351
|
Migliaccio G, Sanchez M, Masiello F, Tirelli V, Varricchio L, Whitsett C, Migliaccio AR. Humanized culture medium for clinical expansion of human erythroblasts. Cell Transplant 2010; 19:453-69. [PMID: 20149301 DOI: 10.3727/096368909x485049] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Ex vivo-generated erythroblasts represent alternative transfusion products. However, inclusion of bovine components in media used for their growth precludes clinical use, highlighting the importance of developing culture media based on pharmaceutical grade reagents. In addition, because adult blood generates ex vivo lower numbers of erythroblasts than cord blood, cord blood has been proposed as the source of choice for ex vivo erythroblast production. To clarify the potential of adult blood to generate erythroblasts ex vivo, experiments were designed to identify growth factors [stem cell factor (SCF), interleukin-3 (IL-3), erythropoietin (EPO), and/or thrombopoietin (TPO)] and the optimal concentration and addition schedule of hormones (dexamethasone and estradiol) sustaining maximal erythroid amplification from adult blood mononuclear cells (MNC) using media with serum previously defined as human erythroid massive amplification culture (HEMA(ser)). Adult MNC stimulated with SCF and IL-3 in combination with EPO generated a 6-12-fold increase in erythroid cells while TPO was ineffective. Dexamethasone and estradiol (both at 10(-6) M) exerted partially overlapping but nonredundant functions. Dexamethasone was indispensable in the first 10 days of culture while estradiol was required from day 10 on. The growth factor and hormone combinations identified in HEMA(ser) were then used to formulate a media composed of dialyzed pharmaceutical grade human albumin, human albumin-lipid liposomes, and iron-saturated recombinant human tranferrin (HEMA(def)). HEMA(def) sustained erythroid amplification as efficiently as HEMA(ser) for cord blood MNC and 10-fold higher than HEMA(ser) for adult blood MNC. In fact, the numbers of erythroblasts generated in HEMA(def) by adult MNC were similar to those generated by cord blood MNC. In conclusion, this study identifies growth factors, hormone combinations, and human protein-based media that allow similar levels of ex vivo erythroid expansion from adult and cord blood MNC, paving the way to evaluate adult blood as a source of ex vivo-expanded erythroblasts for transfusion.
Collapse
Affiliation(s)
- Giovanni Migliaccio
- Division of Hematology and Oncology, Tisch Cancer Institute, New York, NY 10029, USA
| | | | | | | | | | | | | |
Collapse
|
352
|
Lu SJ, Feng Q, Park JS, Lanza R. Directed differentiation of red blood cells from human embryonic stem cells. Methods Mol Biol 2010; 636:105-21. [PMID: 20336519 DOI: 10.1007/978-1-60761-691-7_7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Human embryonic stem cells (hESC) represent a new source of stem cells that can be propagated and expanded in vitro indefinitely, providing a potentially inexhaustible and donorless source of cells for human therapy. The ability to create banks of hESC lines with matched or reduced incompatibility could potentially reduce or eliminate the need for immunosuppressive drugs and/or immunomodulatory protocols altogether, for example, O-type RhD(-) lines for generation of universal red blood cells (RBC). Hematopoietic differentiation of hESCs has been extensively investigated in vitro, and hematopoietic precursors as well as differentiated progeny representing erythroid, myeloid, macrophage, megakaryocytic, and lymphoid lineages have been identified in differentiating hESC cultures. Previous studies also generated primitive erythroid cells from hESCs by embryoid body (EB) formation and coculturing with stromal cells. However, the efficient and controlled differentiation of hESCs into homogeneous RBC populations with oxygen-carrying capacity has not been previously achieved. In this chapter, we describe a robust system that can efficiently generate large numbers of hemangioblasts from multiple hESC lines using well-defined conditions and produce functional homogeneous RBCs with oxygen-carrying capacity in large scale. The homogeneous erythroid cells can be used for further mechanism studies.
Collapse
|
353
|
Seifinejad A, Taei A, Totonchi M, Vazirinasab H, Hassani SN, Aghdami N, Shahbazi E, Yazdi RS, Salekdeh GH, Baharvand H. Generation of human induced pluripotent stem cells from a Bombay individual: moving towards "universal-donor" red blood cells. Biochem Biophys Res Commun 2010; 391:329-34. [PMID: 19912985 DOI: 10.1016/j.bbrc.2009.11.058] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2009] [Accepted: 11/09/2009] [Indexed: 01/22/2023]
Abstract
Bombay phenotype is one of the rare phenotypes in the ABO blood group system that fails to express ABH antigens on red blood cells. Nonsense or missense mutations in fucosyltransfrase1 (FUT1) and fucosyltransfrase2 (FUT2) genes are known to create this phenotype. This blood group is compatible with all other blood groups as a donor, as it does not express the H antigen on the red blood cells. In this study, we describe the establishment of human induced pluripotent stem cells (iPSCs) from the dermal fibroblasts of a Bombay blood-type individual by the ectopic expression of established transcription factors Klf4, Oct4, Sox2, and c-Myc. Sequence analyses of fibroblasts and iPSCs revealed a nonsense mutation 826C to T (276 Gln to Ter) in the FUT1 gene and a missense mutation 739G to A (247 Gly to Ser) in the FUT2 gene in the Bombay phenotype under study. The established iPSCs resemble human embryonic stem cells in morphology, passaging, surface and pluripotency markers, normal karyotype, gene expression, DNA methylation of critical pluripotency genes, and in-vitro differentiation. The directed differentiation of the iPSCs into hematopoietic lineage cells displayed increased expression of the hematopoietic lineage markers such as CD34, CD133, RUNX1, KDR, alpha-globulin, and gamma-globulin. Such specific stem cells provide an unprecedented opportunity to produce a universal blood group donor, in-vitro, thus enabling cellular replacement therapies, once the safety issue is resolved.
Collapse
Affiliation(s)
- Ali Seifinejad
- Department of Stem Cells and Developmental Biology, Royan Institute for Stem Cell Biology and Technology, PO Box 19395-4644, ACECR, Tehran, Iran
| | | | | | | | | | | | | | | | | | | |
Collapse
|
354
|
Abstract
The field of cord blood transplantation has come a long way since the first transplant more than 20 years ago. Advancements in the field will require continuing efforts to better understand hematopoietic stem and progenitor cell function and engraftment. Cautious optimism is inherent in the potential relevance and applicability of nonhematopoietic stem and progenitor cell types found in cord blood, and induced pluripotent stem cells generated from cord blood cells. Rigorous investigations and close interactions between scientific and clinical investigators are required to translate human in vitro and animal in vivo findings into clinical utility.
Collapse
Affiliation(s)
- Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202-5181, USA.
| |
Collapse
|
355
|
Expression of human cytokines dramatically improves reconstitution of specific human-blood lineage cells in humanized mice. Proc Natl Acad Sci U S A 2009; 106:21783-8. [PMID: 19966223 DOI: 10.1073/pnas.0912274106] [Citation(s) in RCA: 193] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Adoptive transfer of human hematopoietic stem cells (HSCs) into mice lacking T, B and natural killer (NK) cells leads to development of human-blood lineage cells in the recipient mice (humanized mice). Although human B cell reconstitution is robust and T cell reconstitution is reasonable in the recipient mice, reconstitution of NK cells and myeloid cells is generally poor or undetectable. Here, we show that the poor reconstitution is mainly the result of a deficiency of appropriate human cytokines that are necessary for the development and maintenance of these cell lineages. When plasmid DNA encoding human IL-15 and Flt-3/Flk-2 ligand were delivered into humanized mice by hydrodynamic tail-vein injection, the expression of the human cytokine lasted for 2 to 3 weeks and elevated levels of NK cells were induced for more than a month. The cytokine-induced NK cells expressed both activation and inhibitory receptors, killed target cells in vitro, and responded robustly to a virus infection in vivo. Similarly, expression of human GM-CSF and IL-4, macrophage colony stimulating factor, or erythropoietin and IL-3 resulted in significantly enhanced reconstitution of dendritic cells, monocytes/macrophages, or erythrocytes, respectively. Thus, human cytokine gene expression by hydrodynamic delivery is a simple and efficient method to improve reconstitution of specific human-blood cell lineages in humanized mice, providing an important tool for studying human immune responses and disease progression in a small animal model.
Collapse
|
356
|
Varricchio L, Fabucci ME, Alfani E, Godbold J, Migliaccio AR. Compensated variability in the expression of globin-related genes in erythroblasts generated ex vivo from different donors. Transfusion 2009; 50:672-84. [PMID: 19891622 DOI: 10.1111/j.1537-2995.2009.02483.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Ex vivo generated erythroblasts are being evaluated for transfusion. Expression of balanced levels of globin mRNA is essential for normal red blood cell function and survival but it is unknown whether the expression of the globin genes in ex vivo expanded cells is balanced. STUDY DESIGN AND METHODS Immature erythroblasts (IEs) were expanded in human erythroid massive amplification cultures from blood mononuclear cells of 19 normal donors and four beta(0)-thalassemia patients (for comparison) and induced to mature for 4 days in the presence of erythropoietin. mRNA was prepared from IEs and mature erythroblasts to evaluate the expression of alpha-, beta-, and gamma-globin genes and of adult hemoglobin-stabilizing protein (AHSP) and BCL11A, two proteins directly controlling globin function and/or production. Results were analyzed using Pearson's correlation coefficient, the Wilcoxon signed rank, and the Mann-Whitney rank sum tests. RESULTS The absolute levels of globin, AHSP, and BCL11A mRNA expressed by erythroblasts generated ex vivo from normal donors were distributed along a 2-log range. With maturation, the levels of gamma-globin and BCL11A mRNA did not decrease while those of alpha-globin, gamma + beta-globins, and AHSP mRNA greatly increased. In normal cells, the modest imbalance (two- to fourfold) observed between alpha- and gamma + beta-globin mRNA was fully compensated by AHSP expression. Thus, the levels of alpha-globin mRNA were correlated with those of gamma + beta-globin (R(2) = 0.93, p < 0.0001) and AHSP (R(2) = 0.86, p < 0.0001). CONCLUSIONS Ex vivo expanded erythroblasts from normal donors express modestly imbalanced levels of alpha-globin and gamma + beta-globin fully compensated by AHSP expression, likely ensuring normal function and survival.
Collapse
|
357
|
Baek EJ, Kim HS, Kim JH, Kim NJ, Kim HO. Stroma-free mass production of clinical-grade red blood cells (RBCs) by using poloxamer 188 as an RBC survival enhancer. Transfusion 2009; 49:2285-95. [DOI: 10.1111/j.1537-2995.2009.02303.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
358
|
|
359
|
Sun H, Dertinger SD, Hyrien O, David Wu JH, Chen Y. Gamma-radiation induces micronucleated reticulocytes in 3D bone marrow bioreactors in vitro. Mutat Res 2009; 680:49-55. [PMID: 19786117 PMCID: PMC2843784 DOI: 10.1016/j.mrgentox.2009.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Revised: 09/12/2009] [Accepted: 09/16/2009] [Indexed: 10/20/2022]
Abstract
Radiation injury to the bone marrow is potentially lethal due to the potent DNA-damaging effects on cells of the hematopoietic system, including bone marrow stem cell, progenitor, and the precursor cell populations. Investigation of radiation genotoxic effects on bone marrow progenitor/precursor cells has been challenged by the lack of optimal in vitro surrogate organ culture systems, and the overall difficulty to sustain lineage-specific proliferation and differentiation of hematopoiesis in vitro. We report the investigation of radiation genotoxic effects in bone marrow cultures of C57Bl/6 mice established in 3D bioreactors, which sustain long-term bone marrow cultures. For these studies, genotoxicity is measured by the induction of micronucleated reticulocytes (MN-RETs). The kinetics and dose-response relationship of MN-RET induction in response to gamma-radiation of bioreactor-maintained bone marrow cultures are presented. Our data showed that 3D long-term bone marrow cultures had sustained erythropoiesis capable of generating reticulocytes up to 8 weeks. The peak time-interval of viable cell output and percentage of reticulocytes increased steadily and reached the initial peak between the 14th and 21st days after inoculations. This was followed by a rebound or staying relatively constant until week 8. The percentage of MN-RET reached the maximum between 24 h and 32 h post 1 Gy gamma-ray. There was a near linear MN-RET induction by gamma-radiation from 0 Gy to 1.0 Gy, followed by an attenuated increase to 1.5-2.0 Gy. The MN-RET response showed a downtrend beyond 2 Gy. Our data suggest that bone marrow culture in the 3D bioreactor may be a useful organ culture system for the investigation of radiation genotoxic effect in vitro.
Collapse
Affiliation(s)
- Hongliang Sun
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY
- Department of Chemical Engineering, University of Rochester, Rochester, NY
| | | | - Ollivier Hyrien
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY
| | - J. H. David Wu
- Department of Chemical Engineering, University of Rochester, Rochester, NY
| | - Yuhchyau Chen
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
360
|
Garderet L, Kobari L, Mazurier C, De Witte C, Giarratana MC, Pérot C, Gorin NC, Lapillonne H, Douay L. Unimpaired terminal erythroid differentiation and preserved enucleation capacity in myelodysplastic 5q(del) clones: a single cell study. Haematologica 2009; 95:398-405. [PMID: 19815832 DOI: 10.3324/haematol.2009.012773] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Anemia is a characteristic of myelodysplastic syndromes, such as the rare 5q- syndrome, but its mechanism remains unclear. In particular, data are lacking on the terminal phase of differentiation of erythroid cells (enucleation) in myelodysplastic syndromes. DESIGN AND METHODS We used a previously published culture model to generate mature red blood cells in vitro from human hematopoietic progenitor cells in order to study the pathophysiology of the 5q- syndrome. Our model enables analysis of cell proliferation and differentiation at a single cell level and determination of the enucleation capacity of erythroid precursors. RESULTS The erythroid commitment of 5q(del) clones was not altered and their terminal differentiation capacity was preserved since they achieved final erythroid maturation (enucleation stage). The drop in red blood cell production was secondary to the decrease in the erythroid progenitor cell pool and to impaired proliferative capacity. RPS14 gene haploinsufficiency was related to defective erythroid proliferation but not to differentiation capacity. CONCLUSIONS The 5q- syndrome should be considered a quantitative rather than qualitative bone marrow defect. This observation might open the way to new therapeutic concepts.
Collapse
|
361
|
Abstract
PURPOSE OF REVIEW Red blood cells (RBCs) transfusion plays a critical role in numerous therapies. Disruption of blood collection by political unrest, natural disasters and emerging infections and implementation of restrictions on the use of erythropoiesis-stimulating agents in cancer may impact blood availability in the near future. These considerations highlight the importance of developing alternative blood products. RECENT FINDINGS Knowledge about the processes that control RBC production has been applied to the establishment of culture conditions allowing ex-vivo generation of RBCs in numbers close to those (2.5 x 10 cells/ml) present in a transfusion, from cord blood, donated blood units or embryonic stem cells. In addition, experimental studies demonstrate that such cells protect mice from lethal bleeding. Therefore, erythroid cells generated ex vivo may be suitable for transfusion provided they can be produced safely in adequate numbers. However, much remains to be done to translate a theoretical production of approximately 2.5 x 10 RBCs in the laboratory into a 'clinical grade production process'. SUMMARY This review summarizes the state-of-the-art in establishing ex-vivo culture conditions for erythroid cells and discusses the most compelling issues to be addressed to translate this progress into a clinical grade transfusion product.
Collapse
|
362
|
The potential of human peripheral blood derived CD34+ cells for ex vivo red blood cell production. J Biotechnol 2009; 144:127-34. [PMID: 19735679 DOI: 10.1016/j.jbiotec.2009.08.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2009] [Revised: 07/16/2009] [Accepted: 08/31/2009] [Indexed: 11/20/2022]
Abstract
The potential of peripheral blood derived CD34+ cells for ex vivo erythropoiesis was investigated in a stroma-free culture system using a novel strategy of daily passaging. By expanding PB-derived CD34+ cells up to 1.5 x 10(6)-fold this method achieved expansion factors previously only reported for CD34+ cells derived from more potent stem cell sources such as cord blood, bone marrow and mobilized peripheral blood. Analysis of cell surface markers showed differentiation of immature CD34+ cells to populations with 80% CD71-/GpA+ cells and up to 45% enucleated cells, indicating a significant amount of terminal maturation. Cell crowdedness was found to have decisive effects on in vitro erythropoiesis. Cell density per surface area rather than cell concentration per media volume determined cell expansion during exponential growth where more crowded cells showed reduced overall expansion. In late stage erythropoiesis, however, when cells no longer proliferating, increased cell density was seen to enhance cell viability. These results indicate that peripheral blood derived haematopoietic stem cells can be an alternative to cells sourced from bone marrow, cord blood or leukapheresis in terms of expansion potential. This provides distinct advantages in terms of availability for studies of conditions for scale-up and maturation, and may have particular clinical applications in the future.
Collapse
|
363
|
Coupled transcription-splicing regulation of mutually exclusive splicing events at the 5' exons of protein 4.1R gene. Blood 2009; 114:4233-42. [PMID: 19729518 DOI: 10.1182/blood-2009-02-206219] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The tightly regulated production of distinct erythrocyte protein 4.1R isoforms involves differential splicing of 3 mutually exclusive first exons (1A, 1B, 1C) to the alternative 3' splice sites (ss) of exon 2'/2. Here, we demonstrate that exon 1 and 2'/2 splicing diversity is regulated by a transcription-coupled splicing mechanism. We also implicate distinctive regulatory elements that promote the splicing of exon 1A to the distal 3' ss and exon 1B to the proximal 3' ss in murine erythroleukemia cells. A hybrid minigene driven by cytomegalovirus promoter mimicked 1B-promoter-driven splicing patterns but differed from 1A-promoter-driven splicing patterns, suggesting that promoter identity affects exon 2'/2 splicing. Furthermore, splicing factor SF2/ASF ultraviolet (UV) cross-linked to the exon 2'/2 junction CAGAGAA, a sequence that overlaps the distal U2AF(35)-binding 3' ss. Consequently, depletion of SF2/ASF allowed exon 1B to splice to the distal 3' ss but had no effect on exon 1A splicing. These findings identify for the first time that an SF2/ASF binding site also can serve as a 3' ss in a transcript-dependent manner. Taken together, our results suggest that 4.1R gene expression involves transcriptional regulation coupled with a complex splicing regulatory network.
Collapse
|
364
|
Richardson BM, Heesom KJ, Parsons SF, Anstee DJ, Frayne J. Analysis of the differential proteome of human erythroblasts duringin vitroerythropoiesis by 2-D DIGE. Proteomics Clin Appl 2009; 3:1123-34. [DOI: 10.1002/prca.200900013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2009] [Revised: 05/07/2009] [Accepted: 05/27/2009] [Indexed: 11/08/2022]
|
365
|
Tight control of MEK-ERK activation is essential in regulating proliferation, survival, and cytokine production of CD34+-derived neutrophil progenitors. Blood 2009; 114:3402-12. [PMID: 19667405 DOI: 10.1182/blood-2008-08-175141] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
A plethora of extracellular stimuli regulate growth, survival, and differentiation responses through activation of the MEK-ERK MAPK signaling module. Using CD34+ hematopoietic progenitor cells, we describe a novel role for the MEK-ERK signaling module in the regulation of proliferation, survival, and cytokine production during neutrophil differentiation. Addition of the specific MEK1/2 inhibitor U0126 resulted in decreased proliferation of neutrophil progenitors. Conversely, transient activation of a conditionally active MEK1 mutant resulted in the expansion of progenitor cells, which thereafter differentiated normally into mature neutrophils. In contrast, chronic MEK1 activation was found to induce cell death of CD34+ neutrophil progenitors. Microarray analysis of CD34+ progenitor cells showed that activation of MEK1 resulted in changes in expression of a variety of cell-cycle modulating genes. Furthermore, conditional activation of MEK1 resulted in a dramatic increase in the expression of mRNA transcripts encoding a large number of hematopoietic cytokines, chemokines, and growth factors. These findings identify a novel role for MEK-ERK signaling in regulating the balance between proliferation and apoptosis during neutrophil differentiation, and they suggest the need for tight control of MEK-ERK activation to prevent the development of bone marrow failure.
Collapse
|
366
|
Toward clinical therapies using hematopoietic cells derived from human pluripotent stem cells. Blood 2009; 114:3513-23. [PMID: 19652198 DOI: 10.1182/blood-2009-03-191304] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) provide remarkable cellular platforms to better understand human hematopoiesis and to develop clinically applicable hematopoietic cell-based therapies. Over the past decade, hESCs have been used to characterize molecular and cellular mechanisms underpinning the differentiation of hematopoietic progenitors and mature, functional hematopoietic cells. These advances are now poised to lead to clinical translation of hESC- and iPSC-derived hematopoietic cells for novel therapies in the next few years. On the basis of areas of recent success, initial clinical use of hematopoietic cells derived from human pluripotent stem cells will probably be in the areas of transfusion therapies (erythrocytes and platelets) and immune therapies (natural killer cells). In contrast, efficient development and isolation of hematopoietic stem cells capable of long-term, multilineage engraftment still remains a significant challenge. Technical, safety, and regulatory concerns related to clinical applications of human PSCs must be appropriately addressed. However, proper consideration of these issues should facilitate and not inhibit clinical translation of new therapies. This review outlines the current status of hematopoietic cell development and what obstacles must be surmounted to bring hematopoietic cell therapies from human PSCs from "bench to bedside."
Collapse
|
367
|
Migliaccio G, Sanchez M, Leblanc A, Masiello F, Tirelli V, Migliaccio AR, Najfeld V, Whitsett C. Long-term storage does not alter functionality of in vitro generated human erythroblasts: implications for ex vivo generated erythroid transfusion products. Transfusion 2009; 49:2668-79. [PMID: 19659677 DOI: 10.1111/j.1537-2995.2009.02329.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Cultured human erythroid cells derived in vitro may represent alternative transfusion products. It is unknown, however, if these ex vivo expanded erythroid cells remain functional or develop genetic abnormalities after storage. STUDY DESIGN AND METHODS Using mononuclear cells from four adult blood donors, erythroblasts were generated ex vivo in expansion cultures supplemented with stem cell factor, interleukin-3, erythropoietin (EPO), dexamethasone, and estradiol. The viability and in vitro function of freshly expanded or short (1-2 months)- and long (8 years)-term-stored erythroblasts cryopreserved in dimethyl sulfoxide were compared. Erythroblast function was defined as ability to proliferate in expansion media and mature in response to EPO. Cell number was determined manually and expressed as fold increase. Viability was assessed by trypan blue and propidium iodide exclusion. Maturation was evaluated by morphologic analyses and CD36/CD235a expression profiling. Cytogenetic evaluation included karyotype and multicolor fluorescence in situ hybridization analyses. RESULTS Equivalent numbers (>80%) of erythroblasts were viable after short- and long-term storage. Freshly expanded and short- and long-term-stored erythroblasts equally doubled in number (fold increase, 2.4) retaining an immature phenotype (23% of the cells were CD36(high)CD235a(neg)) when cultured for 4 days under expansion conditions. The numbers of freshly expanded and short-term-stored erythroblasts that matured when exposed for 4 days to EPO were also similar (approx. 22% of the cells became CD36(neg)CD235a(high)). In spite of the massive amplification, ex vivo generated erythroblasts demonstrated a normal (46,XY) karyotype with no obvious genomic rearrangements. CONCLUSION Ex vivo expanded erythroblasts remain functional and genetically normal after long-term storage.
Collapse
Affiliation(s)
- Giovanni Migliaccio
- Division of Hematology and Oncology, Tisch Cancer Institute, Mount Sinai School of Medicine, New York, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|
368
|
Douay L, Lapillonne H, Turhan AG. Stem cells--a source of adult red blood cells for transfusion purposes: present and future. Crit Care Clin 2009; 25:383-98, Table of Contents. [PMID: 19341915 DOI: 10.1016/j.ccc.2008.12.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We have sufficient knowledge of the biology of hematopoietic stem cells to hope that we might generate human red blood cells in the laboratory. It may soon be possible to produce enough to transfuse "cultured" red blood cells to manufacture human red blood cells from hematopoietic stem cells for transfusion purposes. This article describes progress and the challenges that remain in the search for in vitro generated red blood cells that can be efficiently manufactured in high volumes and given to any recipient.
Collapse
Affiliation(s)
- Luc Douay
- INSERM, UMR_S 893, Proliferation and differentiation of stem cells, Paris, France.
| | | | | |
Collapse
|
369
|
de Vooght KMK, van Solinge WW, van Wesel AC, Kersting S, van Wijk R. First mutation in the red blood cell-specific promoter of hexokinase combined with a novel missense mutation causes hexokinase deficiency and mild chronic hemolysis. Haematologica 2009; 94:1203-10. [PMID: 19608687 DOI: 10.3324/haematol.2008.002881] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Hexokinase is one of the key enzymes of glycolysis and catalyzes the phosphorylation of glucose to glucose-6-phosphate. Red blood cell-specific hexokinase is transcribed from HK1 by use of an erythroid-specific promoter. The aim of this study was to investigate the molecular basis for hexokinase deficiency in a patient with chronic hemolysis. DESIGN AND METHODS Functional studies were performed using transient transfection of HK promoter constructs in human K562 erythroleukemia cells. The DNA-protein interaction at the promoter of hexokinase was studied using electrophoretic mobility shift assays with nuclear extracts from K562 cells. DNA analysis and reverse transcriptase polymerase chain reaction were performed according to standardized procedures. RESULTS On the paternal allele we identified two novel mutations in cis in the erythroid-specific promoter of HKI: -373A>C and -193A>G. Transfection of promoter reporter constructs showed that the -193A>G mutation reduced promoter activity to 8%. Hence, -193A>G is the first mutation reported to affect red blood cell-specific hexokinase specific transcription. By electrophoretic mobility shift assays we showed that in vitro binding of c-jun to an AP-1 binding site was disrupted by this mutation. Subsequent chromatin-immunoprecipitation assays demonstrated that c-jun binds this region of the promoter in vivo. On the maternal allele we identified a novel missense mutation in exon 3: c.278G>A, encoding an arginine to glutamine substitution at residue 93, affecting both hexokinase-1 and red cell specific-hexokinase. In addition, this missense mutation was shown to compromise normal pre-mRNA processing. CONCLUSIONS We postulate that reduced erythroid transcription of HK1 together with aberrant splicing of both hexokinase-1 and red cell specific-hexokinase results in hexokinase deficiency and mild chronic hemolysis.
Collapse
Affiliation(s)
- Karen M K de Vooght
- Department of Clinical Chemistry and Hematology, Laboratory for Red Blood Cell Research, University Medical Center Utrecht, The Netherlands.
| | | | | | | | | |
Collapse
|
370
|
Mahajan MC, Karmakar S, Newburger PE, Krause DS, Weissman SM. Dynamics of alpha-globin locus chromatin structure and gene expression during erythroid differentiation of human CD34(+) cells in culture. Exp Hematol 2009; 37:1143-1156.e3. [PMID: 19607874 DOI: 10.1016/j.exphem.2009.07.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2009] [Revised: 06/05/2009] [Accepted: 07/07/2009] [Indexed: 01/17/2023]
Abstract
OBJECTIVE The aim of the present study has been to establish serum-free culture conditions for ex vivo expansion and differentiation of human CD34(+) cells into erythroid lineage and to study the chromatin structure, gene expression, and transcription factor recruitment at the alpha-globin locus in the developing erythron. MATERIALS AND METHODS A basal Iscove's modified Dulbecco's medium cell culture medium with 1% bovine serum albumin as a serum replacement and a combination of cytokines and growth factors was used for expansion and differentiation of the CD34(+) cells. Expression patterns of the alpha- and beta-like genes at various stages of erythropoiesis was studied by reverse transcriptase quantitative polymerase chain reaction analysis, profile of key erythroid transcription factors was investigated by Western blotting, and the chromatin structure and transcription factor recruitment at the alpha-globin locus was investigated by chromatin immunoprecipitation quantitative polymerase chain reaction analysis. RESULTS Human CD34(+) cells in the serum-free medium undergo near synchronous erythroid differentiation to yield large amount of cells at different differentiation stages. We observe distinct patterns of the histone modifications and transcription factor binding at the alpha-globin locus during erythroid differentiation of CD34(+) cells. Nuclear factor erythroid-derived 2 (NF-E2) was present at upstream activator sites even before addition of erythropoietin (EPO), while bound GATA-1 was only detectable after EPO treatment. After 7 days of EPO treatment, H3K4Me2 modification uniformly increases throughout the alpha-globin locus. Acetylation at H3K9 and binding of Pol II, NF-E2, and GATA-1 were restricted to certain hypersensitive sites of the enhancer and theta gene, and were conspicuously low at the alpha-like globin promoters. Rearrangement of the insulator binding factor CTCF took place at and around the alpha-globin locus as CD34(+) cells differentiated into erythroid pathway. CONCLUSION Our results indicate that remodeling of the upstream elements may be the primary event in activation of alpha-globin gene expression. Activation of alpha-globin genes upon EPO treatment involves initial binding of Pol II, downregulation of pre-existing factors like NF-E2, removal of CTCF from the locus, then rebinding of CTCF in an altered pattern, and concurrent or subsequent binding of transcription factors like GATA-1.
Collapse
Affiliation(s)
- Milind C Mahajan
- Department of Genetics, The Anlyan Center, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | | | |
Collapse
|
371
|
Timmins NE, Nielsen LK. Blood cell manufacture: current methods and future challenges. Trends Biotechnol 2009; 27:415-22. [DOI: 10.1016/j.tibtech.2009.03.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2009] [Revised: 03/27/2009] [Accepted: 03/30/2009] [Indexed: 01/16/2023]
|
372
|
Hintze C, Ströbele C, Rüster B, Göttig S, Bugert P, Seifried E, Henschler R. Erythrocytic precursor cells show potent shear stress resistant adhesion and home to hematopoietic tissue in vivo. Transfusion 2009; 49:2122-30. [PMID: 19500322 DOI: 10.1111/j.1537-2995.2009.02241.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND Transfusion of erythropoietic precursor cells has been suggested as an alternative to conventional red blood cells. However, little is known about the fate of transfused erythrocytic precursors after they enter the bloodstream. STUDY DESIGN AND METHODS Erythrocytic precursors were classified by flow cytometry into different maturation stages. Precursors were enriched using cell surface expression of CD71 and Ter119 antigens and analyzed under shear stress in a parallel plate flow chamber and after fluorescence tagging with PKH and transfusion into anemic mice. RESULTS We found that at all maturation stages, erythrocytic precursors expressed the adhesion receptor very late antigen (VLA)-4 with a frequency decreasing from 90% to approximately 60% during maturation. In contrast, expression of the beta(2)-integrins LFA-1 and Mac-1 and the rolling receptor P-selectin glycoprotein ligand-1 increased from 10% to 20% to approximately 50% during erythrocytic maturation. The chemokine receptor CXCR4 was expressed at low levels during differentiation stages. In vitro shear stress adhesion analysis showed that erythrocytic precursors can efficiently activate VLA-4 such that it binds its cognate ligand, vascular cell adhesion molecule (VCAM)-1. The coimmobilization of stromal cell-derived factor-1 alpha with VCAM-1 strengthened this adhesion. Transfusion of primitive (CD71+) or more mature (Ter119+) erythrocytic precursors into mice showed that both populations selectively and efficiently home to hematopoietic tissues. CONCLUSION Our results demonstrate that erythrocytic precursor cells of different maturation stages are capable of homing to hematopoietic organs. This work has implications for the development of transfusion protocols that use ex vivo expanded, but not fully matured, erythrocytic precursors from cultured stem cell populations.
Collapse
Affiliation(s)
- Christian Hintze
- Institute of Transfusion Medicine and Immune Hematology, German Red Cross Blood Donor Center, Johann Wolfgang Goethe-University Frankfurt, Frankfurt, Germany
| | | | | | | | | | | | | |
Collapse
|
373
|
Bregoli L, Chiarini F, Gambarelli A, Sighinolfi G, Gatti AM, Santi P, Martelli AM, Cocco L. Toxicity of antimony trioxide nanoparticles on human hematopoietic progenitor cells and comparison to cell lines. Toxicology 2009; 262:121-9. [PMID: 19482055 DOI: 10.1016/j.tox.2009.05.017] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2009] [Revised: 05/20/2009] [Accepted: 05/21/2009] [Indexed: 10/20/2022]
Abstract
Nanoparticles (NPs) are materials with one dimension in the range of 1-100 nm. The toxicity of NPs remains widely unknown and still poses concerns, due to the peculiar characteristics of materials in the nano-size range. We analyze the toxicity of seven NPs ((Fe2O3, Fe3O4, Sb2O3, Au, TiO2, Co, and Ag) on primary cultures of human hematopoietic progenitor cells from the bone marrow of healthy donors with CFU assays, and show that antimony oxide (Sb2O3) NPs and cobalt (Co) NPs have a toxic effect, while the other NPs have no effect at the tested concentrations (5, 25 and 100 microg/ml). While Co NPs suspension is toxic to both erythroid and granulocytic-monocytic precursors, Sb2O3 NPs at 5 microg/ml are specifically toxic to erythroid colony development, suggesting a highly selective type of toxicity. With liquid culture assays we show that Sb2O3 NPs impair the proliferation of erythroid progenitors, while no toxic effect is observed when Sb2O3 NPs are added during erythroid differentiation. CFU assays and liquid culture assays on seven human cell lines of hematopoietic origin (K562, HL-60, CEM, CEM-R, Thp-1, Jurkat, and Molt-4) show that, contrary to what observed on primary cultures of bone marrow progenitors, Sb2O3 NPs have no toxic effect on proliferation of any of the cell lines, raising concerns about the use of immortalized cell lines for nanotoxicology tests.
Collapse
Affiliation(s)
- Lisa Bregoli
- Cellular Signaling Laboratory, Department of Anatomical Sciences, University of Bologna, via Irnerio 48, 40126 Bologna, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
374
|
Vlaski M, Lafarge X, Chevaleyre J, Duchez P, Boiron JM, Ivanovic Z. Low oxygen concentration as a general physiologic regulator of erythropoiesis beyond the EPO-related downstream tuning and a tool for the optimization of red blood cell production ex vivo. Exp Hematol 2009; 37:573-84. [PMID: 19375648 DOI: 10.1016/j.exphem.2009.01.007] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Revised: 01/19/2009] [Accepted: 01/20/2009] [Indexed: 01/30/2023]
Abstract
OBJECTIVE The control of mature erythroid progenitors and precursors' production via erythropoietin (EPO) is the major systemic regulatory mechanism in erythropoiesis. However, hypoxia seems to influence erythropoiesis beyond this well-known mechanism. The aim of our study is to test this hypothesis adapting the oxygenation level to each stage of erythropoiesis. MATERIALS AND METHODS We exploited the newly developed ex vivo three-phase protocol for red blood cell (RBC) production starting from the steady-state peripheral blood and cord blood CD34(+) cells exposed to adapted O(2) concentrations. Differentiation and maturation were followed by functional tests, morphology, immunophenotype, and analysis of molecular markers' expression. RESULTS We report here an enhancement of total RBC production if low O(2) concentrations (1.5-5%) were applied, instead of 20% O(2), during the first phase of culture. This results from a comprehensive action of low-O(2) concentration on: 1) amplification of erythroid progenitors, 2) acceleration of their proliferation, 3) differentiation, and 4) maturation of erythroid precursors. In addition, arterial blood O(2) concentration (13%) is critical for stromal cells to fully sustain the differentiation of erythroid precursors. These effects were associated with upregulation of erythroid 5-aminolevulinate synthase and gamma-globin gene expression. CONCLUSION These results imply that integral regulation of erythropoiesis is operated by low O(2) concentrations, beyond the EPO/EPO-responsive cells loop and provide a tool to optimize the technology for ex vivo production of RBC.
Collapse
Affiliation(s)
- Marija Vlaski
- Aquitaine-Limousin Branch of French Blood Institute, Bordeaux, France
| | | | | | | | | | | |
Collapse
|
375
|
Culture de cellules à visée transfusionnelle : le cas des globules rouges. Transfus Clin Biol 2009; 16:134-7. [DOI: 10.1016/j.tracli.2009.03.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Accepted: 03/18/2009] [Indexed: 11/22/2022]
|
376
|
Bagnis C, Chapel S, Chiaroni J, Bailly P. A genetic strategy to control expression of human blood group antigens in red blood cells generated in vitro. Transfusion 2009; 49:967-76. [PMID: 19175544 DOI: 10.1111/j.1537-2995.2008.02078.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND The ability to generate red blood cells of a chosen blood group phenotype would be a major advance in transfusion when considering low- and high-frequency blood group antigens. STUDY DESIGN AND METHODS Cord blood CD34+ cells undergoing erythroid differentiation in vitro were genetically manipulated with human immunodeficiency virus Type 1-derived lentiviral vectors expressing hUT-B1 cDNA (overexpression strategy) or bicistronic vectors expressing both enhanced green fluorescent protein and a short-hairpin RNA (shRNA) designed to silence SLC14A1(JK) gene that encodes hUT-B1 protein (silencing strategy). Resulting cell populations were analyzed by fluorescent-activated cell sorting and gel affinity column assay. RESULTS When transduced with hUT-B1 cDNA lentiviral vectors encoding JK*B and JK*A alleles, respectively, CD34+ cell-derived erythroid populations from Jk(a+b-) and Jk(a-b+) donors exhibited a Jk(a+b+) phenotype different from the original phenotype. In concomitant tests, Jk(a+b+) donor cells transduced with lentiviral vectors carrying a shRNA designed to interfere with hUT-B1 transcription showed a marked decrease in hUT-B1 expression and were assessed as null for Jk antigen by a routine assay. CONCLUSION In this work focusing on the Kidd blood group system that relies on expression of hUT-B1 glycoprotein under the Jk(a) or Jk(b) antigenic configurations, we demonstrated that hematopoietic progenitors could be genetically modified to exhibit a chosen Kidd phenotype. Beyond production of atypical Kidd phenotypes, this genetic strategy could allow generation of rare blood phenotypes from hematopoietic stem cells regardless of initial donor phenotype. Potential applications for genetically modified blood include production of control samples for immunohematologic testing and for resolution of antibody detection in multiply transfused patients.
Collapse
Affiliation(s)
- Claude Bagnis
- Etablissement Français du Sang Alpes Méditerranée, UMR 6578-CNRS/Université de la Méditerranée/EFS, Laboratoire Hématologie Moléculaire, Marseille, France.
| | | | | | | |
Collapse
|
377
|
Tiberghien P, Chabod J, Deconinck E, Pouthier F. Conservation et indications des greffons de sang placentaire. Transfus Clin Biol 2009; 16:204-8. [DOI: 10.1016/j.tracli.2009.03.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2009] [Accepted: 03/18/2009] [Indexed: 10/20/2022]
|
378
|
Biocompatibility Study of Hemoglobin Vesicles, Cellular-Type Artificial Oxygen Carriers, with Human Umbilical Cord Hematopoietic Stem/Progenitor Cells Using an In Vitro Expansion System. ASAIO J 2009; 55:200-5. [DOI: 10.1097/mat.0b013e318198e550] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
379
|
Ex vivo large-scale generation of human red blood cells from cord blood CD34+ cells by co-culturing with macrophages. Int J Hematol 2009; 87:339-350. [PMID: 18369691 DOI: 10.1007/s12185-008-0062-y] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Revised: 01/10/2008] [Accepted: 01/29/2008] [Indexed: 01/10/2023]
Abstract
We generated red blood cells (RBC) from cord blood (CB) CD34+ cells using a four-phase culture system. We first cultured CB CD34+ cells on telomerase gene-transduced human stromal cells in serum-free medium containing stem cell factor (SCF), Flt-3/Flk-2 ligand, and thrombopoietin to expand CD34+ cells (980-fold) and the total cells (10,400-fold) (first phase). Expanded cells from the first phase were liquid-cultured with SCF, interleukin-3 (IL-3), and erythropoietin (EPO) to expand (113-fold) and differentiate them into erythroblasts (second phase). To obtain macrophages for the next phase, we expanded CD34+ cells from a different donor using the same coculture system. Expanded cells from the first phase were liquid-cultured with granulocyte-macrophage colony stimulating factor, macrophage-colony stimulating factor (M-CSF), IL-3, and SCF to generate monocytes/macrophages (75-fold), which were incubated with type AB serum and M-CSF to fully differentiate them into macrophages. Erythroblasts were then co-cultured with macrophages in the presence of EPO to expand (threefold) and fully differentiate them (61% orthochromatic erythroblasts plus 39% RBC) (third phase). RBC were purified from erythroblasts and debris through a deleukocyting filter to generate 6.0 x 10(12) RBC from 1.0 unit of CB (3.0 transfusable units). Qualitatively, these RBC showed a hemoglobin content, oxygenation of hemoglobin, and in vivo clearance similar to those of adult peripheral RBC. Finally, an almost complete enucleation of orthochromatic erythroblasts (99.4%) was achieved by the cultivation method recently described by Miharada et al. in the absence of macrophages and cytokines (fourth phase). RBC were purified from remnant erythroblasts and debris by passage through a deleukocyting filter to generate 1.76 x 10(13) RBC from 1.0 unit of CB (8.8 transfusable units), the highest yield ever reported. Thus, this method may be useful for generating an alternative RBC supply for transfusions, investigating infectious agents that target erythroid cells, and as a general in vitro hematopoietic model system.
Collapse
|
380
|
|
381
|
Bruchova H, Yoon D, Agarwal AM, Swierczek S, Prchal JT. Erythropoiesis in polycythemia vera is hyper-proliferative and has accelerated maturation. Blood Cells Mol Dis 2009; 43:81-7. [PMID: 19264517 DOI: 10.1016/j.bcmd.2009.02.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2009] [Accepted: 02/05/2009] [Indexed: 01/31/2023]
Abstract
Polycythemia vera (PV) is an acquired myeloproliferative clonal disorder, characterized by augmented erythropoiesis. To better define PV pathogenesis, we performed an in vitro erythroid expansion from peripheral blood mononuclear cells of controls and PV patients and evaluated the cells for proliferation, apoptosis, erythroid differentiation, and morphology at the defined time points. PV erythroid progenitors exhibited increased proliferation at days 9-14 and accelerated maturation at days 7-14, with a larger S-phase population (40%) than controls (20%) at day 11; however, the proportion of apoptotic cells was comparable to controls. Previously, we have identified PV-specific dysregulation of several microRNAs (i.e. miR-150, 451, 222, 155, 378). We had analyzed expression profiles of selected target genes of these microRNAs based on in silico prediction and their known function pertinent to the observed PV-specific erythropoiesis differences. p27, cMYB and EPOR showed differential expression in PV erythroid progenitors at the specific stages of erythroid differentiation. In this study, we identified accelerated maturation and hyper-proliferation at early stages of PV erythropoiesis. We speculate that aberrant expression of p27, c-MYB, and EPOR may contribute to these abnormal features in PV erythropoiesis.
Collapse
Affiliation(s)
- Hana Bruchova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | | | | | | | | |
Collapse
|
382
|
Chromatin signatures in multipotent human hematopoietic stem cells indicate the fate of bivalent genes during differentiation. Cell Stem Cell 2009; 4:80-93. [PMID: 19128795 DOI: 10.1016/j.stem.2008.11.011] [Citation(s) in RCA: 464] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2007] [Revised: 09/03/2008] [Accepted: 11/18/2008] [Indexed: 12/28/2022]
Abstract
Histone modifications have been implicated in stem cell maintenance and differentiation. We have analyzed genome-wide changes in gene expression and histone modifications during differentiation of multipotent human primary hematopoietic stem cells/progenitor cells (HSCs/HPCs) into erythrocyte precursors. Our data indicate that H3K4me1, H3K9me1, and H3K27me1 associate with enhancers of differentiation genes prior to their activation and correlate with basal expression, suggesting that these monomethylations are involved in the maintenance of activation potential required for differentiation. In addition, although the majority of genes associated with both H3K4me3 and H3K27me3 in HSCs/HPCs become silent and lose H3K4me3 after differentiation, those that lose H3K27me3 and become activated after differentiation are associated with increased levels of H2A.Z, H3K4me1, H3K9me1, H4K20me1, and RNA polymerase II in HSCs/HPCs. Thus, our data suggest that gene expression changes during differentiation are programmed by chromatin modifications present at the HSC/HPC stage and provide a resource for enhancer and promoter identification.
Collapse
|
383
|
|
384
|
Abstract
We describe a technological approach permitting the massive expansion of CD34(+) stem cells and their 100% conversion ex vivo into mature red blood cells (RBC). The protocol comprises three steps: a first step consisting of cell proliferation and induction of erythroid differentiation in a liquid medium without serum in the presence of growth factors (GF), a second based on a model reconstitution of the medullar microenvironment (ME) (human MSC or murine stromal cells) in the presence of GF, and a third in the presence of the ME alone, without any GF. This work highlights the impact of the ex vivo microenvironment on the terminal maturation of erythroid cells. A critical point is that the RBC generated in vitro have all the characteristics of functional native adult RBC. Moreover, this new concept of 'cultured RBC' (cRBC) is important for basic research into terminal erythropoiesis and has major clinical implications, especially in transfusion medicine. The three-step protocol can be adapted to use hematopoietic stem cells (HSC) from diverse sources: peripheral blood, bone marrow or cord blood.
Collapse
|
385
|
Yamaguchi M, Fujihara M, Wakamoto S, Sakai H, Takeoka S, Tsuchida E, Azuma H, Ikeda H. Influence of hemoglobin vesicles, cellular-type artificial oxygen carriers, on human umbilical cord blood hematopoietic progenitor cellsin vitro. J Biomed Mater Res A 2009; 88:34-42. [DOI: 10.1002/jbm.a.31857] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
386
|
Dijon M, Chabannon C, Tonnelle C. Ikaros, facteur de transcription impliqué, aussi, dans l’érythropoïèse. Med Sci (Paris) 2008; 24:1065-70. [DOI: 10.1051/medsci/200824121065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
387
|
Dynamics of human erythroblast enucleation. Int J Hematol 2008; 88:498-507. [DOI: 10.1007/s12185-008-0200-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Revised: 09/24/2008] [Accepted: 10/12/2008] [Indexed: 10/21/2022]
|
388
|
|
389
|
Diwan A, Koesters AG, Capella D, Geiger H, Kalfa TA, Dorn GW. Targeting erythroblast-specific apoptosis in experimental anemia. Apoptosis 2008; 13:1022-30. [PMID: 18584327 DOI: 10.1007/s10495-008-0236-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Erythrocyte production is regulated by balancing precursor cell apoptosis and survival signaling. Previously, we found that BH3-only proapoptotic factor, Nix, opposed erythroblast-survival signaling by erythropoietin-induced Bcl-xl during normal erythrocyte formation. Since erythropoietin treatment of human anemia has limitations, we explored the therapeutic potential of abrogating Nix-mediated erythroblast apoptosis to enhance erythrocyte production. Nix gene ablation blunted the phenylhydrazine-induced fall in blood count, enhanced hematocrit recovery, and reduced erythroblast apoptosis, despite lower endogenous erythropoietin levels. Similar to erythropoietin, Nix ablation increased early splenic erythroblasts and circulating reticulocytes, while maintaining a pool of mature erythroblasts as erythropoietic reserve. Erythrocytes in Nix-deficient mice showed morphological abnormalities, suggesting that apoptosis during erythropoiesis not only controls red blood cell number, but also serves a "triage" function, preferentially eliminating abnormal erythrocytes. These results support the concept of targeting erythroblast apoptosis to maximize erythrocyte production in acute anemia, which may be of value in erythropoietin resistance.
Collapse
Affiliation(s)
- Abhinav Diwan
- Center for Molecular Cardiovascular Research, University of Cincinnati, Cincinnati, OH, USA
| | | | | | | | | | | |
Collapse
|
390
|
Abstract
Human erythropoiesis is a complex multistep process that involves the differentiation of early erythroid progenitors to mature erythrocytes. Here we show that it is feasible to differentiate and mature human embryonic stem cells (hESCs) into functional oxygen-carrying erythrocytes on a large scale (10(10)-10(11) cells/6-well plate hESCs). We also show for the first time that the oxygen equilibrium curves of the hESC-derived cells are comparable with normal red blood cells and respond to changes in pH and 2,3-diphosphoglyerate. Although these cells mainly expressed fetal and embryonic globins, they also possessed the capacity to express the adult beta-globin chain on further maturation in vitro. Polymerase chain reaction and globin chain specific immunofluorescent analysis showed that the cells increased expression of beta-globin (from 0% to > 16%) after in vitro culture. Importantly, the cells underwent multiple maturation events, including a progressive decrease in size, increase in glycophorin A expression, and chromatin and nuclear condensation. This process resulted in extrusion of the pycnotic nuclei in up to more than 60% of the cells generating red blood cells with a diameter of approximately 6 to 8 mum. The results show that it is feasible to differentiate and mature hESCs into functional oxygen-carrying erythrocytes on a large scale.
Collapse
|
391
|
Block to the production of full-length B19 virus transcripts by internal polyadenylation is overcome by replication of the viral genome. J Virol 2008; 82:9951-63. [PMID: 18684834 DOI: 10.1128/jvi.01162-08] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The pre-mRNA processing strategy of the B19 virus is unique among parvoviruses. B19 virus-generated pre-mRNAs are transcribed from a single promoter and are extensively processed by alternative splicing and alternative polyadenylation to generate 12 transcripts. Blockage of the production of full-length B19 virus transcripts at the internal polyadenylation site [(pA)p] was previously reported to be a limiting step in B19 virus permissiveness. We show here that in the absence of genome replication, internal polyadenylation of B19 virus RNAs at (pA)p is favored in cells which are both permissive and nonpermissive for B19 viral replication. Replication of the B19 virus genome, however, introduced either by viral infection or by transfection of an infectious clone into permissive cells or forced by heterologous replication systems in nonpermissive cells, enhanced readthrough of (pA)p and the polyadenylation of B19 virus transcripts at the distal site [(pA)d]. Therefore, replication of the genome facilitates the generation of sufficient full-length transcripts that encode the viral capsid proteins and the essential 11-kDa nonstructural protein. Furthermore, we show that polyadenylation of B19 viral RNA at (pA)p likely competes with splicing at the second intron. Thus, we conclude that replication of the B19 virus genome is the primary limiting step governing B19 virus tropism.
Collapse
|
392
|
Baek EJ, Kim HS, Kim S, Jin H, Choi TY, Kim HO. In vitro clinical-grade generation of red blood cells from human umbilical cord blood CD34+ cells. Transfusion 2008; 48:2235-45. [PMID: 18673341 DOI: 10.1111/j.1537-2995.2008.01828.x] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND There is no appropriate alternative source of red blood cells (RBCs) to relieve the worsening shortage of blood available for transfusion. Therefore, in vitro generation of clinically available RBCs from hematopoietic stem cells could be a promising new source to supplement the blood supply. However, there have been few studies about the generation of clinical-grade RBCs by coculture on human mesenchymal stem cells (MSCs) and various cytokine supplements, even though the production of pure RBCs requires coculture on stromal cells and proper cytokine supplements. STUDY DESIGN AND METHODS Umbilical cord blood (CB) CD34+ cells were cultured in serum-free medium supplemented with two cytokine sets of stem cell factor (SCF) plus interleukin-3 (IL-3) plus erythropoietin (EPO) and SCF plus IL-3 plus EPO plus thrombopoietin (TPO) plus Flt-3 for 1 week, followed by coculture upon MSCs derived from bone marrow (BM) or CB for 2 weeks. RESULTS Almost pure clinical-grade RBCs could be generated by coculturing with CB-MSCs but not BM-MSCs. Expansion fold and enucleation rate were significantly higher in coculture with CB-MSCs than BM-MSCs. Despite a 2.5-fold expansion of erythroblasts in the presence of TPO and Flt-3 for 8 days, the final RBC count was higher without TPO and Flt-3. CONCLUSIONS This study is the first report on generating clinical-grade RBCs by in vitro culture with human MSCs and compared effectiveness of several cytokines for RBC production. This provides a useful basis for future production of clinically available RBCs and a model of erythropoiesis that is analogous to the in vivo system.
Collapse
Affiliation(s)
- Eun Jung Baek
- The Department of Laboratory Medicine, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
393
|
Abstract
Patients with major trauma present a challenge, often using large quantities of banked blood both at the time of injury and during their hospital stay. Blood transfusion is not without risk and is associated with high costs; it is immunosuppressive, rendering patients more susceptible to infection. In the western world, banked blood is fully screened and relatively safe; the same is not true in parts of the developing world, where high rates of HIV carriage make blood transfusion a risky undertaking. Additionally, blood transfusion as a vector for transmission of illnesses such as prion disease is a distinct possibility, for both the developed and developing world alike. The introduction of artificial blood substitutes would ameliorate some risk and also remove the cost of extensive blood testing. For trauma outside hospital, blood substitutes could compete directly with fluid resuscitation as donated blood is not usually available. Patients with prolonged transport times would appear to be the most obvious beneficiaries and volume expansion, along with improvement in oxygen-carrying capacity would be the ultimate goal. All clinicians confronted with the need for transfusion of homologous blood would welcome the development of a safe and reliable alternative to red blood cells in order to ensure oxygen transport to the tissues. However, even though research on red cell substitutes started more than 100 years ago, even now none of the heavily investigated compounds based on haemoglobin or perfluorocarbons has been released in Europe or the USA for routine clinical use.
Collapse
Affiliation(s)
- Peter J Shirley
- Intensive Care and Anaesthesia, Royal London Hospital, Whitechapel, London, E1 1BB, UK,
| |
Collapse
|
394
|
Dorn I, Lazar-Karsten P, Boie S, Ribbat J, Hartwig D, Driller B, Kirchner H, Schlenke P. In vitro proliferation and differentiation of human CD34+ cells from peripheral blood into mature red blood cells with two different cell culture systems. Transfusion 2008; 48:1122-32. [DOI: 10.1111/j.1537-2995.2008.01653.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
395
|
Wang W, Horner DN, Chen WLK, Zandstra PW, Audet J. Synergy between erythropoietin and stem cell factor during erythropoiesis can be quantitatively described without co-signaling effects. Biotechnol Bioeng 2008; 99:1261-72. [DOI: 10.1002/bit.21677] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
396
|
Development of an enhanced B-specific lentiviral vector expressing BTK: a tool for gene therapy of XLA. Gene Ther 2008; 15:942-52. [PMID: 18323795 DOI: 10.1038/gt.2008.17] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Further development of haematopoietic stem cell (HSC) gene therapy will depend on enhancement of gene transfer safety: ad hoc improvement of vector design relating to each particular disease is thus a crucial issue for HSC gene therapy. We modified a previously described lentiviral vector by adding the Emumar B-specific enhancer to a human CD19 promoter-derived sequence (Mol Ther 2004;10:45-56). We thus significantly improved the level of expression of the green fluorescent protein (GFP) reporter gene while retaining the specificity of expression in B-cell progeny of transduced human CD34+ progenitor cells obtained from cord blood or adult bone marrow. Indeed, GFP was strongly expressed from early medullary pro-B cells to splenic mature B cells whereas transgene expression remained low in transduced immature progenitors as in myeloid and T-lymphoid progeny retrieved from xenografted NOD/SCID/gammac(null) mice. Using this lentiviral vector, we further demonstrated the possibility to express a functional human BTK protein in long-term human CD34+ cell B-lymphoid progeny. This newly designed lentiviral vector fulfils one of the pre-requisites for the development of efficient and safe gene therapy for X-linked agammaglobulinaemia, the most common primary humoral immunodeficiency disorder.
Collapse
|
397
|
Hiroyama T, Miharada K, Sudo K, Danjo I, Aoki N, Nakamura Y. Establishment of mouse embryonic stem cell-derived erythroid progenitor cell lines able to produce functional red blood cells. PLoS One 2008; 3:e1544. [PMID: 18253492 PMCID: PMC2212133 DOI: 10.1371/journal.pone.0001544] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2007] [Accepted: 01/03/2008] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The supply of transfusable red blood cells (RBCs) is not sufficient in many countries. If erythroid cell lines able to produce transfusable RBCs in vitro were established, they would be valuable resources. However, such cell lines have not been established. To evaluate the feasibility of establishing useful erythroid cell lines, we attempted to establish such cell lines from mouse embryonic stem (ES) cells. METHODOLOGY/PRINCIPAL FINDINGS We developed a robust method to obtain differentiated cell lines following the induction of hematopoietic differentiation of mouse ES cells and established five independent hematopoietic cell lines using the method. Three of these lines exhibited characteristics of erythroid cells. Although their precise characteristics varied, each of these lines could differentiate in vitro into more mature erythroid cells, including enucleated RBCs. Following transplantation of these erythroid cells into mice suffering from acute anemia, the cells proliferated transiently, subsequently differentiated into functional RBCs, and significantly ameliorated the acute anemia. In addition, we did not observe formation of any tumors following transplantation of these cells. CONCLUSION/SIGNIFICANCE To the best of our knowledge, this is the first report to show the feasibility of establishing erythroid cell lines able to produce mature RBCs. Considering the number of human ES cell lines that have been established so far, the intensive testing of a number of these lines for erythroid potential may allow the establishment of human erythroid cell lines similar to the mouse erythroid cell lines described here. In addition, our results strongly suggest the possibility of establishing useful cell lines committed to specific lineages other than hematopoietic progenitors from human ES cells.
Collapse
Affiliation(s)
- Takashi Hiroyama
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
| | - Kenichi Miharada
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
| | - Kazuhiro Sudo
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
| | - Inaho Danjo
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
| | - Naoko Aoki
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
398
|
Abstract
Abstract
Ikaros—a factor that positively or negatively controls gene transcription—is active in murine adult erythroid cells, and involved in fetal to adult globin switching. Mice with Ikaros mutations have defects in erythropoiesis and anemia. In this paper, we have studied the role of Ikaros in human erythroid development for the first time. Using a gene-transfer strategy, we expressed Ikaros 6 (Ik6)—a known dominant-negative protein that interferes with normal Ikaros activity—in cord blood or apheresis CD34+ cells that were induced to differentiate along the erythroid pathway. Lentivirally induced Ik6-forced expression resulted in increased cell death, decreased cell proliferation, and decreased expression of erythroid-specific genes, including GATA1 and fetal and adult globins. In contrast, we observed the maintenance of a residual myeloid population that can be detected in this culture system, with a relative increase of myeloid gene expression, including PU1. In secondary cultures, expression of Ik6 favored reversion of sorted and phenotypically defined erythroid cells into myeloid cells, and prevented reversion of myeloid cells into erythroid cells. We conclude that Ikaros is involved in human adult or fetal erythroid differentiation as well as in the commitment between erythroid and myeloid cells.
Collapse
|
399
|
Boyer L, Robert A, Proulx C, Pineault N. Increased production of megakaryocytes near purity from cord blood CD34+ cells using a short two-phase culture system. J Immunol Methods 2008; 332:82-91. [PMID: 18234208 DOI: 10.1016/j.jim.2007.12.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2007] [Revised: 11/13/2007] [Accepted: 12/19/2007] [Indexed: 10/22/2022]
Abstract
Expansion of hematopoietic progenitor cells (HPC) ex vivo remains an important focus in fundamental and clinical research. The aim of this study was to determine whether the implementation of such expansion phase in a two-phase culture strategy prior to the induction of megakaryocyte (Mk) differentiation would increase the yield of Mks produced in cultures. Toward this end, we first characterized the functional properties of five cytokine cocktails to be tested in the expansion phase on the growth and differentiation kinetics of CD34+-enriched cells, and on their capacity to expand clonogenic progenitors in cultures. Three of these cocktails were chosen based on their reported ability to induce HPC expansion ex vivo, while the other two represented new cytokine combinations. These analyses revealed that none of the cocktails tested could prevent the differentiation of CD34+ cells and the rapid expansion of lineage-positive cells. Hence, we sought to determine the optimum length of time for the expansion phase that would lead to the best final Mk yields. Despite greater expansion of CD34+ cells and overall cell growth with a longer expansion phase, the optimal length for the expansion phase that provided greater Mk yield at near maximal purity was found to be 5 days. Under such settings, two functionally divergent cocktails were found to significantly increase the final yield of Mks. Surprisingly, these cocktails were either deprived of thrombopoietin or of stem cell factor, two cytokines known to favor megakaryopoiesis and HPC expansion, respectively. Based on these results, a short resource-efficient two-phase culture protocol for the production of Mks near purity (>95%) from human CD34+ CB cells has been established.
Collapse
Affiliation(s)
- Lucie Boyer
- Héma-Québec R&D Department, Québec City, PQ, Canada
| | | | | | | |
Collapse
|
400
|
Tanabe O, Shen Y, Liu Q, Campbell AD, Kuroha T, Yamamoto M, Engel JD. The TR2 and TR4 orphan nuclear receptors repress Gata1 transcription. Genes Dev 2008; 21:2832-44. [PMID: 17974920 DOI: 10.1101/gad.1593307] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
When the orphan nuclear receptors TR2 and TR4, the DNA-binding subunits of the DRED repressor complex, are forcibly expressed in erythroid cells of transgenic mice, embryos exhibit a transient mid-gestational anemia as a consequence of a reduction in the number of primitive erythroid cells. GATA-1 mRNA is specifically diminished in the erythroid cells of these TR2/TR4 transgenic embryos as it is in human CD34(+) progenitor cells transfected with forcibly expressed TR2/TR4. In contrast, in loss-of-function studies analyzing either Tr2- or Tr4-germline-null mutant mice or human CD34(+) progenitor cells transfected with force-expressed TR2 and TR4 short hairpin RNAs (shRNAs), GATA-1 mRNA is induced. An evolutionarily conserved direct repeat (DR) element, a canonical binding site for nuclear receptors, was identified in the GATA1 hematopoietic enhancer (G1HE), and TR2/TR4 binds to that site in vitro and in vivo. Mutation of that DR element led to elevated Gata1 promoter activity, and reduced promoter responsiveness to cotransfected TR2/TR4. Thus, TR2/TR4 directly represses Gata1/GATA1 transcription in murine and human erythroid progenitor cells through an evolutionarily conserved binding site within a well-characterized, tissue-specific Gata1 enhancer, thereby providing a mechanism by which Gata1 can be directly silenced during terminal erythroid maturation.
Collapse
Affiliation(s)
- Osamu Tanabe
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | | | |
Collapse
|