoglobin content similar to parallel cultures under static conditions.
Collapse
Affiliation(s)
- Joan Sebastián Gallego‐Murillo
- Sanquin Research and Landsteiner Laboratory, Department of HematopoiesisAmsterdam UMCAmsterdamThe Netherlands,Department of Biotechnology, Faculty of Applied SciencesDelft University of TechnologyDelftThe Netherlands,Present address:
MeatableAlexander Fleminglaan 1,2613AX,DelftThe Netherlands
| | - Giulia Iacono
- Sanquin Research and Landsteiner Laboratory, Department of HematopoiesisAmsterdam UMCAmsterdamThe Netherlands
| | - Luuk A. M. van der Wielen
- Department of Biotechnology, Faculty of Applied SciencesDelft University of TechnologyDelftThe Netherlands,Bernal Institute, Faculty of Science and EngineeringUniversity of LimerickLimerickRepublic of Ireland
| | - Emile van den Akker
- Sanquin Research and Landsteiner Laboratory, Department of HematopoiesisAmsterdam UMCAmsterdamThe Netherlands
| | - Marieke von Lindern
- Sanquin Research and Landsteiner Laboratory, Department of HematopoiesisAmsterdam UMCAmsterdamThe Netherlands
| | - Sebastian Aljoscha Wahl
- Department of Biotechnology, Faculty of Applied SciencesDelft University of TechnologyDelftThe Netherlands,Present address:
Lehrstuhl Für BioverfahrenstechnikFriedrich‐Alexander Universität Erlangen‐NürnbergPaul‐Gordan‐Str. 3,91052,ErlangenGermany
| |
Collapse
|
9
|
Sex differences in normal and malignant hematopoiesis. BLOOD SCIENCE 2022; 4:185-191. [PMID: 36311819 PMCID: PMC9592170 DOI: 10.1097/bs9.0000000000000133] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/10/2022] [Indexed: 12/09/2022] Open
Abstract
Hematopoiesis is a continuous and well-regulated process requiring both the capacity for self-renewal and the potential for differentiation of hematopoietic stem cells. Multiple studies indicate that sex hormones exert significant effects on not only hematopoietic stem and progenitor cells, but also the development of hematopoietic lineages, resulting in sexual dimorphisms in normal hematopoiesis. Hematologic malignancies comprise a wide variety of cancers affecting the blood, bone marrow, and lymphatic system, such as leukemia, lymphoma, myeloma, myelodysplastic syndrome, and myeloproliferative diseases. Overall, males are at greater risk and have worse prognosis for most of these malignancies compared with females. A better understanding of the differences between male and female could be of substantial value in research as well as clinical management.
Collapse
|
10
|
Role of Nuclear Receptors in Controlling Erythropoiesis. Int J Mol Sci 2022; 23:ijms23052800. [PMID: 35269942 PMCID: PMC8911257 DOI: 10.3390/ijms23052800] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/25/2022] [Accepted: 02/27/2022] [Indexed: 02/04/2023] Open
Abstract
Nuclear receptors (NRs), are a wide family of ligand-regulated transcription factors sharing a common modular structure composed by an N-terminal domain and a ligand-binding domain connected by a short hinge linker to a DNA-binding domain. NRs are involved in many physiological processes, including metabolism, reproduction and development. Most of them respond to small lipophilic ligands, such as steroids, retinoids, and phospholipids, which act as conformational switches. Some NRs are still "orphan" and the search for their ligands is still ongoing. Upon DNA binding, NRs can act both as transcriptional activators or repressors of their target genes. Theoretically, the possibility to modulate NRs activity with small molecules makes them ideal therapeutic targets, although the complexity of their signaling makes drug design challenging. In this review, we discuss the role of NRs in erythropoiesis, in both homeostatic and stress conditions. This knowledge is important in view of modulating red blood cells production in disease conditions, such as anemias, and for the expansion of erythroid cells in culture for research purposes and for reaching the long-term goal of cultured blood for transfusion.
Collapse
|
11
|
Epigenomic analysis of KLF1 haploinsufficiency in primary human erythroblasts. Sci Rep 2022; 12:336. [PMID: 35013432 PMCID: PMC8748495 DOI: 10.1038/s41598-021-04126-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 12/16/2021] [Indexed: 12/13/2022] Open
Abstract
Haploinsufficiency for the erythroid-specific transcription factor KLF1 is associated with hereditary persistence of fetal hemoglobin (HPFH). Increased HbF ameliorates the symptoms of β-hemoglobinopathies and downregulation of KLF1 activity has been proposed as a potential therapeutic strategy. However, the feasibility of this approach has been challenged by the observation that KLF1 haploinsufficient individuals with the same KLF1 variant, within the same family, display a wide range of HbF levels. This phenotypic variability is not readily explained by co-inheritance of known HbF-modulating variants in the HBB, HBS1L-MYB and/or BCL11A loci. We studied cultured erythroid progenitors obtained from Maltese individuals in which KLF1 p.K288X carriers display HbF levels ranging between 1.3 and 12.3% of total Hb. Using a combination of gene expression analysis, chromatin accessibility assays and promoter activity tests we find that variation in expression of the wildtype KLF1 allele may explain a significant part of the variability in HbF levels observed in KLF1 haploinsufficiency. Our results have general bearing on the variable penetrance of haploinsufficiency phenotypes and on conflicting interpretations of pathogenicity of variants in other transcriptional regulators such as EP300, GATA2 and RUNX1.
Collapse
|
12
|
Javed R, Flores L, Bhave SJ, Jawed A, Mishra DK. The Future of Red Cell Transfusion Lies in Cultured Red Cells. Indian J Med Paediatr Oncol 2021. [DOI: 10.1055/s-0041-1740068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
AbstractBlood is a very important resource for healthcare-based services and there has been a consistently increasing demand for it in most parts of the world. Poor volunteer-based collection system, high-risk of transfusion-transmitted infections, and emergence of new pathogens as evident from the ongoing Coronavirus Disease 2019 (COVID-19) pandemic are potential challenges to the global healthcare systems. It is imperative to explore safe and reliable alternatives to red cell transfusions. Ex vivo culture of red cells (cRBCs) from different sources such as hematopoietic stem cells (HSCs), pluripotent stem cells, and immortalized progenitors (e.g., BELA-2 cells) could revolutionize transfusion medicine. cRBC could be of great diagnostic and therapeutic utility. It may provide a backup in times of acute shortages in patients with rare blood groups, and in cases with multiple antibodies or sickle cell anemia. The CRISP-Cas9 system has been used to develop personalized, multi-compatible RBCs for diagnostic reagents and patients with multiple allo-antibodies. cRBC could be practically feasible for pediatric patients, who require small quantities of red cell transfusions. cRBC produced under good manufacturing practice (GMP) conditions has been reported to survive in human blood circulation for more than 26 days. Recently, a phase I randomized controlled clinical trial called RESTORE was initiated to assess the survival and recovery of cRBCs. However, feasible technological advancement is required to produce enough cRBCs for clinical use. It is crucial to identify sustainable sources for large-scale production of clinically useful cRBCs. Although the potential cost of one unit of cRBC is extrapolated to be around US$ 8000, it is a life-saving product for patients having rare blood groups and is a “ready to use” source of phenotype-matched, homogenous young red cells in emergency situations.
Collapse
Affiliation(s)
- Rizwan Javed
- Department of Clinical Haematology and BMT, TATA Medical Center, Kolkata, West Bengal, India
| | - Lorraine Flores
- Department of Histocompatibility and Immunogenetics, NHS Blood and Transplant, Filton, Bristol, United Kingdom
| | - Saurabh Jayant Bhave
- Department of Clinical Haematology and BMT, TATA Medical Center, Kolkata, West Bengal, India
| | - Asheer Jawed
- Department of Respiratory Medicine at William Harvey Hospital, Ashford, United Kingdom
| | | |
Collapse
|
13
|
Banyeh M, Amidu N, Quaye L. The relationship between 2D:4D ratio and postpartum adult female variables in a Ghanaian population. Am J Hum Biol 2021; 34:e23680. [PMID: 34523769 DOI: 10.1002/ajhb.23680] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 11/08/2022] Open
Abstract
OBJECTIVES Postpartum hematological and anthropometric assessment is a requirement for optimal maternal and child health. The study aimed to determine the relationship between the 2D:4D ratio and postpartum hematological and anthropometric variables in adult females. METHODS The study was cross-sectional from December 2020 to April 2021 involving 272 postpartum adult females, aged between 18 and 36 years. The right (2D:4DR) and the left (2D:4DL) digit ratios were measured using computer-assisted analysis. Fasting venous samples were collected at a median (interquartile range) of 111 (44-210) days postpartum and analyzed for total testosterone (TT), estradiol, sex hormone-binding globulin, and complete blood count. RESULTS The mean ± standard deviation 2D:4DR and 2D:4DL were 0.94 ± 0.04 and 0.93 ± 0.04, respectively. As expected, the TT (r = -0.198, p = .015) and the free androgen index (FAI: r = -0.186, p = .019) were inversely correlated with the 2D:4DL while free testosterone (FT%: r = -0.157, p = .038) was inversely correlated with the 2D:4DR. The absolute basophile count (BASO: r = -0.124, p = .040) and the Platelet-lymphocyte ratio (PLR: r = -0.153, p = .016) were inversely correlated with the 2D:4DL and the 2D:4DR respectively. In addition, the mean cell volume was inversely correlated with the 2D:4DR (r = -0.139, p = .024) and the 2D:4DL (r = -0.122, p = .045). Moreover, the 2D:4DR was inversely correlated with height (r = -0.164, p = .007). Unexpectedly, the red blood cell count (RBC: r = 0.138, p = .025) was positively correlated with the 2D:4DR. CONCLUSION There are significant relationships between the 2D:4D ratio and postpartum female variables. These findings are useful preliminary reference data for postpartum research and subsequent 2D:4D ratio studies.
Collapse
Affiliation(s)
- Moses Banyeh
- Department of Biomedical Laboratory Science, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
| | - Nafiu Amidu
- Department of Biomedical Laboratory Science, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
| | - Lawrence Quaye
- Department of Biomedical Laboratory Science, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
| |
Collapse
|
14
|
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: 2.7] [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
|
15
|
Identification of the transcription factor MAZ as a regulator of erythropoiesis. Blood Adv 2021; 5:3002-3015. [PMID: 34351390 DOI: 10.1182/bloodadvances.2021004609] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/11/2021] [Indexed: 12/28/2022] Open
Abstract
Erythropoiesis requires a combination of ubiquitous and tissue-specific transcription factors (TFs). Here, through DNA affinity purification followed by mass spectrometry, we have identified the widely expressed protein MAZ (Myc-associated zinc finger) as a TF that binds to the promoter of the erythroid-specific human α-globin gene. Genome-wide mapping in primary human erythroid cells revealed that MAZ also occupies active promoters as well as GATA1-bound enhancer elements of key erythroid genes. Consistent with an important role during erythropoiesis, knockdown of MAZ reduces α-globin expression in K562 cells and impairs differentiation in primary human erythroid cells. Genetic variants in the MAZ locus are associated with changes in clinically important human erythroid traits. Taken together, these findings reveal the zinc-finger TF MAZ to be a previously unrecognized regulator of the erythroid differentiation program.
Collapse
|
16
|
Chen L, Zhou D, Li X, Yang B, Xu T. Bioprinting of Human Cord Blood-Derived CD34+ Cells and Exploration of the Multilineage Differentiation Ability in Vitro. ACS Biomater Sci Eng 2021; 7:2592-2604. [PMID: 33939424 DOI: 10.1021/acsbiomaterials.0c01297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The three-dimensional (3D) marrow microenvironment plays an essential role in regulating human cord blood-derived CD34+ cells (hCB-CD34+) migration, proliferation, and differentiation. Extensive in vitro and in vivo studies have aimed to recapitulate the main components of the bone marrow (BM) niche. Nonetheless, the models are limited by a lack of heterogeneity and compound structure. Here, we fabricated coaxial extruded core-shell tubular scaffolds and extrusion-based bioprinted cell-laden mesh scaffolds to mimic the functional niche in vitro. A multicellular mesh scaffold and two different core-shell tubular scaffolds were developed with human bone marrow-derived mesenchymal stromal cells (BMSCs) in comparison with a conventional 2D coculture system. A clear cell-cell connection was established in all three bioprinted constructs. Cell distribution and morphology were observed in different systems with scanning electron microscopy (SEM). Collected hCB-CD34+ cells were characterized by various stem cell-specific and lineage-specific phenotypic parameters. The results showed that compared with hCB-CD34+ cells cocultured with BMSCs in Petri dishes, the self-renewal potential of hCB-CD34+ cells was stronger in the tubular scaffolds after 14 days. Besides, cells in these core-shell constructs tended to obtain stronger differentiation potential of lymphoid and megakaryocytes, while cells encapsulated in mesh scaffolds obtained stronger differentiation tendency into erythroid cells. Consequently, 3D bioprinting technology could partially simulate the niche of human hematopoietic stem cells. The three models have their potential in stemness maintenance and multilineage differentiation. This study can provide initial effective guidance in the directed differentiation research and related screening of drug models for hematological diseases.
Collapse
Affiliation(s)
- Lidan Chen
- Centre of Maxillofacial Surgery and Digital Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100144, People's Republic of China
| | - Dezhi Zhou
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China.,East China Institute of Digital Medical Engineering, Shangrao 334000, People's Republic of China
| | - Xinda Li
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China.,East China Institute of Digital Medical Engineering, Shangrao 334000, People's Republic of China
| | - Bin Yang
- Centre of Maxillofacial Surgery and Digital Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100144, People's Republic of China
| | - Tao Xu
- Biomanufacturing and Rapid Forming Technology Key Laboratory of Beijing, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, People's Republic of China.,Department of Precision Medicine and Healthcare, Tsinghua Berkeley Shenzhen Institute, Shenzhen 518055, People's Republic of China
| |
Collapse
|
17
|
Oxidative DNA Damage, Inflammatory Signature, and Altered Erythrocytes Properties in Diamond-Blackfan Anemia. Int J Mol Sci 2020; 21:ijms21249652. [PMID: 33348919 PMCID: PMC7768356 DOI: 10.3390/ijms21249652] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022] Open
Abstract
Molecular pathophysiology of Diamond-Blackfan anemia (DBA) involves disrupted erythroid-lineage proliferation, differentiation and apoptosis; with the activation of p53 considered as a key component. Recently, oxidative stress was proposed to play an important role in DBA pathophysiology as well. CRISPR/Cas9-created Rpl5- and Rps19-deficient murine erythroleukemia (MEL) cells and DBA patients' samples were used to evaluate proinflammatory cytokines, oxidative stress, DNA damage and DNA damage response. We demonstrated that the antioxidant defense capacity of Rp-mutant cells is insufficient to meet the greater reactive oxygen species (ROS) production which leads to oxidative DNA damage, cellular senescence and activation of DNA damage response signaling in the developing erythroblasts and altered characteristics of mature erythrocytes. We also showed that the disturbed balance between ROS formation and antioxidant defense is accompanied by the upregulation of proinflammatory cytokines. Finally, the alterations detected in the membrane of DBA erythrocytes may cause their enhanced recognition and destruction by reticuloendothelial macrophages, especially during infections. We propose that the extent of oxidative stress and the ability to activate antioxidant defense systems may contribute to high heterogeneity of clinical symptoms and response to therapy observed in DBA patients.
Collapse
|
18
|
Zebrafish Kit ligands cooperate with erythropoietin to promote erythroid cell expansion. Blood Adv 2020; 4:5915-5924. [PMID: 33259600 DOI: 10.1182/bloodadvances.2020001700] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 10/20/2020] [Indexed: 01/09/2023] Open
Abstract
Kit ligand (Kitlg) is pleiotropic cytokine with a prominent role in vertebrate erythropoiesis. Although the role of Kitlg in this process has not been reported in Danio rerio (zebrafish), in the present study we show that its function is evolutionarily conserved. Zebrafish possess 2 copies of Kitlg genes (Kitlga and Kitlgb) as a result of whole-genome duplication. To determine the role of each ligand in zebrafish, we performed a series of ex vivo and in vivo gain- and loss-of-function experiments. First, we tested the biological activity of recombinant Kitlg proteins in suspension culture from zebrafish whole-kidney marrow, and we demonstrate that Kitlga is necessary for expansion of erythroid progenitors ex vivo. To further address the role of kitlga and kitlgb in hematopoietic development in vivo, we performed gain-of-function experiments in zebrafish embryos, showing that both ligands cooperate with erythropoietin (Epo) to promote erythroid cell expansion. Finally, using the kita mutant (kitab5/b5 or sparse), we show that the Kita receptor is crucial for Kitlga/b cooperation with Epo in erythroid cells. In summary, using optimized suspension culture conditions with recombinant cytokines (Epo, Kitlga), we report, for the first time, ex vivo suspension cultures of zebrafish hematopoietic progenitor cells that can serve as an indispensable tool to study normal and aberrant hematopoiesis in zebrafish. Furthermore, we conclude that, although partial functional diversification of Kit ligands has been described in other processes, in erythroid development, both paralogs play a similar role, and their function is evolutionarily conserved.
Collapse
|
19
|
Large-scale in vitro production of red blood cells from human peripheral blood mononuclear cells. Blood Adv 2020; 3:3337-3350. [PMID: 31698463 DOI: 10.1182/bloodadvances.2019000689] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 09/09/2019] [Indexed: 12/11/2022] Open
Abstract
Transfusion of donor-derived red blood cells (RBC) is the most common form of cellular therapy. Donor availability and the potential risk of alloimmunization and other transfusion-related complications may, however, limit the availability of transfusion units, especially for chronically transfused patients. In vitro cultured, customizable RBC would negate these concerns and further increase precision medicine. Large-scale, cost-effective production depends on optimization of culture conditions. We developed a defined medium and adapted our protocols to good manufacturing practice (GMP) culture requirements, which reproducibly provided pure erythroid cultures from peripheral blood mononuclear cells without prior CD34+ isolation, and a 3 × 107-fold increase in erythroblasts in 25 days (or from 100 million peripheral blood mononuclear cells, 2 to 4 mL packed red cells can be produced). Expanded erythroblast cultures could be differentiated to CD71dimCD235a+CD44+CD117-DRAQ5- RBC in 12 days. More than 90% of the cells enucleated and expressed adult hemoglobin as well as the correct blood group antigens. Deformability and oxygen-binding capacity of cultured RBC was comparable to in vivo reticulocytes. Daily RNA sampling during differentiation followed by RNA-sequencing provided a high-resolution map/resource of changes occurring during terminal erythropoiesis. The culture process was compatible with upscaling using a G-Rex bioreactor with a capacity of 1 L per reactor, allowing transition toward clinical studies and small-scale applications.
Collapse
|
20
|
McManus JF, Nguyen NYN, Davey RA, MacLean HE, Pomilio G, McCormack MP, Chiu WS, Wei AH, Zajac JD, Curtis DJ. Androgens stimulate erythropoiesis through the DNA-binding activity of the androgen receptor in non-hematopoietic cells. Eur J Haematol 2020; 105:247-254. [PMID: 32311143 DOI: 10.1111/ejh.13431] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/10/2020] [Accepted: 04/13/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Androgens function through DNA and non-DNA binding-dependent signalling of the androgen receptor (AR). How androgens promote erythropoiesis is not fully understood. DESIGN AND METHODS To identify the androgen signalling pathway, we treated male mice lacking the second zinc finger of the DNA-binding domain of the AR (ARΔZF2 ) with non-aromatizable 5α-dihydrotestosterone (5α-DHT) or aromatizable testosterone. To distinguish direct hematopoietic and non-hematopoietic mechanisms, we performed bone marrow reconstitution experiments. RESULTS In wild-type mice, 5α-DHT had greater erythroid activity than testosterone, which can be aromatized to estradiol. The erythroid response in wild-type mice following 5α-DHT treatment was associated with increased serum erythropoietin (EPO) and its downstream target erythroferrone, and hepcidin suppression. 5α-DHT had no erythroid activity in ARΔZF2 mice, proving the importance of DNA binding by the AR. Paradoxically, testosterone, but not 5α-DHT, suppressed EPO levels in ARΔZF2 mice, suggesting testosterone following aromatization may oppose the erythroid-stimulating effects of androgens. Female wild-type mice reconstituted with ARΔZF2 bone marrow cells remained responsive to 5α-DHT. In contrast, ARΔZF2 mice reconstituted with female wild-type bone marrow cells showed no response to 5α-DHT. CONCLUSION Erythroid promoting effects of androgens are mediated through DNA binding-dependent actions of the AR in non-hematopoietic cells, including stimulating EPO expression.
Collapse
Affiliation(s)
- Julie F McManus
- Central Clinical School, Australian Centre for Blood Diseases, Monash University, Melbourne, Vic., Australia.,Human Molecular Pathology, Alfred Pathology Service, Alfred Health, Melbourne, Vic., Australia
| | - Nhu-Y N Nguyen
- Cartherics Pty Ltd, Melbourne, Vic., Australia.,Hudson Institute of Medical Research, Melbourne, Vic., Australia
| | - Rachel A Davey
- Department of Medicine, Austin Health, The University of Melbourne, Melbourne, Vic., Australia
| | - Helen E MacLean
- Department of Medicine, Austin Health, The University of Melbourne, Melbourne, Vic., Australia
| | - Giovanna Pomilio
- Central Clinical School, Australian Centre for Blood Diseases, Monash University, Melbourne, Vic., Australia.,Department of Clinical Haematology, Alfred Health, Melbourne, Vic., Australia
| | - Matthew P McCormack
- Central Clinical School, Australian Centre for Blood Diseases, Monash University, Melbourne, Vic., Australia
| | - Wan Sze Chiu
- Department of Medicine, Austin Health, The University of Melbourne, Melbourne, Vic., Australia
| | - Andrew H Wei
- Central Clinical School, Australian Centre for Blood Diseases, Monash University, Melbourne, Vic., Australia.,Department of Clinical Haematology, Alfred Health, Melbourne, Vic., Australia
| | - Jeffrey D Zajac
- Department of Medicine, Austin Health, The University of Melbourne, Melbourne, Vic., Australia
| | - David J Curtis
- Central Clinical School, Australian Centre for Blood Diseases, Monash University, Melbourne, Vic., Australia.,Department of Clinical Haematology, Alfred Health, Melbourne, Vic., Australia
| |
Collapse
|
21
|
Bernecker C, Köfeler H, Pabst G, Trötzmüller M, Kolb D, Strohmayer K, Trajanoski S, Holzapfel GA, Schlenke P, Dorn I. Cholesterol Deficiency Causes Impaired Osmotic Stability of Cultured Red Blood Cells. Front Physiol 2019; 10:1529. [PMID: 31920725 PMCID: PMC6933518 DOI: 10.3389/fphys.2019.01529] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 12/04/2019] [Indexed: 11/17/2022] Open
Abstract
Ex vivo generation of red blood cells (cRBCs) is an attractive tool in basic research and for replacing blood components donated by volunteers. As a prerequisite for the survival of cRBCs during storage as well as in the circulation, the quality of the membrane is of utmost importance. Besides the cytoskeleton and embedded proteins, the lipid bilayer is critical for membrane integrity. Although cRBCs suffer from increased fragility, studies investigating the lipid content of their membrane are still lacking. We investigated the membrane lipid profile of cRBCs from CD34+ human stem and progenitor cells compared to native red blood cells (nRBCs) and native reticulocytes (nRETs). Ex vivo erythropoiesis was performed in a well-established liquid assay. cRBCs showed a maturation grade between nRETs and nRBCs. High-resolution mass spectrometry analysis for cholesterol and the major phospholipid classes, phosphatidylcholine, phosphatidylethanolamine, phosphatidylinositol, phosphatidylserine, sphingomyelin and lysophosphatidylcholin, demonstrated severe cholesterol deficiency in cRBCs. Although cRBCs showed normal deformability capacity, they suffered from increased hemolysis due to minimal changes in the osmotic conditions. After additional lipid supplementation, especially cholesterol during culturing, the cholesterol content of cRBCs increased to a subnormal amount. Concurrently, the osmotic resistance recovered completely and became comparable to that of nRETs. Minor differences in the amount of phospholipids in cRBCs compared to native cells could mainly be attributed to the ongoing membrane remodeling process from the reticulocyte to the erythrocyte stage. Obtained results demonstrate severe cholesterol deficiency as a reason for enhanced fragility of cRBCs. Therefore, the supplementation of lipids, especially cholesterol during ex vivo erythropoiesis may overcome this limitation and strengthens the survival of cRBCs ex vivo and in vivo.
Collapse
Affiliation(s)
- Claudia Bernecker
- Department for Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria
| | - Harald Köfeler
- Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Georg Pabst
- Institute of Molecular Biosciences, University of Graz, Biophysics Division, BioTechMed Graz, Graz, Austria
| | | | - Dagmar Kolb
- Center for Medical Research, Medical University of Graz, Graz, Austria.,Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Karl Strohmayer
- Institute of Biomechanics, Graz University of Technology, Graz, Austria
| | - Slave Trajanoski
- Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Gerhard A Holzapfel
- Institute of Biomechanics, Graz University of Technology, Graz, Austria.,Department of Structural Engineering, Norwegian University of Science and Technology, Trondheim, Norway
| | - Peter Schlenke
- Department for Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria
| | - Isabel Dorn
- Department for Blood Group Serology and Transfusion Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
22
|
Fibach E. Erythropoiesis In Vitro-A Research and Therapeutic Tool in Thalassemia. J Clin Med 2019; 8:jcm8122124. [PMID: 31810354 PMCID: PMC6947291 DOI: 10.3390/jcm8122124] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 11/26/2019] [Accepted: 11/29/2019] [Indexed: 12/13/2022] Open
Abstract
Thalassemia (thal) is a hereditary chronic hemolytic anemia due to a partial or complete deficiency in the production of globin chains, in most cases, α or β, which compose, together with the iron-containing porphyrins (hemes), the hemoglobin molecules in red blood cells (RBC). The major clinical symptom of β-thal is severe chronic anemia—a decrease in RBC number and their hemoglobin content. In spite of the improvement in therapy, thal still severely affects the quality of life of the patients and their families and imposes a substantial financial burden on the community. These considerations position β-thal, among other hemoglobinopathies, as a major health and social problem that deserves increased efforts in research and its clinical application. These efforts are based on clinical studies, experiments in animal models and the use of erythroid cells grown in culture. The latter include immortal cell lines and cultures initiated by erythroid progenitor and stem cells derived from the blood and RBC producing (erythropoietic) sites of normal and thal donors, embryonic stem cells, and recently, "induced pluripotent stem cells" generated by manipulation of differentiated somatic cells. The present review summarizes the use of erythroid cultures, their technological aspects and their contribution to the research and its clinical application in thal. The former includes deciphering of the normal and pathological biology of the erythroid cell development, and the latter—their role in developing innovative therapeutics—drugs and methods of gene therapy, as well as providing an alternative source of RBC that may complement or substitute blood transfusions.
Collapse
Affiliation(s)
- Eitan Fibach
- The Hematology Department, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel
| |
Collapse
|
23
|
Klei TRL, Aglialoro F, Mul FPJ, Tol S, Ligthart PC, Seignette IM, Geissler J, van den Akker E, van Bruggen R. Differential interaction between DARC and SDF-1 on erythrocytes and their precursors. Sci Rep 2019; 9:16245. [PMID: 31700087 PMCID: PMC6838059 DOI: 10.1038/s41598-019-52186-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 09/26/2019] [Indexed: 01/21/2023] Open
Abstract
The Duffy Antigen Receptor for Chemokines (DARC) is expressed on erythrocytes and on endothelium of postcapillary venules and splenic sinusoids. Absence of DARC on erythrocytes, but not on endothelium, is referred to as the Duffy negative phenotype and is associated with neutropenia. Here we provide evidence that stromal cell-derived factor 1 (SDF-1), the chemokine that restricts neutrophil precursors to the bone marrow, binds to erythrocyte progenitors in a DARC-dependent manner. Furthermore, we show that SDF-1 binding to DARC is dependent on the conformation of DARC, which gradually changes during erythroid development, resulting in the absence of SDF-1 binding to mature erythrocytes. However, SDF-1 binding to erythrocytes was found to be inducible by pre-treating erythrocytes with IL-8 or with antibodies recognizing specific epitopes on DARC. Taken together, these novel findings identify DARC on erythrocyte precursors as a receptor for SDF-1, which may be of interest in beginning to understand the development of neutropenia in situations where DARC expression is limited.
Collapse
Affiliation(s)
- T R L Klei
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, The Netherlands
| | - F Aglialoro
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, Amsterdam, The Netherlands
| | - F P J Mul
- Research facility, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, The Netherlands
| | - S Tol
- Research facility, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, The Netherlands
| | - P C Ligthart
- Erythrocyte Serology, Sanquin, Amsterdam, The Netherlands
| | - I M Seignette
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, The Netherlands
| | - J Geissler
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, The Netherlands
| | - E van den Akker
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, Amsterdam, The Netherlands
| | - R van Bruggen
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, Amsterdam, The Netherlands.
| |
Collapse
|
24
|
Hansen M, von Lindern M, van den Akker E, Varga E. Human‐induced pluripotent stem cell‐derived blood products: state of the art and future directions. FEBS Lett 2019; 593:3288-3303. [DOI: 10.1002/1873-3468.13599] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/13/2019] [Accepted: 08/14/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Marten Hansen
- Department of Hematopoiesis, Sanquin Research, and Landsteiner Laboratory Academic Medical Center University of Amsterdam The Netherlands
| | - Marieke von Lindern
- Department of Hematopoiesis, Sanquin Research, and Landsteiner Laboratory Academic Medical Center University of Amsterdam The Netherlands
| | - Emile van den Akker
- Department of Hematopoiesis, Sanquin Research, and Landsteiner Laboratory Academic Medical Center University of Amsterdam The Netherlands
| | - Eszter Varga
- Department of Hematopoiesis, Sanquin Research, and Landsteiner Laboratory Academic Medical Center University of Amsterdam The Netherlands
| |
Collapse
|
25
|
Zingariello M, Bardelli C, Sancillo L, Ciaffoni F, Genova ML, Girelli G, Migliaccio AR. Dexamethasone Predisposes Human Erythroblasts Toward Impaired Lipid Metabolism and Renders Their ex vivo Expansion Highly Dependent on Plasma Lipoproteins. Front Physiol 2019; 10:281. [PMID: 31019464 PMCID: PMC6458278 DOI: 10.3389/fphys.2019.00281] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 03/04/2019] [Indexed: 02/04/2023] Open
Abstract
Cultures of stem cells from discarded sources supplemented with dexamethasone, a synthetic glucocorticoid receptor agonist, generate cultured red blood cells (cRBCs) in numbers sufficient for transfusion. According to the literature, however, erythroblasts generated with dexamethasone exhibit low enucleation rates giving rise to cRBCs that survive poorly in vivo. The knowledge that the glucocorticoid receptor regulates lipid metabolism and that lipid composition dictates the fragility of the plasma membrane suggests that insufficient lipid bioavailability restrains generation of cRBCs. To test this hypothesis, we first compared the expression profiling of erythroblasts generated with or without dexamethasone. This analysis revealed differences in expression of 55 genes, 6 of which encoding proteins involved in lipid metabolism. These were represented by genes encoding the mitochondrial proteins 3-Hydroxymethyl-3-Methylglutaryl-CoA lyase, upregulated, and 3-Oxoacid CoA-Transferase1 and glycerol-3-phosphate acyltransferase1, both downregulated, and the proteins ATP-binding cassette transporter1 and Hydroxysteroid-17-Beta-Dehydrogenase7, upregulated, and cAMP-dependent protein kinase catalytic subunit beta, downregulated. This profiling predicts that dexamethasone, possibly by interfering with mitochondrial functions, impairs the intrinsic lipid metabolism making the synthesis of the plasma membrane of erythroid cells depend on lipid-uptake from external sources. Optical and electron microscopy analyses confirmed that the mitochondria of erythroblasts generated with dexamethasone are abnormal and that their plasma membranes present pebbles associated with membrane ruptures releasing exosomes and micro-vesicles. These results indicate that the lipid supplements of media currently available are not adequate for cRBCs. To identify better lipid supplements, we determined the number of erythroblasts generated in synthetic media supplemented with either currently used liposomes or with lipoproteins purified from human plasma [the total lipoprotein fraction (TL) or its high (HDL), low (LDL) and very low (VLDL) density lipoprotein components]. Both LDL and VLDL generated numbers of erythroid cells 3-2-fold greater than that observed in controls. These greater numbers were associated with 2-3-fold greater amplification of erythroid cells due both to increased proliferation and to resistance to stress-induced death. In conclusion, dexamethasone impairs lipid metabolism making ex vivo expansion of erythroid cells highly dependent on lipid absorbed from external sources and the use of LDL and VLDL as lipid supplements improves the generation of cRBCs.
Collapse
Affiliation(s)
- Maria Zingariello
- Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Claudio Bardelli
- Department of Biomedical and NeuroMotor Sciences, Alma Mater Studiorum University, Bologna, Italy
| | - Laura Sancillo
- Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | | | - Maria Luisa Genova
- Department of Biomedical and NeuroMotor Sciences, Alma Mater Studiorum University, Bologna, Italy
| | | | - Anna Rita Migliaccio
- Department of Biomedical and NeuroMotor Sciences, Alma Mater Studiorum University, Bologna, Italy
| |
Collapse
|
26
|
Activation of the vitamin D receptor transcription factor stimulates the growth of definitive erythroid progenitors. Blood Adv 2019; 2:1207-1219. [PMID: 29844206 DOI: 10.1182/bloodadvances.2018017533] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 04/15/2018] [Indexed: 12/30/2022] Open
Abstract
The pathways that regulate the growth of erythroid progenitors are incompletely understood. In a computational analysis of gene expression changes during erythroid ontogeny, the vitamin D receptor (Vdr) nuclear hormone receptor transcription factor gene was identified in fetal and adult stages, but not at the embryonic stage of development. Vdr was expressed in definitive erythroid (EryD) progenitors and was downregulated during their maturation. Activation of Vdr signaling by the vitamin D3 agonist calcitriol increased the outgrowth of EryD colonies from fetal liver and adult bone marrow, maintained progenitor potential, and delayed erythroid maturation, as revealed by clonogenic assays, suspension culture, cell surface phenotype, and gene expression analyses. The early (cKit+CD71lo/neg), but not the late (cKit+CD71hi), EryD progenitor subset of LinnegcKit+ cells was responsive to calcitriol. Culture of cKit+CD71lo/neg progenitors in the presence of both vitamin D3 and glucocorticoid receptor ligands resulted in an increase in proliferation that was at least additive compared with either ligand alone. Lentivirus shRNA-mediated knockdown of Vdr expression abrogated the stimulation of early erythroid progenitor growth by calcitriol. These findings suggest that Vdr has a cell-intrinsic function in early erythroid progenitors. Targeting of downstream components of the Vdr signaling pathway may lead to new approaches for the expansion of erythroid progenitors ex vivo.
Collapse
|
27
|
Allenby MC, Panoskaltsis N, Tahlawi A, Dos Santos SB, Mantalaris A. Dynamic human erythropoiesis in a three-dimensional perfusion bone marrow biomimicry. Biomaterials 2019; 188:24-37. [DOI: 10.1016/j.biomaterials.2018.08.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 08/06/2018] [Indexed: 12/21/2022]
|
28
|
Generation of clinical-grade red blood cells from human umbilical cord blood mononuclear cells. Cell Tissue Res 2018; 375:437-449. [PMID: 30284087 DOI: 10.1007/s00441-018-2919-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 09/03/2018] [Indexed: 01/18/2023]
Abstract
A xeno-free method for ex vivo generation of red blood cells (RBCs) is attempted in order to replicate for large-scale production and clinical applications. An efficient milieu was formulated using injectable drugs substituting the animal-derived components in the culture medium. Unfractionated mononuclear cells isolated from human umbilical cord blood were used hypothesizing that the heterogeneous cell population could effectively contribute to erythroid cell generation. The strategy adopted includes a combination of erythropoietin and other injectable drugs under low oxygen levels, which resulted in an increase in the number of mature RBCs produced in vitro. The novelty in this study is the addition of supplements to the medium in a stage-specific manner for the differentiation of unfractionated umbilical cord blood mononuclear cells (MNCs) into erythropoietic lineage. The erythropoietic lineage was well established by day 21, wherein the mean cell count of RBCs was found to be 21.36 ± 0.9 × 108 and further confirmed by an upregulated expression of CD235a+ specific to RBCs. The rationale was to have a simple method to produce erythroid cells from umbilical cord blood isolates in vitro by mitigating the effects of multiple erythroid-activating agents and batch to batch variability.
Collapse
|
29
|
Lee E, Sivalingam J, Lim ZR, Chia G, Shi LG, Roberts M, Loh YH, Reuveny S, Oh SKW. Review: In vitro generation of red blood cells for transfusion medicine: Progress, prospects and challenges. Biotechnol Adv 2018; 36:2118-2128. [PMID: 30273713 DOI: 10.1016/j.biotechadv.2018.09.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 09/19/2018] [Accepted: 09/27/2018] [Indexed: 02/07/2023]
Abstract
In vitro generation of red blood cells (RBCs) has the potential to circumvent the shortfalls in global demand for blood for transfusion applications. The conventional approach for RBC generation has been from differentiation of hematopoietic stem cells (HSCs) derived from cord blood, adult bone marrow or peripheral blood. More recently, RBCs have been generated from human induced pluripotent stem cells (hiPSCs) as well as from immortalized adult erythroid progenitors. In this review, we highlight the recent advances to RBC generation from these different approaches and discuss the challenges and new strategies that can potentially make large-scale in vitro generation of RBCs a feasible approach.
Collapse
Affiliation(s)
- Esmond Lee
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore 138668, Republic of Singapore; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Palo Alto, CA 94305, USA.
| | - Jaichandran Sivalingam
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore 138668, Republic of Singapore.
| | - Zhong Ri Lim
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore 138668, Republic of Singapore
| | - Gloryn Chia
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore 138668, Republic of Singapore
| | - Low Gin Shi
- Brilliant Research Pte. Ltd, Singapore 139955, Republic of Singapore
| | - Mackenna Roberts
- Oxford-University College London Centre for the Advancement of Sustainable Medical Innovation, University of Oxford, UK
| | - Yuin-Han Loh
- Institute of Molecular and Cellular Biology, Agency for Science, Technology and Research, Singapore 138668, Republic of Singapore; Department of Biological Sciences, National University of Singapore, Singapore 117543, Republic of Singapore
| | - Shaul Reuveny
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore 138668, Republic of Singapore
| | - Steve Kah-Weng Oh
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore 138668, Republic of Singapore
| |
Collapse
|
30
|
Mutant ASXL1 cooperates with BAP1 to promote myeloid leukaemogenesis. Nat Commun 2018; 9:2733. [PMID: 30013160 PMCID: PMC6048047 DOI: 10.1038/s41467-018-05085-9] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 06/13/2018] [Indexed: 12/27/2022] Open
Abstract
ASXL1 mutations occur frequently in myeloid neoplasms and are associated with poor prognosis. However, the mechanisms by which mutant ASXL1 induces leukaemogenesis remain unclear. In this study, we report mutually reinforcing effects between a C-terminally truncated form of mutant ASXL1 (ASXL1-MT) and BAP1 in promoting myeloid leukaemogenesis. BAP1 expression results in increased monoubiquitination of ASXL1-MT, which in turn increases the catalytic function of BAP1. This hyperactive ASXL1-MT/BAP1 complex promotes aberrant myeloid differentiation of haematopoietic progenitor cells and accelerates RUNX1-ETO-driven leukaemogenesis. Mechanistically, this complex induces upregulation of posterior HOXA genes and IRF8 through removal of H2AK119 ubiquitination. Importantly, BAP1 depletion inhibits posterior HOXA gene expression and leukaemogenicity of ASXL1-MT-expressing myeloid leukemia cells. Furthermore, BAP1 is also required for the growth of MLL-fusion leukemia cells with posterior HOXA gene dysregulation. These data indicate that BAP1, which has long been considered a tumor suppressor, in fact plays tumor-promoting roles in myeloid neoplasms. ASXL1 gene is often mutated in myeloid malignancies. Here, the authors show that mutant ASXL1 and BAP1 are in a positive feedback loop such that BAP1 induces monoubiquitination of mutant ASXL1, which in turn enhances BAP1 activity to potentiate myeloid transformation via HOXA clusters and IRF8.
Collapse
|
31
|
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: 6.7] [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
|
32
|
Efficient production of erythroid, megakaryocytic and myeloid cells, using single cell-derived iPSC colony differentiation. Stem Cell Res 2018; 29:232-244. [PMID: 29751281 DOI: 10.1016/j.scr.2018.04.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 04/10/2018] [Accepted: 04/26/2018] [Indexed: 12/16/2022] Open
Abstract
Hematopoietic differentiation of human induced pluripotent stem cells (iPSCs) provide opportunities not only for fundamental research and disease modelling/drug testing but also for large-scale production of blood effector cells for future clinical application. Although there are multiple ways to differentiate human iPSCs towards hematopoietic lineages, there is a need to develop reproducible and robust protocols. Here we introduce an efficient way to produce three major blood cell types using a standardized differentiation protocol that starts with a single hematopoietic initiation step. This system is feeder-free, avoids EB-formation, starts with a hematopoietic initiation step based on a novel single cell-derived iPSC colony differentiation and produces multi-potential progenitors within 8-10 days. Followed by lineage-specific growth factor supplementation these cells can be matured into well characterized erythroid, megakaryocytic and myeloid cells with high-purity, without transcription factor overexpression or any kind of pre-purification step. This standardized differentiation system provides a simple platform to produce specific blood cells in a reproducible manner for hematopoietic development studies, disease modelling, drug testing and the potential for future therapeutic applications.
Collapse
|
33
|
Minetti G, Achilli C, Perotti C, Ciana A. Continuous Change in Membrane and Membrane-Skeleton Organization During Development From Proerythroblast to Senescent Red Blood Cell. Front Physiol 2018; 9:286. [PMID: 29632498 PMCID: PMC5879444 DOI: 10.3389/fphys.2018.00286] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 03/12/2018] [Indexed: 12/11/2022] Open
Abstract
Within the context of erythropoiesis and the possibility of producing artificial red blood cells (RBCs) in vitro, a most critical step is the final differentiation of enucleated erythroblasts, or reticulocytes, to a fully mature biconcave discocyte, the RBC. Reviewed here is the current knowledge about this fundamental maturational process. By combining literature data with our own experimental evidence we propose that the early phase in the maturation of reticulocytes to RBCs is driven by a membrane raft-based mechanism for the sorting of disposable membrane proteins, mostly the no longer needed transferrin receptor (TfR), to the multivesicular endosome (MVE) as cargo of intraluminal vesicles that are subsequently exocytosed as exosomes, consistently with the seminal and original observation of Johnstone and collaborators of more than 30 years ago (Pan BT, Johnstone RM. Cell. 1983;33:967-978). According to a strikingly selective sorting process, the TfR becomes cargo destined to exocytosis while other molecules, including the most abundant RBC transmembrane protein, band 3, are completely retained in the cell membrane. It is also proposed that while this process could be operating in the early maturational steps in the bone marrow, additional mechanism(s) must be at play for the final removal of the excess reticulocyte membrane that is observed to occur in the circulation. This processing will most likely require the intervention of the spleen, whose function is also necessary for the continuous remodeling of the RBC membrane all along this cell's circulatory life.
Collapse
Affiliation(s)
- Giampaolo Minetti
- Laboratori di Biochimica, Dipartimento di Biologia e Biotecnologie, Università degli Studi di Pavia, Pavia, Italy
| | - Cesare Achilli
- Laboratori di Biochimica, Dipartimento di Biologia e Biotecnologie, Università degli Studi di Pavia, Pavia, Italy
| | - Cesare Perotti
- Servizio Immunoematologia e Medicina Trasfusionale, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Annarita Ciana
- Laboratori di Biochimica, Dipartimento di Biologia e Biotecnologie, Università degli Studi di Pavia, Pavia, Italy
| |
Collapse
|
34
|
Migliaccio AR, Varricchio L. Concise Review: Advanced Cell Culture Models for Diamond Blackfan Anemia and Other Erythroid Disorders. Stem Cells 2018; 36:172-179. [PMID: 29124822 PMCID: PMC5785423 DOI: 10.1002/stem.2735] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/14/2017] [Accepted: 11/01/2017] [Indexed: 01/26/2023]
Abstract
In vitro surrogate models of human erythropoiesis made many contributions to our understanding of the extrinsic and intrinsic regulation of this process in vivo and how they are altered in erythroid disorders. In the past, variability among the levels of hemoglobin F produced by adult erythroblasts generated in vitro by different laboratories identified stage of maturation, fetal bovine serum, and accessory cells as "confounding factors," that is, parameters intrinsically wired in the experimental approach that bias the results observed. The discovery of these factors facilitated the identification of drugs that accelerate terminal maturation or activate specific signaling pathways for the treatment of hemoglobinopathies. It also inspired studies to understand how erythropoiesis is regulated by macrophages present in the erythroid islands. Recent cell culture advances have greatly increased the number of human erythroid cells that can be generated in vitro and are used as experimental models to study diseases, such as Diamond Blackfan Anemia, which were previously poorly amenable to investigation. However, in addition to the confounding factors already identified, improvement in the culture models has introduced novel confounding factors, such as possible interactions between signaling from cKIT, the receptor for stem cell factor, and from the glucocorticoid receptor, the cell proliferation potential and the clinical state of the patients. This review will illustrate these new confounding factors and discuss their clinical translation potential to improve our understanding of Diamond Blackfan Anemia and other erythroid disorders. Stem Cells 2018;36:172-179.
Collapse
Affiliation(s)
- Anna Rita Migliaccio
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY, USA
- Department of Biomedical and Neuromotorial Sciences, Alma Mater University, Bologna, Italy
| | - Lilian Varricchio
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY, USA
| |
Collapse
|
35
|
|
36
|
Heideveld E, Hampton-O'Neil LA, Cross SJ, van Alphen FPJ, van den Biggelaar M, Toye AM, van den Akker E. Glucocorticoids induce differentiation of monocytes towards macrophages that share functional and phenotypical aspects with erythroblastic island macrophages. Haematologica 2017; 103:395-405. [PMID: 29284682 PMCID: PMC5830394 DOI: 10.3324/haematol.2017.179341] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 12/27/2017] [Indexed: 12/14/2022] Open
Abstract
The classical central macrophage found in erythroblastic islands plays an important role in erythroblast differentiation, proliferation and enucleation in the bone marrow. Convenient human in vitro models to facilitate the study of erythroid-macrophage interactions are desired. Recently, we demonstrated that cultured monocytes/macrophages enhance in vitro erythropoiesis by supporting hematopoietic stem and progenitor cell survival. Herein, we describe that these specific macrophages also support erythropoiesis. Human monocytes cultured in serum-free media supplemented with stem cell factor, erythropoietin, lipids and dexamethasone differentiate towards macrophages expressing CD16, CD163, CD169, CD206, CXCR4 and the phagocytic TAM-receptor family. Phenotypically, they resemble both human bone marrow and fetal liver resident macrophages. This differentiation is dependent on glucocorticoid receptor activation. Proteomic studies confirm that glucocorticoid receptor activation differentiates monocytes to anti-inflammatory tissue macrophages with a M2 phenotype, termed GC-macrophages. Proteins involved in migration, tissue residence and signal transduction/receptor activity are upregulated whilst lysosome and hydrolase activity GO-categories are downregulated. Functionally, we demonstrate that GC-macrophages are highly mobile and can interact to form clusters with erythroid cells of all differentiation stages and phagocytose the expelled nuclei, recapitulating aspects of erythroblastic islands. In conclusion, glucocorticoid-directed monocyte differentiation to macrophages represents a convenient model system to study erythroid-macrophage interactions.
Collapse
Affiliation(s)
- Esther Heideveld
- Sanquin Research, Department of Hematopoiesis, Amsterdam and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, the Netherlands
| | | | - Stephen J Cross
- Wolfson Bioimaging Facility, School of Medical Sciences, Bristol, UK
| | | | - Maartje van den Biggelaar
- Sanquin Research, Department of Research Facilities, Amsterdam, the Netherlands.,Sanquin Research, Department of Plasma Proteins, Amsterdam, the Netherlands
| | - Ashley M Toye
- Department of Biochemistry, School of Medical Sciences, Bristol, UK.,Bristol Institute for Transfusion Sciences, NHS Blood and Transplant, Filton, Bristol, UK.,National Institute for Health Research (NIHR) Blood and Transplant Research Unit in Red Blood Cell Products, University of Bristol, UK
| | - Emile van den Akker
- Sanquin Research, Department of Hematopoiesis, Amsterdam and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, the Netherlands
| |
Collapse
|
37
|
Dai Y, Chen T, Ijaz H, Cho EH, Steinberg MH. SIRT1 activates the expression of fetal hemoglobin genes. Am J Hematol 2017; 92:1177-1186. [PMID: 28776729 DOI: 10.1002/ajh.24879] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 07/31/2017] [Indexed: 02/06/2023]
Abstract
High fetal hemoglobin (HbF, α2 γ2 ) levels ameliorate the clinical manifestations of sickle cell disease and β thalassemia. The mechanisms that repress HbF expression and silence γ-globin genes in adults are incompletely characterized and only a single HbF inducer, hydroxyurea, is approved for treatment, and only in patients with sickle cell disease. We identified SIRT1, a protein deacetylase, as a new inducer of γ-globin. SIRT1 knockdown decreased, while SIRT1 ectopic expression upregulated γ-globin gene (HBG) expression in primary human erythroid cells and in K562 cells. The small molecule SIRT1 activators SRT2104 and SRT1720 enhanced HBG expression in cord blood human erythroblasts and reactivated silenced HBG in adult human erythroblasts. Furthermore, SIRT1 binds in the β-globin gene cluster locus control region (LCR) and HBG promoters, promotes the looping of the LCR to HBG promoter, and increases the binding of RNA polymerase II and H4K16Ac in the HBG promoter. SIRT1 suppressed the expression of the HBG suppressors BCL11A, KLF1, HDAC1 and HDAC2. Lastly, SIRT1 did not change the proliferation of human erythroid progenitor cells or the expression of differentiation marker CD235a. These data suggest that SIRT1 activates HBG expression through facilitating LCR looping to the HBG promoter, inhibiting the expression of transcriptional suppressors of HBG, and indirectly increasing histone acetylation in the HBG promoter. SIRT1 is a potential therapeutic target for γ-globin gene induction, and small molecule SIRT1 activators might serve as a lead compound for the development of new HbF inducers.
Collapse
Affiliation(s)
- Yan Dai
- Department of Medicine; Boston University School of Medicine; Boston Massachusetts 02118
| | - Tyngwei Chen
- Department of Medicine; Boston University School of Medicine; Boston Massachusetts 02118
| | - Heba Ijaz
- Department of Medicine; Boston University School of Medicine; Boston Massachusetts 02118
| | - Elizabeth H. Cho
- Department of Medicine; Boston University School of Medicine; Boston Massachusetts 02118
| | - Martin H. Steinberg
- Department of Medicine; Boston University School of Medicine; Boston Massachusetts 02118
| |
Collapse
|
38
|
Chondrou V, Kolovos P, Sgourou A, Kourakli A, Pavlidaki A, Kastrinou V, John A, Symeonidis A, Ali BR, Papachatzopoulou A, Katsila T, Patrinos GP. Whole transcriptome analysis of human erythropoietic cells during ontogenesis suggests a role of VEGFA gene as modulator of fetal hemoglobin and pharmacogenomic biomarker of treatment response to hydroxyurea in β-type hemoglobinopathy patients. Hum Genomics 2017; 11:24. [PMID: 29061162 PMCID: PMC5654038 DOI: 10.1186/s40246-017-0120-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 10/16/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Human erythropoiesis is characterized by distinct gene expression profiles at various developmental stages. Previous studies suggest that fetal-to-adult hemoglobin switch is regulated by a complex mechanism, in which many key players still remain unknown. Here, we report our findings from whole transcriptome analysis of erythroid cells, isolated from erythroid tissues at various developmental stages in an effort to identify distinct molecular signatures of each erythroid tissue. RESULTS From our in-depth data analysis, pathway analysis, and text mining, we opted to focus on the VEGFA gene, given its gene expression characteristics. Selected VEGFA genomic variants, identified through linkage disequilibrium analysis, were explored further for their association with elevated fetal hemoglobin levels in β-type hemoglobinopathy patients. Our downstream analysis of non-transfusion-dependent β-thalassemia patients, β-thalassemia major patients, compound heterozygous sickle cell disease/β-thalassemia patients receiving hydroxyurea as fetal hemoglobin augmentation treatment, and non-thalassemic individuals indicated that VEGFA genomic variants were associated with disease severity in β-thalassemia patients and hydroxyurea treatment efficacy in SCD/β-thalassemia compound heterozygous patients. CONCLUSIONS Our findings suggest that VEGFA may act as a modifier gene of human globin gene expression and, at the same time, serve as a genomic biomarker in β-type hemoglobinopathy disease severity and hydroxyurea treatment efficacy.
Collapse
Affiliation(s)
- Vasiliki Chondrou
- Department of Pharmacy, School of Health Sciences, University of Patras, University Campus, Rion, GR-265 04, Patras, Greece
| | - Petros Kolovos
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | | | - Alexandra Kourakli
- Hematology Division, Department of Internal Medicine, Faculty of Medicine, University of Patras, Patras, Greece
| | - Alexia Pavlidaki
- Department of Pharmacy, School of Health Sciences, University of Patras, University Campus, Rion, GR-265 04, Patras, Greece.,Present address: Institut de Génétique et de Biologie Moléculaire et Cellulaire IGBMC/CNRS/INSERM/UDS, 67404 ILLKIRCH, BP 10142, CU de Strasbourg, France
| | - Vlasia Kastrinou
- Department of Pharmacy, School of Health Sciences, University of Patras, University Campus, Rion, GR-265 04, Patras, Greece
| | - Anne John
- Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Argiris Symeonidis
- Hematology Division, Department of Internal Medicine, Faculty of Medicine, University of Patras, Patras, Greece
| | - Bassam R Ali
- Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates.,Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | | | - Theodora Katsila
- Department of Pharmacy, School of Health Sciences, University of Patras, University Campus, Rion, GR-265 04, Patras, Greece
| | - George P Patrinos
- Department of Pharmacy, School of Health Sciences, University of Patras, University Campus, Rion, GR-265 04, Patras, Greece. .,Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates. .,Zayed Bin Sultan Center for Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates.
| |
Collapse
|
39
|
Severn CE, Toye AM. The challenge of growing enough reticulocytes for transfusion. ACTA ACUST UNITED AC 2017. [DOI: 10.1111/voxs.12374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- C. E. Severn
- School of Biochemistry; Biomedical Sciences Building; University Walk; University of Bristol; Bristol UK
- National Institute for Health Research (NIHR) Blood and Transplant Research Unit; University of Bristol; Bristol UK
| | - A. M. Toye
- School of Biochemistry; Biomedical Sciences Building; University Walk; University of Bristol; Bristol UK
- National Institute for Health Research (NIHR) Blood and Transplant Research Unit; University of Bristol; Bristol UK
- Bristol Institute of Transfusion Sciences; NHSBT Filton; Bristol UK
| |
Collapse
|
40
|
van Gucht ALM, Meima ME, Moran C, Agostini M, Tylki-Szymanska A, Krajewska M, Walasek, Chrzanowska K, Efthymiadou A, Chrysis D, Demir K, Visser WE, Visser TJ, Chatterjee K, van Dijk TB, Peeters RP. Anemia in Patients With Resistance to Thyroid Hormone α: A Role for Thyroid Hormone Receptor α in Human Erythropoiesis. J Clin Endocrinol Metab 2017; 102:3517-3525. [PMID: 28911146 PMCID: PMC5587074 DOI: 10.1210/jc.2017-00840] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 07/03/2017] [Indexed: 11/25/2022]
Abstract
CONTEXT Patients with resistance to thyroid hormone (TH) α (RTHα) are characterized by growth retardation, macrocephaly, constipation, and abnormal thyroid function tests. In addition, almost all RTHα patients have mild anemia, the pathogenesis of which is unknown. Animal studies suggest an important role for TH and TH receptor (TR)α in erythropoiesis. OBJECTIVE To investigate whether a defect in TRα affects the maturation of red blood cells in RTHα patients. DESIGN, SETTING, AND PATIENTS Cultures of primary human erythroid progenitor cells (HEPs), from peripheral blood of RTHα patients (n = 11) harboring different inactivating mutations in TRα (P398R, F397fs406X, C392X, R384H, A382fs388X, A263V, A263S), were compared with healthy controls (n = 11). During differentiation, erythroid cells become smaller, accumulate hemoglobin, and express different cell surface markers. We assessed cell number and cell size, and used cell staining and fluorescence-activated cell sorter analysis to monitor maturation at different time points. RESULTS After ∼14 days of ex vivo expansion, both control and patient-derived progenitors differentiated spontaneously. However, RTHα-derived cells differentiated more slowly. During spontaneous differentiation, RTHα-derived HEPs were larger, more positive for c-Kit (a proliferation marker), and less positive for glycophorin A (a differentiation marker). The degree of abnormal spontaneous maturation of RTHα-derived progenitors did not correlate with severity of underlying TRα defect. Both control and RTHα-derived progenitors responded similarly when differentiation was induced. T3 exposure accelerated differentiation of both control- and RTHα patient-derived HEPs. CONCLUSIONS Inactivating mutations in human TRα affect the balance between proliferation and differentiation of progenitor cells during erythropoiesis, which may contribute to the mild anemia seen in most RTHα patients.
Collapse
Affiliation(s)
- Anja L. M. van Gucht
- Department of Internal Medicine, Erasmus University Medical Center, 3000 Rotterdam, The Netherlands
| | - Marcel E. Meima
- Department of Internal Medicine, Erasmus University Medical Center, 3000 Rotterdam, The Netherlands
| | - Carla Moran
- Wellcome–Medical Research Council Institute of Metabolic Science, University of Cambridge, CB2 0QQ Cambridge, United Kingdom
| | - Maura Agostini
- Wellcome–Medical Research Council Institute of Metabolic Science, University of Cambridge, CB2 0QQ Cambridge, United Kingdom
| | | | | | - Walasek
- The Children’s Memorial Health Institute, 04-730 Warsaw, Poland
| | | | - Alexandra Efthymiadou
- Department of Pediatrics, Division of Endocrinology, University of Patras Medical School, 25002 Patras, Greece
| | - Dionisios Chrysis
- Department of Pediatrics, Division of Endocrinology, University of Patras Medical School, 25002 Patras, Greece
| | - Korcan Demir
- Division of Pediatric Endocrinology, Faculty of Medicine, Dokuz Eylül University, 35100 Izmir, Turkey
| | - W. Edward Visser
- Department of Internal Medicine, Erasmus University Medical Center, 3000 Rotterdam, The Netherlands
| | - Theo J. Visser
- Department of Internal Medicine, Erasmus University Medical Center, 3000 Rotterdam, The Netherlands
| | - Krishna Chatterjee
- Wellcome–Medical Research Council Institute of Metabolic Science, University of Cambridge, CB2 0QQ Cambridge, United Kingdom
| | - Thamar B. van Dijk
- Department of Cell Biology, Erasmus University Medical Center, 3000 Rotterdam, The Netherlands
| | - Robin P. Peeters
- Department of Internal Medicine, Erasmus University Medical Center, 3000 Rotterdam, The Netherlands
| |
Collapse
|
41
|
Zhang Y, Wang C, Wang L, Shen B, Guan X, Tian J, Ren Z, Ding X, Ma Y, Dai W, Jiang Y. Large-Scale Ex Vivo Generation of Human Red Blood Cells from Cord Blood CD34 + Cells. Stem Cells Transl Med 2017; 6:1698-1709. [PMID: 28618139 PMCID: PMC5689780 DOI: 10.1002/sctm.17-0057] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 05/08/2017] [Indexed: 12/17/2022] Open
Abstract
The ex vivo generation of human red blood cells on a large scale from hematopoietic stem and progenitor cells has been considered as a potential method to overcome blood supply shortages. Here, we report that functional human erythrocytes can be efficiently produced from cord blood (CB) CD34+ cells using a bottle turning device culture system. Safety and efficiency studies were performed in murine and nonhuman primate (NHP) models. With the selected optimized culture conditions, one human CB CD34+ cell could be induced ex vivo to produce up to 200 million erythrocytes with a purity of 90.1% ± 6.2% and 50% ± 5.7% (mean ± SD) for CD235a+ cells and enucleated cells, respectively. The yield of erythrocytes from one CB unit (5 million CD34+ cells) could be, in theory, equivalent to 500 blood transfusion units in clinical application. Moreover, induced human erythrocytes had normal hemoglobin content and could continue to undergo terminal maturation in the murine xenotransplantation model. In NHP model, xenotransplantation of induced human erythrocytes enhanced hematological recovery and ameliorated the hypoxia situation in the primates with hemorrhagic anemia. These findings suggested that the ex vivo-generated erythrocytes could be an alternative blood source for traditional transfusion products in the clinic. Stem Cells Translational Medicine 2017;6:1698-1709.
Collapse
Affiliation(s)
- Yu Zhang
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| | - Chen Wang
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.,Biopharmagen Corp, Suzhou, China
| | - Lan Wang
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| | - Bin Shen
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| | - Xin Guan
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| | - Jing Tian
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China
| | - Zhihua Ren
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.,Biopharmagen Corp, Suzhou, China
| | - Xinxin Ding
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.,College of Nanoscale Science, SUNY Polytechnic Institute, Albany, New York, USA
| | - Yupo Ma
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.,Department of Pathology, School of Medicine, The State University of New York at Stony Brook, Stony Brook, New York, USA
| | - Wei Dai
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.,Department of Environment Medicine, New York University Langone Medical center, Tuxedo, New York, USA
| | - Yongping Jiang
- Biopharmaceutical R&D Center, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou, China.,Biopharmagen Corp, Suzhou, China
| |
Collapse
|
42
|
Karunasena N, Han TS, Mallappa A, Elman M, Merke DP, Ross RJ, Daniel E. Androgens correlate with increased erythropoiesis in women with congenital adrenal hyperplasia. Clin Endocrinol (Oxf) 2017; 86:19-25. [PMID: 27344964 PMCID: PMC5161698 DOI: 10.1111/cen.13148] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/26/2016] [Accepted: 06/22/2016] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Hyperandrogenism in congenital adrenal hyperplasia (CAH) provides an in vivo model for exploring the effect of androgens on erythropoiesis in women. We investigated the association of androgens with haemoglobin (Hb) and haematocrit (Hct) in women with CAH. DESIGN Cross-validation study. PATIENTS Women with CAH from Sheffield Teaching Hospitals, UK (cohort 1, the training set: n = 23) and National Institutes of Health, USA (cohort 2, the validation set: n = 53). MEASUREMENTS Androgens, full blood count and basic biochemistry, all measured on the same day. Demographic and anthropometric data. RESULTS Significant age-adjusted correlations (P < 0·001) were observed for Ln testosterone with Hb and Hct in cohorts 1 and 2 (Hb r = 0·712 & 0·524 and Hct r = 0·705 & 0·466), which remained significant after adjustments for CAH status, glucocorticoid treatment dose and serum creatinine. In the combined cohorts, Hb correlated with androstenedione (P = 0·002) and 17-hydroxyprogesterone (P = 0·008). Hb and Hct were significantly higher in cohort 1 than those in cohort 2, while there were no group differences in androgen levels, glucocorticoid treatment dose or body mass index. In both cohorts, women with Hb and Hct in the highest tertile had significantly higher testosterone levels than women with Hb and Hct in the lowest tertile. CONCLUSIONS In women with CAH, erythropoiesis may be driven by androgens and could be considered a biomarker for disease control.
Collapse
Affiliation(s)
| | - Thang S. Han
- Institute of Cardiovascular ResearchRoyal Holloway University of London (ICR2UL) and Ashford and St Peter's NHS Foundation TrustSurreyUK
| | - Ashwini Mallappa
- National Institutes of Health Clinical CentreEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentBethesdaMDUSA
| | - Meredith Elman
- National Institutes of Health Clinical CentreEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentBethesdaMDUSA
| | - Deborah P. Merke
- National Institutes of Health Clinical CentreEunice Kennedy Shriver National Institute of Child Health and Human DevelopmentBethesdaMDUSA
| | - Richard J.M. Ross
- Department of Oncology and MetabolismUniversity of SheffieldSheffieldUK
| | - Eleni Daniel
- Department of Oncology and MetabolismUniversity of SheffieldSheffieldUK
| |
Collapse
|
43
|
Shah SN, Gelderman MP, Lewis EMA, Farrel J, Wood F, Strader MB, Alayash AI, Vostal JG. Evaluation of Stem Cell-Derived Red Blood Cells as a Transfusion Product Using a Novel Animal Model. PLoS One 2016; 11:e0166657. [PMID: 27959920 PMCID: PMC5154495 DOI: 10.1371/journal.pone.0166657] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 10/17/2016] [Indexed: 11/18/2022] Open
Abstract
Reliance on volunteer blood donors can lead to transfusion product shortages, and current liquid storage of red blood cells (RBCs) is associated with biochemical changes over time, known as ‘the storage lesion’. Thus, there is a need for alternative sources of transfusable RBCs to supplement conventional blood donations. Extracorporeal production of stem cell-derived RBCs (stemRBCs) is a potential and yet untapped source of fresh, transfusable RBCs. A number of groups have attempted RBC differentiation from CD34+ cells. However, it is still unclear whether these stemRBCs could eventually be effective substitutes for traditional RBCs due to potential differences in oxygen carrying capacity, viability, deformability, and other critical parameters. We have generated ex vivo stemRBCs from primary human cord blood CD34+ cells and compared them to donor-derived RBCs based on a number of in vitro parameters. In vivo, we assessed stemRBC circulation kinetics in an animal model of transfusion and oxygen delivery in a mouse model of exercise performance. Our novel, chronically anemic, SCID mouse model can evaluate the potential of stemRBCs to deliver oxygen to tissues (muscle) under resting and exercise-induced hypoxic conditions. Based on our data, stem cell-derived RBCs have a similar biochemical profile compared to donor-derived RBCs. While certain key differences remain between donor-derived RBCs and stemRBCs, the ability of stemRBCs to deliver oxygen in a living organism provides support for further development as a transfusion product.
Collapse
Affiliation(s)
- Sandeep N. Shah
- Laboratory of Cellular Hematology, Division of Hematology Research and Review, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Monique P. Gelderman
- Laboratory of Cellular Hematology, Division of Hematology Research and Review, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Emily M. A. Lewis
- Laboratory of Cellular Hematology, Division of Hematology Research and Review, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - John Farrel
- Laboratory of Cellular Hematology, Division of Hematology Research and Review, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Francine Wood
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Michael Brad Strader
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Abdu I. Alayash
- Laboratory of Biochemistry and Vascular Biology, Division of Hematology Research and Review, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Jaroslav G. Vostal
- Laboratory of Cellular Hematology, Division of Hematology Research and Review, Office of Blood Research and Review, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland, United States of America
- * E-mail:
| |
Collapse
|
44
|
Digesting the role of bone marrow macrophages on hematopoiesis. Immunobiology 2016; 222:814-822. [PMID: 27890297 DOI: 10.1016/j.imbio.2016.11.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 10/27/2016] [Accepted: 11/12/2016] [Indexed: 01/07/2023]
Abstract
Tissue resident macrophages are found in various tissues like Langerhans cells in the skin or alveolar macrophages in the lung, and their main function is to regulate organ homeostasis. They have also been observed in the bone marrow and these cells in particular have been gaining importance in recent years as they are key players in hematopoiesis. However, as the characterization and classification of these putatively different bone marrow resident macrophages is far from established there is a need to generate an overview of tissue resident macrophages of the bone marrow. Here, we will review the current knowledge of bone marrow resident macrophages both in mouse and human. We will discuss the state of the art on the origin of bone marrow macrophages, specialized microenvironments where they reside and their unique characteristics. We will emphasize the two best studied examples of macrophage homeostatic function in the bone marrow, specifically within erythroblastic islands and the hematopoietic stem cell niche. Although increasing evidence shows that bone marrow resident macrophages are indispensable for hematopoietic stem cell function and bone marrow erythroid output, the field of bone marrow macrophages is in its infancy. This field is in dire need for a unified nomenclature to support functional experiments, model systems, and the identification of niches.
Collapse
|
45
|
Polyurethane scaffolds seeded with CD34(+) cells maintain early stem cells whilst also facilitating prolonged egress of haematopoietic progenitors. Sci Rep 2016; 6:32149. [PMID: 27573994 PMCID: PMC5004174 DOI: 10.1038/srep32149] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/03/2016] [Indexed: 12/15/2022] Open
Abstract
We describe a 3D erythroid culture system that utilises a porous polyurethane (PU) scaffold to mimic the compartmentalisation found in the bone marrow. PU scaffolds seeded with peripheral blood CD34+ cells exhibit a remarkable reproducibility of egress, with an increased output when directly compared to human bone scaffolds over 28 days. Immunofluorescence demonstrated the persistence of CD34+ cells within the scaffolds for the entirety of the culture. To characterise scaffold outputs, we designed a flow cytometry panel that utilises surface marker expression observed in standard 2D erythroid and megakaryocyte cultures. This showed that the egress population is comprised of haematopoietic progenitor cells (CD36+GPA−/low). Control cultures conducted in parallel but in the absence of a scaffold were also generally maintained for the longevity of the culture albeit with a higher level of cell death. The harvested scaffold egress can also be expanded and differentiated to the reticulocyte stage. In summary, PU scaffolds can behave as a subtractive compartmentalised culture system retaining and allowing maintenance of the seeded “CD34+ cell” population despite this population decreasing in amount as the culture progresses, whilst also facilitating egress of increasingly differentiated cells.
Collapse
|
46
|
Geiler C, Andrade I, Clayton A, Greenwald D. Genetically Engineered In Vitro Erythropoiesis. Int J Stem Cells 2016; 9:53-9. [PMID: 27426086 PMCID: PMC4961104 DOI: 10.15283/ijsc.2016.9.1.53] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/23/2016] [Indexed: 01/13/2023] Open
Abstract
Background Engineered blood has the greatest potential to combat a predicted future shortfall in the US blood supply for transfusion treatments. Engineered blood produced from hematopoietic stem cell (HSC) derived red blood cells in a laboratory is possible, but critical barriers exist to the production of clinically relevant quantities of red blood cells required to create a unit of blood. Erythroblasts have a finite expansion capacity and there are many negative regulatory mechanisms that inhibit in vitro erythropoiesis. In order to overcome these barriers and enable mass production, the expansion capacity of erythroblasts in culture will need to be exponentially improved over the current state of art. This work focused on the hypothesis that genetic engineering of HSC derived erythroblasts can overcome these obstacles. Objectives The objective of this research effort was to improve in vitro erythropoiesis efficiency from human adult stem cell derived erythroblasts utilizing genetic engineering. The ultimate goal is to enable the mass production of engineered blood. Methods HSCs were isolated from blood samples and cultured in a liquid media containing growth factors. Cells were transfected using a Piggybac plasmid transposon. Results Cells transfected with SPI-1 continued to proliferate in a liquid culture media. Fluorescence-activated cell sorting (FACS) analysis on culture day 45 revealed a single population of CD71+CD117+ proerythroblast cells. The results of this study suggest that genetically modified erythroblasts could be immortalized in vitro by way of a system modeling murine erythroleukemia. Conclusion Genetic modification can increase erythroblast expansion capacity and potentially enable mass production of red blood cells.
Collapse
Affiliation(s)
- Cristopher Geiler
- Department of Basic Science Research, Cellologi, LLC, California, USA.,Santa Barbara Cottage Hospital, Santa Barbara, California, USA
| | - Inez Andrade
- Department of Basic Science Research, Cellologi, LLC, California, USA
| | - Alexandra Clayton
- Department of Basic Science Research, Cellologi, LLC, California, USA
| | - Daniel Greenwald
- Department of Basic Science Research, Cellologi, LLC, California, USA.,Santa Barbara Cottage Hospital, Santa Barbara, California, USA
| |
Collapse
|
47
|
Kim AR, Sankaran VG. Development of autologous blood cell therapies. Exp Hematol 2016; 44:887-94. [PMID: 27345108 DOI: 10.1016/j.exphem.2016.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 06/07/2016] [Accepted: 06/10/2016] [Indexed: 12/21/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation and blood cell transfusions are performed commonly in patients with a variety of blood disorders. Unfortunately, these donor-derived cell therapies are constrained due to limited supplies, infectious risk factors, a lack of appropriately matched donors, and the risk of immunologic complications from such products. The use of autologous cell therapies has been proposed to overcome these shortcomings. One can derive such therapies directly from hematopoietic stem and progenitor cells of individuals, which can then be manipulated ex vivo to produce the desired modifications or differentiated to produce a particular target population. Alternatively, pluripotent stem cells, which have a theoretically unlimited self-renewal capacity and an ability to differentiate into any desired cell type, can be used as an autologous starting source for such manipulation and differentiation approaches. Such cell products can also be used as a delivery vehicle for therapeutics. In this review, we highlight recent advances and discuss ongoing challenges for the in vitro generation of autologous hematopoietic cells that can be used for cell therapy.
Collapse
Affiliation(s)
- Ah Ram Kim
- Division of Hematology/Oncology, Boston Children's Hospital, and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | - Vijay G Sankaran
- Division of Hematology/Oncology, Boston Children's Hospital, and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.
| |
Collapse
|
48
|
Ding N, Xi J, Li Y, Xie X, Shi J, Zhang Z, Li Y, Fang F, Wang S, Yue W, Pei X, Fang X. Global transcriptome analysis for identification of interactions between coding and noncoding RNAs during human erythroid differentiation. Front Med 2016; 10:297-310. [PMID: 27272188 DOI: 10.1007/s11684-016-0452-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 04/22/2016] [Indexed: 12/26/2022]
Abstract
Studies on coding genes, miRNAs, and lncRNAs during erythroid development have been performed in recent years. However, analysis focusing on the integration of the three RNA types has yet to be done. In the present study, we compared the dynamics of coding genes, miRNA, and lncRNA expression profiles. To explore dynamic changes in erythropoiesis and potential mechanisms that control these changes in the transcriptome level, we took advantage of high throughput sequencing technologies to obtain transcriptome data from cord blood hematopoietic stem cells and the following four erythroid differentiation stages, as well as from mature red blood cells. Results indicated that lncRNAs were promising cell marker candidates for erythroid differentiation. Clustering analysis classified the differentially expressed genes into four subtypes that corresponded to dynamic changes during stemness maintenance, mid-differentiation, and maturation. Integrated analysis revealed that noncoding RNAs potentially participated in controlling blood cell maturation, and especially associated with heme metabolism and responses to oxygen species and DNA damage. These regulatory interactions were displayed in a comprehensive network, thereby inferring correlations between RNAs and their associated functions. These data provided a substantial resource for the study of normal erythropoiesis, which will permit further investigation and understanding of erythroid development and acquired erythroid disorders.
Collapse
Affiliation(s)
- Nan Ding
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiafei Xi
- Lab of Stem Cell and Regenerative Medicine, Beijing Institute of Transfusion Medicine, AMMS, Beijing, 100850, China.,South China Research Center for Stem Cell & Regenerative Medicine, AMMS, Guangzhou, 510300, China
| | - Yanming Li
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiaoyan Xie
- Lab of Stem Cell and Regenerative Medicine, Beijing Institute of Transfusion Medicine, AMMS, Beijing, 100850, China.,South China Research Center for Stem Cell & Regenerative Medicine, AMMS, Guangzhou, 510300, China
| | - Jian Shi
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhaojun Zhang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yanhua Li
- Lab of Stem Cell and Regenerative Medicine, Beijing Institute of Transfusion Medicine, AMMS, Beijing, 100850, China.,South China Research Center for Stem Cell & Regenerative Medicine, AMMS, Guangzhou, 510300, China
| | - Fang Fang
- Lab of Stem Cell and Regenerative Medicine, Beijing Institute of Transfusion Medicine, AMMS, Beijing, 100850, China.,South China Research Center for Stem Cell & Regenerative Medicine, AMMS, Guangzhou, 510300, China
| | - Sihan Wang
- Lab of Stem Cell and Regenerative Medicine, Beijing Institute of Transfusion Medicine, AMMS, Beijing, 100850, China.,South China Research Center for Stem Cell & Regenerative Medicine, AMMS, Guangzhou, 510300, China
| | - Wen Yue
- Lab of Stem Cell and Regenerative Medicine, Beijing Institute of Transfusion Medicine, AMMS, Beijing, 100850, China.,South China Research Center for Stem Cell & Regenerative Medicine, AMMS, Guangzhou, 510300, China
| | - Xuetao Pei
- Lab of Stem Cell and Regenerative Medicine, Beijing Institute of Transfusion Medicine, AMMS, Beijing, 100850, China. .,South China Research Center for Stem Cell & Regenerative Medicine, AMMS, Guangzhou, 510300, China.
| | - Xiangdong Fang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
49
|
Zhou H, Martinez H, Sun B, Li A, Zimmer M, Katsanis N, Davis EE, Kurtzberg J, Lipnick S, Noggle S, Rao M, Chang S. Rapid and Efficient Generation of Transgene-Free iPSC from a Small Volume of Cryopreserved Blood. Stem Cell Rev Rep 2016; 11:652-65. [PMID: 25951995 PMCID: PMC4493720 DOI: 10.1007/s12015-015-9586-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human peripheral blood and umbilical cord blood represent attractive sources of cells for reprogramming to induced pluripotent stem cells (iPSCs). However, to date, most of the blood-derived iPSCs were generated using either integrating methods or starting from T-lymphocytes that have genomic rearrangements thus bearing uncertain consequences when using iPSC-derived lineages for disease modeling and cell therapies. Recently, both peripheral blood and cord blood cells have been reprogrammed into transgene-free iPSC using the Sendai viral vector. Here we demonstrate that peripheral blood can be utilized for medium-throughput iPSC production without the need to maintain cell culture prior to reprogramming induction. Cell reprogramming can also be accomplished with as little as 3000 previously cryopreserved cord blood cells under feeder-free and chemically defined Xeno-free conditions that are compliant with standard Good Manufacturing Practice (GMP) regulations. The first iPSC colonies appear 2–3 weeks faster in comparison to previous reports. Notably, these peripheral blood- and cord blood-derived iPSCs are free of detectable immunoglobulin heavy chain (IGH) and T cell receptor (TCR) gene rearrangements, suggesting they did not originate from B- or T- lymphoid cells. The iPSCs are pluripotent as evaluated by the scorecard assay and in vitro multi lineage functional cell differentiation. Our data show that small volumes of cryopreserved peripheral blood or cord blood cells can be reprogrammed efficiently at a convenient, cost effective and scalable way. In summary, our method expands the reprogramming potential of limited or archived samples either stored at blood banks or obtained from pediatric populations that cannot easily provide large quantities of peripheral blood or a skin biopsy.
Collapse
Affiliation(s)
- Hongyan Zhou
- The New York Stem Cell Foundation Research Institute, New York, NY, 10032, USA,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Patel MD, Lateef A, Das H, Patel AS, Patel AG, Joshi AB. Effect of age, sex and physiological stages on hematological indices of Banni buffalo (Bubalus bubalis). Vet World 2016; 9:38-42. [PMID: 27051182 PMCID: PMC4819347 DOI: 10.14202/vetworld.2016.38-42] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 11/26/2015] [Accepted: 12/04/2015] [Indexed: 11/22/2022] Open
Abstract
Aim: To determine the physiological baseline values for hematological indices of Banni buffalo (Bubalus bubalis) as well as to assess their alteration due to age, sex and physiological stages. Materials and Methods: A total of 42 clinically healthy Banni buffaloes were categorized into seven groups (n=6): Group I (male calves ≤1 year), Group II (bulls >1 year), Group III (female calves ≤1 year), Group IV (pregnant lactating buffaloes), Group V (non-pregnant lactating buffaloes), Group VI (pregnant dry buffaloes), and Group VII (non-pregnant dry buffaloes). Blood samples collected aseptically from all the experimental groups were analyzed employing automated hematology analyzer. The data obtained were statistically analyzed; the mean and standard deviations were calculated and set as the reference values. Results: The erythrocytic indices viz. total erythrocytes count (TEC), hemoglobin, and packed cell volume (PCV) were significantly higher in bulls as compared to that of male calves unlike mean corpuscular volume, mean corpuscular hemoglobin (MCH), and MCH concentration. The female calves had higher TEC and PCV than the adult buffaloes irrespective of sex. The total leukocyte count (TLC) and neutrophil counts in male calves were significantly lower than the bulls unlike the eosinophil, while monocyte and basophil remained unchanged with age. The TLC, differential leukocyte count and platelet count varied non-significantly among the adult female groups at different physiological stages. However, neutrophils were found to be apparently higher in lactating buffaloes. Conclusion: The present study would be helpful for physiological characterization of this unique buffalo breed of Gujarat. Further, data generated may be a tool for monitoring the health and prognosis as well as diagnosis of diseases.
Collapse
Affiliation(s)
- Mehul D Patel
- Department of Physiology and Biochemistry, College of Veterinary Science & Animal Husbandry, Sardarkrushinagar Dantiwada Agricultural University, Sardarkrushinagar, Dantiwada-385506, Gujarat, India
| | - Abdul Lateef
- Department of Physiology and Biochemistry, College of Veterinary Science & Animal Husbandry, Sardarkrushinagar Dantiwada Agricultural University, Sardarkrushinagar, Dantiwada-385506, Gujarat, India
| | - Hemen Das
- Department of Physiology and Biochemistry, College of Veterinary Science & Animal Husbandry, Sardarkrushinagar Dantiwada Agricultural University, Sardarkrushinagar, Dantiwada-385506, Gujarat, India
| | - Ajay S Patel
- Department of Livestock Products Technology, College of Veterinary Science & Animal Husbandry, Anand Agricultural University, Anand- 388110, Gujarat, India
| | - Ajay G Patel
- Department of Physiology and Biochemistry, College of Veterinary Science & Animal Husbandry, Sardarkrushinagar Dantiwada Agricultural University, Sardarkrushinagar, Dantiwada-385506, Gujarat, India
| | - Axay B Joshi
- Department of Physiology and Biochemistry, College of Veterinary Science & Animal Husbandry, Sardarkrushinagar Dantiwada Agricultural University, Sardarkrushinagar, Dantiwada-385506, Gujarat, India
| |
Collapse
|