101
|
|
102
|
Jacobsen SEW, Nerlov C. Haematopoiesis in the era of advanced single-cell technologies. Nat Cell Biol 2019; 21:2-8. [PMID: 30602765 DOI: 10.1038/s41556-018-0227-8] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 10/02/2018] [Indexed: 12/19/2022]
Abstract
The molecular and functional characterization of single cells at scale has emerged as a key driver to unravel tissue biology. Thus, it is important to understand the strengths and limitations of transcriptomic approaches, molecular barcoding and functional assays used to study cellular properties at the single-cell level. Here, we review recent relevant work from the haematopoietic system and discuss how to interpret and integrate data obtained with different technologies.
Collapse
Affiliation(s)
- Sten Eirik W Jacobsen
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK.
- Haematopoietic Stem Cell Laboratory, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK.
- Department of Cell and Molecular Biology, Wallenberg Institute for Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden.
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden.
- Karolinska University Hospital, Stockholm, Sweden.
| | - Claus Nerlov
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK.
| |
Collapse
|
103
|
Vlacil AK, Schuett J, Schieffer B, Grote K. Variety matters: Diverse functions of monocyte subtypes in vascular inflammation and atherogenesis. Vascul Pharmacol 2018; 113:9-19. [PMID: 30553027 DOI: 10.1016/j.vph.2018.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 12/06/2018] [Accepted: 12/07/2018] [Indexed: 12/24/2022]
Abstract
Monocytes are important mediators of the innate immunity by recognizing and attacking especially bacterial pathogens but also play crucial roles in various inflammatory diseases, including vascular inflammation and atherosclerosis. Maturation, differentiation and function of monocytes have been intensively explored for a long time in innumerable experimental and clinical studies. Monocytes do not represent a uniform cell type but could be further subdivided into subpopulations with distinct features and functions. Those subpopulations have been identified in experimental mouse models as well as in humans, albeit distinguished by different cell surface markers. While Ly6C is used for subpopulation differentiation in mice, corresponding human subsets are differentiated by CD14 and CD16. In this review, we specifically focused on new experimental insights from recent years mainly in regard to murine monocyte subpopulations and their roles in vascular inflammation und atherogenesis.
Collapse
Affiliation(s)
| | - Jutta Schuett
- Cardiology and Angiology, Philipps-University Marburg, Marburg, Germany
| | | | - Karsten Grote
- Cardiology and Angiology, Philipps-University Marburg, Marburg, Germany.
| |
Collapse
|
104
|
Bapat A, Keita N, Martelly W, Kang P, Seet C, Jacobsen JR, Stoilov P, Hu C, Crooks GM, Sharma S. Myeloid Disease Mutations of Splicing Factor SRSF2 Cause G2-M Arrest and Skewed Differentiation of Human Hematopoietic Stem and Progenitor Cells. Stem Cells 2018; 36:1663-1675. [PMID: 30004607 PMCID: PMC6283046 DOI: 10.1002/stem.2885] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/17/2018] [Accepted: 06/13/2018] [Indexed: 01/14/2023]
Abstract
Myeloid malignancies, including myelodysplastic syndromes, chronic myelomonocytic leukemia, and acute myeloid leukemia, are characterized by abnormal proliferation and differentiation of hematopoietic stem and progenitor cells (HSPCs). Reports on analysis of bone marrow samples from patients have revealed a high incidence of mutations in splicing factors in early stem and progenitor cell clones, but the mechanisms underlying transformation of HSPCs harboring these mutations remain unknown. Using ex vivo cultures of primary human CD34+ cells as a model, we find that mutations in splicing factors SRSF2 and U2AF1 exert distinct effects on proliferation and differentiation of HSPCs. SRSF2 mutations cause a dramatic inhibition of proliferation via a G2-M phase arrest and induction of apoptosis. U2AF1 mutations, conversely, do not significantly affect proliferation. Mutations in both SRSF2 and U2AF1 cause abnormal differentiation by skewing granulo-monocytic differentiation toward monocytes but elicit diverse effects on megakaryo-erythroid differentiation. The SRSF2 mutations skew differentiation toward megakaryocytes whereas U2AF1 mutations cause an increase in the erythroid cell populations. These distinct functional consequences indicate that SRSF2 and U2AF1 mutations have cell context-specific effects and that the generation of myeloid disease phenotype by mutations in the genes coding these two proteins likely involves different intracellular mechanisms. Stem Cells 2018;36:1663-1675.
Collapse
Affiliation(s)
- Aditi Bapat
- Department of Basic Medical Sciences, College of Medicine—PhoenixUniversity of ArizonaPhoenixArizonaUSA
| | - Nakia Keita
- Department of Basic Medical Sciences, College of Medicine—PhoenixUniversity of ArizonaPhoenixArizonaUSA
| | - William Martelly
- Department of Basic Medical Sciences, College of Medicine—PhoenixUniversity of ArizonaPhoenixArizonaUSA
| | - Paul Kang
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health—PhoenixUniversity of ArizonaPhoenixArizonaUSA
| | - Christopher Seet
- Department of Pathology and Laboratory Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Jeffery R. Jacobsen
- Department of Pathology and Laboratory MedicinePhoenix Children's HospitalPhoenixArizonaUSA
| | - Peter Stoilov
- Department of Biochemistry, School of MedicineWest Virginia UniversityMorgantownWest VirginiaUSA
| | - Chengcheng Hu
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health—PhoenixUniversity of ArizonaPhoenixArizonaUSA
| | - Gay M. Crooks
- Department of Pathology and Laboratory Medicine, David Geffen School of MedicineUniversity of California, Los AngelesLos AngelesCaliforniaUSA
| | - Shalini Sharma
- Department of Basic Medical Sciences, College of Medicine—PhoenixUniversity of ArizonaPhoenixArizonaUSA
| |
Collapse
|
105
|
Belluschi S, Calderbank EF, Ciaurro V, Pijuan-Sala B, Santoro A, Mende N, Diamanti E, Sham KYC, Wang X, Lau WWY, Jawaid W, Göttgens B, Laurenti E. Myelo-lymphoid lineage restriction occurs in the human haematopoietic stem cell compartment before lymphoid-primed multipotent progenitors. Nat Commun 2018; 9:4100. [PMID: 30291229 PMCID: PMC6173731 DOI: 10.1038/s41467-018-06442-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 09/04/2018] [Indexed: 02/02/2023] Open
Abstract
Capturing where and how multipotency is lost is crucial to understand how blood formation is controlled. Blood lineage specification is currently thought to occur downstream of multipotent haematopoietic stem cells (HSC). Here we show that, in human, the first lineage restriction events occur within the CD19-CD34+CD38-CD45RA-CD49f+CD90+ (49f+) HSC compartment to generate myelo-lymphoid committed cells with no erythroid differentiation capacity. At single-cell resolution, we observe a continuous but polarised organisation of the 49f+ compartment, where transcriptional programmes and lineage potential progressively change along a gradient of opposing cell surface expression of CLEC9A and CD34. CLEC9AhiCD34lo cells contain long-term repopulating multipotent HSCs with slow quiescence exit kinetics, whereas CLEC9AloCD34hi cells are restricted to myelo-lymphoid differentiation and display infrequent but durable repopulation capacity. We thus propose that human HSCs gradually transition to a discrete lymphoid-primed state, distinct from lymphoid-primed multipotent progenitors, representing the earliest entry point into lymphoid commitment.
Collapse
Affiliation(s)
- Serena Belluschi
- 0000000121885934grid.5335.0Department of Haematology and Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Emily F. Calderbank
- 0000000121885934grid.5335.0Department of Haematology and Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Valerio Ciaurro
- 0000000121885934grid.5335.0Department of Haematology and Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Blanca Pijuan-Sala
- 0000000121885934grid.5335.0Department of Haematology and Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Antonella Santoro
- 0000000121885934grid.5335.0Department of Haematology and Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Nicole Mende
- 0000000121885934grid.5335.0Department of Haematology and Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Evangelia Diamanti
- 0000000121885934grid.5335.0Department of Haematology and Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Kendig Yen Chi Sham
- 0000000121885934grid.5335.0Department of Haematology and Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Xiaonan Wang
- 0000000121885934grid.5335.0Department of Haematology and Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Winnie W. Y. Lau
- 0000000121885934grid.5335.0Department of Haematology and Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Wajid Jawaid
- 0000000121885934grid.5335.0Department of Haematology and Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Berthold Göttgens
- 0000000121885934grid.5335.0Department of Haematology and Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Elisa Laurenti
- 0000000121885934grid.5335.0Department of Haematology and Wellcome and MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
106
|
Braciak TA, Roskopf CC, Wildenhain S, Fenn NC, Schiller CB, Schubert IA, Jacob U, Honegger A, Krupka C, Subklewe M, Spiekermann K, Hopfner KP, Fey GH, Aigner M, Krause S, Mackensen A, Oduncu FS. Dual-targeting triplebody 33-16-123 (SPM-2) mediates effective redirected lysis of primary blasts from patients with a broad range of AML subtypes in combination with natural killer cells. Oncoimmunology 2018; 7:e1472195. [PMID: 30228941 PMCID: PMC6140553 DOI: 10.1080/2162402x.2018.1472195] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 04/13/2018] [Accepted: 04/29/2018] [Indexed: 12/20/2022] Open
Abstract
A number of agents designed for immunotherapy of Acute Myeloid Leukemia (AML) are in preclinical and early clinical development. Most of them target a single antigen on the surface of AML cells. Here we describe the development and key biological properties of a tri-specific agent, the dual-targeting triplebody SPM-2, with binding sites for target antigens CD33 and CD123, and for CD16 to engage NK cells as cytolytic effectors. Primary blasts of nearly all AML patients carry at least one of these target antigens and the pair is particularly promising for the elimination of blasts and leukemia stem cells (LSCs) from a majority of AML patients by dual-targeting agents. The cytolytic activity of NK cells mediated by SPM-2 was analyzed in vitro for primary leukemic cells from 29 patients with a broad range of AML-subtypes. Blasts from all 29 patients, including patients with genomic alterations associated with an unfavorable genetic subtype, were lysed at nanomolar concentrations of SPM-2. Maximum susceptibility was observed for cells with a combined density of CD33 and CD123 above 10,000 copies/cell. Cell populations enriched for AML-LSCs (CD34pos and CD34pos CD38neg cells) from 2 AML patients carried an increased combined antigen density and were lysed at correspondingly lower concentrations of SPM-2 than unsorted blasts. These initial findings raise the expectation that SPM-2 may also be capable of eliminating AML-LSCs and thus of prolonging survival. In the future, patients with a broad range of AML subtypes may benefit from treatment with SPM-2.
Collapse
Affiliation(s)
- Todd A. Braciak
- Department of Hematology and Oncology, Medizinische Klinik und Poliklinik III, Klinikum der Universität München, Munich, Germany
| | - Claudia C. Roskopf
- Department of Hematology and Oncology, Medizinische Klinik und Poliklinik III, Klinikum der Universität München, Munich, Germany
| | - Sarah Wildenhain
- Department of Biochemistry and Gene Center, Ludwig-Maximilians-Universität, Munich, Germany
| | - Nadja C. Fenn
- Department of Biochemistry and Gene Center, Ludwig-Maximilians-Universität, Munich, Germany
| | - Christian B. Schiller
- Department of Biochemistry and Gene Center, Ludwig-Maximilians-Universität, Munich, Germany
| | - Ingo A. Schubert
- Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| | | | | | - Christina Krupka
- Department of Hematology and Oncology, Medizinische Klinik und Poliklinik III, Klinikum der Universität München, Munich, Germany
- Laboratory of Translational Cancer Immunol ogy, Gene Center of the LMU Munich, Munich, Germany
| | - Marion Subklewe
- Department of Hematology and Oncology, Medizinische Klinik und Poliklinik III, Klinikum der Universität München, Munich, Germany
- Laboratory of Translational Cancer Immunol ogy, Gene Center of the LMU Munich, Munich, Germany
| | - Karsten Spiekermann
- Department of Hematology and Oncology, Medizinische Klinik und Poliklinik III, Klinikum der Universität München, Munich, Germany
| | - Karl-Peter Hopfner
- Department of Biochemistry and Gene Center, Ludwig-Maximilians-Universität, Munich, Germany
| | - Georg H. Fey
- Department of Biology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Michael Aigner
- Department of Internal Medicine 5 - Hematology/Oncology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Stefan Krause
- Department of Internal Medicine 5 - Hematology/Oncology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5 - Hematology/Oncology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Fuat S. Oduncu
- Department of Hematology and Oncology, Medizinische Klinik und Poliklinik III, Klinikum der Universität München, Munich, Germany
| |
Collapse
|
107
|
Yáñez A, Goodridge HS. Identification and Isolation of Oligopotent and Lineage-committed Myeloid Progenitors from Mouse Bone Marrow. J Vis Exp 2018. [PMID: 30102291 DOI: 10.3791/58061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Myeloid progenitors that yield neutrophils, monocytes and dendritic cells (DCs) can be identified in and isolated from the bone marrow of mice for hematological and immunological analyses. For example, studies of the cellular and molecular properties of myeloid progenitor populations can reveal mechanisms underlying leukemic transformation, or demonstrate how the immune system responds to pathogen exposure. Previously described flow cytometry strategies for myeloid progenitor identification have enabled significant advances in many fields, but the fractions they identify are very heterogeneous. The most commonly used gating strategies define bone marrow fractions that are enriched for the desired populations, but also contain large numbers of "contaminating" progenitors. Our recent studies have resolved much of this heterogeneity, and the protocol we present here permits the isolation of 6 subpopulations of oligopotent and lineage-committed myeloid progenitors from 2 previously described bone marrow fractions. The protocol describes 3 stages: 1) isolation of bone marrow cells, 2) enrichment for hematopoietic progenitors by magnetic-activated cell sorting (lineage depletion by MACS), and 3) identification of myeloid progenitor subsets by flow cytometry (including fluorescence-activated cell sorting, FACS, if desired). This approach permits progenitor quantification and isolation for a variety of in vitro and in vivo applications, and has already yielded novel insight into pathways and mechanisms of neutrophil, monocyte, and DC differentiation.
Collapse
Affiliation(s)
- Alberto Yáñez
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center; Research Division of Immunology, Cedars-Sinai Medical Center
| | - Helen S Goodridge
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center; Research Division of Immunology, Cedars-Sinai Medical Center;
| |
Collapse
|
108
|
Radtke S, Adair JE, Giese MA, Chan YY, Norgaard ZK, Enstrom M, Haworth KG, Schefter LE, Kiem HP. A distinct hematopoietic stem cell population for rapid multilineage engraftment in nonhuman primates. Sci Transl Med 2018; 9:9/414/eaan1145. [PMID: 29093179 DOI: 10.1126/scitranslmed.aan1145] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/12/2017] [Accepted: 07/26/2017] [Indexed: 12/12/2022]
Abstract
Hematopoietic reconstitution after bone marrow transplantation is thought to be driven by committed multipotent progenitor cells followed by long-term engrafting hematopoietic stem cells (HSCs). We observed a population of early-engrafting cells displaying HSC-like behavior, which persisted long-term in vivo in an autologous myeloablative transplant model in nonhuman primates. To identify this population, we characterized the phenotype and function of defined nonhuman primate hematopoietic stem and progenitor cell (HSPC) subsets and compared these to human HSPCs. We demonstrated that the CD34+CD45RA-CD90+ cell phenotype is highly enriched for HSCs. This population fully supported rapid short-term recovery and robust multilineage hematopoiesis in the nonhuman primate transplant model and quantitatively predicted transplant success and time to neutrophil and platelet recovery. Application of this cell population has potential in the setting of HSC transplantation and gene therapy/editing approaches.
Collapse
Affiliation(s)
- Stefan Radtke
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen 45147, Germany
| | - Jennifer E Adair
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Morgan A Giese
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Yan-Yi Chan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Zachary K Norgaard
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Mark Enstrom
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Kevin G Haworth
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Lauren E Schefter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Hans-Peter Kiem
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA. .,Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA.,Department of Pathology, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
109
|
Zhang Y, Gao S, Xia J, Liu F. Hematopoietic Hierarchy - An Updated Roadmap. Trends Cell Biol 2018; 28:976-986. [PMID: 29935893 DOI: 10.1016/j.tcb.2018.06.001] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/29/2018] [Accepted: 06/01/2018] [Indexed: 01/08/2023]
Abstract
The classical roadmap of hematopoietic hierarchy has been proposed for nearly 20 years and has become a dogma of stem cell research for most types of adult stem cells, including hematopoietic stem cells (HSCs). However, with the development of new technologies such as omics approaches at single-cell resolution, recent studies in vitro and in vivo have suggested that heterogeneity is a common feature of HSCs and their progenies. While these findings broaden our understanding of hematopoiesis, they also challenge the well-accepted hematopoietic hierarchy roadmap. Here, we review recent advances in the hematopoiesis field and provide an updated view to incorporate these new findings as well as to reflect on the complexity of HSCs and their derivatives in development and adulthood.
Collapse
Affiliation(s)
- Yifan Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China; These authors contributed equally to this work
| | - Shuai Gao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China; These authors contributed equally to this work
| | - Jun Xia
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China; These authors contributed equally to this work
| | - Feng Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China; http://english.biomembrane.ioz.cas.cn/research/groups/liufeng.
| |
Collapse
|
110
|
ASXL1/EZH2 mutations promote clonal expansion of neoplastic HSC and impair erythropoiesis in PMF. Leukemia 2018; 33:99-109. [PMID: 29907810 DOI: 10.1038/s41375-018-0159-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/06/2018] [Accepted: 04/25/2018] [Indexed: 01/02/2023]
Abstract
Primary myelofibrosis (PMF) is a hematopoietic stem cell (HSC) disease, characterized by aberrant differentiation of all myeloid lineages and profound disruption of the bone marrow niche. PMF samples carry several mutations, but their cell origin and hierarchy in regulating the different waves of clonal and aberrant myeloproliferation from the prime HSC compartment is poorly understood. Genotyping of >2000 colonies from CD133+HSC and progenitors from PMF patients confirmed the complex genetic heterogeneity within the neoplastic population. Notably, mutations in chromatin regulators ASXL1 and/or EZH2 were identified as the first genetic lesions, preceding both JAK2-V617F and CALR mutations, and are thus drivers of clonal myelopoiesis in a PMF subset. HSC from PMF patients with double ASXL1/EZH2 mutations exhibited significantly higher engraftment in immunodeficient mice than those from patients without histone modifier mutations. EZH2 mutations correlate with aberrant erythropoiesis in PMF patients, exemplified by impaired maturation and cell cycle arrest of erythroid progenitors. These data underscore the importance of post-transcriptional modifiers of histones in neoplastic stem cells, whose clonal growth sustains aberrant myelopoiesis and expansion of pre-leukemic clones in PMF.
Collapse
|
111
|
Buenrostro JD, Corces MR, Lareau CA, Wu B, Schep AN, Aryee MJ, Majeti R, Chang HY, Greenleaf WJ. Integrated Single-Cell Analysis Maps the Continuous Regulatory Landscape of Human Hematopoietic Differentiation. Cell 2018; 173:1535-1548.e16. [PMID: 29706549 PMCID: PMC5989727 DOI: 10.1016/j.cell.2018.03.074] [Citation(s) in RCA: 425] [Impact Index Per Article: 60.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 01/03/2018] [Accepted: 03/27/2018] [Indexed: 12/26/2022]
Abstract
Human hematopoiesis involves cellular differentiation of multipotent cells into progressively more lineage-restricted states. While the chromatin accessibility landscape of this process has been explored in defined populations, single-cell regulatory variation has been hidden by ensemble averaging. We collected single-cell chromatin accessibility profiles across 10 populations of immunophenotypically defined human hematopoietic cell types and constructed a chromatin accessibility landscape of human hematopoiesis to characterize differentiation trajectories. We find variation consistent with lineage bias toward different developmental branches in multipotent cell types. We observe heterogeneity within common myeloid progenitors (CMPs) and granulocyte-macrophage progenitors (GMPs) and develop a strategy to partition GMPs along their differentiation trajectory. Furthermore, we integrated single-cell RNA sequencing (scRNA-seq) data to associate transcription factors to chromatin accessibility changes and regulatory elements to target genes through correlations of expression and regulatory element accessibility. Overall, this work provides a framework for integrative exploration of complex regulatory dynamics in a primary human tissue at single-cell resolution.
Collapse
Affiliation(s)
- Jason D Buenrostro
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Society of Fellows, Harvard University, Cambridge, MA 02138, USA.
| | - M Ryan Corces
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
| | - Caleb A Lareau
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Beijing Wu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alicia N Schep
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Martin J Aryee
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pathology, Massachusetts General Hospital & Harvard Medical School, Boston, MA 02115, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Ravindra Majeti
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford 94305, CA, USA
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - William J Greenleaf
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Applied Physics, Stanford University, Stanford, CA 94025, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
112
|
Ostendorf BN, Flenner E, Flörcken A, Westermann J. Phenotypic characterization of aberrant stem and progenitor cell populations in myelodysplastic syndromes. PLoS One 2018; 13:e0197823. [PMID: 29799854 PMCID: PMC5969762 DOI: 10.1371/journal.pone.0197823] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 05/09/2018] [Indexed: 01/04/2023] Open
Abstract
Recent reports have revealed myelodysplastic syndromes (MDS) to arise from cancer stem cells phenotypically similar to physiological hematopoietic stem cells. Myelodysplastic hematopoiesis maintains a hierarchical organization, but the proportion of several hematopoietic compartments is skewed and multiple surface markers are aberrantly expressed. These aberrant antigen expression patterns hold diagnostic and therapeutic promise. However, eradication of MDS requires targeting of early myelodysplasia propagating stem cells. This warrants an exact assessment of the differentiation stage at which aberrant expression occurs in transformed hematopoiesis. Here, we report results on the prospective and extensive dissection of the hematopoietic hierarchy in 20 patients with either low-risk MDS or MDS with excess blasts and compare it to hematopoiesis in patients with non-malignancy-associated cytopenia or B cell lymphoma without bone marrow infiltration. We found patients with MDS with excess blasts to exhibit characteristic expansions of specific immature progenitor compartments. We also identified the aberrant expression of several markers including ALDH, CLL-1, CD44, and CD47 to be specific features of hematopoiesis in MDS with excess blasts. We show that amongst these, aberrant CLL-1 expression manifested at the early uncommitted hematopoietic stem cell level, suggesting a potential role as a therapeutic target.
Collapse
Affiliation(s)
- Benjamin N. Ostendorf
- Department of Hematology, Oncology, and Tumor Immunology, Charité –Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
- Labor Berlin Charité Vivantes GmbH, Berlin, Germany
- * E-mail: (BNO); (JW)
| | - Eva Flenner
- Department of Hematology, Oncology, and Tumor Immunology, Charité –Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
- Labor Berlin Charité Vivantes GmbH, Berlin, Germany
| | - Anne Flörcken
- Department of Hematology, Oncology, and Tumor Immunology, Charité –Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
- Labor Berlin Charité Vivantes GmbH, Berlin, Germany
| | - Jörg Westermann
- Department of Hematology, Oncology, and Tumor Immunology, Charité –Universitätsmedizin Berlin, Campus Virchow Klinikum, Berlin, Germany
- Labor Berlin Charité Vivantes GmbH, Berlin, Germany
- * E-mail: (BNO); (JW)
| |
Collapse
|
113
|
Berrun A, Harris E, Stachura DL. Isthmin 1 (ism1) is required for normal hematopoiesis in developing zebrafish. PLoS One 2018; 13:e0196872. [PMID: 29758043 PMCID: PMC5951578 DOI: 10.1371/journal.pone.0196872] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 04/20/2018] [Indexed: 02/06/2023] Open
Abstract
Hematopoiesis is an essential and highly regulated biological process that begins with hematopoietic stem cells (HSCs). In healthy organisms, HSCs are responsible for generating a multitude of mature blood cells every day, yet the molecular pathways that instruct HSCs to self-renew and differentiate into post-mitotic blood cells are not fully known. To understand these molecular pathways, we investigated novel genes expressed in hematopoietic-supportive cell lines from the zebrafish (Danio rerio), a model system increasingly utilized to uncover molecular pathways important in the development of other vertebrate species. We performed RNA sequencing of the transcriptome of three stromal cell lines derived from different stages of embryonic and adult zebrafish and identified hundreds of highly expressed transcripts. For our studies, we focused on isthmin 1 (ism1) due to its shared synteny with its human gene ortholog and because it is a secreted protein. To characterize ism1, we performed loss-of-function experiments to identify if mature blood cell production was disrupted. Myeloid and erythroid lineages were visualized and scored with transgenic zebrafish expressing lineage-specific markers. ism1 knockdown led to reduced numbers of neutrophils, macrophages, and erythrocytes. Analysis of clonal methylcellulose assays from ism1 morphants also showed a reduction in total hematopoietic stem and progenitor cells (HSPCs). Overall, we demonstrate that ism1 is required for normal generation of HSPCs and their downstream progeny during zebrafish hematopoiesis. Further investigation into ism1 and its importance in hematopoiesis may elucidate evolutionarily conserved processes in blood formation that can be further investigated for potential clinical utility.
Collapse
Affiliation(s)
- Arturo Berrun
- Department of Biological Sciences, California State University Chico, Chico, CA, United States of America
| | - Elena Harris
- Department of Computer Sciences, California State University Chico, Chico, CA, United States of America
| | - David L Stachura
- Department of Biological Sciences, California State University Chico, Chico, CA, United States of America
| |
Collapse
|
114
|
Rajasekhar M, Schmitz U, Flamant S, Wong JJL, Bailey CG, Ritchie W, Holst J, Rasko JEJ. Identifying microRNA determinants of human myelopoiesis. Sci Rep 2018; 8:7264. [PMID: 29739970 PMCID: PMC5940821 DOI: 10.1038/s41598-018-24203-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 03/23/2018] [Indexed: 01/05/2023] Open
Abstract
Myelopoiesis involves differentiation of hematopoietic stem cells to cellular populations that are restricted in their self-renewal capacity, beginning with the common myeloid progenitor (CMP) and leading to mature cells including monocytes and granulocytes. This complex process is regulated by various extracellular and intracellular signals including microRNAs (miRNAs). We characterised the miRNA profile of human CD34+CD38+ myeloid progenitor cells, and mature monocytes and granulocytes isolated from cord blood using TaqMan Low Density Arrays. We identified 19 miRNAs that increased in both cell types relative to the CMP and 27 that decreased. miR-125b and miR-10a were decreased by 10-fold and 100-fold respectively in the mature cells. Using in vitro granulopoietic differentiation of human CD34+ cells we show that decreases in both miR-125b and miR-10a correlate with a loss of CD34 expression and gain of CD11b and CD15 expression. Candidate target mRNAs were identified by co-incident predictions between the miRanda algorithm and genes with increased expression during differentiation. Using luciferase assays we confirmed MCL1 and FUT4 as targets of miR-125b and the transcription factor KLF4 as a target of miR-10a. Together, our data identify miRNAs with differential expression during myeloid development and reveal some relevant miRNA-target pairs that may contribute to physiological differentiation.
Collapse
Affiliation(s)
- Megha Rajasekhar
- Gene & Stem Cell Therapy Program, Centenary Institute, University of Sydney, Camperdown, 2050, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
| | - Ulf Schmitz
- Gene & Stem Cell Therapy Program, Centenary Institute, University of Sydney, Camperdown, 2050, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
| | - Stephane Flamant
- Gene & Stem Cell Therapy Program, Centenary Institute, University of Sydney, Camperdown, 2050, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
| | - Justin J-L Wong
- Gene & Stem Cell Therapy Program, Centenary Institute, University of Sydney, Camperdown, 2050, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia.,Gene Regulation in Cancer Laboratory, Centenary Institute, University of Sydney, Camperdown, 2050, Australia
| | - Charles G Bailey
- Gene & Stem Cell Therapy Program, Centenary Institute, University of Sydney, Camperdown, 2050, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
| | - William Ritchie
- Gene & Stem Cell Therapy Program, Centenary Institute, University of Sydney, Camperdown, 2050, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
| | - Jeff Holst
- Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia.,Origins of Cancer Program, Centenary Institute, University of Sydney, Camperdown, 2050, Australia
| | - John E J Rasko
- Gene & Stem Cell Therapy Program, Centenary Institute, University of Sydney, Camperdown, 2050, Australia. .,Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia. .,Cell and Molecular Therapies, Royal Prince Alfred Hospital, Camperdown, 2050, Australia.
| |
Collapse
|
115
|
Xavier-Ferrucio J, Krause DS. Concise Review: Bipotent Megakaryocytic-Erythroid Progenitors: Concepts and Controversies. Stem Cells 2018; 36:1138-1145. [PMID: 29658164 DOI: 10.1002/stem.2834] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 03/28/2018] [Accepted: 04/03/2018] [Indexed: 12/27/2022]
Abstract
Hematopoietic stem and progenitor cells maintain blood formation throughout our lifetime by undergoing long- and short-term self-renewal, respectively. As progenitor cells progress through the hematopoiesis process, their differentiation capabilities narrow, such that the precursors become committed to only one or two lineages. This Review focuses on recent advances in the identification and characterization of bipotent megakaryocytic-erythroid progenitors (MEP), the cells that can further produce two completely different functional outputs: platelets and red blood cells. The existence of MEP has sparked controversy as studies describing the requirement for this intermediate progenitor stage prior to commitment to the erythroid and megakaryocytic lineages have been potentially contradictory. Interpretation of these studies is complicated by the variety of species, cell sources, and analytical approaches used along with inherent challenges in the continuum of hematopoiesis, where hematopoietic progenitors do not stop at discrete steps on single paths as classically drawn in hematopoietic hierarchy models. With the goal of improving our understanding of human hematopoiesis, we discuss findings in both human and murine cells. Based on these data, MEP clearly represent a transitional stage of differentiation in at least one route to the generation of both megakaryocytes and erythroid cells. Stem Cells 2018;36:1138-1145.
Collapse
Affiliation(s)
- Juliana Xavier-Ferrucio
- Yale Stem Cell Center and Department of Laboratory Medicine, Yale University, New Haven, Connecticut, USA
| | - Diane S Krause
- Yale Stem Cell Center and Department of Laboratory Medicine, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
116
|
Megakaryocyte lineage development is controlled by modulation of protein acetylation. PLoS One 2018; 13:e0196400. [PMID: 29698469 PMCID: PMC5919413 DOI: 10.1371/journal.pone.0196400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 04/12/2018] [Indexed: 12/11/2022] Open
Abstract
Treatment with lysine deacetylase inhibitors (KDACi) for haematological malignancies, is accompanied by haematological side effects including thrombocytopenia, suggesting that modulation of protein acetylation affects normal myeloid development, and specifically megakaryocyte development. In the current study, utilising ex-vivo differentiation of human CD34+ haematopoietic progenitor cells, we investigated the effects of two functionally distinct KDACi, valproic acid (VPA), and nicotinamide (NAM), on megakaryocyte differentiation, and lineage choice decisions. Treatment with VPA increased the number of megakaryocyte/erythroid progenitors (MEP), accompanied by inhibition of megakaryocyte differentiation, whereas treatment with NAM accelerated megakaryocyte development, and stimulated polyploidisation. Treatment with both KDACi resulted in no significant effects on erythrocyte differentiation, suggesting that the effects of KDACi primarily affect megakaryocyte lineage development. H3K27Ac ChIP-sequencing analysis revealed that genes involved in myeloid development, as well as megakaryocyte/erythroid (ME)-lineage differentiation are uniquely modulated by specific KDACi treatment. Taken together, our data reveal distinct effects of specific KDACi on megakaryocyte development, and ME-lineage decisions, which can be partially explained by direct effects on promoter acetylation of genes involved in myeloid differentiation.
Collapse
|
117
|
Dimitriou M, Woll PS, Mortera-Blanco T, Karimi M, Wedge DC, Doolittle H, Douagi I, Papaemmanuil E, Jacobsen SEW, Hellström-Lindberg E. Perturbed hematopoietic stem and progenitor cell hierarchy in myelodysplastic syndromes patients with monosomy 7 as the sole cytogenetic abnormality. Oncotarget 2018; 7:72685-72698. [PMID: 27683035 PMCID: PMC5341937 DOI: 10.18632/oncotarget.12234] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 09/16/2016] [Indexed: 11/26/2022] Open
Abstract
The stem and progenitor cell compartments in low- and intermediate-risk myelodysplastic syndromes (MDS) have recently been described, and shown to be highly conserved when compared to those in acute myeloid leukemia (AML). Much less is known about the characteristics of the hematopoietic hierarchy of subgroups of MDS with a high risk of transforming to AML. Immunophenotypic analysis of immature stem and progenitor cell compartments from patients with an isolated loss of the entire chromosome 7 (isolated −7), an independent high-risk genetic event in MDS, showed expansion and dominance of the malignant −7 clone in the granulocyte and macrophage progenitors (GMP), and other CD45RA+ progenitor compartments, and a significant reduction of the LIN−CD34+CD38low/−CD90+CD45RA− hematopoietic stem cell (HSC) compartment, highly reminiscent of what is typically seen in AML, and distinct from low-risk MDS. Established functional in vitro and in vivo stem cell assays showed a poor readout for −7 MDS patients irrespective of marrow blast counts. Moreover, while the −7 clone dominated at all stages of GM differentiation, the −7 clone had a competitive disadvantage in erythroid differentiation. In azacitidine-treated −7 MDS patients with a clinical response, the decreased clonal involvement in mononuclear bone marrow cells was not accompanied by a parallel reduced clonal involvement in the dominant CD45RA+ progenitor populations, suggesting a selective azacitidine-resistance of these distinct −7 progenitor compartments. Our data demonstrate, in a subgroup of high risk MDS with monosomy 7, that the perturbed stem and progenitor cell compartments resemble more that of AML than low-risk MDS.
Collapse
Affiliation(s)
- Marios Dimitriou
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Petter S Woll
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden.,Haematopoietic Stem Cell Biology Laboratory, MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Teresa Mortera-Blanco
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Mohsen Karimi
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - David C Wedge
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom.,Oxford Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Wellcome Trust Centre for Human Genetics Oxford, United Kingdom
| | - Helen Doolittle
- Haematopoietic Stem Cell Biology Laboratory, MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Iyadh Douagi
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Elli Papaemmanuil
- Cancer Genome Project, Wellcome Trust Sanger Institute, Hinxton, United Kingdom.,Computational Oncology, Epidemiology and Biostatistics Memorial Sloan Kettering Cancer Institute, New York, NY, United States of America
| | - Sten Eirik W Jacobsen
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden.,Haematopoietic Stem Cell Biology Laboratory, MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Eva Hellström-Lindberg
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
118
|
Meunier M, Dussiau C, Mauz N, Alary AS, Lefebvre C, Szymanski G, Pezet M, Blanquet F, Kosmider O, Park S. Molecular dissection of engraftment in a xenograft model of myelodysplastic syndromes. Oncotarget 2018; 9:14993-15000. [PMID: 29599920 PMCID: PMC5871091 DOI: 10.18632/oncotarget.24538] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 02/01/2018] [Indexed: 12/27/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are oligoclonal disorders of the hematopoietic stem cells (HSC). Recurrent gene mutations are involved in the MDS physiopathology along with the medullar microenvironment. To better study the heterogeneity of MDS, it is necessary to create patient derived xenograft (PDX). We have reproduced a PDX model by xenografting HSC (CD34+) and mesenchymal stromal cells (MSC) in NOD/SCID/IL2rγ-/- mice with primary samples from one RAEB2, two RAEB1 and one RARS patients harboring karyotype abnormalities and gene mutations. The average human chimerisms ranged from 59.7% to 0.0175% for the 4 patients. Secondary grafts (G2) were only performed for mice derived from the RAEB2 patient and the average human chimerism was 53.33%. G1 mice 1 and 2, and their derived G2 mice showed less than 20% of medullar blasts whereas mouse 3 and the resulting G2 mice transformed to AML. Clonal architecture was dissected in the different hematopoietic progenitors (HP) harvested from G1 and G2 mice. By direct Sanger sequencing, we found the 4 initial mutations in each HP subpopulation and those mutations had the same variant allele frequency in the CD34+ CD38- HSC from G1 and G2 mice by next generation sequencing (NGS). Targeted NGS analysis done in HSC of mouse 3 did not show any additional driver gene mutations explaining the transformation to AML. To conclude, we have generated a PDX mouse model that perfectly reproduces the MDS founder clone which is stable over time, allowing us to consider this system as a powerful tool to test therapeutic approaches.
Collapse
Affiliation(s)
- Mathieu Meunier
- University Clinic of Hematology, CHU Grenoble Alpes, Grenoble, France.,TIMC-Therex, UMR 5525 CNRS, Grenoble Alpes University, Grenoble, France
| | - Charles Dussiau
- Hématologie Biologique, Hôpital Cochin, Assistance Publique-Hôpitaux De Paris, Paris, France
| | - Natacha Mauz
- University Clinic of Hematology, CHU Grenoble Alpes, Grenoble, France.,TIMC-Therex, UMR 5525 CNRS, Grenoble Alpes University, Grenoble, France
| | - Anne Sophie Alary
- Hématologie Biologique, Hôpital Cochin, Assistance Publique-Hôpitaux De Paris, Paris, France
| | - Christine Lefebvre
- Laboratory of Hematology, Onco-Genetic and Immunology, Biology and Pathology Institute, Grenoble, France
| | - Gautier Szymanski
- Laboratory of Hematology, Onco-Genetic and Immunology, Biology and Pathology Institute, Grenoble, France
| | - Mylène Pezet
- Optical Microscopy and Flow cytometry Core, Institute for Advanced Biosciences, Inserm U 1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France
| | - Françoise Blanquet
- Plate-Forme De Haute Technologie Animale, UMR5525, Grenoble Alpes University, Grenoble, France
| | - Olivier Kosmider
- Hématologie Biologique, Hôpital Cochin, Assistance Publique-Hôpitaux De Paris, Paris, France
| | - Sophie Park
- University Clinic of Hematology, CHU Grenoble Alpes, Grenoble, France.,TIMC-Therex, UMR 5525 CNRS, Grenoble Alpes University, Grenoble, France
| |
Collapse
|
119
|
Bill M, B van Kooten Niekerk P, S Woll P, Laine Herborg L, Stidsholt Roug A, Hokland P, Nederby L. Mapping the CLEC12A expression on myeloid progenitors in normal bone marrow; implications for understanding CLEC12A-related cancer stem cell biology. J Cell Mol Med 2018; 22:2311-2318. [PMID: 29411522 PMCID: PMC5867061 DOI: 10.1111/jcmm.13519] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/08/2017] [Indexed: 12/12/2022] Open
Abstract
The C-type lectin domain family 12, member A (CLEC12A) receptor has emerged as a leukaemia-associated and cancer stem cell marker in myeloid malignancies. However, a detailed delineation of its expression in normal haematopoiesis is lacking. Here, we have characterized the expression pattern of CLEC12A on the earliest stem- and myeloid progenitor subsets in normal bone marrow. We demonstrate distinct CLEC12A expression in the classically defined myeloid progenitors, where on average 39.1% (95% CI [32.5;45.7]) of the common myeloid progenitors (CMPs) expressed CLEC12A, while for granulocyte-macrophage progenitors and megakaryocyte-erythroid progenitors (MEPs), the average percentages were 81.0% (95% CI [76.0;85.9]) and 11.9% (95% CI [9.3;14.6]), respectively. In line with the reduced CLEC12A expression on MEPs, functional assessment of purified CLEC12A+/- CMPs and MEPs in the colony-forming unit assay demonstrated CLEC12A+ subsets to favour non-erythroid colony growth. In conclusion, we provide evidence that the earliest CLEC12A+ cell in the haematopoietic tree is the classically defined CMP. Furthermore, we show that CLEC12A-expressing CMPs and MEPs are functionally different than their negative counterparts. Importantly, these data can help determine which cells will be spared during CLEC12A-targeted therapy, and we propose CLEC12A to be included in future studies of myeloid cancer stem cell biology.
Collapse
Affiliation(s)
- Marie Bill
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Petter S Woll
- Department of Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Anne Stidsholt Roug
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark.,Department of Hematology, Aalborg University Hospital, Aalborg, Denmark
| | - Peter Hokland
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark
| | - Line Nederby
- Department of Hematology, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Immunology and Biochemistry, Lillebaelt Hospital, Vejle, Denmark
| |
Collapse
|
120
|
|
121
|
Karamitros D, Stoilova B, Aboukhalil Z, Hamey F, Reinisch A, Samitsch M, Quek L, Otto G, Repapi E, Doondeea J, Usukhbayar B, Calvo J, Taylor S, Goardon N, Six E, Pflumio F, Porcher C, Majeti R, Göttgens B, Vyas P. Single-cell analysis reveals the continuum of human lympho-myeloid progenitor cells. Nat Immunol 2018; 19:85-97. [PMID: 29167569 PMCID: PMC5884424 DOI: 10.1038/s41590-017-0001-2] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 10/03/2017] [Indexed: 12/29/2022]
Abstract
The hierarchy of human hemopoietic progenitor cells that produce lymphoid and granulocytic-monocytic (myeloid) lineages is unclear. Multiple progenitor populations produce lymphoid and myeloid cells, but they remain incompletely characterized. Here we demonstrated that lympho-myeloid progenitor populations in cord blood - lymphoid-primed multi-potential progenitors (LMPPs), granulocyte-macrophage progenitors (GMPs) and multi-lymphoid progenitors (MLPs) - were functionally and transcriptionally distinct and heterogeneous at the clonal level, with progenitors of many different functional potentials present. Although most progenitors had the potential to develop into only one mature cell type ('uni-lineage potential'), bi- and rarer multi-lineage progenitors were present among LMPPs, GMPs and MLPs. Those findings, coupled with single-cell expression analyses, suggest that a continuum of progenitors execute lymphoid and myeloid differentiation, rather than only uni-lineage progenitors' being present downstream of stem cells.
Collapse
Affiliation(s)
- Dimitris Karamitros
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Bilyana Stoilova
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Zahra Aboukhalil
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Fiona Hamey
- Department of Haematology and Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Andreas Reinisch
- Division of Hematology, Stanford Institute for Stem Cell Biology and Regenerative Medicine, California, USA
| | - Marina Samitsch
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Lynn Quek
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
- Department of Hematology, OUH NHS Trust, Oxford, UK
| | - Georg Otto
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Emmanouela Repapi
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Jessica Doondeea
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Batchimeg Usukhbayar
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
- Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Julien Calvo
- UMR967 INSERM/CEA, Université Paris 7/Université Paris 11, Paris, France
| | - Stephen Taylor
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Nicolas Goardon
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Emmanuelle Six
- UMR1163, Imagine Institute, Paris Descartes -Sorbonne Paris Cité University, Paris, France
| | - Francoise Pflumio
- UMR967 INSERM/CEA, Université Paris 7/Université Paris 11, Paris, France
| | - Catherine Porcher
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Ravindra Majeti
- Division of Hematology, Stanford Institute for Stem Cell Biology and Regenerative Medicine, California, USA
| | - Berthold Göttgens
- Department of Haematology and Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Paresh Vyas
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.
- Oxford Biomedical Research Centre, University of Oxford, Oxford, UK.
- Department of Hematology, OUH NHS Trust, Oxford, UK.
| |
Collapse
|
122
|
Identification of MS4A3 as a reliable marker for early myeloid differentiation in human hematopoiesis. Biochem Biophys Res Commun 2018; 495:2338-2343. [DOI: 10.1016/j.bbrc.2017.12.117] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 12/20/2017] [Indexed: 12/12/2022]
|
123
|
Draper JE, Sroczynska P, Fadlullah MZH, Patel R, Newton G, Breitwieser W, Kouskoff V, Lacaud G. A novel prospective isolation of murine fetal liver progenitors to study in utero hematopoietic defects. PLoS Genet 2018; 14:e1007127. [PMID: 29300724 PMCID: PMC5754050 DOI: 10.1371/journal.pgen.1007127] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 11/26/2017] [Indexed: 12/29/2022] Open
Abstract
In recent years, highly detailed characterization of adult bone marrow (BM) myeloid progenitors has been achieved and, as a result, the impact of somatic defects on different hematopoietic lineage fate decisions can be precisely determined. Fetal liver (FL) hematopoietic progenitor cells (HPCs) are poorly characterized in comparison, potentially hindering the study of the impact of genetic alterations on midgestation hematopoiesis. Numerous disorders, for example infant acute leukemias, have in utero origins and their study would therefore benefit from the ability to isolate highly purified progenitor subsets. We previously demonstrated that a Runx1 distal promoter (P1)-GFP::proximal promoter (P2)-hCD4 dual-reporter mouse (Mus musculus) model can be used to identify adult BM progenitor subsets with distinct lineage preferences. In this study, we undertook the characterization of the expression of Runx1-P1-GFP and P2-hCD4 in FL. Expression of P2-hCD4 in the FL immunophenotypic Megakaryocyte-Erythroid Progenitor (MEP) and Common Myeloid Progenitor (CMP) compartments corresponded to increased granulocytic/monocytic/megakaryocytic and decreased erythroid specification. Moreover, Runx1-P2-hCD4 expression correlated with several endogenous cell surface markers' expression, including CD31 and CD45, providing a new strategy for prospective identification of highly purified fetal myeloid progenitors in transgenic mouse models. We utilized this methodology to compare the impact of the deletion of either total RUNX1 or RUNX1C alone and to determine the fetal HPCs lineages most substantially affected. This new prospective identification of FL progenitors therefore raises the prospect of identifying the underlying gene networks responsible with greater precision than previously possible.
Collapse
Affiliation(s)
- Julia E. Draper
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
| | - Patrycja Sroczynska
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
- Centre for Epigenetics, University of Copenhagen, Copenhagen, Denmark
| | - Muhammad Z. H. Fadlullah
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
| | - Rahima Patel
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
| | - Gillian Newton
- Molecular Biology Core Facility, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
| | - Wolfgang Breitwieser
- Molecular Biology Core Facility, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
| | - Valerie Kouskoff
- Division of Developmental Biology & Medicine, Michael Smith Building, The University of Manchester, Manchester, United Kingdom
| | - Georges Lacaud
- Cancer Research UK Stem Cell Biology Group, Cancer Research UK Manchester Institute, Manchester Cancer Research Centre, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
124
|
Sathe P, Pang SHM, Delconte R, Elwood N, Huntington ND. Identification of Novel Human NK Cell Progenitor Subsets. Int J Mol Sci 2017; 18:ijms18122716. [PMID: 29244714 PMCID: PMC5751317 DOI: 10.3390/ijms18122716] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/08/2017] [Accepted: 12/08/2017] [Indexed: 11/16/2022] Open
Abstract
Understanding the pathways and regulation of human haematopoiesis, in particular, lymphopoiesis, is vital to manipulation of these processes for therapeutic purposes. However, although haematopoiesis has been extensively characterised in mice, translation of these findings to human biology remains rudimentary. Here, we describe the isolation of three progenitor subsets from human foetal bone marrow that represent differential stages of commitment to the natural killer (NK) cell lineage based on IL-15 responsiveness. We identify CD7 as a marker of IL-15 responsive progenitors in human bone marrow and find that this expression is maintained throughout commitment and maturation. Within the CD7+ fraction, we focussed on the lineage potential of three subsets based on CD127 and CD117 expression and observed restricted lymphoid and biased NK cell potential amongst subsets. We further demonstrate the presence of subsets similar in both phenotype and function in umbilical cord blood and the bone marrow of humanised mice, validating these as appropriate sources of progenitors for the investigation of human haematopoiesis. Overall, we describe several stages in the process of lymphopoiesis that will form the basis of investigating the regulators of this process in humans.
Collapse
Affiliation(s)
- Priyanka Sathe
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia.
| | - Swee Heng Milon Pang
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia.
| | - Rebecca Delconte
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia.
| | - Ngaire Elwood
- Cord Blood Research, Murdoch Children's Research Institute, Melbourne, VIC 3052, Australia.
- Department of Paediatrics, University of Melbourne, Melbourne, VIC 3010, Australia.
| | - Nicholas D Huntington
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3050, Australia.
| |
Collapse
|
125
|
Schupp J, Krebs FK, Zimmer N, Trzeciak E, Schuppan D, Tuettenberg A. Targeting myeloid cells in the tumor sustaining microenvironment. Cell Immunol 2017; 343:103713. [PMID: 29129292 DOI: 10.1016/j.cellimm.2017.10.013] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 10/26/2017] [Accepted: 10/26/2017] [Indexed: 12/24/2022]
Abstract
Myeloid cells are the most abundant cells in the tumor microenvironment (TME). The tumor recruits and modulates endogenous myeloid cells to tumor-associated macrophages (TAM), dendritic cells (DC), myeloid-derived suppressor cells (MDSC) and neutrophils (TAN), to sustain an immunosuppressive environment. Pathologically overexpressed mediators produced by cancer cells like granulocyte-macrophage colony-stimulating- and vascular endothelial growth factor induce myelopoiesis in the bone marrow. Excess of myeloid cells in the blood, periphery and tumor has been associated with tumor burden. In cancer, myeloid cells are kept at an immature state of differentiation to be diverted to an immunosuppressive phenotype. Here, we review human myeloid cells in the TME and the mechanisms for sustaining the hallmarks of cancer. Simultaneously, we provide an introduction into current and novel therapeutic approaches to redirect myeloid cells from a cancer promoting to a rather inflammatory, cancer inhibiting phenotype. In addition, the role of platelets for tumor promotion is discussed.
Collapse
Affiliation(s)
- Jonathan Schupp
- Department of Dermatology, University Medical Center, Mainz, Germany
| | - Franziska K Krebs
- Department of Dermatology, University Medical Center, Mainz, Germany; German Cancer Consortium (DKTK), partner site Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Niklas Zimmer
- Department of Dermatology, University Medical Center, Mainz, Germany
| | - Emily Trzeciak
- The Edison Biotechnology Institute, Ohio University, Athens, OH, USA
| | - Detlef Schuppan
- Institute of Translational Immunology, University Medical Center, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
126
|
Abstract
I started research in high school, experimenting on immunological tolerance to transplantation antigens. This led to studies of the thymus as the site of maturation of T cells, which led to the discovery, isolation, and clinical transplantation of purified hematopoietic stem cells (HSCs). The induction of immune tolerance with HSCs has led to isolation of other tissue-specific stem cells for regenerative medicine. Our studies of circulating competing germline stem cells in colonial protochordates led us to document competing HSCs. In human acute myelogenous leukemia we showed that all preleukemic mutations occur in HSCs, and determined their order; the final mutations occur in a multipotent progenitor derived from the preleukemic HSC clone. With these, we discovered that CD47 is an upregulated gene in all human cancers and is a "don't eat me" signal; blocking it with antibodies leads to cancer cell phagocytosis. CD47 is the first known gene common to all cancers and is a target for cancer immunotherapy.
Collapse
Affiliation(s)
- Irving Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, and Ludwig Center for Cancer Stem Cell Research and Medicine at Stanford, Stanford, CA 94305
| |
Collapse
|
127
|
Liu FD, Pishesha N, Poon Z, Kaushik T, Van Vliet KJ. Material Viscoelastic Properties Modulate the Mesenchymal Stem Cell Secretome for Applications in Hematopoietic Recovery. ACS Biomater Sci Eng 2017; 3:3292-3306. [DOI: 10.1021/acsbiomaterials.7b00644] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Frances D. Liu
- Department
of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- BioSystems
and Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore−MIT Alliance for Research and Technology, CREATE, Singapore 138602
| | - Novalia Pishesha
- Department
of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Zhiyong Poon
- BioSystems
and Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore−MIT Alliance for Research and Technology, CREATE, Singapore 138602
| | - Tanwi Kaushik
- BioSystems
and Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore−MIT Alliance for Research and Technology, CREATE, Singapore 138602
| | - Krystyn J. Van Vliet
- Department
of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- BioSystems
and Micromechanics (BioSyM) Interdisciplinary Research Group, Singapore−MIT Alliance for Research and Technology, CREATE, Singapore 138602
- Department
of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
128
|
Identification of a Human Clonogenic Progenitor with Strict Monocyte Differentiation Potential: A Counterpart of Mouse cMoPs. Immunity 2017; 46:835-848.e4. [PMID: 28514689 DOI: 10.1016/j.immuni.2017.04.019] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 02/13/2017] [Accepted: 04/27/2017] [Indexed: 12/24/2022]
Abstract
Monocytes give rise to macrophages and dendritic cells (DCs) under steady-state and inflammatory conditions, thereby contributing to host defense and tissue pathology. A common monocyte progenitor (cMoP) that is strictly committed to the monocyte lineage has been recently identified in mice. Here, we identified human cMoPs as a CLEC12AhiCD64hi subpopulation of conventional granulocyte-monocyte progenitors (cGMPs) in umbilical cord blood and in bone marrow. Human cMoPs gave rise to monocyte subsets without showing any potential for differentiating into myeloid or lymphoid cells. Within the cGMP population, we also identified revised GMPs that completely lacked DC and lymphoid potential. Collectively, our findings expand and revise the current understanding of human myeloid cell differentiation pathways.
Collapse
|
129
|
Peterson VM, Zhang KX, Kumar N, Wong J, Li L, Wilson DC, Moore R, McClanahan TK, Sadekova S, Klappenbach JA. Multiplexed quantification of proteins and transcripts in single cells. Nat Biotechnol 2017; 35:936-939. [PMID: 28854175 DOI: 10.1038/nbt.3973] [Citation(s) in RCA: 574] [Impact Index Per Article: 71.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 08/24/2017] [Indexed: 11/09/2022]
Abstract
We present a tool to measure gene and protein expression levels in single cells with DNA-labeled antibodies and droplet microfluidics. Using the RNA expression and protein sequencing assay (REAP-seq), we quantified proteins with 82 barcoded antibodies and >20,000 genes in a single workflow. We used REAP-seq to assess the costimulatory effects of a CD27 agonist on human CD8+ lymphocytes and to identify and characterize an unknown cell type.
Collapse
Affiliation(s)
- Vanessa M Peterson
- Genetics &Pharmacogenomics, Department of Translational Medicine, Merck &Co., Inc., Boston, Massachusetts, USA
| | - Kelvin Xi Zhang
- Informatics IT, Merck &Co., Inc., Boston, Massachusetts, USA
| | - Namit Kumar
- Genetics &Pharmacogenomics, Department of Translational Medicine, Merck &Co., Inc., Boston, Massachusetts, USA
| | - Jerelyn Wong
- Department of Profiling and Expression, Merck &Co., Inc., Palo Alto, California, USA
| | - Lixia Li
- Genetics &Pharmacogenomics, Department of Translational Medicine, Merck &Co., Inc., Boston, Massachusetts, USA
| | - Douglas C Wilson
- Department of Profiling and Expression, Merck &Co., Inc., Palo Alto, California, USA
| | - Renee Moore
- Protein Sciences, Department of Biologics, Merck &Co., Inc., Boston, Massachusetts, USA
| | - Terrill K McClanahan
- Department of Profiling and Expression, Merck &Co., Inc., Palo Alto, California, USA
| | - Svetlana Sadekova
- Department of Profiling and Expression, Merck &Co., Inc., Palo Alto, California, USA
| | - Joel A Klappenbach
- Genetics &Pharmacogenomics, Department of Translational Medicine, Merck &Co., Inc., Boston, Massachusetts, USA
| |
Collapse
|
130
|
Abstract
The hematopoietic stem cell (HSC) is a multipotent stem cell that resides in the bone marrow and has the ability to form all of the cells of the blood and immune system. Since its first purification in 1988, additional studies have refined the phenotype and functionality of HSCs and characterized all of their downstream progeny. The hematopoietic lineage is divided into two main branches: the myeloid and lymphoid arms. The myeloid arm is characterized by the common myeloid progenitor and all of its resulting cell types. The stages of hematopoiesis have been defined in both mice and humans. During embryological development, the earliest hematopoiesis takes place in yolk sac blood islands and then migrates to the fetal liver and hematopoietic organs. Some adult myeloid populations develop directly from yolk sac progenitors without apparent bone marrow intermediates, such as tissue-resident macrophages. Hematopoiesis also changes over time, with a bias of the dominating HSCs toward myeloid development as animals age. Defects in myelopoiesis contribute to many hematologic disorders, and some of these can be overcome with therapies that target the aberrant stage of development. Furthermore, insights into myeloid development have informed us of mechanisms of programmed cell removal. The CD47/SIRPα axis, a myeloid-specific immune checkpoint, limits macrophage removal of HSCs but can be exploited by hematologic and solid malignancies. Therapeutics targeting CD47 represent a new strategy for treating cancer. Overall, an understanding of hematopoiesis and myeloid cell development has implications for regenerative medicine, hematopoietic cell transplantation, malignancy, and many other diseases.
Collapse
|
131
|
Abstract
The maintenance of monocytes, macrophages, and dendritic cells (DCs) involves manifold pathways of ontogeny and homeostasis that have been the subject of intense study in recent years. The concept of a peripheral mononuclear phagocyte system continually renewed by blood-borne monocytes has been modified to include specialized DC pathways of development that do not involve monocytes, and longevity through self-renewal of tissue macrophages. The study of development remains difficult owing to the plasticity of phenotypes and misconceptions about the fundamental structure of hematopoiesis. However, greater clarity has been achieved in distinguishing inflammatory monocyte-derived DCs from DCs arising in the steady state, and new concepts of conjoined lymphomyeloid hematopoiesis more easily accommodate the shared lymphoid and myeloid phenotypes of some DCs. Cross-species comparisons have also yielded coherent systems of nomenclature for all mammalian monocytes, macrophages, and DCs. Finally, the clear relationships between ontogeny and functional specialization offer information about the regulation of immune responses and provide new tools for the therapeutic manipulation of myeloid mononuclear cells in medicine.
Collapse
|
132
|
Mortera-Blanco T, Dimitriou M, Woll PS, Karimi M, Elvarsdottir E, Conte S, Tobiasson M, Jansson M, Douagi I, Moarii M, Saft L, Papaemmanuil E, Jacobsen SEW, Hellström-Lindberg E. SF3B1-initiating mutations in MDS-RSs target lymphomyeloid hematopoietic stem cells. Blood 2017; 130:881-890. [PMID: 28634182 PMCID: PMC5572789 DOI: 10.1182/blood-2017-03-776070] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/13/2017] [Indexed: 12/12/2022] Open
Abstract
Mutations in the RNA splicing gene SF3B1 are found in >80% of patients with myelodysplastic syndrome with ring sideroblasts (MDS-RS). We investigated the origin of SF3B1 mutations within the bone marrow hematopoietic stem and progenitor cell compartments in patients with MDS-RS. Screening for recurrently mutated genes in the mononuclear cell fraction revealed mutations in SF3B1 in 39 of 40 cases (97.5%), combined with TET2 and DNMT3A in 11 (28%) and 6 (15%) patients, respectively. All recurrent mutations identified in mononuclear cells could be tracked back to the phenotypically defined hematopoietic stem cell (HSC) compartment in all investigated patients and were also present in downstream myeloid and erythroid progenitor cells. While in agreement with previous studies, little or no evidence for clonal (SF3B1 mutation) involvement could be found in mature B cells, consistent involvement at the pro-B-cell progenitor stage was established, providing definitive evidence for SF3B1 mutations targeting lymphomyeloid HSCs and compatible with mutated SF3B1 negatively affecting lymphoid development. Assessment of stem cell function in vitro as well as in vivo established that only HSCs and not investigated progenitor populations could propagate the SF3B1 mutated clone. Upon transplantation into immune-deficient mice, SF3B1 mutated MDS-RS HSCs differentiated into characteristic ring sideroblasts, the hallmark of MDS-RS. Our findings provide evidence of a multipotent lymphomyeloid HSC origin of SF3B1 mutations in MDS-RS patients and provide a novel in vivo platform for mechanistically and therapeutically exploring SF3B1 mutated MDS-RS.
Collapse
Affiliation(s)
- Teresa Mortera-Blanco
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Marios Dimitriou
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Petter S Woll
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Haematopoietic Stem Cell Biology Laboratory, MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Mohsen Karimi
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Edda Elvarsdottir
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Simona Conte
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Magnus Tobiasson
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Monika Jansson
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Iyadh Douagi
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Matahi Moarii
- Memorial Sloan Kettering Cancer Center, New York, NY; and
| | - Leonie Saft
- Division of Hematopathology, Department of Pathology, Karolinska University Hospital, Solna, Sweden
| | | | - Sten Eirik W Jacobsen
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Haematopoietic Stem Cell Biology Laboratory, MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Eva Hellström-Lindberg
- Center for Hematology and Regenerative Medicine, Karolinska Institutet, Department of Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
133
|
Cashen AF, Rettig M, Gao F, Smith A, Abboud C, Stockerl-Goldstein K, Vij R, Uy G, Westervelt P, DiPersio J. Phase I/II Study of Intravenous Plerixafor Added to a Mobilization Regimen of Granulocyte Colony-Stimulating Factor in Lymphoma Patients Undergoing Autologous Stem Cell Collection. Biol Blood Marrow Transplant 2017; 23:1282-1289. [PMID: 28476490 DOI: 10.1016/j.bbmt.2017.04.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/24/2017] [Indexed: 11/25/2022]
Abstract
Plerixafor, given subcutaneously with granulocyte colony-stimulating factor (G-CSF), improves autologous stem cell collection in patients with lymphoma and multiple myeloma. Intravenous (i.v.) administration of plerixafor allows administration of plerixafor on the same day as pheresis and it may improve stem cell collection. The primary objectives of this phase I/II study were to determine the maximum tolerated dose of i.v. plerixafor and the efficacy of i.v. plerixafor + G-CSF to mobilize ≥ 2 × 106 CD34+ cells/kg from patients with lymphoma. In phase I, 25 patients were treated with G-CSF + i.v. plerixafor at escalating doses; in phase II, 36 patients were treated with G-CSF + plerixafor .40 mg/kg. The treatment was well tolerated. Fifty-nine of 61 patients (98%) met the collection goal and 47 of 61 patients (77%) collected ≥ 5.0 × 106 CD34+ cells/kg in a median of 2 pheresis days. Analysis of CD34+ hematopoietic stem and progenitor cells (HSPCs) revealed that G-CSF-mobilized grafts were enriched with CD34+CD45RA-CD123+/- primitive HSPCs whereas plerixafor preferentially mobilized CD34+CD45RA+CD123++ plasmacytoid dendritic cell precursors. In conclusion, i.v. plerixafor is well tolerated and effective when added to G-CSF for the mobilization of stem cells from patients with lymphoma, with mobilization kinetics and stem cell collections that compare favorably with subcutaneous dosing.
Collapse
Affiliation(s)
- Amanda F Cashen
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri.
| | - Michael Rettig
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Feng Gao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri
| | - Angela Smith
- Cellular Therapy Laboratory Barnes-Jewish Hospital, St. Louis, Missouri
| | - Camille Abboud
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | | | - Ravi Vij
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Geoffrey Uy
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Peter Westervelt
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - John DiPersio
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
134
|
Barrett TJ, Murphy AJ, Goldberg IJ, Fisher EA. Diabetes-mediated myelopoiesis and the relationship to cardiovascular risk. Ann N Y Acad Sci 2017; 1402:31-42. [PMID: 28926114 PMCID: PMC5659728 DOI: 10.1111/nyas.13462] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/03/2017] [Accepted: 08/07/2017] [Indexed: 12/20/2022]
Abstract
Diabetes is the greatest risk factor for the development of cardiovascular disease, which, in turn, is the most prevalent cause of mortality and morbidity in diabetics. These patients have elevations in inflammatory monocytes, a factor consistently reported to drive the development of atherosclerosis. In preclinical models of both type 1 and type 2 diabetes, studies have demonstrated that the increased production and activation of monocytes is driven by enhanced myelopoiesis, promoted by factors, including hyperglycemia, impaired cholesterol efflux, and inflammasome activation, that affect the proliferation of bone marrow precursor cells. This suggests that continued mechanistic investigations of the enhanced myelopoiesis and the generation of inflammatory monocytes are timely, from the dual perspectives of understanding more deeply the underlying bases of diabetes pathophysiology and identifying therapeutic targets to reduce cardiovascular risk in these patients.
Collapse
Affiliation(s)
- Tessa J. Barrett
- Department of Medicine, Division of Cardiology, New York University
School of Medicine, New York, New York
| | - Andrew J. Murphy
- Haematopoiesis and Leukocyte Biology, Baker Heart and Diabetes
Institute, Melbourne, Australia
- Department of Immunology, Monash University, Melbourne,
Australia
| | - Ira J. Goldberg
- Department of Medicine, Division of Endocrinology, Diabetes and
Metabolism, New York University School of Medicine, New York, New York
| | - Edward A. Fisher
- Department of Medicine, Division of Cardiology, New York University
School of Medicine, New York, New York
| |
Collapse
|
135
|
Yáñez A, Goodridge HS, Grimes HL. Counting the cost of lineage decisions. Nat Immunol 2017; 18:872-873. [PMID: 28722717 DOI: 10.1038/ni.3794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Alberto Yáñez
- Board of Governors Regenerative Medicine Institute and the Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Helen S Goodridge
- Board of Governors Regenerative Medicine Institute and the Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - H Leighton Grimes
- Division of Immunobiology and Center for Systems Immunology and the Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
136
|
Map3k8 controls granulocyte colony-stimulating factor production and neutrophil precursor proliferation in lipopolysaccharide-induced emergency granulopoiesis. Sci Rep 2017; 7:5010. [PMID: 28694430 PMCID: PMC5503936 DOI: 10.1038/s41598-017-04538-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 05/17/2017] [Indexed: 01/15/2023] Open
Abstract
Map3k8 has been proposed as a useful target for the treatment of inflammatory diseases. We show here that during lipopolysaccharide-induced emergency granulopoiesis, Map3k8 deficiency strongly impairs the increase in circulating mature (Ly6GhighCD11b+) and immature (Ly6GlowCD11b+) neutrophils. After chimaeric bone marrow (BM) transplantation into recipient Map3k8−/− mice, lipopolysaccharide treatment did not increase circulating Ly6GhighCD11b+ cells and strongly decreased circulating Ly6GlowCD11b+ cells. Lipopolysaccharide-treated Map3k8−/− mice showed decreased production of granulocyte colony-stimulating factor (G-CSF), a key factor in neutrophil expansion, and a Map3k8 inhibitor blocked lipopolysaccharide-mediated G-CSF expression in endothelial cell lines. Ly6GlowCD11b+ BM cells from lipopolysaccharide-treated Map3k8−/− mice displayed impaired expression of CCAAT-enhancer-binding protein β, which depends on G-CSF for expression and is crucial for cell cycle acceleration in this life-threatening condition. Accordingly, lipopolysaccharide-treated Map3k8−/− mice showed decreased Ly6GlowCD11b+ BM cell proliferation, as evidenced by a decrease in the percentage of the most immature precursors, which have the highest proliferation capacity among this cell population. Thus, Map3k8 expression by non-haematopoietic tissue is required for lipopolysaccharide-induced emergency granulopoiesis. The novel observation that inhibition of Map3k8 activity decreases neutrophilia during life-threatening systemic infection suggests a possible risk in the proposed use of Map3k8 blockade as an anti-inflammatory therapy.
Collapse
|
137
|
Nishikii H, Kurita N, Chiba S. The Road Map for Megakaryopoietic Lineage from Hematopoietic Stem/Progenitor Cells. Stem Cells Transl Med 2017; 6:1661-1665. [PMID: 28682009 PMCID: PMC5689792 DOI: 10.1002/sctm.16-0490] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 05/02/2017] [Indexed: 12/25/2022] Open
Abstract
Megakaryocytes (Mgks) are terminally differentiated blood cells specified to produce platelets, whereas hematopoietic stem cells (HSCs) are the most undifferentiated blood cells that retain multipotency to produce all kinds of blood cells. As such, these two cell types reside at the bottom and the top of the hematopoietic hierarchy, respectively. In spite of this distance, they share several important cell surface molecules as well as transcription factors. In the conventional step‐wise differentiation model, HSCs gradually lose their self‐renewal capacity and differentiate into multipotent progenitors (MPPs), which is the first branch point of myeloid and lymphoid lineage. In this model, common myeloid progenitors can differentiate into bipotent Mgk/erythroid progenitors (MEPs), and MEPs eventually differentiate into unipotent mature Mgks. However, it has been recently reported that a subpopulation within the HSC and MPP compartments demonstrates an Mgk‐biased differentiation potential. These reports imply that revisions to the HSC‐to‐Mgk differentiation pathway should be discussed. In this review, we summarize recent findings about Mgk differentiation from HSCs and discuss future directions in this research field. Stem Cells Translational Medicine2017;6:1661–1665
Collapse
Affiliation(s)
- Hidekazu Nishikii
- Department of Hematology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Naoki Kurita
- Department of Hematology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Shigeru Chiba
- Department of Hematology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
138
|
Lunger I, Fawaz M, Rieger MA. Single-cell analyses to reveal hematopoietic stem cell fate decisions. FEBS Lett 2017; 591:2195-2212. [PMID: 28600837 DOI: 10.1002/1873-3468.12712] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 05/19/2017] [Accepted: 06/02/2017] [Indexed: 12/15/2022]
Abstract
Hematopoietic stem cells (HSCs) are the best studied adult stem cells with enormous clinical value. Most of our knowledge about their biology relies on assays at the single HSC level. However, only the recent advances in developing new single cell technologies allowed the elucidation of the complex regulation of HSC fate decision control. This Review will focus on current attempts to investigate individual HSCs at molecular and functional levels. The advantages of these technologies leading to groundbreaking insights into hematopoiesis will be highlighted, and the challenges facing these technologies will be discussed. The importance of combining molecular and functional assays to enlighten regulatory networks of HSC fate decision control, ideally at high temporal resolution, becomes apparent for future studies.
Collapse
Affiliation(s)
- Ilaria Lunger
- Department of Medicine, Hematology/Oncology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Malak Fawaz
- Department of Medicine, Hematology/Oncology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Michael A Rieger
- Department of Medicine, Hematology/Oncology, Goethe University Frankfurt, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
139
|
Martiáñez Canales T, de Leeuw DC, Vermue E, Ossenkoppele GJ, Smit L. Specific Depletion of Leukemic Stem Cells: Can MicroRNAs Make the Difference? Cancers (Basel) 2017; 9:cancers9070074. [PMID: 28665351 PMCID: PMC5532610 DOI: 10.3390/cancers9070074] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/15/2017] [Accepted: 06/20/2017] [Indexed: 01/22/2023] Open
Abstract
For over 40 years the standard treatment for acute myeloid leukemia (AML) patients has been a combination of chemotherapy consisting of cytarabine and an anthracycline such as daunorubicin. This standard treatment results in complete remission (CR) in the majority of AML patients. However, despite these high CR rates, only 30–40% (<60 years) and 10–20% (>60 years) of patients survive five years after diagnosis. The main cause of this treatment failure is insufficient eradication of a subpopulation of chemotherapy resistant leukemic cells with stem cell-like properties, often referred to as “leukemic stem cells” (LSCs). LSCs co-exist in the bone marrow of the AML patient with residual healthy hematopoietic stem cells (HSCs), which are needed to reconstitute the blood after therapy. To prevent relapse, development of additional therapies targeting LSCs, while sparing HSCs, is essential. As LSCs are rare, heterogeneous and dynamic, these cells are extremely difficult to target by single gene therapies. Modulation of miRNAs and consequently the regulation of hundreds of their targets may be the key to successful elimination of resistant LSCs, either by inducing apoptosis or by sensitizing them for chemotherapy. To address the need for specific targeting of LSCs, miRNA expression patterns in highly enriched HSCs, LSCs, and leukemic progenitors, all derived from the same patients’ bone marrow, were determined and differentially expressed miRNAs between LSCs and HSCs and between LSCs and leukemic progenitors were identified. Several of these miRNAs are specifically expressed in LSCs and/or HSCs and associated with AML prognosis and treatment outcome. In this review, we will focus on the expression and function of miRNAs expressed in normal and leukemic stem cells that are residing within the AML bone marrow. Moreover, we will review their possible prospective as specific targets for anti-LSC therapy.
Collapse
Affiliation(s)
- Tania Martiáñez Canales
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | - David C de Leeuw
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | - Eline Vermue
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | - Gert J Ossenkoppele
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| | - Linda Smit
- Department of Hematology, VU University Medical Center, Cancer Center Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| |
Collapse
|
140
|
Lineage specification of human dendritic cells is marked by IRF8 expression in hematopoietic stem cells and multipotent progenitors. Nat Immunol 2017. [PMID: 28650480 PMCID: PMC5743223 DOI: 10.1038/ni.3789] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The origin and specification of human dendritic cells (DCs) have not been investigated at clonal level. Using clonal assays combined with statistical computation to quantify the yield of granulocytes, monocytes, lymphocytes and three subsets of DCs from single human CD34+ progenitor cells, we show DC lineage specification occurs in parallel with myeloid and lymphoid lineages in HSCs, starting as a lineage bias defined by specific transcriptional programs correlated with the relative IRF8/PU.1 ratios, which is transmitted to most progeny and reinforced by FLT3L-driven IRF8 upregulation over division. We propose a model in which DC lineage specification is driven by parallel and inheritable transcriptional programs in HSCs, and reinforced over cell division by recursive interaction between transcriptional programs and extrinsic signals.
Collapse
|
141
|
Jurkin J, Krump C, Köffel R, Fieber C, Schuster C, Brunner PM, Borek I, Eisenwort G, Lim C, Mages J, Lang R, Bauer W, Mechtcheriakova D, Meshcheryakova A, Elbe-Bürger A, Stingl G, Strobl H. Human skin dendritic cell fate is differentially regulated by the monocyte identity factor Kruppel-like factor 4 during steady state and inflammation. J Allergy Clin Immunol 2017; 139:1873-1884.e10. [PMID: 27742396 PMCID: PMC5538449 DOI: 10.1016/j.jaci.2016.09.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 08/25/2016] [Accepted: 09/09/2016] [Indexed: 11/01/2022]
Abstract
BACKGROUND Langerhans cell (LC) networks play key roles in immunity and tolerance at body surfaces. LCs are established prenatally and can be replenished from blood monocytes. Unlike skin-resident dermal DCs (dDCs)/interstitial-type DCs and inflammatory dendritic epidermal cells appearing in dermatitis/eczema lesions, LCs lack key monocyte-affiliated markers. Inversely, LCs express various epithelial genes critical for their long-term peripheral tissue residency. OBJECTIVE Dendritic cells (DCs) are functionally involved in inflammatory diseases; however, the mechanisms remained poorly understood. METHODS In vitro differentiation models of human DCs, gene profiling, gene transduction, and immunohistology were used to identify molecules involved in DC subset specification. RESULTS Here we identified the monocyte/macrophage lineage identity transcription factor Kruppel-like factor 4 (KLF4) to be inhibited during LC differentiation from human blood monocytes. Conversely, KLF4 is maintained or induced during dermal DC and monocyte-derived dendritic cell/inflammatory dendritic epidermal cell differentiation. We showed that in monocytic cells KLF4 has to be repressed to allow their differentiation into LCs. Moreover, respective KLF4 levels in DC subsets positively correlate with proinflammatory characteristics. We identified epithelial Notch signaling to repress KLF4 in monocytes undergoing LC commitment. Loss of KLF4 in monocytes transcriptionally derepresses Runt-related transcription factor 3 in response to TGF-β1, thereby allowing LC differentiation marked by a low cytokine expression profile. CONCLUSION Monocyte differentiation into LCs depends on activation of Notch signaling and the concomitant loss of KLF4.
Collapse
Affiliation(s)
- Jennifer Jurkin
- Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Corinna Krump
- Institute of Pathophysiology and Immunology, Medical University of Graz, Graz, Austria
| | - René Köffel
- Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Christina Fieber
- Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Christopher Schuster
- Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Patrick M Brunner
- Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Izabela Borek
- Institute of Pathophysiology and Immunology, Medical University of Graz, Graz, Austria
| | - Gregor Eisenwort
- Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Clarice Lim
- Institute of Pathophysiology and Immunology, Medical University of Graz, Graz, Austria; Institute of Immunology, Medical University of Vienna, Vienna, Austria
| | - Jörg Mages
- Institute of Clinical Microbiology, Immunology and Hygiene, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Roland Lang
- Institute of Clinical Microbiology, Immunology and Hygiene, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Wolfgang Bauer
- Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Diana Mechtcheriakova
- Departments of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Anastasia Meshcheryakova
- Departments of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Adelheid Elbe-Bürger
- Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Georg Stingl
- Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Herbert Strobl
- Institute of Pathophysiology and Immunology, Medical University of Graz, Graz, Austria; Institute of Immunology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
142
|
Schroeder MA, Rettig MP, Lopez S, Christ S, Fiala M, Eades W, Mir FA, Shao J, McFarland K, Trinkaus K, Shannon W, Deych E, Yu J, Vij R, Stockerl-Goldstein K, Cashen AF, Uy GL, Abboud CN, Westervelt P, DiPersio JF. Mobilization of allogeneic peripheral blood stem cell donors with intravenous plerixafor mobilizes a unique graft. Blood 2017; 129:2680-2692. [PMID: 28292947 PMCID: PMC5428459 DOI: 10.1182/blood-2016-09-739722] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 03/02/2017] [Indexed: 12/22/2022] Open
Abstract
A single subcutaneous (SC) injection of plerixafor results in rapid mobilization of hematopoietic progenitors, but fails to mobilize 33% of normal allogeneic sibling donors in 1 apheresis. We hypothesized that changing the route of administration of plerixafor from SC to IV may overcome the low stem cell yields and allow collection in 1 day. A phase 1 trial followed by a phase 2 efficacy trial was conducted in allogeneic sibling donors. The optimal dose of IV plerixafor was determined to be 0.32 mg/kg. The primary outcome of reducing the failure to collect ≥2 × 106 CD34+/kg recipient weight in 1 apheresis collection to ≤10% was not reached. The failure rate was 34%. Studies evaluating the stem cell phenotype and gene expression revealed a novel plasmacytoid dendritic cell precursor preferentially mobilized by plerixafor with high interferon-α producing ability. The observed cytomegalovirus (CMV) viremia rate for patients at risk was low (15%), as were the rates of acute grade 2-4 graft-versus-host disease (GVHD) (21%). Day 100 treatment related mortality was low (3%). In conclusion, plerixafor results in rapid stem cell mobilization regardless of route of administration and resulted in novel cellular composition of the graft and favorable recipient outcomes. These trials were registered at clinicaltrials.gov as #NCT00241358 and #NCT00914849.
Collapse
Affiliation(s)
- Mark A Schroeder
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Michael P Rettig
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Sandra Lopez
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Stephanie Christ
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Mark Fiala
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - William Eades
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Fazia A Mir
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, MO; and
| | - Jin Shao
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Kyle McFarland
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | | | | | | | - Jinsheng Yu
- Department of Genetics, Washington University School of Medicine, St. Louis, MO
| | - Ravi Vij
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Keith Stockerl-Goldstein
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Amanda F Cashen
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Geoffrey L Uy
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Camille N Abboud
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Peter Westervelt
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - John F DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
143
|
Amouzegar A, Mittal SK, Sahu A, Sahu SK, Chauhan SK. Mesenchymal Stem Cells Modulate Differentiation of Myeloid Progenitor Cells During Inflammation. Stem Cells 2017; 35:1532-1541. [PMID: 28295880 DOI: 10.1002/stem.2611] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 02/14/2017] [Accepted: 02/26/2017] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) possess distinct immunomodulatory properties and have tremendous potential for use in therapeutic applications in various inflammatory diseases. MSCs have been shown to regulate pathogenic functions of mature myeloid inflammatory cells, such as macrophages and neutrophils. Intriguingly, the capacity of MSCs to modulate differentiation of myeloid progenitors (MPs) to mature inflammatory cells remains unknown to date. Here, we report the novel finding that MSCs inhibit the expression of differentiation markers on MPs under inflammatory conditions. We demonstrate that the inhibitory effect of MSCs is dependent on direct cell-cell contact and that this intercellular contact is mediated through interaction of CD200 expressed by MSCs and CD200R1 expressed by MPs. Furthermore, using an injury model of sterile inflammation, we show that MSCs promote MP frequencies and suppress infiltration of inflammatory cells in the inflamed tissue. We also find that downregulation of CD200 in MSCs correlates with abrogation of their immunoregulatory function. Collectively, our study provides unequivocal evidence that MSCs inhibit differentiation of MPs in the inflammatory environment via CD200-CD200R1 interaction. Stem Cells 2017;35:1532-1541.
Collapse
Affiliation(s)
- Afsaneh Amouzegar
- Massachusetts Eye and Ear, Schepens Eye Research Institute, Boston, Massachusetts, 02114, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, 02114, USA
| | - Sharad K Mittal
- Massachusetts Eye and Ear, Schepens Eye Research Institute, Boston, Massachusetts, 02114, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, 02114, USA
| | - Anuradha Sahu
- Massachusetts Eye and Ear, Schepens Eye Research Institute, Boston, Massachusetts, 02114, USA
| | - Srikant K Sahu
- Massachusetts Eye and Ear, Schepens Eye Research Institute, Boston, Massachusetts, 02114, USA.,L. V. Prasad Eye Institute, Bhubaneswar, Odisha, 751024, India
| | - Sunil K Chauhan
- Massachusetts Eye and Ear, Schepens Eye Research Institute, Boston, Massachusetts, 02114, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, 02114, USA
| |
Collapse
|
144
|
Identification of unipotent megakaryocyte progenitors in human hematopoiesis. Blood 2017; 129:3332-3343. [PMID: 28336526 DOI: 10.1182/blood-2016-09-741611] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 03/13/2017] [Indexed: 12/22/2022] Open
Abstract
The developmental pathway for human megakaryocytes remains unclear, and the definition of pure unipotent megakaryocyte progenitor is still controversial. Using single-cell transcriptome analysis, we have identified a cluster of cells within immature hematopoietic stem- and progenitor-cell populations that specifically expresses genes related to the megakaryocyte lineage. We used CD41 as a positive marker to identify these cells within the CD34+CD38+IL-3RαdimCD45RA- common myeloid progenitor (CMP) population. These cells lacked erythroid and granulocyte-macrophage potential but exhibited robust differentiation into the megakaryocyte lineage at a high frequency, both in vivo and in vitro. The efficiency and expansion potential of these cells exceeded those of conventional bipotent megakaryocyte/erythrocyte progenitors. Accordingly, the CD41+ CMP was defined as a unipotent megakaryocyte progenitor (MegP) that is likely to represent the major pathway for human megakaryopoiesis, independent of canonical megakaryocyte-erythroid lineage bifurcation. In the bone marrow of patients with essential thrombocythemia, the MegP population was significantly expanded in the context of a high burden of Janus kinase 2 mutations. Thus, the prospectively isolatable and functionally homogeneous human MegP will be useful for the elucidation of the mechanisms underlying normal and malignant human hematopoiesis.
Collapse
|
145
|
Quantification and three-dimensional microanatomical organization of the bone marrow. Blood Adv 2017; 1:407-416. [PMID: 29296956 DOI: 10.1182/bloodadvances.2016003194] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 01/05/2017] [Indexed: 12/21/2022] Open
Abstract
Bone marrow (BM) constitutes one of the largest organs in mice and humans, continuously generating, in a highly regulated manner, red blood cells, platelets, and white blood cells that together form the majority of cells of the body. In this review, we provide a quantitative overview of BM cellular composition, we summarize emerging knowledge on its structural organization and cellular niches, and we argue for the need of multidimensional approaches such as recently developed imaging techniques to uncover the complex spatial logic that underlies BM function in health and disease.
Collapse
|
146
|
Han L, Jorgensen JL, Brooks C, Shi C, Zhang Q, Nogueras González GM, Cavazos A, Pan R, Mu H, Wang SA, Zhou J, Ai-Atrash G, Ciurea SO, Rettig M, DiPersio JF, Cortes J, Huang X, Kantarjian HM, Andreeff M, Ravandi F, Konopleva M. Antileukemia Efficacy and Mechanisms of Action of SL-101, a Novel Anti-CD123 Antibody Conjugate, in Acute Myeloid Leukemia. Clin Cancer Res 2017; 23:3385-3395. [PMID: 28096272 DOI: 10.1158/1078-0432.ccr-16-1904] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 12/13/2016] [Accepted: 12/29/2016] [Indexed: 12/11/2022]
Abstract
Purpose: The persistence of leukemia stem cells (LSC)-containing cells after induction therapy may contribute to minimal residual disease (MRD) and relapse in acute myeloid leukemia (AML). We investigated the clinical relevance of CD34+CD123+ LSC-containing cells and antileukemia potency of a novel antibody conjugate SL-101 in targeting CD123+ LSCs.Experimental Methods and Results: In a retrospective study on 86 newly diagnosed AML patients, we demonstrated that a higher proportion of CD34+CD123+ LSC-containing cells in remission was associated with persistent MRD and predicted shorter relapse-free survival in patients with poor-risk cytogenetics. Using flow cytometry, we explored the potential benefit of therapeutic targeting of CD34+CD38-CD123+ cells by SL-101, a novel antibody conjugate comprising an anti-CD123 single-chain Fv fused to Pseudomonas exotoxin A The antileukemia potency of SL-101 was determined by the expression levels of CD123 antigen in a panel of AML cell lines. Colony-forming assay established that SL-101 strongly and selectively suppressed the function of leukemic progenitors while sparing normal counterparts. The internalization, protein synthesis inhibition, and flow cytometry assays revealed the mechanisms underlying the cytotoxic activities of SL-101 involved rapid and efficient internalization of antibody, sustained inhibition of protein synthesis, induction of apoptosis, and blockade of IL3-induced p-STAT5 and p-AKT signaling pathways. In a patient-derived xenograft model using NSG mice, the repopulating capacity of LSCs pretreated with SL-101 in vitro was significantly impaired.Conclusions: Our data define the mechanisms by which SL-101 targets AML and warrant further investigation of the clinical application of SL-101 and other CD123-targeting strategies in AML. Clin Cancer Res; 23(13); 3385-95. ©2017 AACR.
Collapse
Affiliation(s)
- Lina Han
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jeffrey L Jorgensen
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Ce Shi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qi Zhang
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Antonio Cavazos
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rongqing Pan
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hong Mu
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sa A Wang
- Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jin Zhou
- Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Gheath Ai-Atrash
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stefan O Ciurea
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mike Rettig
- Bone Marrow Transplantation and Leukemia Program, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - John F DiPersio
- Bone Marrow Transplantation and Leukemia Program, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Jorge Cortes
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xuelin Huang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hagop M Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael Andreeff
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
147
|
Hasan S, Johnson NB, Mosier MJ, Shankar R, Conrad P, Szilagyi A, Gamelli RL, Muthumalaiappan K. Myelo-erythroid commitment after burn injury is under β-adrenergic control via MafB regulation. Am J Physiol Cell Physiol 2016; 312:C286-C301. [PMID: 28031160 PMCID: PMC5401945 DOI: 10.1152/ajpcell.00139.2016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 12/22/2016] [Accepted: 12/22/2016] [Indexed: 12/12/2022]
Abstract
Severely injured burn patients receive multiple blood transfusions for anemia of critical illness despite the adverse consequences. One limiting factor to consider alternate treatment strategies is the lack of a reliable test platform to study molecular mechanisms of impaired erythropoiesis. This study illustrates how conditions resulting in a high catecholamine microenvironment such as burns can instigate myelo-erythroid reprioritization influenced by β-adrenergic stimulation leading to anemia. In a mouse model of scald burn injury, we observed, along with a threefold increase in bone marrow LSK cells (linneg Sca1+cKit+), that the myeloid shift is accompanied with a significant reduction in megakaryocyte erythrocyte progenitors (MEPs). β-Blocker administration (propranolol) for 6 days after burn, not only reduced the number of LSKs and MafB+ cells in multipotent progenitors, but also influenced myelo-erythroid bifurcation by increasing the MEPs and reducing the granulocyte monocyte progenitors in the bone marrow of burn mice. Furthermore, similar results were observed in burn patients' peripheral blood mononuclear cell-derived ex vivo culture system, demonstrating that commitment stage of erythropoiesis is impaired in burn patients and intervention with propranolol (nonselective β1,2-adrenergic blocker) increases MEPs. Also, MafB+ cells that were significantly increased following standard burn care could be mitigated when propranolol was administered to burn patients, establishing the mechanistic regulation of erythroid commitment by myeloid regulatory transcription factor MafB. Overall, results demonstrate that β-adrenergic blockers following burn injury can redirect the hematopoietic commitment toward erythroid lineage by lowering MafB expression in multipotent progenitors and be of potential therapeutic value to increase erythropoietin responsiveness in burn patients.
Collapse
Affiliation(s)
- Shirin Hasan
- Department of Surgery, Loyola University Chicago, Health Sciences Division, Maywood, Illinois; and.,Burn and Shock Trauma Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, Illinois
| | - Nicholas B Johnson
- Department of Surgery, Loyola University Chicago, Health Sciences Division, Maywood, Illinois; and.,Burn and Shock Trauma Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, Illinois
| | - Michael J Mosier
- Department of Surgery, Loyola University Chicago, Health Sciences Division, Maywood, Illinois; and.,Burn and Shock Trauma Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, Illinois
| | - Ravi Shankar
- Department of Surgery, Loyola University Chicago, Health Sciences Division, Maywood, Illinois; and.,Burn and Shock Trauma Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, Illinois
| | - Peggie Conrad
- Department of Surgery, Loyola University Chicago, Health Sciences Division, Maywood, Illinois; and.,Burn and Shock Trauma Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, Illinois
| | - Andrea Szilagyi
- Burn and Shock Trauma Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, Illinois
| | - Richard L Gamelli
- Department of Surgery, Loyola University Chicago, Health Sciences Division, Maywood, Illinois; and.,Burn and Shock Trauma Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, Illinois
| | - Kuzhali Muthumalaiappan
- Department of Surgery, Loyola University Chicago, Health Sciences Division, Maywood, Illinois; and .,Burn and Shock Trauma Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, Illinois
| |
Collapse
|
148
|
Theocharides APA, Rongvaux A, Fritsch K, Flavell RA, Manz MG. Humanized hemato-lymphoid system mice. Haematologica 2016; 101:5-19. [PMID: 26721800 DOI: 10.3324/haematol.2014.115212] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Over the last decades, incrementally improved xenograft mouse models, supporting the engraftment and development of a human hemato-lymphoid system, have been developed and now represent an important research tool in the field. The most significant contributions made by means of humanized mice are the identification of normal and leukemic hematopoietic stem cells, the characterization of the human hematopoietic hierarchy, and their use as preclinical therapy models for malignant hematopoietic disorders. Successful xenotransplantation depends on three major factors: tolerance by the mouse host, correct spatial location, and appropriately cross-reactive support and interaction factors such as cytokines and major histocompatibility complex molecules. Each of these can be modified. Experimental approaches include the genetic modification of mice to faithfully express human support factors as non-cross-reactive cytokines, to create free niche space, the co-transplantation of human mesenchymal stem cells, the implantation of humanized ossicles or other stroma, and the implantation of human thymic tissue. Besides the source of hematopoietic cells, the conditioning regimen and the route of transplantation also significantly affect human hematopoietic development in vivo. We review here the achievements, most recent developments, and the remaining challenges in the generation of pre-clinically-predictive systems for human hematology and immunology, closely resembling the human situation in a xenogeneic mouse environment.
Collapse
Affiliation(s)
| | - Anthony Rongvaux
- Department of Immunobiology and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Kristin Fritsch
- Hematology, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland
| | - Richard A Flavell
- Department of Immunobiology and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Markus G Manz
- Hematology, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
149
|
Szunyogova E, Zhou H, Maxwell GK, Powis RA, Francesco M, Gillingwater TH, Parson SH. Survival Motor Neuron (SMN) protein is required for normal mouse liver development. Sci Rep 2016; 6:34635. [PMID: 27698380 PMCID: PMC5048144 DOI: 10.1038/srep34635] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 09/12/2016] [Indexed: 01/15/2023] Open
Abstract
Spinal Muscular Atrophy (SMA) is caused by mutation or deletion of the survival motor neuron 1 (SMN1) gene. Decreased levels of, cell-ubiquitous, SMN protein is associated with a range of systemic pathologies reported in severe patients. Despite high levels of SMN protein in normal liver, there is no comprehensive study of liver pathology in SMA. We describe failed liver development in response to reduced SMN levels, in a mouse model of severe SMA. The SMA liver is dark red, small and has: iron deposition; immature sinusoids congested with blood; persistent erythropoietic elements and increased immature red blood cells; increased and persistent megakaryocytes which release high levels of platelets found as clot-like accumulations in the heart. Myelopoiesis in contrast, was unaffected. Further analysis revealed significant molecular changes in SMA liver, consistent with the morphological findings. Antisense treatment from birth with PMO25, increased lifespan and ameliorated all morphological defects in liver by postnatal day 21. Defects in the liver are evident at birth, prior to motor system pathology, and impair essential liver function in SMA. Liver is a key recipient of SMA therapies, and systemically delivered antisense treatment, completely rescued liver pathology. Liver therefore, represents an important therapeutic target in SMA.
Collapse
Affiliation(s)
- Eva Szunyogova
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
- Euan MacDonald Center for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Haiyan Zhou
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College London, London, United Kingdom
| | - Gillian K. Maxwell
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
| | - Rachael A. Powis
- Euan MacDonald Center for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, United Kingdom
- Center for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Muntoni Francesco
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College London, London, United Kingdom
| | - Thomas H. Gillingwater
- Euan MacDonald Center for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, United Kingdom
- Center for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Simon H. Parson
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom
- Euan MacDonald Center for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
150
|
Corces MR, Buenrostro JD, Wu B, Greenside PG, Chan SM, Koenig JL, Snyder MP, Pritchard JK, Kundaje A, Greenleaf WJ, Majeti R, Chang HY. Lineage-specific and single-cell chromatin accessibility charts human hematopoiesis and leukemia evolution. Nat Genet 2016; 48:1193-203. [PMID: 27526324 PMCID: PMC5042844 DOI: 10.1038/ng.3646] [Citation(s) in RCA: 792] [Impact Index Per Article: 88.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 07/18/2016] [Indexed: 02/07/2023]
Abstract
We define the chromatin accessibility and transcriptional landscapes in 13 human primary blood cell types that span the hematopoietic hierarchy. Exploiting the finding that the enhancer landscape better reflects cell identity than mRNA levels, we enable 'enhancer cytometry' for enumeration of pure cell types from complex populations. We identify regulators governing hematopoietic differentiation and further show the lineage ontogeny of genetic elements linked to diverse human diseases. In acute myeloid leukemia (AML), chromatin accessibility uncovers unique regulatory evolution in cancer cells with a progressively increasing mutation burden. Single AML cells exhibit distinctive mixed regulome profiles corresponding to disparate developmental stages. A method to account for this regulatory heterogeneity identified cancer-specific deviations and implicated HOX factors as key regulators of preleukemic hematopoietic stem cell characteristics. Thus, regulome dynamics can provide diverse insights into hematopoietic development and disease.
Collapse
Affiliation(s)
- M Ryan Corces
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, California, USA
| | - Jason D Buenrostro
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, California, USA
- Department of Genetics, Stanford University, Stanford, California, USA
- Broad Institute of MIT and Harvard, Harvard University, Cambridge, Massachusetts, USA
| | - Beijing Wu
- Department of Genetics, Stanford University, Stanford, California, USA
| | - Peyton G Greenside
- Department of Genetics, Stanford University, Stanford, California, USA
- Program in Biomedical Informatics, Stanford University School of Medicine, Stanford, California, USA
| | - Steven M Chan
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Julie L Koenig
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Michael P Snyder
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, California, USA
- Department of Genetics, Stanford University, Stanford, California, USA
| | - Jonathan K Pritchard
- Department of Genetics, Stanford University, Stanford, California, USA
- Department of Biology, Stanford University, Stanford, California, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
| | - Anshul Kundaje
- Department of Genetics, Stanford University, Stanford, California, USA
- Department of Computer Science, Stanford University, Stanford, California, USA
| | - William J Greenleaf
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, California, USA
- Department of Genetics, Stanford University, Stanford, California, USA
| | - Ravindra Majeti
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|