201
|
Silencing and overexpression of human blood group antigens in transfusion: Paving the way for the next steps. Blood Rev 2015; 29:163-9. [DOI: 10.1016/j.blre.2014.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 10/23/2014] [Indexed: 01/25/2023]
|
202
|
Velliou EG, Dos Santos SB, Papathanasiou MM, Fuentes-Gari M, Misener R, Panoskaltsis N, Pistikopoulos EN, Mantalaris A. Towards unravelling the kinetics of an acute myeloid leukaemia model system under oxidative and starvation stress: a comparison between two- and three-dimensional cultures. Bioprocess Biosyst Eng 2015; 38:1589-600. [PMID: 25911423 DOI: 10.1007/s00449-015-1401-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 04/14/2015] [Indexed: 12/26/2022]
Abstract
A great challenge when conducting ex vivo studies of leukaemia is the construction of an appropriate experimental platform that would recapitulate the bone marrow (BM) environment. Such a 3D scaffold system has been previously developed in our group [1]. Additionally to the BM architectural characteristics, parameters such as oxygen and glucose concentration are crucial as their value could differ between patients as well as within the same patient at different stages of treatment, consequently affecting the resistance of leukaemia to chemotherapy. The effect of oxidative and glucose stress-at levels close to human physiologic ones-on the proliferation and metabolic evolution of an AML model system (K-562 cell line) in conventional 2D cultures as well as in 3D scaffolds were studied. We observed that the K-562 cell line can proliferate and remain alive for 2 weeks in medium with glucose close to physiological levels both in 20 and 5% O2. We report interesting differences on the cellular response to the environmental, i.e., oxidative and/or nutritional stress stimuli in 2D and 3D. Higher adaptation to oxidative stress under non-starving conditions is observed in the 3D system. The glucose level in the medium has more impact on the cellular proliferation in the 3D compared to the 2D system. These differences can be of significant importance both when applying chemotherapy in vitro and also when constructing mathematical tools for optimisation of disease treatment.
Collapse
Affiliation(s)
- Eirini G Velliou
- Department of Chemical and Process Engineering, Faculty of Engineering and Physical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK,
| | | | | | | | | | | | | | | |
Collapse
|
203
|
Affiliation(s)
- W. H. Choe
- Department of Laboratory Medicine; School of Medicine; Eulji University; Seoul Korea
| | - E. J. Baek
- Department of Laboratory Medicine; School of Medicine; Hanyang University; Seoul Korea
| |
Collapse
|
204
|
Simamura E, Arikawa T, Ikeda T, Shimada H, Shoji H, Masuta H, Nakajima Y, Otani H, Yonekura H, Hatta T. Melanocortins contribute to sequential differentiation and enucleation of human erythroblasts via melanocortin receptors 1, 2 and 5. PLoS One 2015; 10:e0123232. [PMID: 25860801 PMCID: PMC4393082 DOI: 10.1371/journal.pone.0123232] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 02/17/2015] [Indexed: 11/30/2022] Open
Abstract
In this study, we showed that adrenocorticotropic hormone (ACTH) promoted erythroblast differentiation and increased the enucleation ratio of erythroblasts. Because ACTH was contained in hematopoietic medium as contamination, the ratio decreased by the addition of anti-ACTH antibody (Ab). Addition of neutralizing Abs (nAbs) for melanocortin receptors (MCRs) caused erythroblast accumulation at specific stages, i.e., the addition of anti-MC2R nAb led to erythroblast accumulation at the basophilic stage (baso-E), the addition of anti-MC1R nAb caused accumulation at the polychromatic stage (poly-E), and the addition of anti-MC5R nAb caused accumulation at the orthochromatic stage (ortho-E). During erythroblast differentiation, ERK, STAT5, and AKT were consecutively phosphorylated by erythropoietin (EPO). ERK, STAT5, and AKT phosphorylation was inhibited by blocking MC2R, MC1R, and MC5R, respectively. Finally, the phosphorylation of myosin light chain 2, which is essential for the formation of contractile actomyosin rings, was inhibited by anti-MC5R nAb. Taken together, our study suggests that MC2R and MC1R signals are consecutively required for the regulation of EPO signal transduction in erythroblast differentiation, and that MC5R signal transduction is required to induce enucleation. Thus, melanocortin induces proliferation and differentiation at baso-E, and polarization and formation of an actomyosin contractile ring at ortho-E are required for enucleation.
Collapse
MESH Headings
- Adrenocorticotropic Hormone/antagonists & inhibitors
- Adrenocorticotropic Hormone/metabolism
- Antibodies, Neutralizing
- Cell Differentiation/physiology
- Cells, Cultured
- Erythroblasts/cytology
- Erythroblasts/metabolism
- Erythropoiesis/physiology
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Humans
- Melanocortins/metabolism
- Models, Biological
- Proto-Oncogene Proteins c-akt/metabolism
- Receptor, Melanocortin, Type 1/antagonists & inhibitors
- Receptor, Melanocortin, Type 1/genetics
- Receptor, Melanocortin, Type 1/metabolism
- Receptor, Melanocortin, Type 2/antagonists & inhibitors
- Receptor, Melanocortin, Type 2/genetics
- Receptor, Melanocortin, Type 2/metabolism
- Receptors, Melanocortin/antagonists & inhibitors
- Receptors, Melanocortin/genetics
- Receptors, Melanocortin/metabolism
- STAT5 Transcription Factor/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Eriko Simamura
- Department of Anatomy, Kanazawa Medical University School of Medicine, Uchinada, Ishikawa 920–0293, Japan
| | - Tomohiro Arikawa
- Department of Biology, Kanazawa Medical University School of Medicine, Uchinada, Ishikawa 920–0293, Japan
| | - Takayuki Ikeda
- Department of Biochemistry, Kanazawa Medical University School of Medicine, Uchinada, Ishikawa 920–0293, Japan
| | - Hiroki Shimada
- Department of Anatomy, Kanazawa Medical University School of Medicine, Uchinada, Ishikawa 920–0293, Japan
| | - Hiroki Shoji
- Department of Biology, Kanazawa Medical University School of Medicine, Uchinada, Ishikawa 920–0293, Japan
| | - Hiroko Masuta
- Department of Anatomy, Kanazawa Medical University School of Medicine, Uchinada, Ishikawa 920–0293, Japan
| | - Yuriko Nakajima
- Department of Anatomy, Kanazawa Medical University School of Medicine, Uchinada, Ishikawa 920–0293, Japan
| | - Hiroki Otani
- Department of Developmental Biology, Faculty of Medicine, Shimane University, Izumo 693–8601, Japan
| | - Hideto Yonekura
- Department of Biochemistry, Kanazawa Medical University School of Medicine, Uchinada, Ishikawa 920–0293, Japan
| | - Toshihisa Hatta
- Department of Anatomy, Kanazawa Medical University School of Medicine, Uchinada, Ishikawa 920–0293, Japan
- * E-mail:
| |
Collapse
|
205
|
Zhu F, Shi L, Engel JD, Guan Y. Regulatory network inferred using expression data of small sample size: application and validation in erythroid system. Bioinformatics 2015; 31:2537-44. [PMID: 25840044 DOI: 10.1093/bioinformatics/btv186] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 03/27/2015] [Indexed: 11/13/2022] Open
Abstract
MOTIVATION Modeling regulatory networks using expression data observed in a differentiation process may help identify context-specific interactions. The outcome of the current algorithms highly depends on the quality and quantity of a single time-course dataset, and the performance may be compromised for datasets with a limited number of samples. RESULTS In this work, we report a multi-layer graphical model that is capable of leveraging many publicly available time-course datasets, as well as a cell lineage-specific data with small sample size, to model regulatory networks specific to a differentiation process. First, a collection of network inference methods are used to predict the regulatory relationships in individual public datasets. Then, the inferred directional relationships are weighted and integrated together by evaluating against the cell lineage-specific dataset. To test the accuracy of this algorithm, we collected a time-course RNA-Seq dataset during human erythropoiesis to infer regulatory relationships specific to this differentiation process. The resulting erythroid-specific regulatory network reveals novel regulatory relationships activated in erythropoiesis, which were further validated by genome-wide TR4 binding studies using ChIP-seq. These erythropoiesis-specific regulatory relationships were not identifiable by single dataset-based methods or context-independent integrations. Analysis of the predicted targets reveals that they are all closely associated with hematopoietic lineage differentiation.
Collapse
Affiliation(s)
- Fan Zhu
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lihong Shi
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | | | - Yuanfang Guan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA, Department of Internal Medicine, and Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
206
|
Correction of the sickle cell disease mutation in human hematopoietic stem/progenitor cells. Blood 2015; 125:2597-604. [PMID: 25733580 DOI: 10.1182/blood-2014-12-615948] [Citation(s) in RCA: 249] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 02/07/2015] [Indexed: 12/26/2022] Open
Abstract
Sickle cell disease (SCD) is characterized by a single point mutation in the seventh codon of the β-globin gene. Site-specific correction of the sickle mutation in hematopoietic stem cells would allow for permanent production of normal red blood cells. Using zinc-finger nucleases (ZFNs) designed to flank the sickle mutation, we demonstrate efficient targeted cleavage at the β-globin locus with minimal off-target modification. By co-delivering a homologous donor template (either an integrase-defective lentiviral vector or a DNA oligonucleotide), high levels of gene modification were achieved in CD34(+) hematopoietic stem and progenitor cells. Modified cells maintained their ability to engraft NOD/SCID/IL2rγ(null) mice and to produce cells from multiple lineages, although with a reduction in the modification levels relative to the in vitro samples. Importantly, ZFN-driven gene correction in CD34(+) cells from the bone marrow of patients with SCD resulted in the production of wild-type hemoglobin tetramers.
Collapse
|
207
|
Schmelzer E, Finoli A, Nettleship I, Gerlach JC. Long-term three-dimensional perfusion culture of human adult bone marrow mononuclear cells in bioreactors. Biotechnol Bioeng 2015; 112:801-10. [PMID: 25335987 DOI: 10.1002/bit.25485] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 08/22/2014] [Accepted: 10/13/2014] [Indexed: 12/14/2022]
Abstract
The construction and long-term maintenance of three-dimensional in vitro bone marrow models is of great interest but still quite challenging. Here we describe the use of a multi-compartment hollow-fiber membrane based three-dimensional perfusion bioreactor for long-term culture of whole human bone marrow mononuclear cells. We also investigated bioreactors with incorporated open-porous foamed hydroxyapatite scaffolds, mimicking the in vivo bone matrix. Cells in bioreactors with and without scaffolds were cultured to 6 weeks and compared to Petri dish controls. Cells were analyzed for gene expression, surface markers by flow cytometry, metabolic activity, hematopoietic potential, viability, and attachment by immunocytochemistry. Cells in bioreactors were metabolic active during long-term culture. The percentages of hematopoietic stem cell and mature endothelial cell fractions were maintained in bioreactors. The expression of most of the analyzed genes stabilized and increased after long-term culture of 6 weeks. Compared to Petri dish culture controls, bioreactor perfusion culture improved in both the short and long-term, the colony formation unit capacity of hematopoietic progenitors. Cells attached to the ample surface area provided by hydroxyapatite scaffolds. The implementation of a hydroxyapatite scaffold did not influence colony formation capacity, percentages of cell type specific fractions, gene expression, cell viability or metabolic turnover when compared to control cells cultured in bioreactors without scaffolds. In conclusion, three-dimensional perfusion bioreactor culture enables long-term maintenance of primary human bone marrow cells, with hydroxyapatite scaffolds providing an in vivo-like scaffold for three-dimensional culture.
Collapse
Affiliation(s)
- Eva Schmelzer
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, 3025 E. Carson St., Pittsburgh, Pennsylvania, 15203.
| | | | | | | |
Collapse
|
208
|
Spinelli E, Bartlett RH. Anemia and Transfusion in Critical Care. J Intensive Care Med 2015; 31:295-306. [DOI: 10.1177/0885066615571901] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 12/03/2014] [Indexed: 01/28/2023]
Abstract
Objective: The objective of this report is to review the physiology and management of anemia in critical care. Selected publications on physiology and transfusion related to anemia and critical care, including the modern randomized trials of conservative versus liberal transfusion policy, were used. Anemia is compensated and tolerated in most critically ill patients as long as oxygen delivery is at least twice oxygen consumption. There are risks to blood transfusion which can be minimized by blood banking practice. The availability of cultured red cells may allow correction of anemia without significant risk. The benefit of transfusion in anemia must be weighted against the risk in any specific patient. Conclusion and Recommendation: In a criticially ill patient, anemia should be managed to avoid oxygen supply dependency (oxygen delivery less than twice comsumption) and to maintain moderate oxygen delivery reserve (DO2/VO2 > 3).
Collapse
Affiliation(s)
- Elena Spinelli
- University of Michigan ECLS Laboratory, Ann Arbor, MI, USA
| | | |
Collapse
|
209
|
Yeom J, Kim SJ, Jung H, Namkoong H, Yang J, Hwang BW, Oh K, Kim K, Sung YC, Hahn SK. Supramolecular hydrogels for long-term bioengineered stem cell therapy. Adv Healthc Mater 2015; 4:237-44. [PMID: 25100551 DOI: 10.1002/adhm.201400304] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 07/03/2014] [Indexed: 01/13/2023]
Abstract
Synthetic hydrogels have been extensively investigated as artificial extracellular matrices (ECMs) for tissue engineering in vitro and in vivo. Crucial challenges for such hydrogels are sustaining long-term cytocompatible encapsulation and providing appropriate cues at the right place and time for spatio-temporal control of the cells. Here, in situ supramolecularly assembled and modularly modified hydrogels for long-term engineered mesenchymal stem cell (eMSC) therapy are reported using cucurbit[6]uril-conjugated hyaluronic acid (CB[6]-HA), diaminohexane conjugated HA (DAH-HA), and drug-conjugated CB[6] (drug-CB[6]). The eMSCs producing enhanced green fluorescence protein (EGFP) remain alive and emit the fluorescence within CB[6]/DAH-HA hydrogels in mice for more than 60 d. Furthermore, the long-term expression of mutant interleukin-12 (IL-12M) by eMSCs within the supramolecular hydrogels results in effective inhibition of tumor growth with a significantly enhanced survival rate. Taken together, these findings confirm the feasibility of supramolecular HA hydrogels as 3D artificial ECMs for cell therapies and tissue engineering applications.
Collapse
Affiliation(s)
- Junseok Yeom
- Department of Materials Science and Engineering; 77 Cheongam-ro, Nam-gu Pohang 790-784 Republic of Korea
| | - Su Jin Kim
- Department of Life Sciences; 77 Cheongam-ro, Nam-gu Pohang 790-784 Republic of Korea
| | - Hyuntae Jung
- School of Interdisciplinary Bioscience and Bioengineering; 77 Cheongam-ro, Nam-gu Pohang 790-784 Republic of Korea
| | - Hong Namkoong
- Department of Life Sciences; 77 Cheongam-ro, Nam-gu Pohang 790-784 Republic of Korea
| | - Jeonga Yang
- Department of Materials Science and Engineering; 77 Cheongam-ro, Nam-gu Pohang 790-784 Republic of Korea
| | - Byung Woo Hwang
- Department of Materials Science and Engineering; 77 Cheongam-ro, Nam-gu Pohang 790-784 Republic of Korea
| | - Kyunghoon Oh
- Department of Chemistry; Division of Advanced Materials Science; Center for Self-assembly and Complexity; Institute for Basic Science (IBS); 77 Cheongam-ro, Nam-gu Pohang 790-784 Republic of Korea
| | - Kimoon Kim
- Department of Chemistry; Division of Advanced Materials Science; Center for Self-assembly and Complexity; Institute for Basic Science (IBS); 77 Cheongam-ro, Nam-gu Pohang 790-784 Republic of Korea
| | - Young Chul Sung
- Department of Life Sciences; 77 Cheongam-ro, Nam-gu Pohang 790-784 Republic of Korea
| | - Sei Kwang Hahn
- Department of Materials Science and Engineering; 77 Cheongam-ro, Nam-gu Pohang 790-784 Republic of Korea
| |
Collapse
|
210
|
Srivastava A, Leighton X, Eidelman O, Starr J, Jozwik C, Srivastava M, Pollard HB, Singh VK. Personalized Radioproteomics: Identification of a Protein Biomarker Signature for Preemptive Rescue by Tocopherol Succinate in CD34 + Irradiated Progenitor Cells Isolated from a Healthy Control Donor. ACTA ACUST UNITED AC 2015; 8:23-30. [PMID: 27087761 PMCID: PMC4833407 DOI: 10.4172/jpb.1000349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Tocopherol succinate (TS) has been shown to protect mice against acute radiation syndrome, however, its exact mechanism of action and its possible use in humans has not yet been evaluated. Our approach has been to test the radioprotectant properties of TS on CD34-positive stem cells from healthy volunteers. We hypothesize that a radioproteomics strategy can identify a drug-dependent, personalized proteomics signature for radioprotection. To directly test the radioproteomics hypothesis, we treated human CD34-positive stem cells with 20 μM TS for 24 h, and then exposed the cells to 2 Gy of cobalt-60 gamma-radiation. We isolated protein from all cultures and used a high throughput Antibody Microarray (AbMA) platform to measure concentrations of 725 low abundance proteins. As an in vivo control, we also tested mouse CD34-positive stem cells using the same preemptive TS paradigm on progenitor colony forming units. TS pretreatment of in vitro or in vivo CD34-positive stem cells rescued radiation-induced loss of colony-forming potential of progenitors. We identified 50 of 725 proteins that could be preemptively rescued from radiation-induced reduction by pretreatment with TS. Ingenuity Pathway Analysis (IPA) reveals that the modified proteins fall into categories dominated by epigenetic regulation, DNA repair, and inflammation. Our results suggest that radioproteomics can be used to develop personalized medicine for radioprotection using protein signatures from primary CD34-positive progenitors derived from the patient or victim prior to radiation exposure. The protective effect of TS may be due to its ability to preemptively activate epigenetic mechanisms relevant to radioprotection and to preemptively activate the programs for DNA repair and inflammation leading to cell survival.
Collapse
Affiliation(s)
| | - Ximena Leighton
- Department of Anatomy, Physiology and Genetics, and Center for Medical Proteomics, USA
| | - Ofer Eidelman
- Department of Anatomy, Physiology and Genetics, and Center for Medical Proteomics, USA
| | - Joshua Starr
- Department of Anatomy, Physiology and Genetics, and Center for Medical Proteomics, USA
| | - Catherine Jozwik
- Department of Anatomy, Physiology and Genetics, and Center for Medical Proteomics, USA
| | - Meera Srivastava
- Department of Anatomy, Physiology and Genetics, and Center for Medical Proteomics, USA
| | - Harvey B Pollard
- Department of Anatomy, Physiology and Genetics, and Center for Medical Proteomics, USA
| | - Vijay K Singh
- Armed Forces Radiobiology Research Institute, Bethesda, MD, USA; Department of Radiation Biology, F. Edward Hébert School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
211
|
Compound loss of function of nuclear receptors Tr2 and Tr4 leads to induction of murine embryonic β-type globin genes. Blood 2015; 125:1477-87. [PMID: 25561507 DOI: 10.1182/blood-2014-10-605022] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The orphan nuclear receptors TR2 and TR4 have been shown to play key roles in repressing the embryonic and fetal globin genes in erythroid cells. However, combined germline inactivation of Tr2 and Tr4 leads to periimplantation lethal demise in inbred mice. Hence, we have previously been unable to examine the consequences of their dual loss of function in adult definitive erythroid cells. To circumvent this issue, we generated conditional null mutants in both genes and performed gene inactivation in vitro in adult bone marrow cells. Compound Tr2/Tr4 loss of function led to induced expression of the embryonic εy and βh1 globins (murine counterparts of the human ε- and γ-globin genes). Additionally, TR2/TR4 function is required for terminal erythroid cell maturation. Loss of TR2/TR4 abolished their occupancy on the εy and βh1 gene promoters, and concurrently impaired co-occupancy by interacting corepressors. These data strongly support the hypothesis that the TR2/TR4 core complex is an adult stage-specific, gene-selective repressor of the embryonic globin genes. Detailed mechanistic understanding of the roles of TR2/TR4 and their cofactors in embryonic and fetal globin gene repression may ultimately enhance the discovery of novel therapeutic agents that can effectively inhibit their transcriptional activity and be safely applied to the treatment of β-globinopathies.
Collapse
|
212
|
Kumar AA, Lim C, Moreno Y, Mace CR, Syed A, Van Tyne D, Wirth DF, Duraisingh MT, Whitesides GM. Enrichment of reticulocytes from whole blood using aqueous multiphase systems of polymers. Am J Hematol 2015; 90:31-6. [PMID: 25263455 DOI: 10.1002/ajh.23860] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 09/24/2014] [Indexed: 01/10/2023]
Abstract
This paper demonstrates the enrichment of reticulocytes by centrifuging whole blood through aqueous multiphase systems (AMPSs)-immiscible phases of solutions of polymers that form step-gradients in density. The interfaces of an AMPS concentrate cells; this concentration facilitates the extraction of blood enriched for reticulocytes. AMPS enrich reticulocytes from blood from both healthy and hemochromatosis donors. Varying the osmolality and density of the phases of AMPS provides different levels of enrichment and yield of reticulocytes. A maximum enrichment of reticulocytemia of 64 ± 3% was obtained from donors with hemochromatosis. When used on peripheral blood from normal donors, AMPS can provide a higher yield of enriched reticulocytes and a higher proportion of reticulocytes expressing CD71 than differential centrifugation followed by centrifugation over Percoll. Blood enriched for reticulocytes by AMPS could be useful for research on malaria. Several species of malaria parasites show a preference to invade young erythrocytes and reticulocytes; this preference complicates in vitro cultivation of these species in human blood. Plasmodium knowlesi malaria parasites invade normal human blood enriched for reticulocytes by AMPSs at a rate 2.2 times greater (P < 0.01) than they invade unenriched blood. Parasite invasion in normal blood enriched by AMPS was 1.8 times greater (P < 0.05) than in blood enriched to a similar reticulocytemia by differential centrifugation followed by centrifugation over Percoll. The enrichment of reticulocytes that are invaded by malaria parasites demonstrates that AMPSs can provide a label-free method to enrich cells for biological research.
Collapse
Affiliation(s)
- Ashok A. Kumar
- School of Engineering and Applied Sciences; Harvard University; Cambridge Massachusetts
| | - Caeul Lim
- Harvard School of Public Health; Harvard University; Boston Massachusetts
| | - Yovany Moreno
- Harvard School of Public Health; Harvard University; Boston Massachusetts
| | - Charles R. Mace
- Department of Chemistry and Chemical Biology; Harvard University; Cambridge Massachusetts
| | - Abeer Syed
- Department of Chemistry and Chemical Biology; Harvard University; Cambridge Massachusetts
| | - Daria Van Tyne
- Harvard School of Public Health; Harvard University; Boston Massachusetts
| | - Dyann F. Wirth
- Harvard School of Public Health; Harvard University; Boston Massachusetts
| | | | - George M. Whitesides
- Department of Chemistry and Chemical Biology; Harvard University; Cambridge Massachusetts
- Wyss Institute for Biologically Inspired Engineering; Harvard University; Cambridge Massachusetts
| |
Collapse
|
213
|
Misener R, Fuentes Garí M, Rende M, Velliou E, Panoskaltsis N, Pistikopoulos EN, Mantalaris A. Global superstructure optimisation of red blood cell production in a parallelised hollow fibre bioreactor. Comput Chem Eng 2014. [DOI: 10.1016/j.compchemeng.2014.10.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
214
|
Noulin F, Manesia JK, Rosanas-Urgell A, Erhart A, Borlon C, Van Den Abbeele J, d'Alessandro U, Verfaillie CM. Hematopoietic stem/progenitor cell sources to generate reticulocytes for Plasmodium vivax culture. PLoS One 2014; 9:e112496. [PMID: 25393299 PMCID: PMC4231068 DOI: 10.1371/journal.pone.0112496] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 10/17/2014] [Indexed: 01/31/2023] Open
Abstract
The predilection of Plasmodium vivax (P. vivax) for reticulocytes is a major obstacle for its establishment in a long-term culture system, as this requires a continuous supply of large quantities of reticulocytes, representing only 1-2% of circulating red blood cells. We here compared the production of reticulocytes using an established in vitro culture system from three different sources of hematopoietic stem/progenitor cells (HSPC), i.e. umbilical cord blood (UCB), bone marrow (BM) and adult peripheral blood (PB). Compared to CD34+-enriched populations of PB and BM, CD34+-enriched populations of UCB produced the highest amount of reticulocytes that could be invaded by P. vivax. In addition, when CD34+-enriched cells were first expanded, a further extensive increase in reticulocytes was seen for UCB, to a lesser degree BM but not PB. As invasion by P. vivax was significantly better in reticulocytes generated in vitro, we also suggest that P. vivax may have a preference for invading immature reticulocytes, which should be confirmed in future studies.
Collapse
Affiliation(s)
- Florian Noulin
- Unit of Malariology, Institute of Tropical Medicine, Antwerp, Belgium
- * E-mail:
| | - Javed Karim Manesia
- Department of development and regeneration, Stem Cell Institute, Leuven, Belgium
| | | | - Annette Erhart
- Unit of Malariology, Institute of Tropical Medicine, Antwerp, Belgium
| | - Céline Borlon
- Unit of Malariology, Institute of Tropical Medicine, Antwerp, Belgium
| | - Jan Van Den Abbeele
- Unit of Veterinary Protozoology, Institute of Tropical Medicine, Antwerp, Belgium
| | | | | |
Collapse
|
215
|
Detection of microparticles from human red blood cells by multiparametric flow cytometry. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2014; 13:274-80. [PMID: 25369588 DOI: 10.2450/2014.0136-14] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 08/26/2014] [Indexed: 12/25/2022]
Abstract
BACKGROUND During storage, red blood cells (RBC) undergo chemical and biochemical changes referred to as "storage lesions". These events determine the loss of RBC integrity, resulting in lysis and release of microparticles. There is growing evidence of the clinical importance of microparticles and their role in blood transfusion-related side effects and pathogen transmission. Flow cytometry is currently one of the most common techniques used to quantify and characterise microparticles. Here we propose multiparametric staining to monitor and quantify the dynamic release of microparticles by stored human RBC. MATERIAL AND METHODS RBC units (n=10) were stored under blood bank conditions for up to 42 days. Samples were tested at different time points to detect microparticles and determine the haemolysis rate (HR%). Microparticles were identified by flow cytometry combining carboxyfluorescein diacetate succinimidyl ester (CFSE) dye, annexin V and anti-glycophorin A antibody. RESULTS We demonstrated that CFSE can be successfully used to label closed vesicles with an intact membrane. The combination of CFSE and glycophorin A antibody was effective for monitoring and quantifying the dynamic release of microparticles from RBC during storage. Double staining with CFSE/glycophorin A was a more precise approach, increasing vesicle detection up to 4.7-fold vs the use of glycophorin A/annexin V alone. Moreover, at all the time points tested, we found a robust correlation (R=0.625; p=0.0001) between HR% and number of microparticles detected. DISCUSSION Multiparametric staining, based on a combination of CFSE, glycophorin A antibody and annexin V, was able to detect, characterise and monitor the release of microparticles from RBC units during storage, providing a sensitive approach to labelling and identifying microparticles for transfusion medicine and, more broadly, for cell-based therapies.
Collapse
|
216
|
Dorn I, Klich K, Arauzo-Bravo MJ, Radstaak M, Santourlidis S, Ghanjati F, Radke TF, Psathaki OE, Hargus G, Kramer J, Einhaus M, Kim JB, Kögler G, Wernet P, Schöler HR, Schlenke P, Zaehres H. Erythroid differentiation of human induced pluripotent stem cells is independent of donor cell type of origin. Haematologica 2014; 100:32-41. [PMID: 25326431 DOI: 10.3324/haematol.2014.108068] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Epigenetic memory in induced pluripotent stem cells, which is related to the somatic cell type of origin of the stem cells, might lead to variations in the differentiation capacities of the pluripotent stem cells. In this context, induced pluripotent stem cells from human CD34(+) hematopoietic stem cells might be more suitable for hematopoietic differentiation than the commonly used fibroblast-derived induced pluripotent stem cells. To investigate the influence of an epigenetic memory on the ex vivo expansion of induced pluripotent stem cells into erythroid cells, we compared induced pluripotent stem cells from human neural stem cells and human cord blood-derived CD34(+) hematopoietic stem cells and evaluated their potential for differentiation into hematopoietic progenitor and mature red blood cells. Although genome-wide DNA methylation profiling at all promoter regions demonstrates that the epigenetic memory of induced pluripotent stem cells is influenced by the somatic cell type of origin of the stem cells, we found a similar hematopoietic induction potential and erythroid differentiation pattern of induced pluripotent stem cells of different somatic cell origin. All human induced pluripotent stem cell lines showed terminal maturation into normoblasts and enucleated reticulocytes, producing predominantly fetal hemoglobin. Differences were only observed in the growth rate of erythroid cells, which was slightly higher in the induced pluripotent stem cells derived from CD34(+) hematopoietic stem cells. More detailed methylation analysis of the hematopoietic and erythroid promoters identified similar CpG methylation levels in the induced pluripotent stem cell lines derived from CD34(+) cells and those derived from neural stem cells, which confirms their comparable erythroid differentiation potential.
Collapse
Affiliation(s)
- Isabel Dorn
- Max Planck Institute for Molecular Biomedicine, Münster, Germany Pediatric Hematology and Oncology, University Hospital Münster, Germany
| | - Katharina Klich
- Max Planck Institute for Molecular Biomedicine, Münster, Germany Institute for Transfusion Medicine and Transplantation Immunology, University Hospital Münster, Germany
| | - Marcos J Arauzo-Bravo
- Group of Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, San Sebastián, Spain IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Martina Radstaak
- Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Simeon Santourlidis
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine-University Düsseldorf, Germany
| | - Foued Ghanjati
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine-University Düsseldorf, Germany
| | - Teja F Radke
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine-University Düsseldorf, Germany
| | | | - Gunnar Hargus
- Max Planck Institute for Molecular Biomedicine, Münster, Germany Institute for Neuropathology, University Hospital Münster, Germany
| | - Jan Kramer
- Medical Department I, University of Lübeck, Germany LADR GmbH, Geesthacht, Germany
| | | | - Jeong Beom Kim
- UNIST, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Gesine Kögler
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine-University Düsseldorf, Germany
| | - Peter Wernet
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine-University Düsseldorf, Germany
| | - Hans R Schöler
- Max Planck Institute for Molecular Biomedicine, Münster, Germany Faculty of Medicine, University of Münster, Germany
| | - Peter Schlenke
- Institute for Transfusion Medicine and Transplantation Immunology, University Hospital Münster, Germany Clinics for Blood Group Serology and Transfusion Medicine, Medical University Graz, Austria
| | - Holm Zaehres
- Max Planck Institute for Molecular Biomedicine, Münster, Germany
| |
Collapse
|
217
|
Fernandez-Becerra C, Lelievre J, Ferrer M, Anton N, Thomson R, Peligero C, Almela MJ, Lacerda MV, Herreros E, Del Portillo HA. Red blood cells derived from peripheral blood and bone marrow CD34⁺ human haematopoietic stem cells are permissive to Plasmodium parasites infection. Mem Inst Oswaldo Cruz 2014; 108:801-3. [PMID: 24037205 PMCID: PMC3970681 DOI: 10.1590/0074-0276108062013019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 07/12/2013] [Indexed: 01/17/2023] Open
Abstract
The production of fully functional human red cells in vitro from haematopoietic
stem cells (hHSCs) has been successfully achieved. Recently, the use of hHSCs
from cord blood represented a major improvement to develop the continuous
culture system for Plasmodium vivax. Here, we demonstrated that
CD34+hHSCs from peripheral blood and bone marrow can be expanded and
differentiated to reticulocytes using a novel stromal cell. Moreover, these
reticulocytes and mature red blood cells express surface markers for entrance of
malaria parasites contain adult haemoglobin and are also permissive to invasion
by P. vivax and Plasmodium falciparum
parasites.
Collapse
Affiliation(s)
- Carmen Fernandez-Becerra
- Barcelona Centre for International Health Research, Hospital Clinic, Universitat de Barcelona, BarcelonaSpain, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
218
|
Abstract
Erythropoiesis is a vital process governed through various factors. There is extreme unavailability of suitable donor due to rare phenotypic blood groups and other related complications like hemoglobinopathies, polytransfusion patients, and polyimmunization. Looking at the worldwide scarcity of blood, especially in low income countries and the battlefield, mimicking erythropoiesis using ex vivo methods can provide an efficient answer to various problems associated with present donor derived blood supply system. Fortunately, there are many ex vivo erythropoiesis methodologies being developed by various research groups using stem cells as the major source material for large scale blood production. Most of these ex vivo protocols use a cocktail of similar growth factors under overlapping growth conditions. Erythropoietin (EPO) is a key regulator in most ex vivo protocols along with other growth factors such as SCF, IL-3, IGF-1, and Flt-3. Now transfusable units of blood can be produced by using these protocols with their set of own limitations. The present paper focuses on the molecular mechanism and significance of various growth factors in these protocols that shall remain helpful for large scale production.
Collapse
|
219
|
Zhu F, Shi L, Li H, Eksi R, Engel JD, Guan Y. Modeling dynamic functional relationship networks and application to ex vivo human erythroid differentiation. ACTA ACUST UNITED AC 2014; 30:3325-33. [PMID: 25115705 DOI: 10.1093/bioinformatics/btu542] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
MOTIVATION Functional relationship networks, which summarize the probability of co-functionality between any two genes in the genome, could complement the reductionist focus of modern biology for understanding diverse biological processes in an organism. One major limitation of the current networks is that they are static, while one might expect functional relationships to consistently reprogram during the differentiation of a cell lineage. To address this potential limitation, we developed a novel algorithm that leverages both differentiation stage-specific expression data and large-scale heterogeneous functional genomic data to model such dynamic changes. We then applied this algorithm to the time-course RNA-Seq data we collected for ex vivo human erythroid cell differentiation. RESULTS Through computational cross-validation and literature validation, we show that the resulting networks correctly predict the (de)-activated functional connections between genes during erythropoiesis. We identified known critical genes, such as HBD and GATA1, and functional connections during erythropoiesis using these dynamic networks, while the traditional static network was not able to provide such information. Furthermore, by comparing the static and the dynamic networks, we identified novel genes (such as OSBP2 and PDZK1IP1) that are potential drivers of erythroid cell differentiation. This novel method of modeling dynamic networks is applicable to other differentiation processes where time-course genome-scale expression data are available, and should assist in generating greater understanding of the functional dynamics at play across the genome during development. AVAILABILITY AND IMPLEMENTATION The network described in this article is available at http://guanlab.ccmb.med.umich.edu/stageSpecificNetwork.
Collapse
Affiliation(s)
- Fan Zhu
- Department of Computational Medicine and Bioinformatics, Department of Cell and Developmental Biology, Department of Internal Medicine and Department of Computer Science and Engineering, University of Michigan, MI48109, USA
| | - Lihong Shi
- Department of Computational Medicine and Bioinformatics, Department of Cell and Developmental Biology, Department of Internal Medicine and Department of Computer Science and Engineering, University of Michigan, MI48109, USA
| | - Hongdong Li
- Department of Computational Medicine and Bioinformatics, Department of Cell and Developmental Biology, Department of Internal Medicine and Department of Computer Science and Engineering, University of Michigan, MI48109, USA
| | - Ridvan Eksi
- Department of Computational Medicine and Bioinformatics, Department of Cell and Developmental Biology, Department of Internal Medicine and Department of Computer Science and Engineering, University of Michigan, MI48109, USA
| | - James Douglas Engel
- Department of Computational Medicine and Bioinformatics, Department of Cell and Developmental Biology, Department of Internal Medicine and Department of Computer Science and Engineering, University of Michigan, MI48109, USA
| | - Yuanfang Guan
- Department of Computational Medicine and Bioinformatics, Department of Cell and Developmental Biology, Department of Internal Medicine and Department of Computer Science and Engineering, University of Michigan, MI48109, USA Department of Computational Medicine and Bioinformatics, Department of Cell and Developmental Biology, Department of Internal Medicine and Department of Computer Science and Engineering, University of Michigan, MI48109, USA Department of Computational Medicine and Bioinformatics, Department of Cell and Developmental Biology, Department of Internal Medicine and Department of Computer Science and Engineering, University of Michigan, MI48109, USA
| |
Collapse
|
220
|
Browne SM, Daud H, Murphy WG, Al-Rubeai M. Measuring dissolved oxygen to track erythroid differentiation of hematopoietic progenitor cells in culture. J Biotechnol 2014; 187:135-8. [PMID: 25107508 DOI: 10.1016/j.jbiotec.2014.07.433] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 07/16/2014] [Accepted: 07/18/2014] [Indexed: 10/24/2022]
Abstract
As stem cell technologies move from the developmental to the commercial stage strategies must be developed to monitor culture operations. These will ensure consistency of differentiation programs and maintenance of optimum cell viability during production runs. Due to the sensitivity of stem cells to their environment, and their variability in response to external stimuli, accurate monitoring of in vitro conditions will be crucial for effective large-scale culturing of therapeutic stem cells. Here we describe a simple method to monitor the expansion and maturation of adult human haematopoietic stem/progenitor cells into red blood cells in vitro by measuring the oxygen consumption rate of cultures. Cell cultures followed a characteristic pattern of oxygen consumption that is reflective of in vivo erythroid maturation. This method could be easily developed as an online system to map erythroid differentiation and maturation of cultured cells as effectively as the more time consuming process of flow cytometric analysis of surface marker expression patterns.
Collapse
Affiliation(s)
- Susan M Browne
- School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin 4, Ireland
| | - Hasbullah Daud
- School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin 4, Ireland
| | - William G Murphy
- School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland; Health Service Executive, Dublin, Ireland
| | - Mohamed Al-Rubeai
- School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
221
|
Simply red: A novel spectrophotometric erythroid proliferation assay as a tool for erythropoiesis and erythrotoxicity studies. ACTA ACUST UNITED AC 2014. [PMID: 28626660 PMCID: PMC5466125 DOI: 10.1016/j.btre.2014.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Most mammalian cell proliferation assays rely on manual or automated cell counting or the assessment of metabolic activity in colorimetric assays, with the former being either labor and time intensive or expensive and the latter being multistep procedures requiring the addition of several reagents. The proliferation of erythroid cells from hematopoietic stem cells and their differentiation into mature red blood cells is characterized by the accumulation of large amounts of hemoglobin. Hemoglobin concentrations are easily quantifiable using spectrophotometric methods due to the specific absorbance peak of the molecule’s heme moiety between 400 and 420 nm. Erythroid proliferation can therefore be readily assessed using spectrophotometric measurement in this range. We have used this feature of erythroid cells to develop a simple erythroid proliferation assay that is minimally labor/time- and reagent-intensive and could easily be automated for use in high-throughput screening. Such an assay can be a valuable tool for investigations into hematological disorders where erythropoiesis is dysregulated, i.e., either inhibited or enhanced, into the development of anemia as a side-effect of primary diseases such as parasitic infections and into cyto-(particularly erythro-) toxicity of chemical agents or drugs.
Collapse
|
222
|
MerTK-mediated engulfment of pyrenocytes by central macrophages in erythroblastic islands. Blood 2014; 123:3963-71. [DOI: 10.1182/blood-2014-01-547976] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Key Points
An in vitro system for the engulfment of pyrenocytes was established using erythroblastic islands. MerTK, a receptor kinase, was essential for the engulfment of pyrenocytes by the central macrophages at erythroblastic islands.
Collapse
|
223
|
Singh VK, Saini A, Tsuji K, Sharma PB, Chandra R. Manufacturing blood ex vivo: a futuristic approach to deal with the supply and safety concerns. Front Cell Dev Biol 2014; 2:26. [PMID: 25364733 PMCID: PMC4206981 DOI: 10.3389/fcell.2014.00026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 05/26/2014] [Indexed: 12/13/2022] Open
Abstract
Blood transfusions are routinely done in every medical regimen and a worldwide established collection, processing/storage centers provide their services for the same. There have been extreme global demands for both raising the current collections and supply of safe/adequate blood due to increasingly demanding population. With, various risks remain associated with the donor derived blood, and a number of post collection blood screening and processing methods put extreme constraints on supply system especially in the underdeveloped countries. A logistic approach to manufacture erythrocytes ex-vivo by using modern tissue culture techniques have surfaced in the past few years. There are several reports showing the possibilities of RBCs (and even platelets/neutrophils) expansion under tightly regulated conditions. In fact, ex vivo synthesis of the few units of clinical grade RBCs from a single dose of starting material such as umbilical cord blood (CB) has been well established. Similarly, many different sources are also being explored for the same purpose, such as embryonic stem cells, induced pluripotent stem cells. However, the major concerns remain elusive before the manufacture and clinical use of different blood components may be used to successfully replace the present system of donor derived blood transfusion. The most important factor shall include the large scale of RBCs production from each donated unit within a limited time period and cost of their production, both of these issues need to be handled carefully since many of the recipients among developing countries are unable to pay even for the freely available donor derived blood. Anyways, keeping these issues in mind, present article shall be focused on the possibilities of blood production and their use in the near future.
Collapse
Affiliation(s)
- Vimal K Singh
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Abhishek Saini
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Kohichiro Tsuji
- Departments of Pediatric Hematology/Oncology, Research Hospital, The Institute of Medical Science, The University of Tokyo Hospital Tokyo, Japan
| | - P B Sharma
- Stem Cell Research Laboratory, Department of Biotechnology, Delhi Technological University Delhi, India
| | - Ramesh Chandra
- Dr B. R. Ambedkar Center for Biomedical Research, University of Delhi Delhi, India
| |
Collapse
|
224
|
Konstantinidis DG, Pushkaran S, Giger K, Manganaris S, Zheng Y, Kalfa TA. Identification of a murine erythroblast subpopulation enriched in enucleating events by multi-spectral imaging flow cytometry. J Vis Exp 2014. [PMID: 24962543 DOI: 10.3791/50990] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Erythropoiesis in mammals concludes with the dramatic process of enucleation that results in reticulocyte formation. The mechanism of enucleation has not yet been fully elucidated. A common problem encountered when studying the localization of key proteins and structures within enucleating erythroblasts by microscopy is the difficulty to observe a sufficient number of cells undergoing enucleation. We have developed a novel analysis protocol using multiparameter high-speed cell imaging in flow (Multi-Spectral Imaging Flow Cytometry), a method that combines immunofluorescent microscopy with flow cytometry, in order to identify efficiently a significant number of enucleating events, that allows to obtain measurements and perform statistical analysis. We first describe here two in vitro erythropoiesis culture methods used in order to synchronize murine erythroblasts and increase the probability of capturing enucleation at the time of evaluation. Then, we describe in detail the staining of erythroblasts after fixation and permeabilization in order to study the localization of intracellular proteins or lipid rafts during enucleation by multi-spectral imaging flow cytometry. Along with size and DNA/Ter119 staining which are used to identify the orthochromatic erythroblasts, we utilize the parameters "aspect ratio" of a cell in the bright-field channel that aids in the recognition of elongated cells and "delta centroid XY Ter119/Draq5" that allows the identification of cellular events in which the center of Ter119 staining (nascent reticulocyte) is far apart from the center of Draq5 staining (nucleus undergoing extrusion), thus indicating a cell about to enucleate. The subset of the orthochromatic erythroblast population with high delta centroid and low aspect ratio is highly enriched in enucleating cells.
Collapse
Affiliation(s)
- Diamantis G Konstantinidis
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine
| | - Suvarnamala Pushkaran
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine
| | - Katie Giger
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine
| | | | - Yi Zheng
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine
| | - Theodosia A Kalfa
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine;
| |
Collapse
|
225
|
Xie X, Li Y, Pei X. From stem cells to red blood cells: how far away from the clinical application? SCIENCE CHINA-LIFE SCIENCES 2014; 57:581-5. [PMID: 24829108 DOI: 10.1007/s11427-014-4667-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Accepted: 04/05/2014] [Indexed: 12/23/2022]
Abstract
The generation of red blood cells (RBCs) from stem cells provides a solution for deficiencies in blood transfusion. Currently, primary hematopoietic stem cells, embryonic stem cells and induced pluripotent stem cells have shown the potential to produce fully mature RBCs. Here, we discuss the advantages, induction protocols, progress and possible clinical applications of stem cells in RBC production.
Collapse
Affiliation(s)
- XiaoYan Xie
- Stem Cell and Regenerative Medicine Lab, Beijing Institute of Transfusion Medicine, Beijing, 100850, China
| | | | | |
Collapse
|
226
|
Wensveen FM, Geest CR, Libregts SFWM, Derks IAM, Ekert PG, Labi V, Villunger A, Nolte MA, Eldering E. BH3-only protein Noxa contributes to apoptotic control of stress-erythropoiesis. Apoptosis 2014; 18:1306-1318. [PMID: 23975731 PMCID: PMC3825139 DOI: 10.1007/s10495-013-0890-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Apoptosis plays an essential role in the control of erythropoiesis under normal and pathological conditions. However, the contribution of individual proteins within cell death signalling pathways remains poorly defined. Here, we investigated the role of the pro-apoptotic Bcl-2 family member Noxa in the regulation of erythropoiesis. We found that expression of Noxa is induced during erythroid differentiation of human and murine precursor cells. Using in vitro model systems for erythroid progenitors, we observed rapid induction of Noxa upon cytokine deprivation. Knockdown or deletion of Noxa conferred significant protection against apoptosis upon cytokine withdrawal. In vivo, Noxa deficiency did not affect hematological blood parameters or erythroid progenitor composition of bone marrow and spleen under steady-state conditions. In contrast, in a model of acute haemolytic anemia, Noxa-deficiency enhanced hematocrit recovery. Moreover, in a model of chronic inflammation-induced anemia, Noxa-ablation resulted in a dramatic increase of erythroblast expansion. Our data indicate that induction of Noxa in erythroid progenitors sets a survival threshold that limits expansion beyond the number of cells that can be sustained by the available cytokines, which becomes apparent under conditions of induced anemia.
Collapse
Affiliation(s)
- Felix M. Wensveen
- Department of Experimental Immunology, Academic Medical Center, Meibergdreef 8, 1105 AZ Amsterdam, The Netherlands
| | - Christian R. Geest
- Department of Experimental Immunology, Academic Medical Center, Meibergdreef 8, 1105 AZ Amsterdam, The Netherlands
| | - Sten F. W. M. Libregts
- Department of Experimental Immunology, Academic Medical Center, Meibergdreef 8, 1105 AZ Amsterdam, The Netherlands
| | - Ingrid A. M. Derks
- Department of Experimental Immunology, Academic Medical Center, Meibergdreef 8, 1105 AZ Amsterdam, The Netherlands
| | - Paul G. Ekert
- Division of Cell Signaling and Cell Death, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Melbourne, VIC Australia
| | - Verena Labi
- Division of Developmental Immunology, BIOCENTER, Innsbruck Medical University, Innrain 80-82, 6020 Innsbruck, Austria
| | - Andreas Villunger
- Division of Developmental Immunology, BIOCENTER, Innsbruck Medical University, Innrain 80-82, 6020 Innsbruck, Austria
| | - Martijn A. Nolte
- Department of Experimental Immunology, Academic Medical Center, Meibergdreef 8, 1105 AZ Amsterdam, The Netherlands
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory AMC/UvA, Amsterdam, The Netherlands
| | - Eric Eldering
- Department of Experimental Immunology, Academic Medical Center, Meibergdreef 8, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
227
|
Biased, non-equivalent gene-proximal and -distal binding motifs of orphan nuclear receptor TR4 in primary human erythroid cells. PLoS Genet 2014; 10:e1004339. [PMID: 24811540 PMCID: PMC4014424 DOI: 10.1371/journal.pgen.1004339] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 03/17/2014] [Indexed: 02/04/2023] Open
Abstract
We previously reported that TR2 and TR4 orphan nuclear receptors bind to direct repeat (DR) elements in the ε- and γ-globin promoters, and act as molecular anchors for the recruitment of epigenetic corepressors of the multifaceted DRED complex, thereby leading to ε- and γ-globin transcriptional repression during definitive erythropoiesis. Other than the ε- and γ-globin and the GATA1 genes, TR4-regulated target genes in human erythroid cells remain unknown. Here, we identified TR4 binding sites genome-wide using chromatin immunoprecipitation followed by massively parallel sequencing (ChIP-seq) as human primary CD34+ hematopoietic progenitors differentiated progressively to late erythroid precursors. We also performed whole transcriptome analyses by RNA-seq to identify TR4 downstream targets after lentiviral-mediated TR4 shRNA knockdown in erythroid cells. Analyses from combined ChIP-seq and RNA-seq datasets indicate that DR1 motifs are more prevalent in the proximal promoters of TR4 direct target genes, which are involved in basic biological functions (e.g., mRNA processing, ribosomal assembly, RNA splicing and primary metabolic processes). In contrast, other non-DR1 repeat motifs (DR4, ER6 and IR1) are more prevalent at gene-distal TR4 binding sites. Of these, approximately 50% are also marked with epigenetic chromatin signatures (such as P300, H3K27ac, H3K4me1 and H3K27me3) associated with enhancer function. Thus, we hypothesize that TR4 regulates gene transcription via gene-proximal DR1 sites as TR4/TR2 heterodimers, while it can associate with novel nuclear receptor partners (such as RXR) to bind to distant non-DR1 consensus sites. In summary, this study reveals that the TR4 regulatory network is far more complex than previously appreciated and that TR4 regulates basic, essential biological processes during the terminal differentiation of human erythroid cells. Sequential genome-wide binding studies investigated by deep sequencing (ChIP-seq) represent a powerful tool for investigating the temporal sequence of gene activation and repression events that take place as cells differentiate. Here, we report the binding of an “orphan” nuclear receptor (one for which no ligand has been identified) to its cognate genomic regulatory sites and perform the functional analysis to validate its downstream targets as precursor cells differentiate from very early human hematopoietic progenitors into red blood cells. We discovered that when this receptor is bound at gene proximal promoters, it recognizes a different DNA sequence than when it binds to more distant regulatory sites (enhancers and silencers). Since this receptor can either activate or repress specific target genes, the data suggest the intriguing possibility that the two different modes of DNA recognition may reflect association of the receptor with different partner molecules when regulating gene expression from proximal or distal sequences.
Collapse
|
228
|
Erythropoietic potential of CD34+ hematopoietic stem cells from human cord blood and G-CSF-mobilized peripheral blood. BIOMED RESEARCH INTERNATIONAL 2014; 2014:435215. [PMID: 24883313 PMCID: PMC4026878 DOI: 10.1155/2014/435215] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Accepted: 03/30/2014] [Indexed: 01/02/2023]
Abstract
Red blood cell (RBC) supply for transfusion has been severely constrained by the limited availability of donor blood and the emergence of infection and contamination issues. Alternatively, hematopoietic stem cells (HSCs) from human organs have been increasingly considered as safe and effective blood source. Several methods have been studied to obtain mature RBCs from CD34+ hematopoietic stem cells via in vitro culture. Among them, human cord blood (CB) and granulocyte colony-stimulating factor-mobilized adult peripheral blood (mPB) are common adult stem cells used for allogeneic transplantation. Our present study focuses on comparing CB- and mPB-derived stem cells in differentiation from CD34+ cells into mature RBCs. By using CD34+ cells from cord blood and G-CSF mobilized peripheral blood, we showed in vitro RBC generation of artificial red blood cells. Our results demonstrate that CB- and mPB-derived CD34+ hematopoietic stem cells have similar characteristics when cultured under the same conditions, but differ considerably with respect to expression levels of various genes and hemoglobin development. This study is the first to compare the characteristics of CB- and mPB-derived erythrocytes. The results support the idea that CB and mPB, despite some similarities, possess different erythropoietic potentials in in vitro culture systems.
Collapse
|
229
|
Larochelle A. Generation of red blood cells in vitro: monitoring the process for improved efficiency. Cytotherapy 2014; 15:1043-5. [PMID: 23911006 DOI: 10.1016/j.jcyt.2013.06.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Andre Larochelle
- National Heart, Lung, and Blood Institute, Hematology Branch, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
230
|
Lee EJ, Godara P, Haylock D. Biomanufacture of human platelets for transfusion: Rationale and approaches. Exp Hematol 2014; 42:332-46. [DOI: 10.1016/j.exphem.2014.02.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 02/07/2014] [Accepted: 02/10/2014] [Indexed: 12/21/2022]
|
231
|
Shi L, Lin YH, Sierant MC, Zhu F, Cui S, Guan Y, Sartor MA, Tanabe O, Lim KC, Engel JD. Developmental transcriptome analysis of human erythropoiesis. Hum Mol Genet 2014; 23:4528-42. [PMID: 24781209 DOI: 10.1093/hmg/ddu167] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
To globally survey the changes in transcriptional landscape during terminal erythroid differentiation, we performed RNA sequencing (RNA-seq) on primary human CD34(+) cells after ex vivo differentiation from the earliest into the most mature erythroid cell stages. This analysis identified thousands of novel intergenic and intronic transcripts as well as novel alternative transcript isoforms. After rigorous data filtering, 51 (presumptive) novel protein-coding transcripts, 5326 long and 679 small non-coding RNA candidates remained. The analysis also revealed two clear transcriptional trends during terminal erythroid differentiation: first, the complexity of transcript diversity was predominantly achieved by alternative splicing, and second, splicing junctional diversity diminished during erythroid differentiation. Finally, 404 genes that were not known previously to be differentially expressed in erythroid cells were annotated. Analysis of the most extremely differentially expressed transcripts revealed that these gene products were all closely associated with hematopoietic lineage differentiation. Taken together, this study will serve as a comprehensive platform for future in-depth investigation of human erythroid development that, in turn, may reveal new insights into multiple layers of the transcriptional regulatory hierarchy that controls erythropoiesis.
Collapse
Affiliation(s)
- Lihong Shi
- Department of Cell and Developmental Biology and
| | - Yu-Hsuan Lin
- Department of Cell and Developmental Biology and Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - M C Sierant
- Department of Cell and Developmental Biology and
| | - Fan Zhu
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | - Yuanfang Guan
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Maureen A Sartor
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Osamu Tanabe
- Department of Cell and Developmental Biology and Department of Integrative Genomics, Tohoku Medical Megabank, Tohoku University, 2-1 Seiryo-machi, Sendai 980-8573, Japan
| | - Kim-Chew Lim
- Department of Cell and Developmental Biology and
| | | |
Collapse
|
232
|
van Veen T, Hunt JA. Tissue engineering red blood cells: a therapeutic. J Tissue Eng Regen Med 2014; 9:760-70. [DOI: 10.1002/term.1885] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 01/14/2014] [Accepted: 02/18/2014] [Indexed: 01/10/2023]
Affiliation(s)
- Theun van Veen
- Clinical Engineering, Institute of Ageing and Chronic Disease; University of Liverpool; UK
| | - John A. Hunt
- Clinical Engineering, Institute of Ageing and Chronic Disease; University of Liverpool; UK
| |
Collapse
|
233
|
Abstract
To date, the use of red blood cells (RBCs) produced from stem cells in vitro has not proved practical for routine transfusion. However, the perpetual and widespread shortage of blood products, problems related to transfusion-transmitted infections, and new emerging pathogens elicit an increasing demand for artificial blood. Worldwide efforts to achieve the goal of RBC production through stem cell research have received vast attention; however, problems with large-scale production and cost effectiveness have yet to prove practical usefulness. Some progress has been made, though, as cord blood stem cells and embryonic stem cells have shown an ability to differentiate and proliferate, and induced pluripotent stem cells have been shown to be an unlimited source for RBC production. However, transfusion of stem cell-derived RBCs still presents a number of challenges to overcome. This paper will summarize an up to date account of research and advances in stem cell-derived RBCs, delineate our laboratory protocol in producing RBCs from cord blood, and introduce the technological developments and limitations to current RBC production practices.
Collapse
Affiliation(s)
- Hyun Ok Kim
- Department of Laboratory Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 120-752, Korea.
| |
Collapse
|
234
|
Li Y, Liu M, Yang ST. Dendritic cells derived from pluripotent stem cells: Potential of large scale production. World J Stem Cells 2014; 6:1-10. [PMID: 24567783 PMCID: PMC3927009 DOI: 10.4252/wjsc.v6.i1.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 10/23/2013] [Accepted: 11/05/2013] [Indexed: 02/06/2023] Open
Abstract
Human pluripotent stem cells (hPSCs), including human embryonic stem cells and human induced pluripotent stem cells, are promising sources for hematopoietic cells due to their unlimited growth capacity and the pluripotency. Dendritic cells (DCs), the unique immune cells in the hematopoietic system, can be loaded with tumor specific antigen and used as vaccine for cancer immunotherapy. While autologous DCs from peripheral blood are limited in cell number, hPSC-derived DCs provide a novel alternative cell source which has the potential for large scale production. This review summarizes recent advances in differentiating hPSCs to DCs through the intermediate stage of hematopoietic stem cells. Step-wise growth factor induction has been used to derive DCs from hPSCs either in suspension culture of embryoid bodies (EBs) or in co-culture with stromal cells. To fulfill the clinical potential of the DCs derived from hPSCs, the bioprocess needs to be scaled up to produce a large number of cells economically under tight quality control. This requires the development of novel bioreactor systems combining guided EB-based differentiation with engineered culture environment. Hence, recent progress in using bioreactors for hPSC lineage-specific differentiation is reviewed. In particular, the potential scale up strategies for the multistage DC differentiation and the effect of shear stress on hPSC differentiation in bioreactors are discussed in detail.
Collapse
|
235
|
Karalyan Z, Simonyan L, Misakyan A, Abroyan L, Hakobyan L, Avetisyan A, Saroyan D. Cell Development in Primary Culture of Porcine Bone Marrow. Cell 2014. [DOI: 10.4236/cellbio.2014.32005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
236
|
Shah S, Huang X, Cheng L. Concise review: stem cell-based approaches to red blood cell production for transfusion. Stem Cells Transl Med 2013; 3:346-55. [PMID: 24361925 DOI: 10.5966/sctm.2013-0054] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Blood transfusion is a common procedure in modern medicine, and it is practiced throughout the world; however, many countries report a less than sufficient blood supply. Even in developed countries where the supply is currently adequate, projected demographics predict an insufficient supply as early as 2050. The blood supply is also strained during occasional widespread disasters and crises. Transfusion of blood components such as red blood cells (RBCs), platelets, or neutrophils is increasingly used from the same blood unit for multiple purposes and to reduce alloimmune responses. Even for RBCs and platelets lacking nuclei and many antigenic cell-surface molecules, alloimmunity could occur, especially in patients with chronic transfusion requirements. Once alloimmunization occurs, such patients require RBCs from donors with a different blood group antigen combination, making it a challenge to find donors after every successive episode of alloimmunization. Alternative blood substitutes such as synthetic oxygen carriers have so far proven unsuccessful. In this review, we focus on current research and technologies that permit RBC production ex vivo from hematopoietic stem cells, pluripotent stem cells, and immortalized erythroid precursors.
Collapse
Affiliation(s)
- Siddharth Shah
- Division of Hematology, Department of Medicine, and Stem Cell Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | |
Collapse
|
237
|
Kouhkan F, Hafizi M, Mobarra N, Mossahebi-Mohammadi M, Mohammadi S, Behmanesh M, Soufi Zomorrod M, Alizadeh S, Lahmy R, Daliri M, Soleimani M. miRNAs: a new method for erythroid differentiation of hematopoietic stem cells without the presence of growth factors. Appl Biochem Biotechnol 2013; 172:2055-69. [PMID: 24326679 DOI: 10.1007/s12010-013-0633-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2013] [Accepted: 10/30/2013] [Indexed: 12/23/2022]
Abstract
Micro RNAs (miRNAs) are a novel class of non-coding regulatory RNA molecules that contribute to post-transcriptional gene regulation. Recent studies have demonstrated that specific miRNAs such as miR-150, miR-154, and miR-451 have key roles in erythropoiesis. To date, stimulatory cytokines are considered as unique effectors for in vitro differentiation of HSCs to erythropoietic lineage. However, the use of these factors is not cost-effective for clinical applications and therapeutic strategies. Here, we present a novel and cost-effective strategy in which miRNAs expression modulation promotes erythroid differentiation in HSCs in the absence of any extrinsic factors. Thus, CD133(+) hematopoietic stem cells purified from human umbilical cord blood were treated with pre-miR-451 containing lentiviruses, anti-miR-150 and anti-miR-154 in the absence of growth factors and cytokines. Obtained results indicated that miR-451 upregulation and miR-150 downregulation have positive effect on GATA-1, FOG-1, and EKLF, CD71 and CD235a genes expression and induce hemoglobinization efficiently. However, downregulation of miR-154 had no effect on erythropoiesis indexes compared to that observed in the control group. In conclusion, the data presented here for the first time demonstrate that expression modulation of miR-451 and miR-150 could be an efficient alternative to stimulatory cytokines for CD133(+) differentiation into erythroid lineage. Modulation of erythropoiesis in stem cells via miRNA holds promising potential for vascular tissue engineering and regenerative medicine applications.
Collapse
Affiliation(s)
- Fatemeh Kouhkan
- Department of Molecular Biology and Genetic Engineering, Stem Cell Technology Research Center, Tehran, Iran
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
238
|
Hirose SI, Takayama N, Nakamura S, Nagasawa K, Ochi K, Hirata S, Yamazaki S, Yamaguchi T, Otsu M, Sano S, Takahashi N, Sawaguchi A, Ito M, Kato T, Nakauchi H, Eto K. Immortalization of erythroblasts by c-MYC and BCL-XL enables large-scale erythrocyte production from human pluripotent stem cells. Stem Cell Reports 2013; 1:499-508. [PMID: 24371805 PMCID: PMC3871399 DOI: 10.1016/j.stemcr.2013.10.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 10/22/2013] [Accepted: 10/22/2013] [Indexed: 02/08/2023] Open
Abstract
The lack of knowledge about the mechanism of erythrocyte biogenesis through self-replication makes the in vitro generation of large quantities of cells difficult. We show that transduction of c-MYC and BCL-XL into multipotent hematopoietic progenitor cells derived from pluripotent stem cells and gene overexpression enable sustained exponential self-replication of glycophorin A(+) erythroblasts, which we term immortalized erythrocyte progenitor cells (imERYPCs). In an inducible expression system, turning off the overexpression of c-MYC and BCL-XL enabled imERYPCs to mature with chromatin condensation and reduced cell size, hemoglobin synthesis, downregulation of GCN5, upregulation of GATA1, and endogenous BCL-XL and RAF1, all of which appeared to recapitulate normal erythropoiesis. imERYPCs mostly displayed fetal-type hemoglobin and normal oxygen dissociation in vitro and circulation in immunodeficient mice following transfusion. Using critical factors to induce imERYPCs provides a model of erythrocyte biogenesis that could potentially contribute to a stable supply of erythrocytes for donor-independent transfusion.
Collapse
Affiliation(s)
- Sho-Ichi Hirose
- Laboratory of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Naoya Takayama
- Laboratory of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan ; Clinical Application Department, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Sou Nakamura
- Laboratory of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan ; Clinical Application Department, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Kazumichi Nagasawa
- Graduate School of Advanced Science and Engineering, Center for Advanced Life and Medical Science, Waseda University, Tokyo 162-8480, Japan
| | - Kiyosumi Ochi
- Laboratory of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan ; Clinical Application Department, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Shinji Hirata
- Clinical Application Department, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| | - Satoshi Yamazaki
- Laboratory of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Tomoyuki Yamaguchi
- Laboratory of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Makoto Otsu
- Laboratory of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Shinya Sano
- Laboratory of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Nobuyasu Takahashi
- Department of Anatomy, Ultrastructural Cell Biology, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Akira Sawaguchi
- Department of Anatomy, Ultrastructural Cell Biology, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Mamoru Ito
- Central Institute for Experimental Animals, Kawasaki 210-0821, Japan
| | - Takashi Kato
- Department of Anatomy, Ultrastructural Cell Biology, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Hiromitsu Nakauchi
- Laboratory of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Koji Eto
- Laboratory of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan ; Clinical Application Department, Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
239
|
Reticulocytes from cryopreserved erythroblasts support Plasmodium vivax infection in vitro. Parasitol Int 2013; 63:278-84. [PMID: 24291603 DOI: 10.1016/j.parint.2013.11.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 11/19/2013] [Accepted: 11/21/2013] [Indexed: 11/20/2022]
Abstract
Plasmodium vivax is the most widely distributed human malaria parasite. Despite its importance, both clinical research and basic research have been hampered by lack of a convenient in vitro culture system, in part due to the parasite's infection preference of reticulocytes rather than mature erythrocytes. The use of reticulocyte-producing hematopoietic stem cell culture has been proposed for the maintenance of the parasite, but good numbers of reticulocytes and P. vivax parasites sufficient for practical use in research have been difficult to produce from this system. Here, we report an improved method of hematopoietic stem cell culture for P. vivax infection, which requires less time and produces higher or equivalent percentage of reticulocytes than previously reported systems. Reticulocytes were cultured from cryopreserved erythroblasts that were frozen after 8day-cultivation of purified CD34+ cells from human umbilical cord blood. This method of production allowed the recovery of reticulocytes in a shorter time than with continuous stem cell culture. We obtained a relatively high percentage of peak reticulocyte production by using co-cultivation with a mouse stromal cell line. Using P. vivax mature stage parasites obtained from infected Aotus monkeys, we observed substantial numbers (up to 0.8% of the total number of the cells) of newly invaded reticulocytes 24h after initial cultivation. The addition of fresh reticulocytes after 48h culture, however, did not result in significant increase of second cycle reticulocyte invasion. Assays of invasion inhibition with specific antibodies were successful with this system, demonstrating potential for study of biological processes as well as the conditions necessary for long-term maintenance of P. vivax in vitro.
Collapse
|
240
|
Sousa BR, Parreira RC, Fonseca EA, Amaya MJ, Tonelli FMP, Lacerda SMSN, Lalwani P, Santos AK, Gomes KN, Ulrich H, Kihara AH, Resende RR. Human adult stem cells from diverse origins: An overview from multiparametric immunophenotyping to clinical applications. Cytometry A 2013; 85:43-77. [DOI: 10.1002/cyto.a.22402] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 09/27/2013] [Accepted: 10/01/2013] [Indexed: 02/06/2023]
Affiliation(s)
- Bruna R. Sousa
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Ricardo C. Parreira
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Emerson A Fonseca
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Maria J. Amaya
- Department of Internal Medicine, Section of Digestive Diseases; Yale University School of Medicine; New Haven Connecticut
| | - Fernanda M. P. Tonelli
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Samyra M. S. N. Lacerda
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Pritesh Lalwani
- Faculdade de Ciências Farmacêuticas; Universidade Federal do Amazonas; Manaus AM Brazil
| | - Anderson K. Santos
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Katia N. Gomes
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| | - Henning Ulrich
- Departamento de Bioquímica; Instituto de Química, Universidade de São Paulo; São Paulo SP Brazil
| | - Alexandre H. Kihara
- Núcleo de Cognição e Sistemas Complexos, Centro de Matemática, Computação e Cognição; Universidade Federal do ABC; Santo André SP Brazil
| | - Rodrigo R. Resende
- Department of Biochemistry and Immunology, Cell Signaling and Nanobiotechnology Laboratory; Federal University of Minas Gerais; Belo Horizonte MG Brazil
| |
Collapse
|
241
|
Sjeklocha LM, Wong PYP, Belcher JD, Vercellotti GM, Steer CJ. β-Globin sleeping beauty transposon reduces red blood cell sickling in a patient-derived CD34(+)-based in vitro model. PLoS One 2013; 8:e80403. [PMID: 24260386 PMCID: PMC3832362 DOI: 10.1371/journal.pone.0080403] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2013] [Accepted: 10/02/2013] [Indexed: 11/18/2022] Open
Abstract
The ultimate goal of gene therapy for sickle cell anemia (SCA) is an improved phenotype for the patient. In this study, we utilized bone marrow from a sickle cell patient as a model of disease in an in vitro setting for the hyperactive Sleeping Beauty transposon gene therapy system. We demonstrated that mature sickle red blood cells containing hemoglobin-S and sickling in response to metabisulfite can be generated in vitro from SCA bone marrow. These cells showed the characteristic morphology and kinetics of hemoglobin-S polymerization, which we quantified using video microscopy and imaging cytometry. Using video assessment, we showed that delivery of an IHK-βT87Q antisickling globin gene by Sleeping Beauty via nucleofection improves metrics of sickling, decreasing percent sickled from 53.2 ± 2.2% to 43.9 ± 2.0%, increasing the median time to sickling from 8.5 to 9.6 min and decreasing the maximum rate of sickling from 2.3 x 10-3 sickling cells/total cells/sec in controls to 1.26 x 10-3 sickling cells/total cells/sec in the IHK-βT87Q-globin group (p < 0.001). Using imaging cytometry, the percentage of elongated sickled cells decreased from 34.8 ± 4.5% to 29.5 ± 3.0% in control versus treated (p < 0.05). These results support the potential use of Sleeping Beauty as a clinical gene therapy vector and provide a useful tool for studying sickle red blood cells in vitro.
Collapse
Affiliation(s)
- Lucas M. Sjeklocha
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Phillip Y.-P. Wong
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - John D. Belcher
- Vascular Biology Center, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Gregory M. Vercellotti
- Vascular Biology Center, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Clifford J. Steer
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
242
|
Rousseau GF, Giarratana MC, Douay L. Large-scale production of red blood cells from stem cells: what are the technical challenges ahead? Biotechnol J 2013; 9:28-38. [PMID: 24408610 DOI: 10.1002/biot.201200368] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 08/05/2013] [Accepted: 09/12/2013] [Indexed: 12/11/2022]
Abstract
Blood-transfusion centers regularly face the challenge of donor blood shortages, especially for rare blood groups. The possibility of producing universal red blood cells from stem cells industrially has become a possible alternative since the successful injection of blood generated in vitro into a human being in 2011. Although there remains many biological and regulatory issues concerning the efficacy and safety of this new product, the major challenge today for future clinical applications is switching from the current limited 2-dimensional production techniques to large-scale 3-dimensional bioreactors. In addition to requiring technological breakthroughs, the whole process also has to become at least five-fold more cost-efficient to match the current prices of high-quality blood products. The current review sums up the main biological advances of the past decade, outlines the key biotechnological challenges for the large-scale cost-effective production of red blood cells, proposes solutions based on strategies used in the bioindustry and presents the state-of-the-art of large-scale blood production.
Collapse
Affiliation(s)
- Guillaume F Rousseau
- UPMC University Paris 6, UMR_S938, Proliferation and Differentiation of Stem Cells, Paris, France; INSERM, UMR_S938, Proliferation and Differentiation of Stem Cells, Paris, France; Université Paris Diderot, Paris, France
| | | | | |
Collapse
|
243
|
Huang X, Shah S, Wang J, Ye Z, Dowey SN, Tsang KM, Mendelsohn LG, Kato GJ, Kickler TS, Cheng L. Extensive ex vivo expansion of functional human erythroid precursors established from umbilical cord blood cells by defined factors. Mol Ther 2013; 22:451-463. [PMID: 24002691 DOI: 10.1038/mt.2013.201] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 08/21/2013] [Indexed: 12/15/2022] Open
Abstract
There is a constant shortage of red blood cells (RBCs) from sufficiently matched donors for patients who need chronic transfusion. Ex vivo expansion and maturation of human erythroid precursors (erythroblasts) from the patients or optimally matched donors could represent a potential solution. Proliferating erythroblasts can be expanded from umbilical cord blood mononuclear cells (CB MNCs) ex vivo for 10(6)-10(7)-fold (in ~50 days) before proliferation arrest and reaching sufficient number for broad application. Here, we report that ectopic expression of three genetic factors (Sox2, c-Myc, and an shRNA against TP53 gene) associated with iPSC derivation enables CB-derived erythroblasts to undergo extended expansion (~10(68)-fold in ~12 months) in a serum-free culture condition without change of cell identity or function. These expanding erythroblasts maintain immature erythroblast phenotypes and morphology, a normal diploid karyotype and dependence on a specific combination of growth factors for proliferation throughout expansion period. When being switched to a terminal differentiation condition, these immortalized erythroblasts gradually exit cell cycle, decrease cell size, accumulate hemoglobin, condense nuclei and eventually give rise to enucleated hemoglobin-containing erythrocytes that can bind and release oxygen. Our result may ultimately lead to an alternative approach to generate unlimited numbers of RBCs for personalized transfusion medicine.
Collapse
Affiliation(s)
- Xiaosong Huang
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Stem Cell Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Siddharth Shah
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Stem Cell Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jing Wang
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Stem Cell Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zhaohui Ye
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Stem Cell Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sarah N Dowey
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Stem Cell Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kit Man Tsang
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Stem Cell Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Laurel G Mendelsohn
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Gregory J Kato
- Hematology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Thomas S Kickler
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Linzhao Cheng
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Stem Cell Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
244
|
Li X, Wu Z, Fu X, Han W. How Far Are Stem-Cell-Derived Erythrocytes from the Clinical Arena? Bioscience 2013. [DOI: 10.1525/bio.2013.63.8.6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
245
|
Bustos RI, Jensen EL, Ruiz LM, Rivera S, Ruiz S, Simon F, Riedel C, Ferrick D, Elorza AA. Copper deficiency alters cell bioenergetics and induces mitochondrial fusion through up-regulation of MFN2 and OPA1 in erythropoietic cells. Biochem Biophys Res Commun 2013; 437:426-32. [DOI: 10.1016/j.bbrc.2013.06.095] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Accepted: 06/25/2013] [Indexed: 12/23/2022]
|
246
|
Romero Z, Urbinati F, Geiger S, Cooper AR, Wherley J, Kaufman ML, Hollis RP, Ruiz de Assin R, Senadheera S, Sahagian A, Jin X, Gellis A, Wang X, Gjertson D, DeOliveira S, Kempert P, Shupien S, Abdel-Azim H, Walters MC, Meiselman HJ, Wenby RB, Gruber T, Marder V, Coates TD, Kohn DB. β-globin gene transfer to human bone marrow for sickle cell disease. J Clin Invest 2013; 123:67930. [PMID: 23863630 PMCID: PMC4011030 DOI: 10.1172/jci67930] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Accepted: 05/02/2013] [Indexed: 12/20/2022] Open
Abstract
Autologous hematopoietic stem cell gene therapy is an approach to treating sickle cell disease (SCD) patients that may result in lower morbidity than allogeneic transplantation. We examined the potential of a lentiviral vector (LV) (CCL-βAS3-FB) encoding a human hemoglobin (HBB) gene engineered to impede sickle hemoglobin polymerization (HBBAS3) to transduce human BM CD34+ cells from SCD donors and prevent sickling of red blood cells produced by in vitro differentiation. The CCL-βAS3-FB LV transduced BM CD34+ cells from either healthy or SCD donors at similar levels, based on quantitative PCR and colony-forming unit progenitor analysis. Consistent expression of HBBAS3 mRNA and HbAS3 protein compromised a fourth of the total β-globin-like transcripts and hemoglobin (Hb) tetramers. Upon deoxygenation, a lower percentage of HBBAS3-transduced red blood cells exhibited sickling compared with mock-transduced cells from sickle donors. Transduced BM CD34+ cells were transplanted into immunodeficient mice, and the human cells recovered after 2-3 months were cultured for erythroid differentiation, which showed levels of HBBAS3 mRNA similar to those seen in the CD34+ cells that were directly differentiated in vitro. These results demonstrate that the CCL-βAS3-FB LV is capable of efficient transfer and consistent expression of an effective anti-sickling β-globin gene in human SCD BM CD34+ progenitor cells, improving physiologic parameters of the resulting red blood cells.
Collapse
Affiliation(s)
- Zulema Romero
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Fabrizia Urbinati
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Sabine Geiger
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Aaron R. Cooper
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Jennifer Wherley
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Michael L. Kaufman
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Roger P. Hollis
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Rafael Ruiz de Assin
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Shantha Senadheera
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Arineh Sahagian
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Xiangyang Jin
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Alyse Gellis
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Xiaoyan Wang
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - David Gjertson
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Satiro DeOliveira
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Pamela Kempert
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Sally Shupien
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Hisham Abdel-Azim
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Mark C. Walters
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Herbert J. Meiselman
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Rosalinda B. Wenby
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Theresa Gruber
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Victor Marder
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Thomas D. Coates
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Donald B. Kohn
- Department of Microbiology, Immunology and Molecular Genetics,
Molecular Biology Interdepartmental Ph.D. Program,
Department of Medicine Statistics Core,
Department of Biostatistics, School of Public Health, and
Division of Pediatric Hematology/Oncology, Department of Pediatrics, UCLA, Los Angeles, California, USA.
Division of Research Immunology/Bone Marrow Transplantation, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Children’s Hospital and Research Center, Oakland, California, USA.
Department of Physiology and Biophysics, University of Southern California Keck School of Medicine, Los Angeles, California, USA.
Division of Hematology and Medical Oncology, Department of Medicine, UCLA, Los Angeles, California, USA.
Division of Hematology/Oncology, Department of Pediatrics, Childrens Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| |
Collapse
|
247
|
Zaid T, Frömmel C, Lun A, Moldenhauer A. Erythropoietin-stimulated endothelial cells support erythroid cell differentiation of CD34(+) haematopoietic progenitors. Vox Sang 2013; 105:253-8. [PMID: 23773054 DOI: 10.1111/vox.12046] [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: 05/31/2012] [Revised: 01/22/2013] [Accepted: 03/20/2013] [Indexed: 11/30/2022]
Abstract
BACKGROUND AND OBJECTIVES Endothelial cells provide a unique medium for the proliferation and white lineage differentiation of haematopoietic progenitor cells (HPC). Whether this quality can be exploited to facilitate the differentiation of erythroid precursors is not yet known. MATERIALS AND METHODS Haematopoietic progenitor cells derived from cord blood were cultured for 3 weeks in erythropoietin-stimulated supernatants with (n = 6) or without cyclosporine A (CSA, n = 6). Cell count, phenotype and morphology were assessed on a weekly basis, and the haemoglobin content was analysed. These cultures were compared with erythroid differentiation induced by cytokines only (n = 6). RESULTS Endothelial supernatants combined with CSA led to equivalent numbers of CD71(+) erythroblasts after 1 week as cytokines only. The purity of glycophorin-positive, CD45-negative cells was higher in cells generated in endothelial supernatants than in cytokine-based media. Additional prostaglandin E2 induced a change from fetal to adult haemoglobin. CONCLUSION For the generation of erythroblasts from HPC, endothelial supernatants are a simple and cost-effective alternative to culture conditions based on cytokines.
Collapse
Affiliation(s)
- T Zaid
- Institute for Transfusion Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | |
Collapse
|
248
|
Juutistenaho S, Möttönen S, Eskola M, Aranko K, Kekomäki R. Growth of erythroid cells from thawed unseparated cord blood in vitro without exogenous erythropoietin. Transfus Apher Sci 2013; 49:193-9. [PMID: 23683500 DOI: 10.1016/j.transci.2013.04.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2011] [Revised: 01/21/2013] [Accepted: 04/25/2013] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Previous erythroid cell cultures have depended on added serum or erythropoietin. In this paper, the growth of erythroid cells from thawed unseparated cord blood units in vitro without serum or exogenous erythropoietin is reported. METHODS Thawed volume-reduced cord blood was cultured in conditions designed to support the megakaryocytic lineage, with thrombopoietin and interleukins 3 and 6. Erythroid cells were detected with glycophorin A (GlyA), CD71, and benzidine (flow cytometry and immunocytochemistry). RESULTS Nucleated and anucleated GlyA-positive, as well as benzidine-positive cells were observed from day 9. In flow cytometry, at days 0 and 9, 5.9% and 14% of all events were GlyA+, and 14% and 53% were CD71+, respectively. At days 0 and 9, 4.5% and 12% of the events were double-positive for GlyA and CD71, respectively. By day 14, the percentages of GlyA+, CD71+ and double-positive events had started to decrease (9.7%, 35%, and 5.3%, respectively). CONCLUSIONS Erythroid cells were generated from thawed unseparated cord blood units without exogenous erythropoietin. Thawed cord blood possesses the potential for erythroid growth in vitro in a culture medium designed for other cell types.
Collapse
Affiliation(s)
- Sari Juutistenaho
- Finnish Red Cross Blood Service, Kivihaantie 7, 00310 Helsinki, Finland.
| | | | | | | | | |
Collapse
|
249
|
Boehm D, Mazurier C, Giarratana MC, Darghouth D, Faussat AM, Harmand L, Douay L. Caspase-3 is involved in the signalling in erythroid differentiation by targeting late progenitors. PLoS One 2013; 8:e62303. [PMID: 23658722 PMCID: PMC3642196 DOI: 10.1371/journal.pone.0062303] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 03/23/2013] [Indexed: 02/07/2023] Open
Abstract
A role for caspase activation in erythroid differentiation has been established, yet its precise mode of action remains elusive. A drawback of all previous investigations on caspase activation in ex vivo erythroid differentiation is the lack of an in vitro model producing full enucleation of erythroid cells. Using a culture system which renders nearly 100% enucleated red cells from human CD34(+) cells, we investigated the role of active caspase-3 in erythropoiesis. Profound effects of caspase-3 inhibition were found on erythroid cell growth and differentiation when inhibitors were added to CD34(+) cells at the start of the culture and showed dose-response to the concentration of inhibitor employed. Enucleation was only reduced as a function of the reduced maturity of the culture and the increased cell death of mature cells while the majority of cells retained their ability to extrude their nuclei. Cell cycle analysis after caspase-3 inhibition showed caspase-3 to play a critical role in cell proliferation and highlighted a novel function of this protease in erythroid differentiation, i.e. its contribution to cell cycle regulation at the mitotic phase. While the effect of caspase-3 inhibitor treatment on CD34(+) derived cells was not specific to the erythroid lineage, showing a similar reduction of cell expansion in myeloid cultures, the mechanism of action in both lineages appeared to be distinct with a strong induction of apoptosis causing the decreased yield of myeloid cells. Using a series of colony-forming assays we were able to pinpoint the stage at which cells were most sensitive to caspase-3 inhibition and found activated caspase-3 to play a signalling role in erythroid differentiation by targeting mature BFU-E and CFU-E but not early BFU-E.
Collapse
Affiliation(s)
- Daniela Boehm
- Université Pierre et Marie Curie - Paris 6, UMR_S938 CDR Saint-Antoine, Prolifération et Différentiation des Cellules Souches, Paris, France
- INSERM, UMR_S938, Prolifération et Différentiation des Cellules Souches, Paris, France
| | - Christelle Mazurier
- Université Pierre et Marie Curie - Paris 6, UMR_S938 CDR Saint-Antoine, Prolifération et Différentiation des Cellules Souches, Paris, France
- INSERM, UMR_S938, Prolifération et Différentiation des Cellules Souches, Paris, France
- Etablissement Français du Sang Ile de France, Unité d'Ingénierie et de Thérapie Cellulaire, Créteil, France
| | - Marie-Catherine Giarratana
- Université Pierre et Marie Curie - Paris 6, UMR_S938 CDR Saint-Antoine, Prolifération et Différentiation des Cellules Souches, Paris, France
- INSERM, UMR_S938, Prolifération et Différentiation des Cellules Souches, Paris, France
| | - Dhouha Darghouth
- Université Pierre et Marie Curie - Paris 6, UMR_S938 CDR Saint-Antoine, Prolifération et Différentiation des Cellules Souches, Paris, France
- INSERM, UMR_S938, Prolifération et Différentiation des Cellules Souches, Paris, France
| | - Anne-Marie Faussat
- Université Pierre et Marie Curie - Paris 6, UMR_S938 CDR Saint-Antoine, Prolifération et Différentiation des Cellules Souches, Paris, France
- IFR 65-St Antoine, Université Pierre et Marie Curie - Paris 6, Plateforme de Cytométrie, Paris, France
| | - Laurence Harmand
- Université Pierre et Marie Curie - Paris 6, UMR_S938 CDR Saint-Antoine, Prolifération et Différentiation des Cellules Souches, Paris, France
- INSERM, UMR_S938, Prolifération et Différentiation des Cellules Souches, Paris, France
- Etablissement Français du Sang Ile de France, Unité d'Ingénierie et de Thérapie Cellulaire, Créteil, France
| | - Luc Douay
- Université Pierre et Marie Curie - Paris 6, UMR_S938 CDR Saint-Antoine, Prolifération et Différentiation des Cellules Souches, Paris, France
- INSERM, UMR_S938, Prolifération et Différentiation des Cellules Souches, Paris, France
- Etablissement Français du Sang Ile de France, Unité d'Ingénierie et de Thérapie Cellulaire, Créteil, France
- IFR 65-St Antoine, Université Pierre et Marie Curie - Paris 6, Plateforme de Cytométrie, Paris, France
- Assistance Publique - Hôpitaux de Paris, Hôpital St Antoine et Hôpital Trousseau, Service d'Hématologie Biologique, Paris, France
- * E-mail:
| |
Collapse
|
250
|
Noulin F, Borlon C, Van Den Abbeele J, D'Alessandro U, Erhart A. 1912-2012: a century of research on Plasmodium vivax in vitro culture. Trends Parasitol 2013; 29:286-94. [PMID: 23623759 DOI: 10.1016/j.pt.2013.03.012] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 03/22/2013] [Accepted: 03/29/2013] [Indexed: 01/17/2023]
Abstract
The development of a continuous Plasmodium vivax blood cycle in vitro was first attempted 100 years ago. Since then, and despite the use of different methods, only short-term cultures have been achieved so far. The available literature has been reviewed in order to provide a critical overview of the currently available knowledge on P. vivax blood cycle culture systems and identify some unexplored ways forward. Results show that data accumulated over the past century remain fragmented and often contradictory, making it difficult to draw conclusions. There is the need for an international consortium on P. vivax culture able to collect, update, and share new evidence, including negative results, and thus better coordinate current efforts towards the establishment of a continuous P. vivax culture.
Collapse
Affiliation(s)
- Florian Noulin
- Unit of Malariology, Institute of Tropical Medicine, Antwerp, 2000, Belgium.
| | | | | | | | | |
Collapse
|