1
|
Schippel N, Wei J, Ma X, Kala M, Qiu S, Stoilov P, Sharma S. Erythropoietin-dependent Acquisition of CD71 hi CD105 hi Phenotype within CD235a - Early Erythroid Progenitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.29.610192. [PMID: 39257831 PMCID: PMC11383684 DOI: 10.1101/2024.08.29.610192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
The development of committed erythroid progenitors and their continued maturation into mature erythrocytes requires the cytokine erythropoietin (Epo). Here, we describe the immunophenotypic identification of a unique Epo-dependent colony-forming unit-erythroid (CFU-E) cell subtype that forms during early erythropoiesis (EE). This previously undescribed CFU-E subtype, termed late-CFU-E (lateC), lacks surface expression of the characteristic erythroid marker CD235a (glycophorin A) but has high levels of CD71 and CD105. LateCs could be prospectively detected in human bone marrow (BM) cells and, upon isolation and reculture, exhibited the potential to form CFU-E colonies in medium containing only Epo (no other cytokines) and continued differentiation along the erythroid trajectory. Analysis of ex vivo cultures of BM CD34 + cells showed that acquisition of the CD7 hi CD105 hi phenotype in lateCs is gradual and occurs through the formation of four EE cell subtypes. Of these, two are CD34 + burst-forming unit-erythroid (BFU-E) cells, distinguishable as CD7 lo CD105 lo early BFU-E and CD7 hi CD105 lo late BFU-E, and two are CD34 - CFU-Es, also distinguishable as CD71 lo CD105 lo early CFU-E and CD7 hi CD105 lo mid-CFU-E. The transition of these EE populations is accompanied by a rise in CD36 expression, such that all lateCs are CD36 + . Single cell RNA-sequencing analysis confirmed Epo-dependent formation of a CFU-E cluster that exhibits high coexpression of CD71, CD105, and CD36 transcripts. Gene set enrichment analysis revealed the involvement of genes specific to fatty acid and cholesterol metabolism in lateC formation. Overall, in addition to identifying a key Epo-dependent EE cell stage, this study provides a framework for investigation into mechanisms underlying other erythropoiesis-stimulating agents.
Collapse
|
2
|
Boccacci Y, Dumont N, Doyon Y, Laganière J. Accessory-cell-free differentiation of hematopoietic stem and progenitor cells into mature red blood cells. Cytotherapy 2023; 25:1242-1248. [PMID: 37598334 DOI: 10.1016/j.jcyt.2023.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 07/14/2023] [Accepted: 07/25/2023] [Indexed: 08/21/2023]
Abstract
BACKGROUND AIMS The culture and ex vivo engineering of red blood cells (RBCs) can help characterize genetic variants, model diseases, and may eventually spur the development of applications in transfusion medicine. In the last decade, improvements to the in vitro production of RBCs have enabled efficient erythroid progenitor proliferation and high enucleation levels from several sources of hematopoietic stem and progenitor cells (HSPCs). Despite these advances, there remains a need for refining the terminal step of in vitro human erythropoiesis, i.e., the terminal maturation of reticulocytes into erythrocytes, so that it can occur without feeder or accessory cells and animal-derived components. METHODS Here, we describe the near-complete erythroid differentiation of cultured RBCs (cRBCs) from adult HSPCs in accessory-cell-free and xeno-free conditions. RESULTS The approach improves post-enucleation cell integrity and cell survival, and it enables subsequent storage of cRBCs for up to 42 days in classical additive solution conditions without any specialized equipment. CONCLUSIONS We foresee that these improvements will facilitate the characterization of RBCs derived from gene-edited HSPCs.
Collapse
Affiliation(s)
- Yelena Boccacci
- Medical Affairs and Innovation, Héma-Qubec, Québec, Quebec, Canada; Centre Hospitalier Universitaire de Québec Research Center, Université Laval, Québec, Quebec, Canada
| | - Nellie Dumont
- Medical Affairs and Innovation, Héma-Qubec, Québec, Quebec, Canada
| | - Yannick Doyon
- Centre Hospitalier Universitaire de Québec Research Center, Université Laval, Québec, Quebec, Canada
| | - Josée Laganière
- Medical Affairs and Innovation, Héma-Qubec, Québec, Quebec, Canada.
| |
Collapse
|
3
|
The Application of Ethnomedicine in Modulating Megakaryocyte Differentiation and Platelet Counts. Int J Mol Sci 2023; 24:ijms24043168. [PMID: 36834579 PMCID: PMC9961075 DOI: 10.3390/ijms24043168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/28/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023] Open
Abstract
Megakaryocytes (MKs), a kind of functional hematopoietic stem cell, form platelets to maintain platelet balance through cell differentiation and maturation. In recent years, the incidence of blood diseases such as thrombocytopenia has increased, but these diseases cannot be fundamentally solved. The platelets produced by MKs can treat thrombocytopenia-associated diseases in the body, and myeloid differentiation induced by MKs has the potential to improve myelosuppression and erythroleukemia. Currently, ethnomedicine is extensively used in the clinical treatment of blood diseases, and the recent literature has reported that many phytomedicines can improve the disease status through MK differentiation. This paper reviewed the effects of botanical drugs on megakaryocytic differentiation covering the period 1994-2022, and information was obtained from PubMed, Web of Science and Google Scholar. In conclusions, we summarized the role and molecular mechanism of many typical botanical drugs in promoting megakaryocyte differentiation in vivo, providing evidence as much as possible for botanical drugs treating thrombocytopenia and other related diseases in the future.
Collapse
|
4
|
African Plasmodium vivax malaria improbably rare or benign. Trends Parasitol 2022; 38:683-696. [PMID: 35667992 DOI: 10.1016/j.pt.2022.05.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 11/23/2022]
Abstract
The overwhelming dominance of Duffy blood group negativity among most people living in sub-Saharan Africa has been considered the basis of their protection from endemic Plasmodium vivax malaria. New evidence demonstrates widespread transmission of P. vivax in Duffy-negative Africa, though currently of unknown distribution, magnitude, or consequences. Other new evidence from outside of Africa demonstrates marked tropisms of P. vivax for extravascular tissues of bone marrow and spleen. Those establish states of proliferative infection with low-grade or undetectable parasitemia of peripheral blood causing acute and chronic disease. This review examines the plausibility of those infectious processes also operating in Duffy-negative Africans and causing harm of unrecognized origin.
Collapse
|
5
|
Simionato G, Rabe A, Gallego-Murillo JS, van der Zwaan C, Hoogendijk AJ, van den Biggelaar M, Minetti G, Bogdanova A, Mairbäurl H, Wagner C, Kaestner L, van den Akker E. In Vitro Erythropoiesis at Different pO 2 Induces Adaptations That Are Independent of Prior Systemic Exposure to Hypoxia. Cells 2022; 11:cells11071082. [PMID: 35406648 PMCID: PMC8997720 DOI: 10.3390/cells11071082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/20/2022] [Accepted: 03/22/2022] [Indexed: 12/23/2022] Open
Abstract
Hypoxia is associated with increased erythropoietin (EPO) release to drive erythropoiesis. At high altitude, EPO levels first increase and then decrease, although erythropoiesis remains elevated at a stable level. The roles of hypoxia and related EPO adjustments are not fully understood, which has contributed to the formulation of the theory of neocytolysis. We aimed to evaluate the role of oxygen exclusively on erythropoiesis, comparing in vitro erythroid differentiation performed at atmospheric oxygen, a lower oxygen concentration (three percent oxygen) and with cultures of erythroid precursors isolated from peripheral blood after a 19-day sojourn at high altitude (3450 m). Results highlight an accelerated erythroid maturation at low oxygen and more concave morphology of reticulocytes. No differences in deformability were observed in the formed reticulocytes in the tested conditions. Moreover, hematopoietic stem and progenitor cells isolated from blood affected by hypoxia at high altitude did not result in different erythroid development, suggesting no retention of a high-altitude signature but rather an immediate adaptation to oxygen concentration. This adaptation was observed during in vitro erythropoiesis at three percent oxygen by a significantly increased glycolytic metabolic profile. These hypoxia-induced effects on in vitro erythropoiesis fail to provide an intrinsic explanation of the concept of neocytolysis.
Collapse
Affiliation(s)
- Greta Simionato
- Department of Experimental Physics, University Campus, Building E2.6, Saarland University, 66123 Saarbrücken, Germany; (A.R.); (C.W.); (L.K.)
- Department of Experimental Surgery, Campus University Hospital, Building 65, Saarland University, 66421 Homburg, Germany
- Correspondence: (G.S.); (E.v.d.A.)
| | - Antonia Rabe
- Department of Experimental Physics, University Campus, Building E2.6, Saarland University, 66123 Saarbrücken, Germany; (A.R.); (C.W.); (L.K.)
| | - Joan Sebastián Gallego-Murillo
- Sanquin Research, Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, The Netherlands;
- Department of Biotechnology, Faculty of Applied Sciences, Delft University of Technology, 2629 HZ Delft, The Netherlands
| | - Carmen van der Zwaan
- Sanquin Research, Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, The Netherlands; (C.v.d.Z.); (A.J.H.); (M.v.d.B.)
| | - Arie Johan Hoogendijk
- Sanquin Research, Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, The Netherlands; (C.v.d.Z.); (A.J.H.); (M.v.d.B.)
| | - Maartje van den Biggelaar
- Sanquin Research, Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, The Netherlands; (C.v.d.Z.); (A.J.H.); (M.v.d.B.)
| | - Giampaolo Minetti
- Department of Biology and Biotechnology “L. Spallanzani”, Laboratories of Biochemistry, University of Pavia, I-27100 Pavia, Italy;
| | - Anna Bogdanova
- Red Blood Cell Research Group, Institute of Veterinary Physiology, University of Zurich, CH-8057 Zurich, Switzerland;
| | - Heimo Mairbäurl
- University Hospital Heidelberg, Medical Clinic VII, Sports Medicine, 69120 Heidelberg, Germany;
- Translational Lung Research Centre Heidelberg (TLRC), Part of the German Centre for Lung Research (DZL), 69120 Heidelberg, Germany
- Translational Pneumology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Christian Wagner
- Department of Experimental Physics, University Campus, Building E2.6, Saarland University, 66123 Saarbrücken, Germany; (A.R.); (C.W.); (L.K.)
- Physics and Materials Science Research Unit, University of Luxembourg, L-1511 Luxembourg City, Luxembourg
| | - Lars Kaestner
- Department of Experimental Physics, University Campus, Building E2.6, Saarland University, 66123 Saarbrücken, Germany; (A.R.); (C.W.); (L.K.)
- Theoretical Medicine and Biosciences, Campus University Hospital, Building 61.4, Saarland University, 66421 Homburg, Germany
| | - Emile van den Akker
- Sanquin Research, Landsteiner Laboratory, Department of Hematopoiesis, Amsterdam UMC, University of Amsterdam, 1066 CX Amsterdam, The Netherlands;
- Correspondence: (G.S.); (E.v.d.A.)
| |
Collapse
|
6
|
Ghosh S, Raundhal M, Myers SA, Carr SA, Chen X, Petsko GA, Glimcher LH. Identification of RIOK2 as a master regulator of human blood cell development. Nat Immunol 2022; 23:109-121. [PMID: 34937919 DOI: 10.1038/s41590-021-01079-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 10/25/2021] [Indexed: 11/09/2022]
Abstract
Anemia is a major comorbidity in aging, chronic kidney and inflammatory diseases, and hematologic malignancies. However, the transcriptomic networks governing hematopoietic differentiation in blood cell development remain incompletely defined. Here we report that the atypical kinase RIOK2 (right open reading frame kinase 2) is a master transcription factor (TF) that not only drives erythroid differentiation, but also simultaneously suppresses megakaryopoiesis and myelopoiesis in primary human stem and progenitor cells. Our study reveals the previously uncharacterized winged helix-turn-helix DNA-binding domain and two transactivation domains of RIOK2 that are critical to regulate key hematopoietic TFs GATA1, GATA2, SPI1, RUNX3 and KLF1. This establishes RIOK2 as an integral component of the transcriptional regulatory network governing human hematopoietic differentiation. Importantly, RIOK2 mRNA expression significantly correlates with these TFs and other hematopoietic genes in myelodysplastic syndromes, acute myeloid leukemia and chronic kidney disease. Further investigation of RIOK2-mediated transcriptional pathways should yield therapeutic approaches to correct defective hematopoiesis in hematologic disorders.
Collapse
Affiliation(s)
- Shrestha Ghosh
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Mahesh Raundhal
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Department of Immunology, Harvard Medical School, Boston, MA, USA.,Jnana Therapeutics, Boston, MA, USA
| | - Samuel A Myers
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Steven A Carr
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Xi Chen
- Department of Molecular & Cellular Biology, Lester and Sue Smith Breast Center, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Gregory A Petsko
- Department of Neurology, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Laurie H Glimcher
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA. .,Department of Immunology, Harvard Medical School, Boston, MA, USA. .,Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Wan G, Medina S, Zhang H, Pan R, Zhou X, Bolt AM, Luo L, Burchiel SW, Liu KJ. Arsenite exposure inhibits the erythroid differentiation of human hematopoietic progenitor CD34 + cells and causes decreased levels of hemoglobin. Sci Rep 2021; 11:22121. [PMID: 34764389 PMCID: PMC8586241 DOI: 10.1038/s41598-021-01643-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 11/01/2021] [Indexed: 02/02/2023] Open
Abstract
Arsenic exposure poses numerous threats to human health. Our previous work in mice has shown that arsenic causes anemia by inhibiting erythropoiesis. However, the impacts of arsenic exposure on human erythropoiesis remain largely unclear. We report here that low-dose arsenic exposure inhibits the erythroid differentiation of human hematopoietic progenitor cells (HPCs). The impacts of arsenic (in the form of arsenite; As3+) on red blood cell (RBC) development was evaluated using a long-term culture of normal human bone marrow CD34+-HPCs stimulated in vitro to undergo erythropoiesis. Over the time course studied, we analyzed the expression of the cell surface antigens CD34, CD71 and CD235a, which are markers commonly used to monitor the progression of HPCs through the stages of erythropoiesis. Simultaneously, we measured hemoglobin content, which is an important criterion used clinically for diagnosing anemia. As compared to control, low-dose As3+ exposure (100 nM and 500 nM) inhibited the expansion of CD34+-HPCs over the time course investigated; decreased the number of committed erythroid progenitors (BFU-E and CFU-E) and erythroblast differentiation in the subsequent stages; and caused a reduction of hemoglobin content. These findings demonstrate that low-dose arsenic exposure impairs human erythropoiesis, likely by combined effects on various stages of RBC formation.
Collapse
Affiliation(s)
- Guanghua Wan
- Department of Pharmaceutical Sciences, The University of New Mexico College of Pharmacy, Albuquerque, NM, 87131, USA
| | - Sebastian Medina
- Department of Pharmaceutical Sciences, The University of New Mexico College of Pharmacy, Albuquerque, NM, 87131, USA
- Department of Biology, New Mexico Highlands University, Las Vegas, NM, 87701, USA
| | - Haikun Zhang
- Department of Pharmaceutical Sciences, The University of New Mexico College of Pharmacy, Albuquerque, NM, 87131, USA
| | - Rong Pan
- Department of Pharmaceutical Sciences, The University of New Mexico College of Pharmacy, Albuquerque, NM, 87131, USA
| | - Xixi Zhou
- Department of Pharmaceutical Sciences, The University of New Mexico College of Pharmacy, Albuquerque, NM, 87131, USA
| | - Alicia M Bolt
- Department of Pharmaceutical Sciences, The University of New Mexico College of Pharmacy, Albuquerque, NM, 87131, USA
| | - Li Luo
- Division of Epidemiology, Biostatistics and Preventive Medicine at the University of New Mexico, Albuquerque, NM, 87131, USA
| | - Scott W Burchiel
- Department of Pharmaceutical Sciences, The University of New Mexico College of Pharmacy, Albuquerque, NM, 87131, USA
| | - Ke Jian Liu
- Department of Pharmaceutical Sciences, The University of New Mexico College of Pharmacy, Albuquerque, NM, 87131, USA.
| |
Collapse
|
8
|
Leclerc CJ, Cooper TT, Bell GI, Lajoie GA, Flynn LE, Hess DA. Decellularized adipose tissue scaffolds guide hematopoietic differentiation and stimulate vascular regeneration in a hindlimb ischemia model. Biomaterials 2021; 274:120867. [PMID: 33992837 DOI: 10.1016/j.biomaterials.2021.120867] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 04/29/2021] [Accepted: 04/29/2021] [Indexed: 12/30/2022]
Abstract
Cellular therapies to stimulate therapeutic angiogenesis in individuals with critical limb ischemia (CLI) remain under intense investigation. In this context, the efficacy of cell therapy is dependent on the survival, biodistribution, and pro-angiogenic paracrine signaling of the cells transplanted. Hematopoietic progenitor cells (HPC) purified from human umbilical cord blood using high aldehyde dehydrogenase-activity (ALDHhi cells) and expanded ex vivo, represent a heterogeneous mixture of progenitor cells previously shown to support limb revascularization in mouse models of CLI. The objectives of this study were to investigate the utility of bioscaffolds derived from human decellularized adipose tissue (DAT) to guide the differentiation of seeded HPC in vitro and harness the pro-angiogenic capacity of HPC at the site of ischemia after implantation in vivo. Probing whether the DAT scaffolds altered HPC differentiation, label-free quantitative mass spectrometry and flow cytometric phenotype analyses indicated that culturing the HPC on the DAT scaffolds supported their differentiation towards the pro-angiogenic monocyte/macrophage lineage at the expense of megakaryopoiesis. Moreover, implantation of HPC in DAT scaffolds within a unilateral hindlimb ischemia model in NOD/SCID mice increased cell retention at the site of ischemia relative to intramuscular injection, and accelerated the recovery of limb perfusion, improved functional limb use and augmented CD31+ capillary density when compared to DAT implantation alone or saline-injected controls. Collectively, these data indicate that cell-instructive DAT scaffolds can direct therapeutic HPC differentiation towards the monocyte/macrophage lineage and represent a promising delivery platform for improving the efficacy of cell therapies for CLI.
Collapse
Affiliation(s)
- Christopher J Leclerc
- School of Biomedical Engineering, Amit Chakma Engineering Building, The University of Western Ontario, London, Ontario, Canada, N6A 5B9; Krembil Centre for Stem Cell Biology, Molecular Medicine Research Laboratories, Robarts Research Institute, London, Ontario, N6A 5B6, Canada
| | - Tyler T Cooper
- Krembil Centre for Stem Cell Biology, Molecular Medicine Research Laboratories, Robarts Research Institute, London, Ontario, N6A 5B6, Canada; Don Rix Protein Identification Facility, Department of Biochemistry, University of Western Ontario, London, Ontario, N6G 2V4, Canada
| | - Gillian I Bell
- Krembil Centre for Stem Cell Biology, Molecular Medicine Research Laboratories, Robarts Research Institute, London, Ontario, N6A 5B6, Canada
| | - Gilles A Lajoie
- Don Rix Protein Identification Facility, Department of Biochemistry, University of Western Ontario, London, Ontario, N6G 2V4, Canada
| | - Lauren E Flynn
- School of Biomedical Engineering, Amit Chakma Engineering Building, The University of Western Ontario, London, Ontario, Canada, N6A 5B9; Department of Chemical and Biochemical Engineering, Thompson Engineering Building, The University of Western Ontario, London, Ontario, N6A 5B9, Canada; Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, N6A 3K7, Canada
| | - David A Hess
- Krembil Centre for Stem Cell Biology, Molecular Medicine Research Laboratories, Robarts Research Institute, London, Ontario, N6A 5B6, Canada; Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, N6A 5C1, Canada.
| |
Collapse
|
9
|
Hussein E, DeFor T, Wagner JE, Sumstad D, Brunstein CG, McKenna DH. Evaluation of post-thaw CFU-GM: clinical utility and role in quality assessment of umbilical cord blood in patients receiving single unit transplant. Transfusion 2019; 60:144-154. [PMID: 31756003 DOI: 10.1111/trf.15592] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/06/2019] [Accepted: 10/06/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND The CFU assay is considered the only in vitro assay that assesses the biologic function of hematopoietic stem and progenitor cells (HSPC). STUDY DESIGN AND METHODS To investigate the impact of post-thaw CFU-GM counts on the quality of umbilical cord blood (UCB), we studied transplant outcomes in 269 patients receiving single UCB transplant. We also correlated the post-thaw CFU-GM counts of 1912 units with the pre-freeze and post-thaw graft characteristics, hoping to optimize selection criteria of UCB. Data analysis included: total nucleated cells, viability, CD34+, nucleated red blood cells (NRBC), hematocrit, frozen storage time, and cord blood bank (CBB). RESULTS We demonstrated an association between post-thaw CFU-GM dose and the speed of neutrophil and platelet engraftment (p < 0.01). Higher post-thaw CFU-GM dose showed an increased benefit for neutrophil and platelet engraftment (p < 0.01). Post-thaw CD34+ cell dose and CFU-GM dose were strongly correlated (r = 0.78). However, CFU-GM dose showed additional benefit for patients receiving the lowest quartile of CD34+ dose. HLA disparity did not adversely impact either neutrophil or platelet engraftment. Post-thaw CFU-GM/million nucleated cells plated showed moderate correlation with pre-freeze and post-thaw CD34+ and weak correlation with other parameters. Post-thaw CFU-GM was not influenced by storage time, but was impacted by the CBB from which the unit is obtained (p < 0.01). CONCLUSION Post-thaw CFU-GM is an effective measure of the quality and efficacy of the UCB graft, particularly adding valuable clinical information when the CD34+ cell dose is low. Consideration of pre-freeze CD34+ cell content and CBB as additional selection criteria is warranted.
Collapse
Affiliation(s)
- Eiman Hussein
- Department of Laboratory medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
| | - Todd DeFor
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - John E Wagner
- Blood and Marrow Transplant Program, Department of Pediatrics, Minneapolis, Minnesota
| | - Darin Sumstad
- Department of Laboratory medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
| | - Claudio G Brunstein
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - David H McKenna
- Department of Laboratory medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
10
|
Nimker S, Sharma K, Saraswathy R, Chandna S. Delineating the Effects of Ionizing Radiation on Erythropoietic Lineage-Implications for Radiation Biodosimetry. HEALTH PHYSICS 2019; 116:677-693. [PMID: 30720544 DOI: 10.1097/hp.0000000000000975] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The overall lethality/morbidity of ionizing radiation exposure involves multiple forms of inhibitory or cytotoxic effects that may manifest in different tissues with a varying dose and time response. One of the major systemic effects leading to lethality of radiation includes its suppressive effect on hematopoiesis, which could be observed even at doses as low as 1-2 Gy, whereas effects on gastrointestinal and nervous systems appear at relatively higher doses in the same order. This article reviews the effects of radiation on the three distinct stages of erythropoiesis-formation of erythroid progenitor cells, differentiation of erythroid precursor cells, and terminal maturation. During these stepwise developmental processes, erythroid progenitor cells undergo rapid expansion to form terminally differentiated red blood cells that are continuously replenished from bone marrow into the circulating peripheral blood stream. Cellular radiation response depends upon many factors such as cell lineage, rate of proliferation, and differentiation status. Therefore, we discuss radiation-induced alterations during the progenitor, precursor, and terminal maturation stages and the implications thereof. Since biomarkers of ionizing radiation exposure in human populations are of great interest for assessing normal tissue injury as well as for biodosimetry in the event of accidental or incidental radiation exposures, we also highlight blood-based biomarkers that have potential utility for medical management.
Collapse
Affiliation(s)
- Shwetanjali Nimker
- Division of Natural Radiation Response Mechanisms, Institute of Nuclear Medicine and Allied Sciences, Timarpur, Delhi, India
- School of Biosciences and Technology, Vellore Institiute of Technology, Vellore, Tamil Nadu, India
| | - Kanupriya Sharma
- Division of Natural Radiation Response Mechanisms, Institute of Nuclear Medicine and Allied Sciences, Timarpur, Delhi, India
| | - Radha Saraswathy
- School of Biosciences and Technology, Vellore Institiute of Technology, Vellore, Tamil Nadu, India
| | - Sudhir Chandna
- Division of Natural Radiation Response Mechanisms, Institute of Nuclear Medicine and Allied Sciences, Timarpur, Delhi, India
| |
Collapse
|
11
|
Rotolo A, Karadimitris A, Ruella M. Building upon the success of CART19: chimeric antigen receptor T cells for hematologic malignancies. Leuk Lymphoma 2018; 59:2040-2055. [PMID: 29165008 PMCID: PMC6814196 DOI: 10.1080/10428194.2017.1403024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Chimeric antigen receptor T cell (CART) therapy has dramatically changed the therapeutic prospects for B cell malignancies. Over the last decade CD19-redirected CART have demonstrated the ability to induce deep, long-lasting remissions and possibly cure patients with relapsing B cell neoplasms. Such impressive results with CART19 fostered efforts to expand this technology to other incurable malignancies that naturally do not express CD19, such as acute myeloid leukemia (AML), Hodgkin lymphoma (HL) and multiple myeloma (MM). However, to reach this goal, several hurdles have to be overcome, in particular: (i) the apparent lack of suitable targets as effective as CD19; (ii) the immunosuppressive tumor microenvironment; (iii) intra-tumoral heterogeneity and antigen-negative relapses. Therefore, new strategies that allow safer and more potent CART platforms are under development and may provide grounds for new exciting breakthroughs in the field.
Collapse
Affiliation(s)
- Antonia Rotolo
- Centre for Haematology, Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - Anastasios Karadimitris
- Centre for Haematology, Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - Marco Ruella
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
12
|
Minagawa K, Jamil MO, AL-Obaidi M, Pereboeva L, Salzman D, Erba HP, Lamb LS, Bhatia R, Mineishi S, Di Stasi A. In Vitro Pre-Clinical Validation of Suicide Gene Modified Anti-CD33 Redirected Chimeric Antigen Receptor T-Cells for Acute Myeloid Leukemia. PLoS One 2016; 11:e0166891. [PMID: 27907031 PMCID: PMC5132227 DOI: 10.1371/journal.pone.0166891] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 11/04/2016] [Indexed: 12/20/2022] Open
Abstract
Background Approximately fifty percent of patients with acute myeloid leukemia can be cured with current therapeutic strategies which include, standard dose chemotherapy for patients at standard risk of relapse as assessed by cytogenetic and molecular analysis, or high-dose chemotherapy with allogeneic hematopoietic stem cell transplant for high-risk patients. Despite allogeneic hematopoietic stem cell transplant about 25% of patients still succumb to disease relapse, therefore, novel strategies are needed to improve the outcome of patients with acute myeloid leukemia. Methods and findings We developed an immunotherapeutic strategy targeting the CD33 myeloid antigen, expressed in ~ 85–90% of patients with acute myeloid leukemia, using chimeric antigen receptor redirected T-cells. Considering that administration of CAR T-cells has been associated with cytokine release syndrome and other potential off-tumor effects in patients, safety measures were here investigated and reported. We genetically modified human activated T-cells from healthy donors or patients with acute myeloid leukemia with retroviral supernatant encoding the inducible Caspase9 suicide gene, a ΔCD19 selectable marker, and a humanized third generation chimeric antigen receptor recognizing human CD33. ΔCD19 selected inducible Caspase9-CAR.CD33 T-cells had a 75±3.8% (average ± standard error of the mean) chimeric antigen receptor expression, were able to specifically lyse CD33+ targets in vitro, including freshly isolated leukemic blasts from patients, produce significant amount of tumor-necrosis-factor-alpha and interferon-gamma, express the CD107a degranulation marker, and proliferate upon antigen specific stimulation. Challenging ΔCD19 selected inducible Caspase9-CAR.CD33 T-cells with programmed-death-ligand-1 enriched leukemia blasts resulted in significant killing like observed for the programmed-death-ligand-1 negative leukemic blasts fraction. Since the administration of 10 nanomolar of a non-therapeutic dimerizer to activate the suicide gene resulted in the elimination of only 76.4±2.0% gene modified cells in vitro, we found that co-administration of the dimerizer with either the BCL-2 inhibitor ABT-199, the pan-BCL inhibitor ABT-737, or mafosfamide, resulted in an additive effect up to complete cell elimination. Conclusions This strategy could be investigated for the safety of CAR T-cell applications, and targeting CD33 could be used as a ‘bridge” therapy for patients coming to allogeneic hematopoietic stem cell transplant, as anti-leukemia activity from infusing CAR.CD33 T-cells has been demonstrated in an ongoing clinical trial. Albeit never performed in the clinical setting, our future plan is to investigate the utility of iC9-CAR.CD33 T-cells as part of the conditioning therapy for an allogeneic hematopoietic stem cell transplant for acute myeloid leukemia, together with other myelosuppressive agents, whilst the activation of the inducible Caspase9 suicide gene would grant elimination of the infused gene modified T-cells prior to stem cell infusion to reduce the risk of engraftment failure as the CD33 is also expressed on a proportion of the donor stem cell graft.
Collapse
MESH Headings
- B7-H1 Antigen/pharmacology
- Biphenyl Compounds/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Caspase 9/genetics
- Caspase 9/immunology
- Cell Engineering
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cellular Reprogramming
- Clinical Trials as Topic
- Cyclophosphamide/analogs & derivatives
- Cyclophosphamide/pharmacology
- Cytotoxicity, Immunologic
- Genetic Vectors
- Humans
- Interferon-gamma/biosynthesis
- Interferon-gamma/immunology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Lysosomal-Associated Membrane Protein 1/genetics
- Lysosomal-Associated Membrane Protein 1/immunology
- Myeloid Cells/drug effects
- Myeloid Cells/immunology
- Myeloid Cells/pathology
- Nitrophenols/pharmacology
- Piperazines/pharmacology
- Primary Cell Culture
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Sialic Acid Binding Ig-like Lectin 3/antagonists & inhibitors
- Sialic Acid Binding Ig-like Lectin 3/genetics
- Sialic Acid Binding Ig-like Lectin 3/immunology
- Sulfonamides/pharmacology
- T-Lymphocytes/cytology
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- Tumor Necrosis Factor-alpha/biosynthesis
- Tumor Necrosis Factor-alpha/immunology
Collapse
Affiliation(s)
- Kentaro Minagawa
- Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Muhammad O. Jamil
- Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Mustafa AL-Obaidi
- Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Larisa Pereboeva
- Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Donna Salzman
- Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Harry P. Erba
- Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Lawrence S. Lamb
- Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Ravi Bhatia
- Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Shin Mineishi
- Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Antonio Di Stasi
- Hematology/Oncology, University of Alabama at Birmingham, Birmingham, AL, United States of America
- * E-mail:
| |
Collapse
|
13
|
Hoehn RS, Jernigan PL, Chang AL, Edwards MJ, Caldwell CC, Gulbins E, Pritts TA. Acid Sphingomyelinase Inhibition Prevents Hemolysis During Erythrocyte Storage. Cell Physiol Biochem 2016; 39:331-40. [PMID: 27352097 PMCID: PMC5731776 DOI: 10.1159/000445627] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2016] [Indexed: 12/29/2022] Open
Abstract
Background/Aims During storage, units of human red blood cells (pRBCs) experience membrane destabilization and hemolysis which may cause harm to transfusion recipients. This study investigates whether inhibition of acid sphingomyelinase could stabilize erythrocyte membranes and prevent hemolysis during storage. Methods Human and murine pRBCs were stored under standard blood banking conditions with and without the addition of amitriptyline, a known acid sphingomyelinase inhibitor. Hemoglobin was measured with an electronic hematology analyzer and flow cytometry was used to measure erythrocyte size, complexity, phosphatidylserine externalization, and band 3 protein expression. Results Cell-free hemoglobin, a marker of hemolysis, increased during pRBC storage. Amitriptyline treatment decreased hemolysis in a dose-dependent manner. Standard pRBC storage led to loss of erythrocyte size and membrane complexity, increased phosphatidylserine externalization, and decreased band 3 protein integrity as determined by flow cytometry. Each of these changes was reduced by treatment with amitriptyline. Transfusion of amitriptyline-treated pRBCs resulted in decreased circulating free hemoglobin. Conclusion Erythrocyte storage is associated with changes in cell size, complexity, membrane molecular composition, and increased hemolysis. Acid sphingomyelinase inhibition reduced these changes in a dose-dependent manner. Our data suggest a novel mechanism to attenuate the harmful effects after transfusion of aged blood products.
Collapse
Affiliation(s)
- Richard S Hoehn
- Department of Surgery and Institute for Military Medicine, University of Cincinnati, Cincinnati, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Li H, Hasserjian RP, Kroft SH, Harrington AM, Wheaton SE, Pildain A, Ewalt MD, Gratzinger D, Hosking P, Olteanu H. Pure Erythroid Leukemia and Erythroblastic Sarcoma Evolving From Chronic Myeloid Neoplasms. Am J Clin Pathol 2016; 145:538-51. [PMID: 27124944 DOI: 10.1093/ajcp/aqw033] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES Pure erythroid leukemia (PEL) is an extremely rare entity that may, even more rarely, evolve from a preexisting chronic myeloid neoplasm (CMN); there is minimal literature regarding this latter phenomenon. METHODS We describe 14 patients with PEL that represented progression from a preexisting myelodysplastic syndrome (MDS, n = 8) or myeloproliferative neoplasm (MPN, n = 6), three of which manifested as erythroblastic sarcoma (EBS), a rare entity. These patients had a highly complex karyotype with prominent clonal evolution and a very aggressive clinical course. RESULTS Patients with PEL from MDS showed a more rapid progression time to PEL and had lower platelet counts compared with PEL from MPN. No other significant differences were found between the two groups. CONCLUSIONS These data represent the largest cohort of patients with PEL and an antecedent CMN, as well as the largest series of EBS reported to date, and underscore the unique morphologic, cytogenetic, immunophenotypic, and clinical features of this uncommon entity.
Collapse
Affiliation(s)
- Hongmei Li
- From the Department of Pathology, Medical College of Wisconsin, Milwaukee
| | | | - Steven H Kroft
- From the Department of Pathology, Medical College of Wisconsin, Milwaukee
| | | | | | - Alex Pildain
- Department of Pathology, Texas Health Presbyterian Hospital, Dallas
| | - Mark D Ewalt
- Department of Pathology, Stanford University, Stanford, CA
| | | | - Paul Hosking
- From the Department of Pathology, Medical College of Wisconsin, Milwaukee
| | - Horatiu Olteanu
- From the Department of Pathology, Medical College of Wisconsin, Milwaukee
| |
Collapse
|
15
|
Interleukin-2 critically regulates bone marrow erythropoiesis and prevents anemia development. Eur J Immunol 2015; 45:3362-74. [DOI: 10.1002/eji.201545596] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 07/30/2015] [Accepted: 09/21/2015] [Indexed: 12/18/2022]
|
16
|
Rich IN. Improving Quality and Potency Testing for Umbilical Cord Blood: A New Perspective. Stem Cells Transl Med 2015; 4:967-73. [PMID: 26160959 DOI: 10.5966/sctm.2015-0036] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 06/01/2015] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED This article critically reviews current methods to test and characterize umbilical cord blood (UCB) for hematopoietic stem cell transplantation. These tests include total nucleated cell (TNC) count, viability, viable CD34-positive content, and the colony-forming unit assay. It is assumed that the data obtained are sufficient to perform a UCB stem cell transplant without actually determining the quality and potency of the stem cells responsible for engraftment. This assumption has led not only to a high graft failure rate attributed to low or lack of potency, but also to noncompliance with present statutes that require UCB stem cells to be of high quality and, indeed, potency for a transplant to be successful. New evidence now calls into question the quality of the data, based on the UCB processed TNC fraction because using this impure fraction masks and significantly underestimates the functionality of the stem cells in both the segment and the unit. It is proposed that UCB units should be processed to the mononuclear cell fraction and that new cost-effective technology that measures the quality and potency of UCB stem cells be implemented to achieve better practices in UCB testing. These changes would provide the transplant physician with the assurance that the stem cells will perform as intended and would reduce risk and increase safety and efficacy for the patient. SIGNIFICANCE Current stem cell transplantation of umbilical cord blood cells requires testing that includes four basic parameters that do not determine whether the stem cells are of high quality, as required by the Stem Cell Therapeutic and Research Act of 2005. No cord blood units collected or transplanted so far have been tested for stem cell quality or potency. New scientific evidence calls into question cord blood processing and testing practices required by regulatory agencies and standards organizations. A new perspective is described that includes stem cell quality and potency testing that could reduce graft failure rates.
Collapse
Affiliation(s)
- Ivan N Rich
- HemoGenix, Inc., Colorado Springs, Colorado, USA
| |
Collapse
|
17
|
Pelleri MC, Piovesan A, Caracausi M, Berardi AC, Vitale L, Strippoli P. Integrated differential transcriptome maps of Acute Megakaryoblastic Leukemia (AMKL) in children with or without Down Syndrome (DS). BMC Med Genomics 2014; 7:63. [PMID: 25476127 PMCID: PMC4304173 DOI: 10.1186/s12920-014-0063-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 11/12/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The incidence of Acute Megakaryoblastic Leukemia (AMKL) is 500-fold higher in children with Down Syndrome (DS) compared with non-DS children, but the relevance of trisomy 21 as a specific background of AMKL in DS is still an open issue. Several Authors have determined gene expression profiles by microarray analysis in DS and/or non-DS AMKL. Due to the rarity of AMKL, these studies were typically limited to a small group of samples. METHODS We generated integrated quantitative transcriptome maps by systematic meta-analysis from any available gene expression profile dataset related to AMKL in pediatric age. This task has been accomplished using a tool recently described by us for the generation and the analysis of quantitative transcriptome maps, TRAM (Transcriptome Mapper), which allows effective integration of data obtained from different experimenters, experimental platforms and data sources. This allowed us to explore gene expression changes involved in transition from normal megakaryocytes (MK, n=19) to DS (n=43) or non-DS (n=45) AMKL blasts, including the analysis of Transient Myeloproliferative Disorder (TMD, n=20), a pre-leukemia condition. RESULTS We propose a biological model of the transcriptome depicting progressive changes from MK to TMD and then to DS AMKL. The data indicate the repression of genes involved in MK differentiation, in particular the cluster on chromosome 4 including PF4 (platelet factor 4) and PPBP (pro-platelet basic protein); the gene for the mitogen-activated protein kinase MAP3K10 and the thrombopoietin receptor gene MPL. Moreover, comparing both DS and non-DS AMKL with MK, we identified three potential clinical markers of progression to AMKL: TMEM241 (transmembrane protein 241) was the most over-expressed single gene, while APOC2 (apolipoprotein C-II) and ZNF587B (zinc finger protein 587B) appear to be the most discriminant markers of progression, specifically to DS AMKL. Finally, the chromosome 21 (chr21) genes resulted to be the most over-expressed in DS and non-DS AMKL, as well as in TMD, pointing out a key role of chr21 genes in differentiating AMKL from MK. CONCLUSIONS Our study presents an integrated original model of the DS AMLK transcriptome, providing the identification of genes relevant for its pathophysiology which can potentially be new clinical markers.
Collapse
Affiliation(s)
- Maria Chiara Pelleri
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Unit of Histology, Embryology and Applied Biology, University of Bologna, Via Belmeloro 8, 40126, Bologna, BO, Italy.
| | - Allison Piovesan
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Unit of Histology, Embryology and Applied Biology, University of Bologna, Via Belmeloro 8, 40126, Bologna, BO, Italy.
| | - Maria Caracausi
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Unit of Histology, Embryology and Applied Biology, University of Bologna, Via Belmeloro 8, 40126, Bologna, BO, Italy.
| | - Anna Concetta Berardi
- Research Laboratory Stem Cells, U.O.C. Immunohematology-Transfusion Medicine and Laboratory of Hematology, Santo Spirito's Hospital, Via del Circuito, 65100, Pescara, Italy.
| | - Lorenza Vitale
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Unit of Histology, Embryology and Applied Biology, University of Bologna, Via Belmeloro 8, 40126, Bologna, BO, Italy.
| | - Pierluigi Strippoli
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Unit of Histology, Embryology and Applied Biology, University of Bologna, Via Belmeloro 8, 40126, Bologna, BO, Italy. .,Interdepartmental Center for Cancer Research Giorgio Prodi (CIRC), S. Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138, Bologna, BO, Italy.
| |
Collapse
|
18
|
A hyperactive Mpl-based cell growth switch drives macrophage-associated erythropoiesis through an erythroid-megakaryocytic precursor. Blood 2014; 125:1025-33. [PMID: 25343958 DOI: 10.1182/blood-2014-02-555318] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Several approaches for controlling hematopoietic stem and progenitor cell expansion, lineage commitment, and maturation have been investigated for improving clinical interventions. We report here that amino acid substitutions in a thrombopoietin receptor (Mpl)--containing cell growth switch (CGS) extending receptor stability improve the expansion capacity of human cord blood CD34(+) cells in the absence of exogenous cytokines. Activation of this CGS with a chemical inducer of dimerization (CID) expands total cells 99-fold, erythrocytes 70-fold, megakaryocytes 0.5-fold, and CD34(+) stem/progenitor cells 4.4-fold by 21 days of culture. Analysis of cells in these expanded populations identified a CID-dependent bipotent erythrocyte-megakaryocyte precursor (PEM) population, and a CID-independent macrophage population. The CD235a(+)/CD41a(+) PEM population constitutes up to 13% of the expansion cultures, can differentiate into erythrocytes or megakaryocytes, exhibits very little expansion capacity, and exists at very low levels in unexpanded cord blood. The CD206(+) macrophage population constitutes up to 15% of the expansion cultures, exhibits high-expansion capacity, and is physically associated with differentiating erythroblasts. Taken together, these studies describe a fundamental enhancement of the CGS expansion platform, identify a novel precursor population in the erythroid/megakaryocytic differentiation pathway of humans, and implicate an erythropoietin-independent, macrophage-associated pathway supporting terminal erythropoiesis in this expansion system.
Collapse
|
19
|
Treatment of CD33-directed chimeric antigen receptor-modified T cells in one patient with relapsed and refractory acute myeloid leukemia. Mol Ther 2014; 23:184-91. [PMID: 25174587 DOI: 10.1038/mt.2014.164] [Citation(s) in RCA: 329] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Accepted: 08/25/2014] [Indexed: 12/20/2022] Open
Abstract
We conducted a clinical trial to assess the feasibility and efficacy of CD33-directed chimeric antigen receptor-modified T cells (CART-33) for the treatment of refractory acute myeloid leukemia (AML). A 41-year-old male patient with AML was enrolled and received a total of 1.12 × 10(9) autologous CART-33 cells, of which ~38% were transduced with CAR. The CART-33 infusion alone induced rigorous chills and fevers; drastic fluctuations of his preexisting pancytopenia; elevated serum cytokine levels, including interleukin (IL)-6, IL-8, tumor necrosis factor-α, and interferon-γ; slight transient hyperbilirubinemia within 2 weeks; a subsequent intermittent moderate fever; and reversed fluctuation of the pancytopenia. A marked decrease of blasts in the bone marrow was observed on examination 2 weeks after therapy, and there was a gradual increase until florid disease progression occurred at 9 weeks after the cell infusion. These observations warrant further research on CART-33 treatment in refractory AML and may spur efforts to extend the CART-33-induced tumor burden to the preparation of other intensive strategies, such as hematopoietic stem cell transplantation. This study is registered at www.ClinicalTrials.gov as NCT01864902.
Collapse
|
20
|
Coller BS. The platelet: life on the razor's edge between hemorrhage and thrombosis. Transfusion 2014; 54:2137-46. [PMID: 25092268 DOI: 10.1111/trf.12806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 06/24/2014] [Indexed: 12/26/2022]
Affiliation(s)
- Barry S Coller
- Laboratory of Blood and Vascular Biology, The Rockefeller University, New York, New York
| |
Collapse
|
21
|
Wang W, Grier DD, Woo J, Ward M, Sui G, Torti SV, Torti FM, Beaty MW. Ferritin H is a novel marker of early erythroid precursors and macrophages. Histopathology 2013; 62:931-40. [PMID: 23611361 DOI: 10.1111/his.12101] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 01/15/2013] [Indexed: 11/29/2022]
Abstract
AIMS Macrophages play a critical role in iron homeostasis by recycling iron from red cells and storing it in ferritin, an iron storage protein. The recycled iron is delivered to erythroid precursors for erythropoiesis. In this study, we aimed to determine whether ferritin is highly expressed in macrophages and erythroid precursors, and whether it can be used as a marker for these two cell types. METHODS AND RESULTS A ferritin monoclonal antibody was developed, and immunohistochemistry was performed. In normal bone marrows, ferritin antibody stained early erythroid precursors and macrophages. In contrast, myeloid cells, lymphoid cells and megakaryocytes lacked ferritin expression. In leukaemic bone marrows, ferritin was selectively expressed in erythroid blasts (M6), whereas all other blasts were negative. In lymph nodes, ferritin was highly and specifically expressed in macrophages, whereas lymphocytes completely lacked ferritin expression. In non-haematopoietic tissues, ferritin antibody highlighted alveolar macrophages in the lung, as well as sinus macrophages in the liver and spleen. CONCLUSIONS We conclude that ferritin is a novel and reliable marker for macrophages and early erythroid precursors, and may be of clinical utility in the diagnosis of diseases associated with these two cell types.
Collapse
Affiliation(s)
- Wei Wang
- Department of Pathology, Wake Forest Baptist Health, Winston Salem, NC 27157, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Frascione N, Pinto V, Daniel B. Development of a biosensor for human blood: new routes to body fluid identification. Anal Bioanal Chem 2012; 404:23-8. [DOI: 10.1007/s00216-012-6111-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 05/04/2012] [Accepted: 05/13/2012] [Indexed: 11/30/2022]
|
23
|
In Vitro and In Vivo Antitumor Effect of Anti-CD33 Chimeric Receptor-Expressing EBV-CTL against CD33 Acute Myeloid Leukemia. Adv Hematol 2012; 2012:683065. [PMID: 22272203 PMCID: PMC3261457 DOI: 10.1155/2012/683065] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 10/05/2011] [Indexed: 11/18/2022] Open
Abstract
Genetic engineering of T cells with chimeric T-cell receptors (CARs) is an attractive strategy to treat malignancies. It extends the range of antigens for adoptive T-cell immunotherapy, and major mechanisms of tumor escape are bypassed. With this strategy we redirected immune responses towards the CD33 antigen to target acute myeloid leukemia. To improve in vivo T-cell persistence, we modified human Epstein Barr Virus-(EBV-) specific cytotoxic T cells with an anti-CD33.CAR. Genetically modified T cells displayed EBV and HLA-unrestricted CD33 bispecificity in vitro. In addition, though showing a myeloablative activity, they did not irreversibly impair the clonogenic potential of normal CD34(+) hematopoietic progenitors. Moreover, after intravenous administration into CD33(+) human acute myeloid leukemia-bearing NOD-SCID mice, anti-CD33-EBV-specific T cells reached the tumor sites exerting antitumor activity in vivo. In conclusion, targeting CD33 by CAR-modified EBV-specific T cells may provide additional therapeutic benefit to AML patients as compared to conventional chemotherapy or transplantation regimens alone.
Collapse
|
24
|
Ossenkoppele GJ, van de Loosdrecht AA, Schuurhuis GJ. Review of the relevance of aberrant antigen expression by flow cytometry in myeloid neoplasms. Br J Haematol 2011; 153:421-36. [PMID: 21385170 DOI: 10.1111/j.1365-2141.2011.08595.x] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This article reviews the use of aberrant antigen expression detected by flow cytometry in the diagnosis and clinical handling of acute myeloid leukaemia (AML) and the myelodysplastic syndromes (MDS). Such aberrancies offer a valuable tool for the proper classification of these myeloid malignancies according the World Health Organization 2008 classification. Aberrant antigen expression by flow cytometry is also important for prognostification. This review supports the view, that minimal residual disease detection methods that make use of such aberrancies should be part of the routine management of AML patients to guide therapy, but also suggests the introduction of flow cytometry in MDS for diagnosis and treatment decisions in the near future.
Collapse
Affiliation(s)
- Gert J Ossenkoppele
- Department of Haematology, VU University Medical Center, De Boelelaan 1117, Amsterdam, the Netherlands.
| | | | | |
Collapse
|
25
|
Immunophenotyping of acute leukemia and lymphoproliferative disorders: a consensus proposal of the European LeukemiaNet Work Package 10. Leukemia 2011; 25:567-74. [DOI: 10.1038/leu.2010.312] [Citation(s) in RCA: 169] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
26
|
Zein S, Li W, Ramakrishnan V, Lou TF, Sivanand S, Mackie A, Pace B. Identification of fetal hemoglobin-inducing agents using the human leukemia KU812 cell line. Exp Biol Med (Maywood) 2010; 235:1385-94. [PMID: 20975082 DOI: 10.1258/ebm.2010.010129] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Fetal hemoglobin (HbF) ameliorates the clinical severity of sickle cell disease; therefore continued research to identify efficacious HbF-inducing agents is desirable. In this study, we investigated KU812 leukemia cells that express the fetal γ-globin and adult β-globin genes, as a system for screening and discovery of novel HbF inducers. KU812 cells were analyzed in the presence or absence of fetal bovine serum and then expression levels of the globin genes, cell surface markers and transcription factors were quantified by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). For comparison, primary erythroid cells were grown in a two-phase liquid culture system. After drug inductions for 48-72 h, globin mRNA and HbF levels were quantified by RT-qPCR and enzyme-linked immunosorbent assay, respectively. Erythroid markers and transcription factors expression levels in KU812 cells were comparable to days 7-14 erythroid cells. We also tested several drugs including butyrate, trichostatin A, scriptaid, suberoylanilide hydroxamic acid and hydroxyurea, which induced γ-globin in KU812 cells; however, some agents also induced β-globin. A novel agent STI-571 was studied in the system, which non-selectively induced the globin genes. Additional studies showed comparable globin gene response patterns in KU812 and primary erythroid cells after treatments with the various drug inducers. Mechanisms of drug-mediated γ-globin induction in KU812 cells require signaling through the p38 mitogen-activated protein kinase pathway similar to that previously demonstrated in primary erythroid cells. These data suggest that KU812 cells serve as a good screening system to identify potential HbF inducers for the treatment of β-hemoglobinopathies.
Collapse
Affiliation(s)
- Sima Zein
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Marsee DK, Pinkus GS, Yu H. CD71 (transferrin receptor): an effective marker for erythroid precursors in bone marrow biopsy specimens. Am J Clin Pathol 2010; 134:429-35. [PMID: 20716799 DOI: 10.1309/ajcpcrk3moaoj6at] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Accurate analysis of the erythroid lineage is essential in evaluating bone marrow biopsy specimens and can be particularly challenging in the setting of dyserythropoiesis. Transferrin receptor (CD71) mediates the uptake of transferrin-iron complexes and is highly expressed on the surface of cells of the erythroid lineage. Although CD71 has been used for flow cytometric analysis, its usefulness in paraffin-embedded bone marrow biopsy specimens has not been examined. This study defined the immunohistochemical profile of CD71, as compared with glycophorin A (CD235a) and hemoglobin, in 65 bone marrow biopsy specimens, including normal marrow specimens and cases of myelodysplastic syndrome, acute myeloid leukemia, acute lymphoblastic leukemia, plasma cell neoplasm, and metastatic carcinoma. Immunoreactivity for CD71 was restricted to erythroid precursors in normal and dyspoietic marrow samples and exhibited a membranous and cytoplasmic staining pattern. The vast majority of mature erythrocytes lack expression of CD71, greatly facilitating interpretation. CD71 is a highly effective marker for the detection of cells of erythroid lineage in bone marrow biopsy specimens.
Collapse
|
28
|
Liu B, Ohishi K, Yamamura K, Suzuki K, Monma F, Ino K, Masuya M, Sekine T, Heike Y, Takaue Y, Katayama N. A potential activity of valproic acid in the stimulation of interleukin-3−mediated megakaryopoiesis and erythropoiesis. Exp Hematol 2010; 38:685-95. [DOI: 10.1016/j.exphem.2010.03.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2009] [Revised: 02/28/2010] [Accepted: 03/25/2010] [Indexed: 10/19/2022]
|
29
|
Mountford J, Olivier E, Turner M. Prospects for the manufacture of red cells for transfusion. Br J Haematol 2010; 149:22-34. [DOI: 10.1111/j.1365-2141.2010.08079.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
30
|
Filippone C, Franssila R, Kumar A, Saikko L, Kovanen PE, Söderlund-Venermo M, Hedman K. Erythroid progenitor cells expanded from peripheral blood without mobilization or preselection: molecular characteristics and functional competence. PLoS One 2010; 5:e9496. [PMID: 20209110 PMCID: PMC2830487 DOI: 10.1371/journal.pone.0009496] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Accepted: 01/19/2010] [Indexed: 11/18/2022] Open
Abstract
Background Continued development of in-vitro procedures for expansion and differentiation of erythroid progenitor cells (EPC) is essential not only in hematology and stem cell research but also virology, in light of the strict erythrotropism of the clinically important human parvovirus B19. Methodology/Principal Findings We cultured EPC directly from ordinary blood samples, without ex vivo stem cell mobilization or CD34+ cell in vitro preselection. Profound increase in the absolute cell number and clustering activity were observed during culture. The cells obtained expressed the EPC marker combination CD36, CD71 and glycophorin, but none of the lymphocyte, monocyte or NK markers. The functionality of the generated EPC was examined by an in vitro infection assay with human parvovirus B19, tropic for BFU-E and CFU-E cells. Following infection (i) viral DNA replication and mRNA production were confirmed by quantitative PCR, and (ii) structural and nonstructural proteins were expressed in >50% of the cells. As the overall cell number increased 100–200 fold, and the proportion of competent EPC (CD34+ to CD36+) rose from <0.5% to >50%, the in vitro culture procedure generated the EPC at an efficiency of >10 000-fold. Comparative culturing of unselected PBMC and ex vivo-preselected CD34+ cells produced qualitatively and quantitatively similar yields of EPC. Conclusions/Significance This approach yielding EPC directly from unmanipulated peripheral blood is gratifyingly robust and will facilitate the study of myeloid infectious agents such as the B19 virus, as well as the examination of erythropoiesis and its cellular and molecular mechanisms.
Collapse
Affiliation(s)
- Claudia Filippone
- Department of Virology, Haartman Institute, University of Helsinki and Helsinki University Laboratory Division, Helsinki, Finland
- * E-mail:
| | - Rauli Franssila
- Department of Virology, Haartman Institute, University of Helsinki and Helsinki University Laboratory Division, Helsinki, Finland
| | - Arun Kumar
- Department of Virology, Haartman Institute, University of Helsinki and Helsinki University Laboratory Division, Helsinki, Finland
| | - Leena Saikko
- Department of Pathology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Panu E. Kovanen
- Department of Pathology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Maria Söderlund-Venermo
- Department of Virology, Haartman Institute, University of Helsinki and Helsinki University Laboratory Division, Helsinki, Finland
| | - Klaus Hedman
- Department of Virology, Haartman Institute, University of Helsinki and Helsinki University Laboratory Division, Helsinki, Finland
| |
Collapse
|
31
|
Martin MG, Augustin KM, Uy GL, Welch JS, Hladnik L, Goyal S, Tiwari D, Monahan RS, Reichley RM, Cashen AF, Stockerl-Goldstein K, Westervelt P, Abboud CN, Dipersio JF, Vij R. Salvage therapy for acute myeloid leukemia with fludarabine, cytarabine, and idarubicin with or without gemtuzumab ozogamicin and with concurrent or sequential G-CSF. Am J Hematol 2009; 84:733-7. [PMID: 19806665 DOI: 10.1002/ajh.21545] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The current salvage therapies for relapsed/refractory acute myeloid leukemia (AML) are unsatisfactory. Over the past 7 years, we have used two salvage regimens: fludarabine, cytarabine, and idarubicin with (FLAG-IM) or without gemtuzumab ozogamicin (GO) (9 mg/m(2) on Day 8) (FLAG-I) in relapsed/refractory AML. Three-quarters of patients also received concurrent G-CSF. Seventy-one patients were treated, 23 with FLAG-I and 48 with FLAG-IM. The median duration of follow-up was 30.6 months. The treatment groups were well balanced with median ages of 48 years (range 18-70) and 47 years (range 20-68), unfavorable cytogenetics in 57% and 35%, prior allogeneic stem cell transplant in 43% and 42%, and CR1 duration <1 year in 60% and 67%, respectively, for FLAG-I and FLAG-IM. The complete remission (CR) rate in the FLAG-I group was 39% with an additional 13% achieving a CRp [overall response rate (ORR) 52%]; the CR rate in the FLAG-IM group was 29% with an additional 27% achieving a CRp (ORR 56%). The median duration of response (DOR; 16.8 vs. 8.3 months), event-free survival (EFS; 7.4 vs. 4.1 months), and overall survival (OS; 8.8 vs. 5.0 months) trended to favor FLAG-I over FLAG-IM. The patients who received G-CSF concurrent with chemotherapy had superior overall response rate (ORR; 62% vs. 29%, P = 0.026), median EFS (6.2 vs. 3.4 months, P = 0.010), and OS (8.8 vs. 3.9 months, P = 0.004) when compared with those who sequentially received G-CSF and chemotherapy, regardless of chemotherapy regimen. The addition of GO, at this dose and schedule, to FLAG-I failed to improve the outcomes in patients with relapsed/refractory AML. The patients who received G-CSF concurrently with chemotherapy had improved outcomes. Am. J. Hematol., 2009. (c) 2009 Wiley-Liss, Inc.
Collapse
Affiliation(s)
- Mike G Martin
- Section of Leukemia and Bone Marrow Transplantation, Division of Oncology, Washington University School of Medicine, Saint Louis, Missouri, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
A novel fluorescence-based method in forensic science for the detection of blood in situ. Forensic Sci Int Genet 2008; 2:363-71. [DOI: 10.1016/j.fsigen.2008.06.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Revised: 05/20/2008] [Accepted: 06/04/2008] [Indexed: 11/23/2022]
|
33
|
Abstract
The study of thrombopoiesis has evolved greatly since an era when platelets were termed "the dust of the blood," only about 100 years ago. During this time megakaryocytes were identified as the origin of blood platelets; marrow-derived megakaryocytic progenitor cells were functionally defined and then purified; and the primary regulator of the process, thrombopoietin, was cloned and characterized and therapeutic thrombopoietic agents developed. During this journey we continue to learn that the physiologic mechanisms that drive proplatelet formation can be recapitulated in cell-free systems and their biochemistry evaluated; the molecular underpinnings of endomitosis are being increasingly understood; the intracellular signals sent by engagement of a large number of megakaryocyte surface receptors have been defined; and many of the transcription factors that drive megakaryocytic fate determination have been identified and experimentally manipulated. While some of these biologic processes mimic those seen in other cell types, megakaryocytes and platelets possess enough unique developmental features that we are virtually assured that continued study of thrombopoiesis will yield innumerable clinical and scientific insights for many decades to come.
Collapse
|
34
|
Ex vivo-generated CD36+ erythroid progenitors are highly permissive to human parvovirus B19 replication. J Virol 2007; 82:2470-6. [PMID: 18160440 DOI: 10.1128/jvi.02247-07] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The pathogenic parvovirus B19 (B19V) has an extreme tropism for human erythroid progenitor cells. In vitro, only a few erythroid leukemic cell lines (JK-1 and KU812Ep6) or megakaryoblastoid cell lines (UT7/Epo and UT7/Epo-S1) with erythroid characteristics support B19V replication, but these cells are only semipermissive. By using recent advances in generating large numbers of human erythroid progenitor cells (EPCs) ex vivo from hematopoietic stem cells (HSCs), we produced a pure population of CD36(+) EPCs expanded and differentiated from CD34(+) HSCs and assessed the CD36(+) EPCs for their permissiveness to B19V infection. Over more than 3 weeks, cells grown in serum-free medium expanded more than 800,000-fold, and 87 to 96% of the CD36(+) EPCs were positive for globoside, the cellular receptor for B19V. Immunofluorescence (IF) staining showed that about 77% of the CD36(+) EPCs were positive for B19V infection, while about 9% of UT7/Epo-S1 cells were B19V positive. Viral DNA detected by real-time PCR increased by more than 3 logs in CD36(+) EPCs; the increase was 1 log in UT7/Epo-S1 cells. Due to the extensive permissivity of CD36(+) EPCs, we significantly improved the sensitivity of detection of infectious B19V by real-time reverse transcription-PCR and IF staining 100- and 1,000-fold, respectively, which is greater than the sensitivity of UT7/Epo-S1 cell-based methods. This is the first description of an ex vivo method to produce large numbers of EPCs that are highly permissive to B19V infection and replication, offering a cellular system that mimics in vivo infection with this pathogenic human virus.
Collapse
|
35
|
Malfitano AM, Toruner GA, Gazzerro P, Laezza C, Husain S, Eletto D, Orlando P, De Petrocellis L, Terskiy A, Schwalb M, Vitale E, Bifulco M. Arvanil and anandamide up-regulate CD36 expression in human peripheral blood mononuclear cells. Immunol Lett 2007; 109:145-54. [PMID: 17360047 DOI: 10.1016/j.imlet.2007.02.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2006] [Revised: 02/05/2007] [Accepted: 02/05/2007] [Indexed: 11/26/2022]
Abstract
In this study we analysed the regulation of gene expression by arvanil and anandamide in human peripheral blood mononuclear cells (PBMCs) to clarify their immunosuppressive properties. PBMCs were activated, leading to CD36 down regulation, that was normalized by arvanil and anandamide. We used microarray technology to identify a regulatory pattern associated with cell proliferation in the presence of both substances. CD3-CD28 stimulated PBMCs showed a pattern of up-regulated and down-regulated genes after treatment with these substances. We selected and analysed several genes chosen by their function in the regulation of cell proliferation. We showed a transcriptional control of the CD36 gene by arvanil and anandamide associated with an increased protein expression, thus suggesting a possible role of CD36 in anandamide and arvanil anti-inflammatory pattern.
Collapse
Affiliation(s)
- Anna Maria Malfitano
- Dipartimento di Scienze Farmaceutiche, Università di Salerno, Fisciano (SA), Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Bowles KM, Vallier L, Smith JR, Alexander MRJ, Pedersen RA. HOXB4 overexpression promotes hematopoietic development by human embryonic stem cells. Stem Cells 2006; 24:1359-69. [PMID: 16410392 DOI: 10.1634/stemcells.2005-0210] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Human embryonic stem cells (hESCs) are a potential source of hematopoietic cells for therapeutic transplantation and can provide a model for human hematopoiesis. Culture of hESCs on murine stromal layers or in stromal-free conditions as embryoid bodies results in low levels of hematopoietic cells. Here we demonstrate that overexpression of the transcription factor HOXB4 considerably augments hematopoietic development of hESCs. Stable HOXB4-expressing hESC clones were generated by lipofection and could be maintained in the undifferentiated state for prolonged passages. Moreover, differentiation of hESCs as embryoid bodies in serum-containing medium without the use of additional cytokines led to sequential expansion of first erythroid and then myeloid and monocytic progenitors from day 10 of culture. These cells retained the capacity to develop into formed blood elements during in vitro culture. Consistent with the development of committed hematopoietic cells, we observed the expression of transcription factors known to be critical for hematopoietic development. We thus demonstrate successful use of enforced gene expression to promote the differentiation of hESCs into a terminally differentiated tissue, thereby revealing an important role for HOXB4 in supporting their in vitro development along the hematopoietic pathway.
Collapse
Affiliation(s)
- Kristian M Bowles
- Department of Surgery, University of Cambridge, Cambridge Institute for Medical Research, Cambridge CB2 2XY, UK.
| | | | | | | | | |
Collapse
|
37
|
Zhang Y, Payne KJ, Zhu Y, Price MA, Parrish YK, Zielinska E, Barsky LW, Crooks GM. SCL expression at critical points in human hematopoietic lineage commitment. Stem Cells 2005; 23:852-60. [PMID: 15917481 DOI: 10.1634/stemcells.2004-0260] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The stem cell leukemia (SCL or tal-1) gene was initially identified as a translocation partner in a leukemia that possessed both lymphoid and myeloid differentiation potential. Mice that lacked SCL expression showed a complete block in hematopoiesis; thus, SCL was associated with hematopoietic stem cell (HSC) function. More recent studies show a role for SCL in murine erythroid differentiation. However, the expression pattern and the role of SCL during early stages of human hematopoietic differentiation are less clear. In this study we chart the pattern of human SCL expression from HSCs, through developmentally sequential populations of lymphoid and myeloid progenitors to mature cells of the hematopoietic lineages. Using recently defined surface immunophenotypes, we fluorescence-activated cell-sorted (FACS) highly purified populations of primary human hematopoietic progenitors for reverse transcription-polymerase chain reaction (RT-PCR) analysis of SCL expression. Our data show that SCL mRNA is easily detectable in all hematopoietic populations with erythroid potential, including HSCs, multipotential progenitors, common myeloid progenitors, megakaryocyte/erythrocyte progenitors, and nucleated erythroid lineage cells. SCL mRNA expression was present but rapidly downregulated in the common lymphoid progenitor and granulocyte/monocyte progenitor populations that lack erythroid potential. SCL expression was undetectable in immature cells of nonerythroid lineages, including pro-B cells, early thymic progenitors, and myeloid precursors expressing the M-CSF receptor. SCL expression was also absent from all mature cells of the nonerythroid lineages. Although low levels of SCL were detected in lymphoid- and myeloid-restricted progenitors, our studies show that abundant SCL expression is normally tightly linked with erythroid differentiation potential.
Collapse
Affiliation(s)
- Yanjia Zhang
- Childrens Hospital Los Angeles, M.S.#62, 4650 Sunset Boulevard, Los Angeles, CA 90027, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Crooks DM, Pacheco-Rodriguez G, DeCastro RM, McCoy JP, Wang JA, Kumaki F, Darling T, Moss J. Molecular and genetic analysis of disseminated neoplastic cells in lymphangioleiomyomatosis. Proc Natl Acad Sci U S A 2004; 101:17462-7. [PMID: 15583138 PMCID: PMC536045 DOI: 10.1073/pnas.0407971101] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2003] [Indexed: 01/19/2023] Open
Abstract
Lymphangioleiomyomatosis (LAM) is a multisystem disorder of women, characterized by cystic degeneration of the lungs, renal angiomyolipomas (AML), and lymphatic abnormalities. LAM lesions result from the proliferation of benign-appearing, smooth muscle-like LAM cells, which are characterized by loss of heterozygosity (LOH) of one of the tuberous sclerosis complex (TSC) genes. LAM cells are believed to migrate among the involved organs. Because of the apparently metastatic behavior of LAM, we tried to isolate LAM cells from body fluids. A cell fraction separated by density gradient centrifugation from blood had TSC2 LOH in 33 of 60 (55%) LAM patients. Cells with TSC2 LOH were also found in urine from 11 of 14 (79%) patients with AML and in chylous fluid from 1 of 3 (33%) patients. Identification of LAM cells with TSC2 LOH in body fluids was not correlated with severity of lung disease or extrapulmonary involvement and was found in one patient after double lung transplantation. These studies are compatible with a multisite origin for LAM cells. They establish the existence of disseminated, potentially metastatic LAM cells through a relatively simple, noninvasive procedure that should be valuable for molecular and genetic studies of somatic mutations in LAM and perhaps other metastatic diseases.
Collapse
Affiliation(s)
- Denise M Crooks
- Pulmonary-Critical Care Medicine Branch and Flow Cytometry Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Drexler HG, Matsuo Y, MacLeod RAF. Malignant hematopoietic cell lines: in vitro models for the study of erythroleukemia. Leuk Res 2004; 28:1243-51. [PMID: 15475063 DOI: 10.1016/j.leukres.2004.03.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2004] [Accepted: 03/22/2004] [Indexed: 10/26/2022]
Abstract
A panel of leukemia cell lines has been assembled over the last 30 years representing a spectrum of erythroid cells arrested at various stages of differentiation. The oldest cell line is K-562 which is one of the most prolific in use. Most cell lines have been established from acute myeloid leukemia M6 or chronic myeloid leukemia in blast crisis and generally express immunoprofiles typically seen in immature erythroid cells. Several cell lines are constitutively growth factor-dependent, responding proliferatively to a variety of cytokines. The predominant cytogenetic abnormalities are the t(9;22)(q34;q11) found exclusively in CML-derived cell lines, and rearrangements of chromosomes 5 and 7 which occur in all disease subtypes. Ph+ve cell lines consistently displayed structural and numerical changes associated with disease evolution, including +8, -17/17p-/i(17q), and +19. It is striking that many cell lines though committed to either the erythroid or megakaryocytic lineage tend to co-express features of the other lineage, consistent with the concept of a common erythroid-megakaryocytic progenitor. Several cell lines may be induced to differentiate along the erythroid, megakaryocytic or monocytic pathway by treatment with pharmacological or physiological reagents. Notable functional features include expression of various globin chains or the complete hemoglobins as erythroid attributes. Taken together, this class of cell lines is relatively well characterized and affords useful model systems for immature erythroid cells.
Collapse
|
40
|
Cerdan C, Rouleau A, Bhatia M. VEGF-A165 augments erythropoietic development from human embryonic stem cells. Blood 2003; 103:2504-12. [PMID: 14656883 DOI: 10.1182/blood-2003-07-2563] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Combinations of hematopoietic cytokines and the ventral mesoderm inducer BMP-4 have recently been shown to augment hematopoietic cell fate of human embryonic stem cells (hESCs) during embryoid body (EB) development. However, factors capable of regulating lineage commitment of hESC-derived hematopoiesis have yet to be reported. Here we show that vascular endothelial growth factor (VEGF-A165) selectively promotes erythropoietic development from hESCs. Effects of VEGF-A165 were dependent on the presence of hematopoietic cytokines and BMP-4, and could be augmented by addition of erythropoietin (EPO). Treatment of human EBs with VEGF-A165 increased the frequency of cells coexpressing CD34 and the VEGF-A165 receptor KDR, as well as cells expressing erythroid markers. Although fetal/adult globins were unaffected, VEGF-A165 induced the expression of embryonic zeta (zeta) and epsilon (epsilon) globins, and was accompanied by expression of the hematopoietic transcription factor SCL/Tal-1. In addition to promoting erythropoietic differentiation from hESCs, the presence of VEGF-A165 enhanced the in vitro self-renewal potential of primitive hematopoietic cells capable of erythroid progenitor capacity. Our study demonstrates a role for VEGF-A165 during erythropoiesis of differentiating hESCs, thereby providing the first evidence for a factor capable of regulating hematopoietic lineage development of hESCs.
Collapse
Affiliation(s)
- Chantal Cerdan
- Robarts Research Institute, Stem Cell Biology and Regenerative Medicine, The University of Western Ontario, London, ON, Canada
| | | | | |
Collapse
|
41
|
Schwartz GN, Vance BA, Levine BM, Fukazawa M, Telford WG, Cesar D, Hellerstein M, Gress RE. Proliferation kinetics of subpopulations of human marrow cells determined by quantifying in vivo incorporation of [2H2]-glucose into DNA of S-phase cells. Blood 2003; 102:2068-73. [PMID: 12763933 DOI: 10.1182/blood-2003-01-0139] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
This report investigated in vivo turnover kinetics of marrow hematopoietic progenitors and precursors using a recently developed stable isotope-mass spectrometric technique (SIMST). Human subjects were administered a 2-day infusion of 6,6-[2H2]-glucose, a nontoxic stable isotope-labeled form of glucose, which becomes incorporated into DNA of all S-phase cells. The percent [2H2]-glucose incorporated into DNA in the form of [2H2]-deoxyadenosine (%[2H2]-dA enrichment) was determined by gas chromatography-mass spectrometry. The rate constant of replacement of unlabeled by labeled DNA strands (labeling kinetics) was used to calculate population turnover kinetics of CD34+ cells, CD133+ cells, and CD133-CD34+ cells. The observed mean replacement half-life (t1/2) was 2.6 days for CD34+ cells, 2.5 days for CD133-CD34+ cells, and 6.2 days for CD133+ cells. Results from the estimated rate constant of replacement of labeled by unlabeled DNA (delabeling kinetics) also demonstrated slower turnover rates for CD133+ cells than for CD133-CD34+ cells. Although there was a relatively rapid initial decrease in the %[2H2]-dA enrichment, low levels of labeled DNA persisted in CD34+ cells for at least 4 weeks. The results indicate the presence of subpopulations of CD34+ cells with relatively rapid turnover rates and subpopulations with a slower t1/2 of 28 days. Results also demonstrate that in vivo [2H2]-glucose-SIMST is sensitive enough to detect differences in turnover kinetics between erythroid and megakaryocyte lineage cells. These studies are the first to demonstrate the use of in vivo [2H2]-glucose-SIMST to measure in vivo turnover kinetics of subpopulations of CD34+ cells and precursors in healthy human subjects.
Collapse
Affiliation(s)
- Gretchen N Schwartz
- Experimental Transplantation and Immunology Branch, National Cancer Institute/NIH, Bldg 10, Room 12S237, MSC 1907, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Somervaille TCP, Linch DC, Khwaja A. Different levels of p38 MAP kinase activity mediate distinct biological effects in primary human erythroid progenitors. Br J Haematol 2003; 120:876-86. [PMID: 12614225 DOI: 10.1046/j.1365-2141.2003.04204.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
There have been conflicting reports regarding the role of p38 mitogen-activated protein kinase (MAPK) in the regulation of differentiation, proliferation and apoptosis in erythroid cell lines. We have, therefore, examined the functions of this kinase in primary human erythroid progenitors. Cells in steady-state culture showed low-level p38 MAPK activity, which decreased further within 1 h of growth factor withdrawal and increased over a limited range within minutes of re-exposure of cells to erythropoietin or stem cell factor, demonstrating the link between low-level p38 MAPK activity and the prevailing growth factor milieu. Use of the p38 MAPK-specific inhibitor SB203580 demonstrated that this level of activity was necessary for (1) optimal proliferation, (2) erythroid burst-forming unit migration and (3) full upregulation of E-cadherin and CD36 expression, but not haemoglobin A or glycophorin A expression, during human erythroid differentiation. In contrast, cells deprived of growth factors for an 8-h period, following a transient decrease in p38 MAPK activity, demonstrated sustained, substantial and caspase-independent increases in p38 MAPK activity, and its blockade using SB203580 reduced the proportion of erythroblasts undergoing apoptosis by 40 +/- 7%, demonstrating a role for p38 MAPK in apoptosis induction in human erythroblasts. Thus, in primary human erythroblasts, different environmental conditions induce different levels of p38 MAPK activity, which have distinct functions.
Collapse
Affiliation(s)
- Tim C P Somervaille
- Department of Haematology, Royal Free and University College Medical School, London, UK.
| | | | | |
Collapse
|
43
|
Harashima A, Suzuki M, Okochi A, Yamamoto M, Matsuo Y, Motoda R, Yoshioka T, Orita K. CD45 tyrosine phosphatase inhibits erythroid differentiation of umbilical cord blood CD34+ cells associated with selective inactivation of Lyn. Blood 2002; 100:4440-5. [PMID: 12393728 DOI: 10.1182/blood-2002-03-0864] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
CD45 is a membrane-associated tyrosine phosphatase that dephosphorylates Src family kinases and Janus kinases (JAKs). To clarify the role of CD45 in hematopoietic differentiation, we examined the effects of anti-CD45 monoclonal antibody NU-L(PAN) on the proliferation and differentiation of umbilical cord blood CD34(+) cells. NU-L(PAN) showed a prominent inhibition of the proliferation of CD34(+) cells induced by the mouse bone marrow stromal cell line MS-5 or erythropoietin (EPO). However, NU-L(PAN) did not affect the proliferation induced by interleukin 3. NU-L(PAN) also inhibited MS-5-induced or EPO-induced erythroid differentiation of CD34(+) cells. The cells stimulated with EPO in the presence of NU-L(PAN) morphologically showed differentiation arrest at the stage of basophilic erythroblasts after 11 days of culture, whereas the cells treated with EPO without NU-L(PAN) differentiated into mature red blood cells. The Src family kinase Lyn and JAK2 were phosphorylated when erythroblasts obtained after 4 days of culture of CD34(+) cells in the presence of EPO were restimulated with EPO. Overnight NU-L(PAN) treatment before addition of EPO reduced the phosphorylation of Lyn but not that of JAK2. Simultaneously, the enhancement of Lyn kinase activity after restimulation with EPO was reduced by NU-L(PAN) treatment. These results indicate selective inactivation of Lyn by CD45 activated with NU-L(PAN) and could partly explain the inhibitory mechanism on erythropoiesis exhibited by EPO. These findings suggest that CD45 may play a pivotal role in erythropoiesis.
Collapse
Affiliation(s)
- Akira Harashima
- Fujisaki Cell Center, Hayashibara Biochemical Labs, and the Kurashiki Medical Center, Kurashiki, Okayama, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Clay D, Rubinstein E, Mishal Z, Anjo A, Prenant M, Jasmin C, Boucheix C, Le Bousse-Kerdilès MC. CD9 and megakaryocyte differentiation. Blood 2001; 97:1982-9. [PMID: 11264162 DOI: 10.1182/blood.v97.7.1982] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
It is shown that the tetraspanin CD9 has a complex pattern of distribution in hematopoietic cells and is heterogeneously expressed on human bone marrow CD34(+) cells. CD34(high)CD38(low)Thy1(+) primitive progenitors are contained in the population with intermediate CD9 expression, thus suggesting that CD9 expression may precede CD38 appearance. Cell sorting shows that colony-forming unit (CFU)-GEMM and CFU-GM are present in high proportions in this fraction and in the fraction with the lowest CD9 expression. Cells with the highest level of CD9 are committed to the B-lymphoid or megakaryocytic (MK) lineages, as shown by the co-expression of either CD19 or CD41/GPIIb and by their strong potential to give rise to CFU-MK. In liquid cultures, CD9(high)CD41(neg) cells give rise to cells with high CD41 expression as early as 2 days, and this was delayed by at least 3 to 4 days for the CD9(mid) cells; few CD41(high) cells could be detected in the CD9(low) cell culture, even after 6 days. Antibody ligation of cell surface CD9 increased the number of human CFU-MK progenitors and reduced the production of CD41(+) megakaryocytic cells in liquid culture. This was associated with a decreased expression of MK differentiation antigens and with an alteration of the membrane structure of MK cells. Altogether these data show a precise regulation of CD9 during hematopoiesis and suggest a role for this molecule in megakaryocytic differentiation, possibly by participation in membrane remodeling. (Blood. 2001;97:1982-1989)
Collapse
MESH Headings
- ADP-ribosyl Cyclase
- ADP-ribosyl Cyclase 1
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antigens, CD/biosynthesis
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antigens, CD/physiology
- Antigens, Differentiation/biosynthesis
- B-Lymphocytes/cytology
- Cell Differentiation
- Cell Lineage
- Cell Membrane/immunology
- Cell Membrane/ultrastructure
- Cells, Cultured
- Colony-Forming Units Assay
- Flow Cytometry
- Gene Expression Regulation, Developmental
- Humans
- Immunophenotyping
- Megakaryocytes/cytology
- Megakaryocytes/metabolism
- Megakaryocytes/ultrastructure
- Membrane Glycoproteins
- Microscopy, Electron
- NAD+ Nucleosidase/biosynthesis
- Platelet Glycoprotein GPIIb-IIIa Complex/biosynthesis
- Tetraspanin 29
Collapse
Affiliation(s)
- D Clay
- INSERM U268, Institut André LWOFF, Hôpital Paul Brousse, Villejuif, France
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Kowal-Vern A, Mazzella FM, Cotelingam JD, Shrit MA, Rector JT, Schumacher HR. Diagnosis and characterization of acute erythroleukemia subsets by determining the percentages of myeloblasts and proerythroblasts in 69 cases. Am J Hematol 2000; 65:5-13. [PMID: 10936857 DOI: 10.1002/1096-8652(200009)65:1<5::aid-ajh2>3.0.co;2-u] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Acute erythroleukemia (FAB M6) is a rare heterogeneous disease with an increase in red cell precursors and myeloblasts. Three subsets have been described: M6A (myeloblast-rich erythroleukemia); M6B (proerythroblast-rich erythroleukemia); and M6C (myeloblast- and proerythroblast-rich mixed variant). This study was undertaken to define and compare the clinical courses and survival outcomes among M6A, M6B, and M6C variants of erythroleukemia. Sixty-nine cases of M6 leukemia were categorized as consisting of >/=50% erythroid of all nucleated cells and M6A with >/=30% myeloblasts/nonerythroid component; M6B with >/=30% proerythroblasts/erythroid component; and M6C with >/=30% myeloblasts and >/=30% proerythroblasts. The demographics, cell type distribution, and survival (mean +/- sd) of these groups were compared. There were 32 M6A, 26 M6B, and 11 M6C patients. No significant difference was seen among the groups in age, sex, or treatment. Compared to M6A, both the M6B (P< 0.0001) and M6C (P< 0.0001) variants showed a statistically significant increase in the percentage of bone marrow erythroid cells, proerythroblasts, and the proerythroblasts/erythroid ratios. Comparing the groups for survival, M6B (3 +/- 3.6 months) versus M6A (25 +/- 28 months), P< 0. 002, and M6C (10 +/- 13 months) versus M6A, P< 0.01 had a poorer prognosis. Calculating the proerythroblasts as a component of total bone marrow erythroids provides a complimentary method for delineating the pure red cell erythroleukemia (M6B) and mixed variant (M6C), similar to that for the myeloid/erythroid (M6A) leukemia. Now that it is possible to delineate erythroleukemia subtypes, innovative treatments are indicated to target the malignant erythroid population, which is resistant to myeloid-based therapies.
Collapse
Affiliation(s)
- A Kowal-Vern
- Department of Pathology, Loyola University Medical Center, Maywood, Illinois, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Girodon F, Favre B, Couillaud G, Carli PM, Parmeland C, Maynadié M. Immunophenotype of a transient myeloproliferative disorder in a newborn with trisomy 21. CYTOMETRY 2000; 42:118-22. [PMID: 10797450 DOI: 10.1002/(sici)1097-0320(20000415)42:2<118::aid-cyto6>3.0.co;2-e] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Cytologic, immunologic, and cytogenetic studies were performed on the blast cells of a newborn with Down syndrome and transient myeloproliferative disease. This hematologic disorder is uncommon, and occurs primarily in infants with Down syndrome. This boy presented with a high white blood cell count and a high percentage of blast cells, without anemia or thrombocytopenia. Chromosome analysis showed a constitutional trisomy 21 without any other clonal abnormality. A three-color flow cytometric analysis was performed and revealed two different CD45 dim, CD34(+), CD117(+), CD56(+) immature subpopulations: the normal immature myeloid precursor and an immature blast cell population that expressed CD41, CD42, CD61, CD36, CD13, CD1a, and CD2. We postulate that this population could be the leukemic precursor involved in the acute megakaryoblastic leukemia frequently observed in children with Down syndrome.
Collapse
Affiliation(s)
- F Girodon
- Hematology Laboratory, University Hospital, Dijon, France
| | | | | | | | | | | |
Collapse
|
47
|
Berman E, Jhanwar S, McBride M, Strife A, Wisniewski D, Lambek C, Clarkson B. Characterization of two novel sublines established from a human megakaryoblastic leukemia cell line transfected with p210(BCR-ABL). Leuk Res 2000; 24:289-97. [PMID: 10713326 DOI: 10.1016/s0145-2126(99)00179-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Disease progression in chronic myelogenous leukemia (CML) is usually accompanied by chromosomal abnormalities such as an additional Ph chromosome, trisomies of chromosome 8 or 19, or i(17) in addition to the standard translocation t(9;22) (q34;q11). However, detailed studies of the various steps involved during this evolution are difficult to perform, thereby making the study of cell lines that contain the transposed genes BCR-ABL, especially those of human origin, an important focus. In this analysis we investigated the human megakaryoblastic cell line MO7e and its subline transfected with BCR-ABL, MO7e/p210. Initial studies demonstrated that the phenotype of the MO7e line was consistent with a megakaryocytic lineage as originally described and was growth factor dependent in liquid culture. The MO7e/p210 subline, however, was growth factor independent and could be further separated into two distinct sublines based on expression of glycophorin A using the monoclonal antibody R10. The subline R10 negative (R10-) was similar to the parent line MO7e but R10 positive (R10+) cells had a distinct erythroid phenotype. In addition, the R10- and R10+ sublines demonstrated strikingly different colony morphology when cultured in semisolid medium. Furthermore, R10+ cells had additional chromosomal abnormalities not detected in the R10- population. These results demonstrate that the insertion of the BCR-ABL in this human leukemia cell line resulted in two distinct subpopulations of cells, each now growth factor independent, but one with a phenotype and karyotype identical to the parent cell line and the other with a different phenotype and additional chromosomal abnormalities. These two subpopulations derived from the MO7e/p210 transfected cell line may prove useful in further understanding the multistep events that occur in the progression of this disease.
Collapse
MESH Headings
- Chromosome Aberrations
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/physiology
- Humans
- Immunophenotyping
- Leukemia, Megakaryoblastic, Acute/genetics
- Leukemia, Megakaryoblastic, Acute/immunology
- Leukemia, Megakaryoblastic, Acute/pathology
- Polymorphism, Restriction Fragment Length
- Transfection
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- E Berman
- Department of Medicine, The Laboratory of Hematopoietic Cell Kinetics, Division of Hematologic Oncology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Kern P, Kolowos W, Hagenhofer M, Frank C, Kalden JR, Herrmann M. Alternatively spliced mRNA molecules of the thrombospondin receptor (CD36) in human PBMC. EUROPEAN JOURNAL OF IMMUNOGENETICS : OFFICIAL JOURNAL OF THE BRITISH SOCIETY FOR HISTOCOMPATIBILITY AND IMMUNOGENETICS 1999; 26:337-42. [PMID: 10553499 DOI: 10.1046/j.1365-2370.1999.00167.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We employed reverse transcription polymerase chain reaction (RT-PCR) to detect alternatively spliced CD36 mRNA in human peripheral blood mononuclear cells (PBMC). Sequencing of cloned cDNA revealed alternatively spliced mRNA molecules in 13 out of 39 clones. We observed exon skipping of up to 10 out of 12 coding exons in eight alternative transcripts. Additionally, in five of the transcripts, alternative splice donor or acceptor sites were used during mRNA maturation. Considering the CD36 molecule serves many functions in coagulation, host defence, lipid metabolism, and scavenging, we speculate that the proteins encoded by the alternatively spliced mRNA molecules may be involved in regulation of both CD36 gene expression and function.
Collapse
Affiliation(s)
- P Kern
- Department of Internal Medicine III, Friedrich-Alexander-University of Erlangen-Nuremberg, Germany
| | | | | | | | | | | |
Collapse
|
49
|
Freyssinier JM, Lecoq-Lafon C, Amsellem S, Picard F, Ducrocq R, Mayeux P, Lacombe C, Fichelson S. Purification, amplification and characterization of a population of human erythroid progenitors. Br J Haematol 1999; 106:912-22. [PMID: 10519992 DOI: 10.1046/j.1365-2141.1999.01639.x] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In humans, studies of the erythroid cell lineage are hampered by difficulties in obtaining sufficient numbers of erythroid progenitors. In fact, these progenitors in bone marrow or peripheral blood are scarce and no specific antibodies are available. We describe a new method which allows proliferation in liquid culture of large numbers of pure normal human erythroid progenitors. CD34+ cells were cultured for 7 d in serum-free conditions with the cytokine mixture interleukin (IL)-3/IL-6/stem cell factor (SCF). This resulted in cell expansion and the appearance of a high proportion of CD36+ cells which were purified on day 7. Methylcellulose clones from these cells were composed of 96.6% late BFU-E and 3.4% CFU-GM. These CD36+ cells could be recultured with the same cytokine mixture plus or minus erythropoietin (Epo) for a further 2-7 d. In both conditions further amplification of CD36+ cells was observed, but Epo induced a more dramatic cell expansion. Glycophorin-positive mature cells appeared only in the presence of Epo, and terminal red cell differentiation was observed after 7 d of secondary culture. Cells obtained from adult CD34+ progenitors mostly contained adult haemoglobin, whereas cord blood-derived cells contained equal proportions of adult and fetal haemoglobin. Activation of STAT5 and tyrosine phosphorylation of the Epo receptor and JAK2 were observed after Epo stimulation of these cells. This new method represents a straightforward alternative to the procedures previously described for the purification of normal erythroid progenitors and is useful in the study of erythropoietic regulation.
Collapse
Affiliation(s)
- J M Freyssinier
- Laboratoire d'Hématopoïèse, ETS de 1'AP-HP, Site Transfusionnel Cochin, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Gubin AN, Njoroge JM, Bouffard GG, Miller JL. Gene expression in proliferating human erythroid cells. Genomics 1999; 59:168-77. [PMID: 10409428 DOI: 10.1006/geno.1999.5855] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A complete understanding of human erythropoiesis will require a robust description of transcriptional activity in hematopoietic cells that proliferate and differentiate in response to erythropoietin (EPO). For this purpose, we cultured peripheral blood mononuclear cells in the presence or in the absence of EPO and examined the transcriptional profile of those cells arising only in response to EPO. A distinct population of CD71( +) cells that demonstrated an average of six additional doublings in suspension culture and erythroid colony formation in methylcellulose was isolated. Suppression subtractive hybridization of mRNA isolated from those cells permitted the identification of transcribed genes. A summary of 719 expressed sequence tags (ESTs) describing 505 independent transcripts is provided here with a full analysis of each EST available at http://hembase.niddk.nih.gov. Several transcripts that matched genes previously reported in the context of erythroid differentiation including 4 cell surface proteins were expressed at this developmental stage. Active chromatin remodeling was suggested by the identification of 4 histone proteins, 4 high-mobility group proteins, 13 transcription factors, and 6 genes involved in DNA recombination and repair. Numerous genes associated with leukemic translocations were also recognized including topoisomerases I and II, nucleophosmin, Translin, EGR1, dek, pim-1, TFG, and MLL. In addition to known transcripts, 44 novel EST were discovered. This transcriptional profile provides the first genomic-scale description of gene activity in erythroid progenitor cells.
Collapse
Affiliation(s)
- A N Gubin
- Laboratory of Chemical Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, Maryland 20892, USA
| | | | | | | |
Collapse
|