1
|
Deng LH, Li MZ, Huang XJ, Zhao XY. Single-cell lineage tracing techniques in hematology: unraveling the cellular narrative. J Transl Med 2025; 23:270. [PMID: 40038725 DOI: 10.1186/s12967-025-06318-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 02/23/2025] [Indexed: 03/06/2025] Open
Abstract
Lineage tracing is a valuable technique that has greatly facilitated the exploration of cell origins and behavior. With the continuous development of single-cell sequencing technology, lineage tracing technology based on the single-cell level has become an important method to study biological development. Single-cell Lineage tracing technology plays an important role in the hematological system. It can help to answer many important questions, such as the heterogeneity of hematopoietic stem cell function and structure, and the heterogeneity of malignant tumor cells in the hematological system. Many studies have been conducted to explore the field of hematology by applying this technology. This review focuses on the superiority of the emerging single-cell lineage tracing technologies of Integration barcodes, CRISPR barcoding, and base editors, and summarizes their applications in the hematology system. These studies have suggested the vast potential in unraveling complex cellular behaviors and lineage dynamics in both normal and pathological contexts.
Collapse
Affiliation(s)
- Lu-Han Deng
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No. 11 Xizhimen South Street, Beijing, 100044, China
| | - Mu-Zi Li
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No. 11 Xizhimen South Street, Beijing, 100044, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No. 11 Xizhimen South Street, Beijing, 100044, China
| | - Xiang-Yu Zhao
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No. 11 Xizhimen South Street, Beijing, 100044, China.
| |
Collapse
|
2
|
Shaban D, Najm N, Droin L, Nijnik A. Hematopoietic Stem Cell Fates and the Cellular Hierarchy of Mammalian Hematopoiesis: from Transplantation Models to New Insights from in Situ Analyses. Stem Cell Rev Rep 2025; 21:28-44. [PMID: 39222178 DOI: 10.1007/s12015-024-10782-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2024] [Indexed: 09/04/2024]
Abstract
Hematopoiesis is the process that generates the cells of the blood and immune system from hematopoietic stem and progenitor cells (HSPCs) and represents the system with the most rapid cell turnover in a mammalian organism. HSPC differentiation trajectories, their underlying molecular mechanisms, and their dysfunctions in hematologic disorders are the focal research questions of experimental hematology. While HSPC transplantations in murine models are the traditional tool in this research field, recent advances in genome editing and next generation sequencing resulted in the development of many fundamentally new approaches for the analyses of mammalian hematopoiesis in situ and at single cell resolution. The current review will cover many recent developments in this field in murine models, from the bulk lineage tracing studies of HSPC differentiation to the barcoding of individual HSPCs with Cre-recombinase, Sleeping Beauty transposase, or CRISPR/Cas9 tools, to map hematopoietic cell fates, together with their transcriptional and epigenetic states. We also address studies of the clonal dynamics of human hematopoiesis, from the tracing of HSPC clonal behaviours based on viral integration sites in gene therapy patients to the recent analyses of unperturbed human hematopoiesis based on naturally accrued mutations in either nuclear or mitochondrial genomes. Such studies are revolutionizing our understanding of HSPC biology and hematopoiesis both under homeostatic conditions and in the response to various forms of physiological stress, reveal the mechanisms responsible for the decline of hematopoietic function with age, and in the future may advance the understanding and management of the diverse disorders of hematopoiesis.
Collapse
Affiliation(s)
- Dania Shaban
- Department of Physiology, McGill University, 368 Bellini Life Sciences Complex, 3649 Promenade Sir William Osler, Montreal, QC, H3G 0B1, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, QC, Canada
| | - Nay Najm
- Department of Physiology, McGill University, 368 Bellini Life Sciences Complex, 3649 Promenade Sir William Osler, Montreal, QC, H3G 0B1, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, QC, Canada
| | - Lucie Droin
- Department of Physiology, McGill University, 368 Bellini Life Sciences Complex, 3649 Promenade Sir William Osler, Montreal, QC, H3G 0B1, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, QC, Canada
| | - Anastasia Nijnik
- Department of Physiology, McGill University, 368 Bellini Life Sciences Complex, 3649 Promenade Sir William Osler, Montreal, QC, H3G 0B1, Canada.
- McGill University Research Centre on Complex Traits, McGill University, Montreal, QC, Canada.
| |
Collapse
|
3
|
Merle C, Fre S. Recording Lineage History with Cellular Barcodes in the Mammary Epithelium and in Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:77-94. [PMID: 39821021 DOI: 10.1007/978-3-031-70875-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Lineage tracing methods have extensively advanced our understanding of physiological cell behaviour in vivo and in situ and have vastly contributed to decipher the phylogeny and cellular hierarchies during normal and tumour development. In recent years, increasingly complex systems have been developed to track thousands of cells within a given tissue or even entire organisms. Cellular barcoding comprises all techniques designed to genetically label single cells with unique DNA sequences or with a combination of fluorescent proteins, in order to trace their history and lineage production in space and time. We distinguish these two types of cellular barcoding as genetic or optical barcodes. Furthermore, transcribed cellular barcodes can integrate the lineage information with single-cell profiling of each barcoded cell. This enables the potential identification of specific markers or signalling pathways defining distinct stem cell states during development, but also signals promoting tumour growth and metastasis or conferring therapy resistance.In this chapter, we describe recent advances in cellular barcoding technologies and outline experimental and computational challenges. We discuss the biological questions that can be addressed using single-cell dynamic lineage tracing, with a focus on the study of cellular hierarchies in the mammary epithelium and in breast cancer.
Collapse
Affiliation(s)
- Candice Merle
- Laboratory of Genetics and Developmental Biology, Institut Curie, INSERM U934, CNRS UMR3215, Paris, France
| | - Silvia Fre
- Laboratory of Genetics and Developmental Biology, Institut Curie, INSERM U934, CNRS UMR3215, Paris, France.
| |
Collapse
|
4
|
Zhang X, Huang Y, Yang Y, Wang QE, Li L. Advancements in prospective single-cell lineage barcoding and their applications in research. Genome Res 2024; 34:2147-2162. [PMID: 39572229 PMCID: PMC11694748 DOI: 10.1101/gr.278944.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 10/03/2024] [Indexed: 12/25/2024]
Abstract
Single-cell lineage tracing (scLT) has emerged as a powerful tool, providing unparalleled resolution to investigate cellular dynamics, fate determination, and the underlying molecular mechanisms. This review thoroughly examines the latest prospective lineage DNA barcode tracing technologies. It further highlights pivotal studies that leverage single-cell lentiviral integration barcoding technology to unravel the dynamic nature of cell lineages in both developmental biology and cancer research. Additionally, the review navigates through critical considerations for successful experimental design in lineage tracing and addresses challenges inherent in this field, including technical limitations, complexities in data analysis, and the imperative for standardization. It also outlines current gaps in knowledge and suggests future research directions, contributing to the ongoing advancement of scLT studies.
Collapse
Affiliation(s)
- Xiaoli Zhang
- College of Nursing, University of South Florida, Tampa, Florida 33620, USA;
| | - Yirui Huang
- College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, USA
| | - Yajing Yang
- Department of Radiation Oncology, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - Qi-En Wang
- Department of Radiation Oncology, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, USA
| | - Lang Li
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, Ohio 43210, USA
| |
Collapse
|
5
|
Quesenberry P, Dooner M, Pereira M, Oulhen N, Wen S. The Essence of Quiescence. Stem Cells Dev 2024; 33:149-152. [PMID: 38445379 PMCID: PMC11036883 DOI: 10.1089/scd.2024.0032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 02/28/2024] [Indexed: 03/07/2024] Open
Abstract
Historically hematopoietic stem cells are believed to be predominantly dormant but could be induced into active cell cycle under specific conditions. This review, coupled with years of research from our laboratory, challenges this belief by demonstrating a significant portion of hematopoietic stem cells are actively cycling rather than quiescent. This addresses a major heuristic error in the understanding of hematopoietic stem cells that has shaped this field for decades. By evaluating the cycle status of engraftable hematopoietic stem cells in whole unseparated bone marrow, we demonstrated that a significant portion of these cells are actively cycling, and further confirmed by tritiated thymidine suicide and bromodeoxyuridine labeling assays. Moreover, by analyzing both whole unseparated bone marrow and purified lineage-negative hematopoietic stem cells in murine models, our findings indicate that lineage-positive cells, usually discarded during purification, actually contain actively cycling stem cells. Taken together, our findings highlight that hematopoietic stem cells are characterized as actively cycling and expressing differentiation epitopes. This corrects a basic mistake in stem cell biology. Furthermore, these findings provide valuable insights for a better understanding of the actively cycling hematopoietic stem cells in the field of stem cell biology.
Collapse
Affiliation(s)
- Peter Quesenberry
- Department of Medicine, Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Mark Dooner
- Department of Medicine, Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Mandy Pereira
- Department of Medicine, Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Nathalie Oulhen
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, USA
| | - Sicheng Wen
- Department of Medicine, Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, Rhode Island, USA
| |
Collapse
|
6
|
Cui Z, Wei H, Goding C, Cui R. Stem cell heterogeneity, plasticity, and regulation. Life Sci 2023; 334:122240. [PMID: 37925141 DOI: 10.1016/j.lfs.2023.122240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 11/06/2023]
Abstract
As a population of homogeneous cells with both self-renewal and differentiation potential, stem cell pools are highly compartmentalized and contain distinct subsets that exhibit stable but limited heterogeneity during homeostasis. However, their striking plasticity is showcased under natural or artificial stress, such as injury, transplantation, cancer, and aging, leading to changes in their phenotype, constitution, metabolism, and function. The complex and diverse network of cell-extrinsic niches and signaling pathways, together with cell-intrinsic genetic and epigenetic regulators, tightly regulate both the heterogeneity during homeostasis and the plasticity under perturbation. Manipulating these factors offers better control of stem cell behavior and a potential revolution in the current state of regenerative medicine. However, disruptions of normal regulation by genetic mutation or excessive plasticity acquisition may contribute to the formation of tumors. By harnessing innovative techniques that enhance our understanding of stem cell heterogeneity and employing novel approaches to maximize the utilization of stem cell plasticity, stem cell therapy holds immense promise for revolutionizing the future of medicine.
Collapse
Affiliation(s)
- Ziyang Cui
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing 100034, China.
| | - Hope Wei
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, United States of America
| | - Colin Goding
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Headington, Oxford OX37DQ, UK
| | - Rutao Cui
- Skin Disease Research Institute, The 2nd Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
7
|
Urbanus J, Cosgrove J, Beltman JB, Elhanati Y, Moral RA, Conrad C, van Heijst JW, Tubeuf E, Velds A, Kok L, Merle C, Magnusson JP, Guyonnet L, Frisén J, Fre S, Walczak AM, Mora T, Jacobs H, Schumacher TN, Perié L. DRAG in situ barcoding reveals an increased number of HSPCs contributing to myelopoiesis with age. Nat Commun 2023; 14:2184. [PMID: 37069150 PMCID: PMC10110593 DOI: 10.1038/s41467-023-37167-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 03/03/2023] [Indexed: 04/19/2023] Open
Abstract
Ageing is associated with changes in the cellular composition of the immune system. During ageing, hematopoietic stem and progenitor cells (HSPCs) that produce immune cells are thought to decline in their regenerative capacity. However, HSPC function has been mostly assessed using transplantation assays, and it remains unclear how HSPCs age in the native bone marrow niche. To address this issue, we present an in situ single cell lineage tracing technology to quantify the clonal composition and cell production of single cells in their native niche. Our results demonstrate that a pool of HSPCs with unequal output maintains myelopoiesis through overlapping waves of cell production throughout adult life. During ageing, the increased frequency of myeloid cells is explained by greater numbers of HSPCs contributing to myelopoiesis rather than the increased myeloid output of individual HSPCs. Strikingly, the myeloid output of HSPCs remains constant over time despite accumulating significant transcriptomic changes throughout adulthood. Together, these results show that, unlike emergency myelopoiesis post-transplantation, aged HSPCs in their native microenvironment do not functionally decline in their regenerative capacity.
Collapse
Affiliation(s)
- Jos Urbanus
- Division of Molecular Oncology & Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jason Cosgrove
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico Chimie Curie, 75005, Paris, France
| | - Joost B Beltman
- Division of Drug Discovery & Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | | | - Rafael A Moral
- Department of Mathematics and Statistics, Maynooth University, Maynooth, Ireland
| | - Cecile Conrad
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico Chimie Curie, 75005, Paris, France
| | - Jeroen W van Heijst
- Division of Molecular Oncology & Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Emilie Tubeuf
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico Chimie Curie, 75005, Paris, France
| | - Arno Velds
- Division of Molecular Oncology & Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Lianne Kok
- Division of Molecular Oncology & Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Candice Merle
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215, Paris, France
| | - Jens P Magnusson
- Department of Bioengineering, Stanford University, Stanford, USA
| | - Léa Guyonnet
- Cytometry Platform, Institut Curie, 75005, Paris, France
| | - Jonas Frisén
- Department of Cell and Molecular Biology, Karolinska Institute, Solna, Sweden
| | - Silvia Fre
- Institut Curie, Laboratory of Genetics and Developmental Biology, PSL Research University, INSERM U934, CNRS UMR3215, Paris, France
| | - Aleksandra M Walczak
- Laboratoire de Physique de l'École Normale Supérieure (PSL University), CNRS, Sorbonne Université, and Université de Paris, Paris, France
| | - Thierry Mora
- Laboratoire de Physique de l'École Normale Supérieure (PSL University), CNRS, Sorbonne Université, and Université de Paris, Paris, France
| | - Heinz Jacobs
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ton N Schumacher
- Division of Molecular Oncology & Immunology, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Leïla Perié
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico Chimie Curie, 75005, Paris, France.
| |
Collapse
|
8
|
Purton LE. Adult murine hematopoietic stem cells and progenitors: an update on their identities, functions, and assays. Exp Hematol 2022; 116:1-14. [PMID: 36283572 DOI: 10.1016/j.exphem.2022.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/16/2022] [Accepted: 10/20/2022] [Indexed: 12/29/2022]
Abstract
The founder of all blood cells are hematopoietic stem cells (HSCs), which are rare stem cells that undergo key cell fate decisions to self-renew to generate more HSCs or to differentiate progressively into a hierarchy of different immature hematopoietic cell types to ultimately produce mature blood cells. These decisions are influenced both intrinsically and extrinsically, the latter by microenvironment cells in the bone marrow (BM). In recent decades, notable progress in our ability to identify, isolate, and study key properties of adult murine HSCs and multipotent progenitor (MPP) cells has challenged our prior understanding of the hierarchy of these primitive hematopoietic cells. These studies have revealed the existence of at least two distinct HSC types in adults: one that generates all hematopoietic cell lineages with almost equal potency and one that is platelet/myeloid-biased and increases with aging. These studies have also revealed distinct MPP cell types that have different functional potential. This review provides an update to these murine HSCs and MPP cells, their key functional properties, and the assays that have been used to assess their potential.
Collapse
Affiliation(s)
- Louise E Purton
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
9
|
Quesenberry PJ, Wen S, Goldberg LR, Dooner MS. The universal stem cell. Leukemia 2022; 36:2784-2792. [PMID: 36307485 PMCID: PMC9712109 DOI: 10.1038/s41375-022-01715-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/26/2022] [Accepted: 09/22/2022] [Indexed: 11/08/2022]
Abstract
Current dogma is that there exists a hematopoietic pluripotent stem cell, resident in the marrow, which is quiescent, but with tremendous proliferative and differentiative potential. Furthermore, the hematopoietic system is essentially hierarchical with progressive differentiation from the pluripotent stem cells to different classes of hematopoietic cells. However, results summarized here indicate that the marrow pluripotent hematopoietic stem cell is actively cycling and thus continually changing phenotype. As it progresses through cell cycle differentiation potential changes as illustrated by sequential changes in surface expression of B220 and GR-1 epitopes. Further data indicated that the potential of purified hematopoietic stem cells extends to multiple other non-hematopoietic cells. It appears that marrow stem cells will give rise to epithelial pulmonary cells at certain points in cell cycle. Thus, it appears that the marrow "hematopoietic" stem cell is also a stem cell for other non-hematopoietic tissues. These observations give rise to the concept of a universal stem cell. The marrow stem cell is not limited to hematopoiesis and its differentiation potential continually changes as it transits cell cycle. Thus, there is a universal stem cell in the marrow which alters its differentiation potential as it progresses through cell cycle. This potential is expressed when it resides in tissues compatible with its differentiation potential, at a particular point in cell cycle transit, or when it interacts with vesicles from that tissue.
Collapse
Affiliation(s)
- Peter J Quesenberry
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, RI, 02903, USA.
| | - Sicheng Wen
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, RI, 02903, USA
| | - Laura R Goldberg
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, RI, 02903, USA
- Division of Hematology, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Mark S Dooner
- Division of Hematology/Oncology, Brown University, Rhode Island Hospital, Providence, RI, 02903, USA
| |
Collapse
|
10
|
Nakamuta A, Yoshido K, Naoki H. Stem cell homeostasis regulated by hierarchy and neutral competition. Commun Biol 2022; 5:1268. [PMID: 36400843 PMCID: PMC9674595 DOI: 10.1038/s42003-022-04218-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 11/03/2022] [Indexed: 11/19/2022] Open
Abstract
Tissue stem cells maintain themselves through self-renewal while constantly supplying differentiating cells. Two distinct models have been proposed as mechanisms of stem cell homeostasis. According to the classical model, there is hierarchy among stem cells, and master stem cells produce stem cells by asymmetric division; whereas, according to the recent model, stem cells are equipotent and neutrally compete. However, the mechanism remains controversial in several tissues and species. Here, we developed a mathematical model linking the two models, named the hierarchical neutral competition (hNC) model. Our theoretical analysis showed that the combination of the hierarchy and neutral competition exhibited bursts in clonal expansion, which was consistent with experimental data of rhesus macaque hematopoiesis. Furthermore, the scaling law in clone size distribution, considered a unique characteristic of the recent model, was satisfied even in the hNC model. Based on the findings above, we proposed the criterion for distinguishing the three models based on experiments.
Collapse
Affiliation(s)
- Asahi Nakamuta
- grid.258799.80000 0004 0372 2033Laboratory of Theoretical Biology, Graduate School of Biostudies, Kyoto University, Yoshidakonoecho, Sakyo, Kyoto, 606-8315 Japan ,grid.258799.80000 0004 0372 2033Faculty of Science, Kyoto University, Yoshidakonoecho, Sakyo, Kyoto, 606-8315 Japan
| | - Kana Yoshido
- grid.258799.80000 0004 0372 2033Laboratory of Theoretical Biology, Graduate School of Biostudies, Kyoto University, Yoshidakonoecho, Sakyo, Kyoto, 606-8315 Japan
| | - Honda Naoki
- grid.258799.80000 0004 0372 2033Laboratory of Theoretical Biology, Graduate School of Biostudies, Kyoto University, Yoshidakonoecho, Sakyo, Kyoto, 606-8315 Japan ,grid.257022.00000 0000 8711 3200Laboratory of Data-driven Biology, Graduate School of Integrated Sciences for Life, Hiroshima University, Kagamiyama, Higashi-hiroshima, Hiroshima, 739-8526 Japan ,grid.257022.00000 0000 8711 3200Kansei-Brain Informatics Group, Center for Brain, Mind and Kansei Sciences Research (BMK Center), Hiroshima University, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan ,grid.250358.90000 0000 9137 6732Theoretical Biology Research Group, Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, Aichi 444-8787 Japan
| |
Collapse
|
11
|
Serrano A, Berthelet J, Naik SH, Merino D. Mastering the use of cellular barcoding to explore cancer heterogeneity. Nat Rev Cancer 2022; 22:609-624. [PMID: 35982229 DOI: 10.1038/s41568-022-00500-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/13/2022] [Indexed: 11/09/2022]
Abstract
Tumours are often composed of a multitude of malignant clones that are genomically unique, and only a few of them may have the ability to escape cancer therapy and grow as symptomatic lesions. As a result, tumours with a large degree of genomic diversity have a higher chance of leading to patient death. However, clonal fate can be driven by non-genomic features. In this context, new technologies are emerging not only to track the spatiotemporal fate of individual cells and their progeny but also to study their molecular features using various omics analysis. In particular, the recent development of cellular barcoding facilitates the labelling of tens to millions of cancer clones and enables the identification of the complex mechanisms associated with clonal fate in different microenvironments and in response to therapy. In this Review, we highlight the recent discoveries made using lentiviral-based cellular barcoding techniques, namely genetic and optical barcoding. We also emphasize the strengths and limitations of each of these technologies and discuss some of the key concepts that must be taken into consideration when one is designing barcoding experiments. Finally, we suggest new directions to further improve the use of these technologies in cancer research.
Collapse
Affiliation(s)
- Antonin Serrano
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Parkville, Victoria, Australia
| | - Jean Berthelet
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia
| | - Shalin H Naik
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Parkville, Victoria, Australia
| | - Delphine Merino
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia.
- School of Cancer Medicine, La Trobe University, Bundoora, Victoria, Australia.
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- Department of Medical Biology, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
12
|
Hadj Abed L, Tak T, Cosgrove J, Perié L. CellDestiny: A RShiny application for the visualization and analysis of single-cell lineage tracing data. Front Med (Lausanne) 2022; 9:919345. [PMID: 36275810 PMCID: PMC9581332 DOI: 10.3389/fmed.2022.919345] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 09/05/2022] [Indexed: 11/25/2022] Open
Abstract
Single-cell lineage tracing permits the labeling of individual cells with a heritable marker to follow the fate of each cell's progeny. Over the last twenty years, several single-cell lineage tracing methods have emerged, enabling major discoveries in developmental biology, oncology and gene therapies. Analytical tools are needed to draw meaningful conclusions from lineage tracing measurements, which are characterized by high variability, sparsity and technical noise. However, the single cell lineage tracing field lacks versatile and easy-to-use tools for standardized and reproducible analyses, in particular tools accessible to biologists. Here we present CellDestiny, a RShiny app and associated web application developed for experimentalists without coding skills to perform visualization and analysis of single cell lineage-tracing datasets through a graphical user interface. We demonstrate the functionality of CellDestiny through the analysis of (i) lentiviral barcoding datasets of murine hematopoietic progenitors; (ii) published integration site data from Wiskott-Aldrich Symdrome patients undergoing gene-therapy treatment; and (iii) simultaneous barcoding and transcriptomic analysis of murine hematopoietic progenitor differentiation in vitro. In summary, CellDestiny is an easy-to-use and versatile toolkit that enables biologists to visualize and analyze single-cell lineage tracing data.
Collapse
Affiliation(s)
- Louisa Hadj Abed
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico Chimie Curie, Paris, France
- Centre de Bio-Informatique, MINES ParisTech, Institut Curie, PSL University, Paris, France
| | - Tamar Tak
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico Chimie Curie, Paris, France
| | - Jason Cosgrove
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico Chimie Curie, Paris, France
| | - Leïla Perié
- Institut Curie, Université PSL, Sorbonne Université, CNRS UMR168, Laboratoire Physico Chimie Curie, Paris, France
| |
Collapse
|
13
|
Konturek-Ciesla A, Bryder D. Stem Cells, Hematopoiesis and Lineage Tracing: Transplantation-Centric Views and Beyond. Front Cell Dev Biol 2022; 10:903528. [PMID: 35573680 PMCID: PMC9091331 DOI: 10.3389/fcell.2022.903528] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 04/12/2022] [Indexed: 12/26/2022] Open
Abstract
An appropriate production of mature blood cells, or hematopoiesis, is essential for organismal health and homeostasis. In this developmental cascade, hematopoietic stem cells (HSCs) differentiate into intermediate progenitor types, that subsequently give rise to the many distinct blood cell lineages. Here, we describe tools and methods that permit for temporal and native clonal-level HSC lineage tracing in the mouse, and that can now be combined with emerging single-cell molecular analyses. We integrate new insights derived from such experimental paradigms with past knowledge, which has predominantly been derived from transplantation-based approaches. Finally, we outline current knowledge and novel strategies derived from studies aimed to trace human HSC-derived hematopoiesis.
Collapse
|
14
|
Yamashita M, Iwama A. Aging and Clonal Behavior of Hematopoietic Stem Cells. Int J Mol Sci 2022; 23:1948. [PMID: 35216063 PMCID: PMC8878540 DOI: 10.3390/ijms23041948] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 12/19/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are the only cell population that possesses both a self-renewing capacity and multipotency, and can give rise to all lineages of blood cells throughout an organism's life. However, the self-renewal capacity of HSCs is not infinite, and cumulative evidence suggests that HSCs alter their function and become less active during organismal aging, leading ultimately to the disruption of hematopoietic homeostasis, such as anemia, perturbed immunity and increased propensity to hematological malignancies. Thus, understanding how HSCs alter their function during aging is a matter of critical importance to prevent or overcome these age-related changes in the blood system. Recent advances in clonal analysis have revealed the functional heterogeneity of murine HSC pools that is established upon development and skewed toward the clonal expansion of functionally poised HSCs during aging. In humans, next-generation sequencing has revealed age-related clonal hematopoiesis that originates from HSC subsets with acquired somatic mutations, and has highlighted it as a significant risk factor for hematological malignancies and cardiovascular diseases. In this review, we summarize the current fate-mapping strategies that are used to track and visualize HSC clonal behavior during development or after stress. We then review the age-related changes in HSCs that can be inherited by daughter cells and act as a cellular memory to form functionally distinct clones. Altogether, we link aging of the hematopoietic system to HSC clonal evolution and discuss how HSC clones with myeloid skewing and low regenerative potential can be expanded during aging.
Collapse
Affiliation(s)
- Masayuki Yamashita
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai Minato-ku, Tokyo 108-8639, Japan;
| | | |
Collapse
|
15
|
Lafage-Pochitaloff M, Gerby B, Baccini V, Largeaud L, Fregona V, Prade N, Juvin PY, Jamrog L, Bories P, Hébrard S, Lagarde S, Mansat-De Mas V, Dovey OM, Yusa K, Vassiliou GS, Jansen JH, Tekath T, Rombaut D, Ameye G, Barin C, Bidet A, Boudjarane J, Collonge-Rame MA, Gervais C, Ittel A, Lefebvre C, Luquet I, Michaux L, Nadal N, Poirel HA, Radford-Weiss I, Ribourtout B, Richebourg S, Struski S, Terré C, Tigaud I, Penther D, Eclache V, Fontenay M, Broccardo C, Delabesse, E. The CADM1 tumor suppressor gene is a major candidate gene in MDS with deletion of the long arm of chromosome 11. Blood Adv 2022; 6:386-398. [PMID: 34638130 PMCID: PMC8791575 DOI: 10.1182/bloodadvances.2021005311] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/10/2021] [Indexed: 11/24/2022] Open
Abstract
Myelodysplastic syndromes (MDS) represent a heterogeneous group of clonal hematopoietic stem cell disorders characterized by ineffective hematopoiesis leading to peripheral cytopenias and in a substantial proportion of cases to acute myeloid leukemia. The deletion of the long arm of chromosome 11, del(11q), is a rare but recurrent clonal event in MDS. Here, we detail the largest series of 113 cases of MDS and myelodysplastic syndromes/myeloproliferative neoplasms (MDS/MPN) harboring a del(11q) analyzed at clinical, cytological, cytogenetic, and molecular levels. Female predominance, a survival prognosis similar to other MDS, a low monocyte count, and dysmegakaryopoiesis were the specific clinical and cytological features of del(11q) MDS. In most cases, del(11q) was isolated, primary and interstitial encompassing the 11q22-23 region containing ATM, KMT2A, and CBL genes. The common deleted region at 11q23.2 is centered on an intergenic region between CADM1 (also known as Tumor Suppressor in Lung Cancer 1) and NXPE2. CADM1 was expressed in all myeloid cells analyzed in contrast to NXPE2. At the functional level, the deletion of Cadm1 in murine Lineage-Sca1+Kit+ cells modifies the lymphoid-to-myeloid ratio in bone marrow, although not altering their multilineage hematopoietic reconstitution potential after syngenic transplantation. Together with the frequent simultaneous deletions of KMT2A, ATM, and CBL and mutations of ASXL1, SF3B1, and CBL, we show that CADM1 may be important in the physiopathology of the del(11q) MDS, extending its role as tumor-suppressor gene from solid tumors to hematopoietic malignancies.
Collapse
Affiliation(s)
- Marina Lafage-Pochitaloff
- Groupe Francophone de Cytogénétique Hématologique (GFCH)
- Laboratoire de Cytogénétique Hématologique, Centre Hospitalier Universitaire (CHU) de Marseille, Aix-Marseille University, Marseille, France
| | - Bastien Gerby
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Team 16, Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
| | - Véronique Baccini
- Groupe Francophone d’Hématologie Cellulaire (GFHC) and
- Laboratoire d’hématologie, CHU de Guadeloupe, Inserm Unité Mixte de Recherche 1134, Pointe à Pitre, France
| | - Laetitia Largeaud
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Team 16, Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Laboratoire d’Hématologie, Institut Universitaire de Cancérologie de Toulouse, CHU Toulouse, France
- Department of Hematology, University Toulouse III, Toulouse, France
| | - Vincent Fregona
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Team 16, Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
| | - Naïs Prade
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Team 16, Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Laboratoire d’Hématologie, Institut Universitaire de Cancérologie de Toulouse, CHU Toulouse, France
| | - Pierre-Yves Juvin
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Team 16, Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
| | - Laura Jamrog
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Team 16, Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
| | - Pierre Bories
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Team 16, Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
| | - Sylvie Hébrard
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Team 16, Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
| | - Stéphanie Lagarde
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Team 16, Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Laboratoire d’Hématologie, Institut Universitaire de Cancérologie de Toulouse, CHU Toulouse, France
| | - Véronique Mansat-De Mas
- Laboratoire d’Hématologie, Institut Universitaire de Cancérologie de Toulouse, CHU Toulouse, France
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Team 8, Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
| | - Oliver M. Dovey
- Gene Editing, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Kosuke Yusa
- Stem Cell Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - George S. Vassiliou
- Wellcome Sanger Institute, Hinxton, UK
- Department of Haematology, Cambridge University Hospitals National Health Service Trust, Cambridge, UK
- Wellcome-Medical Research Council Stem Cell Institute, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Joop H. Jansen
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tobias Tekath
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - David Rombaut
- Institut Cochin, Université de Paris, Inserm U1016, Centre National de la Recherche Scientifique UMR8104, Paris, France
| | - Geneviève Ameye
- Groupe Francophone de Cytogénétique Hématologique (GFCH)
- Belgium Cancer Registry, Brussels, Belgium
- Department of Human Genetics, Katholieke Universiteit Leuven and Universitair Ziekenhuis, Leuven, Belgium
| | - Carole Barin
- Groupe Francophone de Cytogénétique Hématologique (GFCH)
- Laboratoire de Cytogénétique, CHU de Tours, France
| | - Audrey Bidet
- Groupe Francophone de Cytogénétique Hématologique (GFCH)
- Laboratoire d’Hématologie, CHU de Bordeaux, Bordeaux, France
| | - John Boudjarane
- Laboratoire de Cytogénétique Hématologique, Centre Hospitalier Universitaire (CHU) de Marseille, Aix-Marseille University, Marseille, France
| | - Marie-Agnès Collonge-Rame
- Groupe Francophone de Cytogénétique Hématologique (GFCH)
- Laboratoire de Cytogénétique, CHU de Besançon, Besançon, France
| | - Carine Gervais
- Groupe Francophone de Cytogénétique Hématologique (GFCH)
- Laboratoire de Cytogénétique, CHU de Strasbourg, Strasbourg, France
| | - Antoine Ittel
- Groupe Francophone de Cytogénétique Hématologique (GFCH)
- Département de Biopathologie, Institut Paoli-Calmettes, Marseille, France
| | - Christine Lefebvre
- Groupe Francophone de Cytogénétique Hématologique (GFCH)
- Laboratoire de Cytogénétique, CHU de Grenoble, Grenoble, France
| | - Isabelle Luquet
- Groupe Francophone de Cytogénétique Hématologique (GFCH)
- Laboratoire d’Hématologie, Institut Universitaire de Cancérologie de Toulouse, CHU Toulouse, France
- Laboratoire de Cytogénétique, CHU de Reims, Reims, France
| | - Lucienne Michaux
- Groupe Francophone de Cytogénétique Hématologique (GFCH)
- Department of Human Genetics, Katholieke Universiteit Leuven and Universitair Ziekenhuis, Leuven, Belgium
| | - Nathalie Nadal
- Groupe Francophone de Cytogénétique Hématologique (GFCH)
- Laboratoire de Cytogénétique, CHU de Saint-Etienne, Saint-Etienne, France
| | - Hélène A. Poirel
- Groupe Francophone de Cytogénétique Hématologique (GFCH)
- Belgium Cancer Registry, Brussels, Belgium
| | - Isabelle Radford-Weiss
- Groupe Francophone de Cytogénétique Hématologique (GFCH)
- Laboratoire de Cytogénétique, CHU de Paris-Necker, Paris, France
| | - Bénédicte Ribourtout
- Groupe Francophone de Cytogénétique Hématologique (GFCH)
- Laboratoire d'Hématologie, CHU d'Angers, Angers, France
| | - Steven Richebourg
- Groupe Francophone de Cytogénétique Hématologique (GFCH)
- Laboratoire de Cytogénétique, CHU de Nantes, Nantes, France
| | - Stéphanie Struski
- Groupe Francophone de Cytogénétique Hématologique (GFCH)
- Laboratoire d’Hématologie, Institut Universitaire de Cancérologie de Toulouse, CHU Toulouse, France
| | - Christine Terré
- Groupe Francophone de Cytogénétique Hématologique (GFCH)
- Laboratoire de Cytogénétique, CH de Versailles, Le Chesnay, France
| | - Isabelle Tigaud
- Groupe Francophone de Cytogénétique Hématologique (GFCH)
- Laboratoire de Cytogénétique, CHU de Lyon, Lyon, France
| | - Dominique Penther
- Groupe Francophone de Cytogénétique Hématologique (GFCH)
- Laboratoire de Cytogénétique, Centre Henri-Becquerel, Rouen, France
| | - Virginie Eclache
- Groupe Francophone de Cytogénétique Hématologique (GFCH)
- Laboratoire d’Hématologie, CHU Avicenne, Bobigny, France
- Groupe Francophone des Myélodysplasies (GFM); and
| | - Michaela Fontenay
- Institut Cochin, Université de Paris, Inserm U1016, Centre National de la Recherche Scientifique UMR8104, Paris, France
- Groupe Francophone des Myélodysplasies (GFM); and
- Laboratoire d’hématologie, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Centre-Université de Paris, Paris, France
| | - Cyril Broccardo
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Team 16, Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
| | - Eric Delabesse,
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Team 16, Institut National de la Santé et de la Recherche Médicale (INSERM), Toulouse, France
- Laboratoire d’Hématologie, Institut Universitaire de Cancérologie de Toulouse, CHU Toulouse, France
| |
Collapse
|
16
|
Benderitter M, Herrera-Reyes E, Gigov Y, Souleau B, Huet JC, Trompier F, Fagot T, Grégoire E, Malfuson JV, Konopacki-Potet J, Buglova E, Lataillade JJ, Tamarat R, Gourmelon P, de Revel T. Hematopoietic Recovery using Multi-Cytokine Therapy in 8 Patients Presenting Radiation-Induced Myelosuppression after Radiological Accidents. Radiat Res 2021; 196:668-679. [PMID: 34554263 DOI: 10.1667/rade-21-00169.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 08/27/2021] [Indexed: 11/03/2022]
Abstract
Treatment of accidental radiation-induced myelosuppression is primarily based on supportive care and requires specific treatment based on hematopoietic growth factors injection or hematopoietic cell transplantation for the most severe cases. The cytokines used consisted of pegylated erythropoietin (darbepoetin alfa) 500 IU once per week, pegylated G-CSF (pegfilgrastim) 6 mg × 2 once, stem cell factor 20 μg.kg-1 for five days, and romiplostim (TPO analog) 10 μg.kg-1 once per week, with different combinations depending on the accidents. As the stem cell factor did not have regulatory approval for clinical use in France, the French regulatory authorities (ANSM, formerly, AFSSAPS) approved their compassionate use as an investigational drug "on a case-by-case basis". According to the evolution and clinical characteristics, each patient's treatment was adopted on an individual basis. Daily blood count allows initiating G-CSF and SCF delivery when granulocyte <1,000/mm3, TPO delivery when platelets <50,000/mm3, and EPO when Hb<80 g/L. The length of each treatment was based on blood cell recovery criteria. The concept of "stimulation strategy" is linked to each patient's residual hematopoiesis, which varies among them, depending on the radiation exposure's characteristics and heterogeneity. This paper reports the medical management of 8 overexposed patients to ionizing radiation. The recovery of bone marrow function after myelosuppression was accelerated using growth factors, optimized by multiple-line combinations. Particularly in the event of prolonged exposure to ionizing radiation in dose ranges inducing severe myelosuppression (in the order of 5 to 8 Gy), with no indication of hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Marc Benderitter
- Institute of Radioprotection and Nuclear Safety (IRSN), Health Division, BP17, 92262 Fontenay-aux-Roses, France
| | - Eduardo Herrera-Reyes
- Institute of Radioprotection and Nuclear Safety (IRSN), Health Division, BP17, 92262 Fontenay-aux-Roses, France
| | - Yvan Gigov
- Military Hospital, Department of Hematology and Chemotherapy for Solid Tumors, Sofia, Bulgaria
| | - Bertrand Souleau
- Percy Armed Forces Hospital, Department of Hematology, 92140 Clamart, France
| | - Jean Christelle Huet
- Institute of Radioprotection and Nuclear Safety (IRSN), Health Division, BP17, 92262 Fontenay-aux-Roses, France
| | - François Trompier
- Institute of Radioprotection and Nuclear Safety (IRSN), Health Division, BP17, 92262 Fontenay-aux-Roses, France
| | - Thierry Fagot
- Percy Armed Forces Hospital, Department of Hematology, 92140 Clamart, France
| | - Eric Grégoire
- Institute of Radioprotection and Nuclear Safety (IRSN), Health Division, BP17, 92262 Fontenay-aux-Roses, France
| | - Jean Valère Malfuson
- Percy Armed Forces Hospital, Department of Hematology, 92140 Clamart, France.,Blood Transfusion Center, Percy Armed Forces Hospital, 101 avenue Henri Barbusse, 92140 Clamart, France
| | | | - Elena Buglova
- International Atomic Energy Agency, Wagramerstrasse 5, A-1400 Vienna, Austria
| | - Jean-Jacques Lataillade
- Ecole du Val de Grâce, 75005 Paris, France.,Blood Transfusion Center, Percy Armed Forces Hospital, 101 avenue Henri Barbusse, 92140 Clamart, France
| | - Radia Tamarat
- Institute of Radioprotection and Nuclear Safety (IRSN), Health Division, BP17, 92262 Fontenay-aux-Roses, France
| | - Patrick Gourmelon
- Institute of Radioprotection and Nuclear Safety (IRSN), Health Division, BP17, 92262 Fontenay-aux-Roses, France
| | - Thierry de Revel
- Percy Armed Forces Hospital, Department of Hematology, 92140 Clamart, France.,Ecole du Val de Grâce, 75005 Paris, France
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW In the last few decades, revolutionary advances in next-generation sequencing have led to single-cell lineage tracing technologies that now enable researchers to identify and quantify hematopoietic cell behavior with unprecedented detail. Combined readouts of cell lineage and cell state from the same cell mitigate the need to prospectively isolate populations of interest, and allow a system-level understanding of dynamic developmental processes. We will discuss the advantages and shortcomings of these technologies, the intriguing discoveries that stemmed from lineage tracing hematopoiesis at the single-cell level and the directions toward which the field is moving. RECENT FINDINGS Single-cell lineage tracing studies unveiled extensive functional heterogeneity within discrete immunophenotypic populations. Recently, several groups merged lineage tracing with single-cell RNA sequencing to visualize clonal relationships directly on transcriptional landscapes without the requirement for prospective isolation of cell types by FACS. To study the cell dynamics of hematopoiesis, without perturbation in their native niche, researchers have developed mouse models with endogenous single-cell lineage tracing systems, which can simultaneously trace thousands of hematopoietic progenitor cells in a single mouse, without transplantation. The emerging picture is that multiple hematopoietic hierarchies coexist within a single individual, each with distinct regulatory features. These hierarchies are imprinted during development much earlier than previously predicted, persisting well into adulthood and even after injury and transplantation. SUMMARY Clone-tracking experiments allow stem-cell researchers to characterize lineage hierarchies during blood development and regeneration. Combined with single-cell genomics analyses, these studies are allowing system-level description of hematopoiesis in mice and humans. Early exploratory studies have unveiled features with important implications for human biology and disease. VIDEO ABSTRACT.
Collapse
|
18
|
Cordes S, Wu C, Dunbar CE. Clonal tracking of haematopoietic cells: insights and clinical implications. Br J Haematol 2021; 192:819-831. [PMID: 33216985 PMCID: PMC9927566 DOI: 10.1111/bjh.17175] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 09/16/2020] [Indexed: 01/03/2023]
Abstract
Recent advances in high-throughput genomics have enabled the direct tracking of outputs from many cell types, greatly accelerating the study of developmental processes and tissue regeneration. The capacity for long-term self-renewal with multilineage differentiation potential characterises the cellular dynamics of a special set of developmental states that are critical for maintaining homeostasis. In haematopoiesis, the archetypal model for development, lineage-tracing experiments have elucidated the roles of haematopoietic stem cells to ongoing blood production and the importance of long-lived immune cells to immunological memory. An understanding of the biology and clonal dynamics of these cellular fates and states can provide clues to the response of haematopoiesis to ageing, the process of malignant transformation, and are key to designing more efficacious and durable clinical gene and cellular therapies.
Collapse
Affiliation(s)
- Stefan Cordes
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Chuanfeng Wu
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| | - Cynthia E Dunbar
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, Bethesda, MD, USA
| |
Collapse
|
19
|
Dong S, Wang Q, Kao YR, Diaz A, Tasset I, Kaushik S, Thiruthuvanathan V, Zintiridou A, Nieves E, Dzieciatkowska M, Reisz JA, Gavathiotis E, D’Alessandro A, Will B, Cuervo AM. Chaperone-mediated autophagy sustains haematopoietic stem-cell function. Nature 2021; 591:117-123. [PMID: 33442062 PMCID: PMC8428053 DOI: 10.1038/s41586-020-03129-z] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 12/09/2020] [Indexed: 01/29/2023]
Abstract
The activation of mostly quiescent haematopoietic stem cells (HSCs) is a prerequisite for life-long production of blood cells1. This process requires major molecular adaptations to allow HSCs to meet the regulatory and metabolic requirements for cell division2-4. The mechanisms that govern cellular reprograming upon stem-cell activation, and the subsequent return of stem cells to quiescence, have not been fully characterized. Here we show that chaperone-mediated autophagy (CMA)5, a selective form of lysosomal protein degradation, is involved in sustaining HSC function in adult mice. CMA is required for protein quality control in stem cells and for the upregulation of fatty acid metabolism upon HSC activation. We find that CMA activity in HSCs decreases with age and show that genetic or pharmacological activation of CMA can restore the functionality of old mouse and human HSCs. Together, our findings provide mechanistic insights into a role for CMA in sustaining quality control, appropriate energetics and overall long-term HSC function. Our work suggests that CMA may be a promising therapeutic target for enhancing HSC function in conditions such as ageing or stem-cell transplantation.
Collapse
Affiliation(s)
- S Dong
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, NY, USA;,Institute for Aging Studies, Albert Einstein College of Medicine, NY, USA
| | - Q Wang
- Department of Cell Biology, Albert Einstein College of Medicine, NY, USA
| | - YR Kao
- Department of Cell Biology, Albert Einstein College of Medicine, NY, USA
| | - A Diaz
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, NY, USA;,Institute for Aging Studies, Albert Einstein College of Medicine, NY, USA
| | - I Tasset
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, NY, USA;,Institute for Aging Studies, Albert Einstein College of Medicine, NY, USA
| | - S Kaushik
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, NY, USA;,Institute for Aging Studies, Albert Einstein College of Medicine, NY, USA
| | - V Thiruthuvanathan
- Department of Cell Biology, Albert Einstein College of Medicine, NY, USA
| | - A Zintiridou
- Department of Cell Biology, Albert Einstein College of Medicine, NY, USA
| | - E Nieves
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, NY, USA
| | - M Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Anschutz Medical Campus, CO, USA
| | - JA Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Anschutz Medical Campus, CO, USA
| | - E Gavathiotis
- Institute for Aging Studies, Albert Einstein College of Medicine, NY, USA;,Department of Biochemistry, Albert Einstein College of Medicine, NY, USA;,Department of Medicine (Oncology), Albert Einstein College of Medicine, NY, USA
| | - A D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver – Anschutz Medical Campus, CO, USA
| | - B Will
- Institute for Aging Studies, Albert Einstein College of Medicine, NY, USA;,Department of Cell Biology, Albert Einstein College of Medicine, NY, USA;,Department of Medicine (Oncology), Albert Einstein College of Medicine, NY, USA;,Ruth L. and David S. Gottesman Institute for Stem Cell Biology, Albert Einstein College of Medicine, NY, USA,Corresponding authors: Ana Maria Cuervo MD PhD, Dept. Developmental Mol Biol, Institute for Aging Studies, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, Phone: +1 718 430 2689, , Britta Will PhD, Department of Cell Biology, Institute for Aging Studies, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, Phone: +1 718 430 3786,
| | - AM Cuervo
- Department of Development and Molecular Biology, Albert Einstein College of Medicine, NY, USA;,Institute for Aging Studies, Albert Einstein College of Medicine, NY, USA;,Corresponding authors: Ana Maria Cuervo MD PhD, Dept. Developmental Mol Biol, Institute for Aging Studies, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, Phone: +1 718 430 2689, , Britta Will PhD, Department of Cell Biology, Institute for Aging Studies, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, Phone: +1 718 430 3786,
| |
Collapse
|
20
|
Fraint E, Ulloa BA, Feliz Norberto M, Potts KS, Bowman TV. Advances in preclinical hematopoietic stem cell models and possible implications for improving therapeutic transplantation. Stem Cells Transl Med 2020; 10:337-345. [PMID: 33058566 PMCID: PMC7900582 DOI: 10.1002/sctm.20-0294] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/04/2020] [Accepted: 09/20/2020] [Indexed: 12/11/2022] Open
Abstract
Hematopoietic stem cell transplantation (HSCT) is a treatment for many malignant, congenital, and acquired hematologic diseases. Some outstanding challenges in the HSCT field include the paucity of immunologically‐matched donors, our inability to effectively expand hematopoeitic stem cells (HSCs) ex vivo, and the high infection risk during engraftment. Scientists are striving to develop protocols to generate, expand, and maintain HSCs ex vivo, however these are not yet ready for clinical application. Given these problems, advancing our understanding of HSC specification, regulation, and differentiation in preclinical models is essential to improve the therapeutic utility of HSCT. In this review, we link biomedical researchers and transplantation clinicians by discussing the potential therapeutic implications of recent fundamental HSC research in model organisms. We consider deficiencies in current HSCT practice, such as problems achieving adequate cell dose for successful and rapid engraftment, immense inflammatory cascade activation after myeloablation, and graft‐vs‐host disease. Furthermore, we discuss recent advances in the field of HSC biology and transplantation made in preclinical models of zebrafish, mouse, and nonhuman primates that could inform emerging practice for clinical application.
Collapse
Affiliation(s)
- Ellen Fraint
- Department of Pediatrics, Children's Hospital at Montefiore, Bronx, New York, USA
| | - Bianca A Ulloa
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, USA.,Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - María Feliz Norberto
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, USA.,Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Kathryn S Potts
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, USA.,Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Teresa V Bowman
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York, USA.,Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, USA.,Department of Medicine (Oncology), Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, USA
| |
Collapse
|
21
|
Emerging single-cell tools are primed to reveal functional and molecular heterogeneity in malignant hematopoietic stem cells. Curr Opin Hematol 2020; 26:214-221. [PMID: 31170109 DOI: 10.1097/moh.0000000000000512] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW The recent emergence of single-cell technologies has permitted unprecedented insight into the molecular drivers of fate choice in blood stem and progenitor cells. This review gives a broad overview of current efforts to understand the molecular regulators of malignant hematopoietic stem cells (HSCs) at the single-cell level. RECENT FINDINGS The large-scale adoption of single-cell approaches has allowed extensive description of the transcriptional profiles and functional properties of single HSCs. These techniques are now beginning to be applied to malignant HSCs isolated directly from patients or from mouse models of malignancy. However, these studies have generally struggled to pinpoint the functional regulators of malignant characteristics, since malignant HSCs often differ in more than one property when compared with normal HSCs. Moreover, both normal and malignant populations are complicated by HSC heterogeneity. SUMMARY Despite the existence of single-cell gene expression profiling tools, relatively few publications have emerged. Here, we review these studies from recent years with a specific focus on those undertaking single-cell measurements in malignant stem and progenitor cells. We anticipate this to be the tip of the iceberg, expecting the next 2-3 years to produce datasets that will facilitate a much broader understanding of malignant HSCs.
Collapse
|
22
|
Preclinical Development of Autologous Hematopoietic Stem Cell-Based Gene Therapy for Immune Deficiencies: A Journey from Mouse Cage to Bed Side. Pharmaceutics 2020; 12:pharmaceutics12060549. [PMID: 32545727 PMCID: PMC7357087 DOI: 10.3390/pharmaceutics12060549] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/05/2020] [Accepted: 06/09/2020] [Indexed: 02/08/2023] Open
Abstract
Recent clinical trials using patient’s own corrected hematopoietic stem cells (HSCs), such as for primary immunodeficiencies (Adenosine deaminase (ADA) deficiency, X-linked Severe Combined Immunodeficiency (SCID), X-linked chronic granulomatous disease (CGD), Wiskott–Aldrich Syndrome (WAS)), have yielded promising results in the clinic; endorsing gene therapy to become standard therapy for a number of diseases. However, the journey to achieve such a successful therapy is not easy, and several challenges have to be overcome. In this review, we will address several different challenges in the development of gene therapy for immune deficiencies using our own experience with Recombinase-activating gene 1 (RAG1) SCID as an example. We will discuss product development (targeting of the therapeutic cells and choice of a suitable vector and delivery method), the proof-of-concept (in vitro and in vivo efficacy, toxicology, and safety), and the final release steps to the clinic (scaling up, good manufacturing practice (GMP) procedures/protocols and regulatory hurdles).
Collapse
|
23
|
Adair JE, Enstrom MR, Haworth KG, Schefter LE, Shahbazi R, Humphrys DR, Porter S, Tam K, Porteus MH, Kiem HP. DNA Barcoding in Nonhuman Primates Reveals Important Limitations in Retrovirus Integration Site Analysis. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:796-809. [PMID: 32355868 PMCID: PMC7184234 DOI: 10.1016/j.omtm.2020.03.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 03/24/2020] [Indexed: 12/25/2022]
Abstract
In vivo tracking of retrovirus-tagged blood stem and progenitor cells is used to study hematopoiesis. Two techniques are used most frequently: sequencing the locus of retrovirus insertion, termed integration site analysis, or retrovirus DNA barcode sequencing. Of these, integration site analysis is currently the only available technique for monitoring clonal pools in patients treated with retrovirus-modified blood cells. A key question is how these two techniques compare in their ability to detect and quantify clonal contributions. In this study, we assessed both methods simultaneously in a clinically relevant nonhuman primate model of autologous, myeloablative transplantation. Our data demonstrate that both methods track abundant clones; however, DNA barcode sequencing is at least 5-fold more efficient than integration site analysis. Using computational simulation to identify the sources of low efficiency, we identify sampling depth as the major factor. We show that the sampling required for integration site analysis to achieve minimal coverage of the true clonal pool is likely prohibitive, especially in cases of low gene-modified cell engraftment. We also show that early subsampling of different blood cell lineages adds value to clone tracking information in terms of safety and hematopoietic biology. Our analysis demonstrates DNA barcode sequencing as a useful guide to maximize integration site analysis interpretation in gene therapy patients.
Collapse
Affiliation(s)
- Jennifer E Adair
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,School of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Mark R 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
| | - Reza Shahbazi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Daniel R Humphrys
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Shaina Porter
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Kenric Tam
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Matthew H Porteus
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA.,Institute of Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Hans-Peter Kiem
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.,School of Medicine, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
24
|
Ganuza M, Hall T, Obeng EA, McKinney-Freeman S. Clones assemble! The clonal complexity of blood during ontogeny and disease. Exp Hematol 2020; 83:35-47. [PMID: 32006606 PMCID: PMC8343955 DOI: 10.1016/j.exphem.2020.01.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/13/2020] [Accepted: 01/21/2020] [Indexed: 01/30/2023]
Abstract
Hematopoietic stem and progenitor cells (HSPCs) govern the daily expansion and turnover of billions of specialized blood cells. Given their clinical utility, much effort has been made toward understanding the dynamics of hematopoietic production from this pool of stem cells. An understanding of hematopoietic stem cell clonal dynamics during blood ontogeny could yield important insights into hematopoietic regulation, especially during aging and repeated exposure to hematopoietic stress-insults that may predispose individuals to the development of hematopoietic disease. Here, we review the current state of research regarding the clonal complexity of the hematopoietic system during embryogenesis, adulthood, and hematologic disease.
Collapse
Affiliation(s)
- Miguel Ganuza
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN
| | - Trent Hall
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN
| | - Esther A Obeng
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN
| | | |
Collapse
|
25
|
Zarrer J, Haider MT, Smit DJ, Taipaleenmäki H. Pathological Crosstalk between Metastatic Breast Cancer Cells and the Bone Microenvironment. Biomolecules 2020; 10:biom10020337. [PMID: 32092997 PMCID: PMC7072692 DOI: 10.3390/biom10020337] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 02/15/2020] [Accepted: 02/17/2020] [Indexed: 12/12/2022] Open
Abstract
Bone is the most common metastatic site in breast cancer. Upon arrival to the bone, disseminated tumor cells can undergo a period of dormancy but often eventually grow and hijack the bone microenvironment. The bone marrow microenvironment consists of multiple cell types including the bone cells, adipocytes, endothelial cells, and nerve cells that all have crucial functions in the maintenance of bone homeostasis. Tumor cells severely disturb the tightly controlled cellular and molecular interactions in the bone marrow fueling their own survival and growth. While the role of bone resorbing osteoclasts in breast cancer bone metastases is well established, the function of other bone cells, as well as adipocytes, endothelial cells, and nerve cells is less understood. In this review, we discuss the composition of the physiological bone microenvironment and how the presence of tumor cells influences the microenvironment, creating a pathological crosstalk between the cells. A better understanding of the cellular and molecular events that occur in the metastatic bone microenvironment could facilitate the identification of novel cellular targets to treat this devastating disease.
Collapse
Affiliation(s)
- Jennifer Zarrer
- Molecular Skeletal Biology Laboratory, Department of Trauma, Hand and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Marie-Therese Haider
- Molecular Skeletal Biology Laboratory, Department of Trauma, Hand and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Daniel J. Smit
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Hanna Taipaleenmäki
- Molecular Skeletal Biology Laboratory, Department of Trauma, Hand and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Correspondence:
| |
Collapse
|
26
|
Interleukin-12 supports in vitro self-renewal of long-term hematopoietic stem cells. BLOOD SCIENCE 2019; 1:92-101. [PMID: 35402790 PMCID: PMC8974953 DOI: 10.1097/bs9.0000000000000002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 02/07/2019] [Indexed: 11/25/2022] Open
Abstract
Hematopoietic stem cells (HSCs) self-renew or differentiate through division. Cytokines are essential for inducing HSC division, but the optimal cytokine combination to control self-renewal of HSC in vitro remains unclear. In this study, we compared the effects of interleukin-12 (IL-12) and thrombopoietin (TPO) in combination with stem cell factor (SCF) on in vitro self-renewal of HSCs. Single-cell assays were used to overcome the heterogeneity issue of HSCs, and serum-free conditions were newly established to permit reproduction of data. In single-cell cultures, CD150+CD48−CD41−CD34−c-Kit+Sca-1+lineage− HSCs divided significantly more slowly in the presence of SCF+IL-12 compared with cells in the presence of SCF+TPO. Serial transplantation of cells from bulk and clonal cultures revealed that TPO was more effective than IL-12 at supporting in vitro self-renewal of short-term (<6 months) HSCs, resulting in a monophasic reconstitution wave formation, whereas IL-12 was more effective than TPO at supporting the in vitro self-renewal of long-term (>6 months) HSCs, resulting in a biphasic reconstitution wave formation. The control of division rate in HSCs appeared to be crucial for preventing the loss of self-renewal potential from their in vitro culture.
Collapse
|
27
|
The global clonal complexity of the murine blood system declines throughout life and after serial transplantation. Blood 2019; 133:1927-1942. [PMID: 30782612 DOI: 10.1182/blood-2018-09-873059] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 02/18/2019] [Indexed: 01/02/2023] Open
Abstract
Although many recent studies describe the emergence and prevalence of "clonal hematopoiesis of indeterminate potential" in aged human populations, a systematic analysis of the numbers of clones supporting steady-state hematopoiesis throughout mammalian life is lacking. Previous efforts relied on transplantation of "barcoded" hematopoietic stem cells (HSCs) to track the contribution of HSC clones to reconstituted blood. However, ex vivo manipulation and transplantation alter HSC function and thus may not reflect the biology of steady-state hematopoiesis. Using a noninvasive in vivo color-labeling system, we report the first comprehensive analysis of the changing global clonal complexity of steady-state hematopoiesis during the natural murine lifespan. We observed that the number of clones (ie, clonal complexity) supporting the major blood and bone marrow hematopoietic compartments decline with age by ∼30% and ∼60%, respectively. Aging dramatically reduced HSC in vivo-repopulating activity and lymphoid potential while increasing functional heterogeneity. Continuous challenge of the hematopoietic system by serial transplantation provoked the clonal collapse of both young and aged hematopoietic systems. Whole-exome sequencing of serially transplanted aged and young hematopoietic clones confirmed oligoclonal hematopoiesis and revealed mutations in at least 27 genes, including nonsense, missense, and deletion mutations in Bcl11b, Hist1h2ac, Npy2r, Notch3, Ptprr, and Top2b.
Collapse
|
28
|
Clonal-level lineage commitment pathways of hematopoietic stem cells in vivo. Proc Natl Acad Sci U S A 2019; 116:1447-1456. [PMID: 30622181 DOI: 10.1073/pnas.1801480116] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
While the aggregate differentiation of the hematopoietic stem cell (HSC) population has been extensively studied, little is known about the lineage commitment process of individual HSC clones. Here, we provide lineage commitment maps of HSC clones under homeostasis and after perturbations of the endogenous hematopoietic system. Under homeostasis, all donor-derived HSC clones regenerate blood homogeneously throughout all measured stages and lineages of hematopoiesis. In contrast, after the hematopoietic system has been perturbed by irradiation or by an antagonistic anti-ckit antibody, only a small fraction of donor-derived HSC clones differentiate. Some of these clones dominantly expand and exhibit lineage bias. We identified the cellular origins of clonal dominance and lineage bias and uncovered the lineage commitment pathways that lead HSC clones to different levels of self-renewal and blood production under various transplantation conditions. This study reveals surprising alterations in HSC fate decisions directed by conditioning and identifies the key hematopoiesis stages that may be manipulated to control blood production and balance.
Collapse
|
29
|
Lyne AM, Kent DG, Laurenti E, Cornils K, Glauche I, Perié L. A track of the clones: new developments in cellular barcoding. Exp Hematol 2018; 68:15-20. [DOI: 10.1016/j.exphem.2018.11.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/09/2018] [Accepted: 11/13/2018] [Indexed: 11/30/2022]
|
30
|
Fluorescent genetic barcoding for cellular multiplex analyses. Exp Hematol 2018; 67:10-17. [DOI: 10.1016/j.exphem.2018.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 12/11/2022]
|
31
|
Xu S, Kim S, Chen ISY, Chou T. Modeling large fluctuations of thousands of clones during hematopoiesis: The role of stem cell self-renewal and bursty progenitor dynamics in rhesus macaque. PLoS Comput Biol 2018; 14:e1006489. [PMID: 30335762 PMCID: PMC6218102 DOI: 10.1371/journal.pcbi.1006489] [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: 03/23/2017] [Revised: 11/05/2018] [Accepted: 09/05/2018] [Indexed: 01/13/2023] Open
Abstract
In a recent clone-tracking experiment, millions of uniquely tagged hematopoietic stem cells (HSCs) and progenitor cells were autologously transplanted into rhesus macaques and peripheral blood containing thousands of tags were sampled and sequenced over 14 years to quantify the abundance of hundreds to thousands of tags or “clones.” Two major puzzles of the data have been observed: consistent differences and massive temporal fluctuations of clone populations. The large sample-to-sample variability can lead clones to occasionally go “extinct” but “resurrect” themselves in subsequent samples. Although heterogeneity in HSC differentiation rates, potentially due to tagging, and random sampling of the animals’ blood and cellular demographic stochasticity might be invoked to explain these features, we show that random sampling cannot explain the magnitude of the temporal fluctuations. Moreover, we show through simpler neutral mechanistic and statistical models of hematopoiesis of tagged cells that a broad distribution in clone sizes can arise from stochastic HSC self-renewal instead of tag-induced heterogeneity. The very large clone population fluctuations that often lead to extinctions and resurrections can be naturally explained by a generation-limited proliferation constraint on the progenitor cells. This constraint leads to bursty cell population dynamics underlying the large temporal fluctuations. We analyzed experimental clone abundance data using a new statistic that counts clonal disappearances and provided least-squares estimates of two key model parameters in our model, the total HSC differentiation rate and the maximum number of progenitor-cell divisions. Hematopoiesis of virally tagged cells in rhesus macaques is analyzed in the context of a mechanistic and statistical model. We find that the clone size distribution and the temporal variability in the abundance of each clone (viral tag) in peripheral blood are consistent with (i) stochastic HSC self-renewal during bone marrow repair, (ii) clonal aging that restricts the number of generations of progenitor cells, and (iii) infrequent and small-size samples. By fitting data, we infer two key parameters that control the level of fluctuations of clone sizes in our model: the total HSC differentiation rate and the maximum proliferation capacity of progenitor cells. Our analysis provides insight into the mechanisms of hematopoiesis and a framework to guide future multiclone barcoding/lineage tracking measurements.
Collapse
Affiliation(s)
- Song Xu
- Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, California, United States of America
| | - Sanggu Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, United States of America
| | - Irvin S. Y. Chen
- UCLA AIDS Institute and Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Tom Chou
- Center for Biomedical Informatics Research, Department of Medicine, Stanford University, Stanford, California, United States of America
- Department of Mathematics, University of California, Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
32
|
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
|
33
|
Wang X, Dong F, Zhang S, Yang W, Yu W, Wang Z, Zhang S, Wang J, Ma S, Wu P, Gao Y, Dong J, Tang F, Cheng T, Ema H. TGF-β1 Negatively Regulates the Number and Function of Hematopoietic Stem Cells. Stem Cell Reports 2018; 11:274-287. [PMID: 29937145 PMCID: PMC6067088 DOI: 10.1016/j.stemcr.2018.05.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 05/25/2018] [Accepted: 05/25/2018] [Indexed: 12/29/2022] Open
Abstract
Transforming growth factor β1 (TGF-β1) plays a role in the maintenance of quiescent hematopoietic stem cells (HSCs) in vivo. We asked whether TGF-β1 controls the cell cycle status of HSCs in vitro to enhance the reconstitution activity. To examine the effect of TGF-β1 on the HSC function, we used an in vitro culture system in which single HSCs divide with the retention of their short- and long-term reconstitution ability. Extensive single-cell analyses showed that, regardless of its concentration, TGF-β1 slowed down the cell cycle progression of HSCs but consequently suppressed their self-renewal potential. Cycling HSCs were not able to go back to quiescence with TGF-β1. This study revealed a negative role of TGF-β1 in the regulation of the HSC number and reconstitution activity. TGF-β1 slows down the cell cycle progression of HSCs Cycling HSCs were unable to go back to the G0 state with TGF-β1 TGF-β1 suppresses the self-renewal potential in HSCs The reduced division rate with TGF-β1 is reversible
Collapse
Affiliation(s)
- Xiaofang Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing Road 288, Tianjin 300020, China
| | - Fang Dong
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing Road 288, Tianjin 300020, China
| | - Sen Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing Road 288, Tianjin 300020, China
| | - Wanzhu Yang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing Road 288, Tianjin 300020, China
| | - Wenying Yu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing Road 288, Tianjin 300020, China
| | - Zhao Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing Road 288, Tianjin 300020, China
| | - Shanshan Zhang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing Road 288, Tianjin 300020, China
| | - Jinhong Wang
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing Road 288, Tianjin 300020, China
| | - Shihui Ma
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing Road 288, Tianjin 300020, China
| | - Peng Wu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing Road 288, Tianjin 300020, China
| | - Yun Gao
- Biodynamic Optical Imaging Center, College of Life Sciences, Peking University, Beijing 100871, China
| | - Ji Dong
- Biodynamic Optical Imaging Center, College of Life Sciences, Peking University, Beijing 100871, China
| | - Fuchou Tang
- Biodynamic Optical Imaging Center, College of Life Sciences, Peking University, Beijing 100871, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing Road 288, Tianjin 300020, China
| | - Hideo Ema
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing Road 288, Tianjin 300020, China.
| |
Collapse
|
34
|
Nguyen L, Wang Z, Chowdhury AY, Chu E, Eerdeng J, Jiang D, Lu R. Functional compensation between hematopoietic stem cell clones in vivo. EMBO Rep 2018; 19:embr.201745702. [PMID: 29848511 DOI: 10.15252/embr.201745702] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 05/10/2018] [Accepted: 05/13/2018] [Indexed: 02/05/2023] Open
Abstract
In most organ systems, regeneration is a coordinated effort that involves many stem cells, but little is known about whether and how individual stem cells compensate for the differentiation deficiencies of other stem cells. Functional compensation is critically important during disease progression and treatment. Here, we show how individual hematopoietic stem cell (HSC) clones heterogeneously compensate for the lymphopoietic deficiencies of other HSCs in a mouse. This compensation rescues the overall blood supply and influences blood cell types outside of the deficient lineages in distinct patterns. We find that highly differentiating HSC clones expand their cell numbers at specific differentiation stages to compensate for the deficiencies of other HSCs. Some of these clones continue to expand after transplantation into secondary recipients. In addition, lymphopoietic compensation involves gene expression changes in HSCs that are characterized by increased lymphoid priming, decreased myeloid priming, and HSC self-renewal. Our data illustrate how HSC clones coordinate to maintain the overall blood supply. Exploiting the innate compensation capacity of stem cell networks may improve the prognosis and treatment of many diseases.
Collapse
Affiliation(s)
- Lisa Nguyen
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Zheng Wang
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Adnan Y Chowdhury
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Elizabeth Chu
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jiya Eerdeng
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Du Jiang
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Rong Lu
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
35
|
Ge Y, Fuchs E. Stretching the limits: from homeostasis to stem cell plasticity in wound healing and cancer. Nat Rev Genet 2018; 19:311-325. [PMID: 29479084 PMCID: PMC6301069 DOI: 10.1038/nrg.2018.9] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Stem cells (SCs) govern tissue homeostasis and wound repair. They reside within niches, the special microenvironments within tissues that control SC lineage outputs. Upon injury or stress, new signals emanating from damaged tissue can divert nearby cells into adopting behaviours that are not part of their homeostatic repertoire. This behaviour, known as SC plasticity, typically resolves as wounds heal. However, in cancer, it can endure. Recent studies have yielded insights into the orchestrators of maintenance and lineage commitment for SCs belonging to three mammalian tissues: the haematopoietic system, the skin epithelium and the intestinal epithelium. We delineate the multifactorial determinants and general principles underlying the remarkable facets of SC plasticity, which lend promise for regenerative medicine and cancer therapeutics.
Collapse
Affiliation(s)
- Yejing Ge
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Elaine Fuchs
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| |
Collapse
|
36
|
Ball CR, Oppel F, Ehrenberg KR, Dubash TD, Dieter SM, Hoffmann CM, Abel U, Herbst F, Koch M, Werner J, Bergmann F, Ishaque N, Schmidt M, von Kalle C, Scholl C, Fröhling S, Brors B, Weichert W, Weitz J, Glimm H. Succession of transiently active tumor-initiating cell clones in human pancreatic cancer xenografts. EMBO Mol Med 2018; 9:918-932. [PMID: 28526679 PMCID: PMC5494525 DOI: 10.15252/emmm.201607354] [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] [Indexed: 01/05/2023] Open
Abstract
Although tumor-initiating cell (TIC) self-renewal has been postulated to be essential in progression and metastasis formation of human pancreatic adenocarcinoma (PDAC), clonal dynamics of TICs within PDAC tumors are yet unknown. Here, we show that long-term progression of PDAC in serial xenotransplantation is driven by a succession of transiently active TICs producing tumor cells in temporally restricted bursts. Clonal tracking of individual, genetically marked TICs revealed that individual tumors are generated by distinct sets of TICs with very little overlap between subsequent xenograft generations. An unexpected functional and phenotypic plasticity of pancreatic TICs in vivo underlies the recruitment of inactive TIC clones in serial xenografts. The observed clonal succession of TIC activity in serial xenotransplantation is in stark contrast to the continuous activity of limited numbers of self-renewing TICs within a fixed cellular hierarchy observed in other epithelial cancers and emphasizes the need to target TIC activation, rather than a fixed TIC population, in PDAC.
Collapse
Affiliation(s)
- Claudia R Ball
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Felix Oppel
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karl Roland Ehrenberg
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Medical Oncology, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Taronish D Dubash
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sebastian M Dieter
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), University of Heidelberg, Heidelberg, Germany
| | - Christopher M Hoffmann
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ulrich Abel
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Friederike Herbst
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Moritz Koch
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany.,Department of Visceral, Thoracic and Vascular Surgery, University Hospital Dresden, Dresden, Germany
| | - Jens Werner
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany.,Department of Surgery, University of Munich, Munich, Germany
| | - Frank Bergmann
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Naveed Ishaque
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Heidelberg Center for Personalized Oncology, DKFZ-HIPO, DKFZ, Heidelberg, Germany
| | - Manfred Schmidt
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), University of Heidelberg, Heidelberg, Germany
| | - Christof von Kalle
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), University of Heidelberg, Heidelberg, Germany
| | - Claudia Scholl
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan Fröhling
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany.,German Cancer Consortium (DKTK), University of Heidelberg, Heidelberg, Germany.,Heidelberg University Hospital, Heidelberg, Germany
| | - Benedikt Brors
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Wilko Weichert
- German Cancer Consortium (DKTK), University of Heidelberg, Heidelberg, Germany.,Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Jürgen Weitz
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany.,Department of Visceral, Thoracic and Vascular Surgery, University Hospital Dresden, Dresden, Germany
| | - Hanno Glimm
- Department of Translational Oncology, National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany .,German Cancer Consortium (DKTK), University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
37
|
Dharampuriya PR, Scapin G, Wong C, John Wagner K, Cillis JL, Shah DI. Tracking the origin, development, and differentiation of hematopoietic stem cells. Curr Opin Cell Biol 2018; 49:108-115. [PMID: 29413969 DOI: 10.1016/j.ceb.2018.01.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 12/26/2017] [Accepted: 01/09/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW The hierarchical nature of the hematopoietic system provides an ideal model system to illustrate the features of lineage tracing. We have outlined the utility of lineage tracing methods in establishing the origin and development of hematopoietic cells. RECENT FINDINGS Methods such as CRISPR/Cas9, Polylox barcoding, and single-cell RNA-sequencing have improved our understanding of hematopoiesis. SUMMARY This review chronicles the fate of the hematopoietic cells emerging from the mesoderm that subsequently develops into the adult blood lineages. Specifically, we explain classic techniques utilized in lineage tracing for the hematopoietic system, as well as novel state-of-the-art methods to elucidate clonal hematopoiesis and cell fate mapping at a single-cell level.
Collapse
Affiliation(s)
- Priyanka R Dharampuriya
- Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Giorgia Scapin
- Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02138, USA
| | - Colline Wong
- Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Boston College, Chestnut Hill, MA 02467, USA
| | - K John Wagner
- Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Boston College, Chestnut Hill, MA 02467, USA
| | - Jennifer L Cillis
- Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02138, USA
| | - Dhvanit I Shah
- Division of Hematology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02138, USA; Dana-Farber/Harvard Cancer Center, Boston, MA 02115, USA.
| |
Collapse
|
38
|
Migliaccio AR, Hoffman R. Remembering Ihor Lemischka—The scientist's scientist. Exp Hematol 2018. [DOI: 10.1016/j.exphem.2017.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
39
|
The impact of aging on primate hematopoiesis as interrogated by clonal tracking. Blood 2018; 131:1195-1205. [PMID: 29295845 DOI: 10.1182/blood-2017-08-802033] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 12/21/2017] [Indexed: 01/04/2023] Open
Abstract
Age-associated changes in hematopoietic stem and progenitor cells (HSPCs) have been carefully documented in mouse models but poorly characterized in primates and humans. To investigate clinically relevant aspects of hematopoietic aging, we compared the clonal output of thousands of genetically barcoded HSPCs in aged vs young macaques after autologous transplantation. Aged macaques showed delayed emergence of output from multipotent (MP) clones, with persistence of lineage-biased clones for many months after engraftment. In contrast to murine aging models reporting persistence of myeloid-biased HSPCs, aged macaques demonstrated persistent output from both B-cell and myeloid-biased clones. Clonal expansions of MP, myeloid-biased, and B-biased clones occurred in aged macaques, providing a potential model for human clonal hematopoiesis of indeterminate prognosis. These results suggest that long-term MP HSPC output is impaired in aged macaques, resulting in differences in the kinetics and lineage reconstitution patterns between young and aged primates in an autologous transplantation setting.
Collapse
|
40
|
Rossmann MP, Orkin SH, Chute JP. Hematopoietic Stem Cell Biology. Hematology 2018. [DOI: 10.1016/b978-0-323-35762-3.00009-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
41
|
Ganuza M, Hall T, Finkelstein D, Chabot A, Kang G, McKinney-Freeman S. Lifelong haematopoiesis is established by hundreds of precursors throughout mammalian ontogeny. Nat Cell Biol 2017; 19:1153-1163. [PMID: 28920953 PMCID: PMC5705075 DOI: 10.1038/ncb3607] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 08/09/2017] [Indexed: 12/17/2022]
Abstract
Current dogma asserts that mammalian lifelong blood production is established by a small number of blood progenitors. However, this model is based on assays that require the disruption, transplantation and/or culture of embryonic tissues. Here, we used the sample-to-sample variance of a multicoloured lineage trace reporter to assess the frequency of emerging lifelong blood progenitors while avoiding the disruption, culture or transplantation of embryos. We find that approximately 719 Flk1+ mesodermal precursors, 633 VE-cadherin+ endothelial precursors and 545 Vav1+ nascent blood stem and progenitor cells emerge to establish the haematopoietic system at embryonic days (E)7-E8.5, E8.5-E11.5 and E11.5-E14.5, respectively. We also determined that the spatio-temporal recruitment of endothelial blood precursors begins at E8.5 and ends by E10.5, and that many c-Kit+ clusters of newly specified blood progenitors in the aorta are polyclonal in origin. Our work illuminates the dynamics of the developing mammalian blood system during homeostasis.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Cadherins/genetics
- Cadherins/metabolism
- Cell Differentiation
- Cell Lineage
- Cell Tracking/methods
- Cells, Cultured
- Coculture Techniques
- Endothelial Cells/metabolism
- Endothelial Cells/transplantation
- Gene Expression Regulation, Developmental
- Genotype
- Gestational Age
- Hematopoiesis
- Hematopoietic Stem Cell Transplantation
- Hematopoietic Stem Cells/metabolism
- Integrases/genetics
- Integrases/metabolism
- Linear Models
- Luminescent Proteins/genetics
- Luminescent Proteins/metabolism
- Mice, Inbred C57BL
- Mice, Transgenic
- Microscopy, Fluorescence
- Models, Biological
- Phenotype
- Proto-Oncogene Proteins c-kit/genetics
- Proto-Oncogene Proteins c-kit/metabolism
- Proto-Oncogene Proteins c-vav/genetics
- Proto-Oncogene Proteins c-vav/metabolism
- RNA, Untranslated/genetics
- RNA, Untranslated/metabolism
- Signal Transduction
- Time Factors
- Vascular Endothelial Growth Factor Receptor-2/genetics
- Vascular Endothelial Growth Factor Receptor-2/metabolism
Collapse
Affiliation(s)
- Miguel Ganuza
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, 38105
| | - Trent Hall
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, 38105
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105
| | - Ashley Chabot
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, 38105
| | - Guolian Kang
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN, 38105
| | | |
Collapse
|
42
|
Kohlscheen S, Bonig H, Modlich U. Promises and Challenges in Hematopoietic Stem Cell Gene Therapy. Hum Gene Ther 2017; 28:782-799. [DOI: 10.1089/hum.2017.141] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Affiliation(s)
- Saskia Kohlscheen
- Research Group for Gene Modification in Stem Cells, Center for Cell and Gene Therapy Frankfurt, Paul-Ehrlich-Institute, Langen, Germany
| | - Halvard Bonig
- Institute for Transfusion Medicine and Immunohematology, Goethe University, Frankfurt, Germany
- German Red Cross Blood Service Baden-Württemberg-Hessen, Institute Frankfurt, Germany
- Department of Medicine/Division of Hematology, University of Washington, Seattle, Washington
| | - Ute Modlich
- Research Group for Gene Modification in Stem Cells, Center for Cell and Gene Therapy Frankfurt, Paul-Ehrlich-Institute, Langen, Germany
| |
Collapse
|
43
|
Limitations and challenges of genetic barcode quantification. Sci Rep 2017; 7:43249. [PMID: 28256524 PMCID: PMC5335698 DOI: 10.1038/srep43249] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 01/23/2017] [Indexed: 12/16/2022] Open
Abstract
Genetic barcodes are increasingly used to track individual cells and to quantitatively assess their clonal contributions over time. Although barcode quantification relies entirely on counting sequencing reads, detailed studies about the method’s accuracy are still limited. We report on a systematic investigation of the relation between barcode abundance and resulting read counts after amplification and sequencing using cell-mixtures that contain barcodes with known frequencies (“miniBulks”). We evaluated the influence of protocol modifications to identify potential sources of error and elucidate possible limitations of the quantification approach. Based on these findings we designed an advanced barcode construct (BC32) to improved barcode calling and quantification, and to ensure a sensitive detection of even highly diluted barcodes. Our results emphasize the importance of using curated barcode libraries to obtain interpretable quantitative data and underline the need for rigorous analyses of any utilized barcode library in terms of reliability and reproducibility.
Collapse
|
44
|
Yu VW, Yusuf RZ, Oki T, Wu J, Saez B, Wang X, Cook C, Baryawno N, Ziller MJ, Lee E, Gu H, Meissner A, Lin CP, Kharchenko PV, Scadden DT. Epigenetic Memory Underlies Cell-Autonomous Heterogeneous Behavior of Hematopoietic Stem Cells. Cell 2017; 168:944-945. [PMID: 28235203 PMCID: PMC5510238 DOI: 10.1016/j.cell.2017.02.010] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Stem cells determine homeostasis and repair of many tissues and are increasingly recognized as functionally heterogeneous. To define the extent of—and molecular basis for—heterogeneity, we overlaid functional, transcriptional, and epigenetic attributes of hematopoietic stem cells (HSCs) at a clonal level using endogenous fluorescent tagging. Endogenous HSC had clone-specific functional attributes in vivo. The intra-clonal behaviors were highly stereotypic, conserved under the stress of transplantation, inflammation, and genotoxic injury, and associated with distinctive transcriptional, DNA methylation, and chromatin accessibility patterns. Further, HSC function corresponded to epigenetic configuration but not always to transcriptional state. Therefore, hematopoiesis under homeostatic and stress conditions represents the integrated action of highly heterogeneous clones of HSC with epigenetically scripted behaviors. This high degree of epigenetically driven cell autonomy among HSCs implies that refinement of the concepts of stem cell plasticity and of the stem cell niche is warranted.
Collapse
Affiliation(s)
- Vionnie W.C. Yu
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Rushdia Z. Yusuf
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Toshihiko Oki
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Juwell Wu
- Broad Institute of Harvard and MIT, Cambridge, MA 02138, USA
| | - Borja Saez
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Xin Wang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Colleen Cook
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Ninib Baryawno
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| | - Michael J. Ziller
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02138, USA
| | - Eunjung Lee
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Hongcang Gu
- Broad Institute of Harvard and MIT, Cambridge, MA 02138, USA
| | - Alexander Meissner
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02138, USA
| | - Charles P. Lin
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Peter V. Kharchenko
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - David T. Scadden
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
45
|
Quantitative stability of hematopoietic stem and progenitor cell clonal output in rhesus macaques receiving transplants. Blood 2017; 129:1448-1457. [PMID: 28087539 DOI: 10.1182/blood-2016-07-728691] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 01/04/2017] [Indexed: 02/07/2023] Open
Abstract
Autologous transplantation of hematopoietic stem and progenitor cells lentivirally labeled with unique oligonucleotide barcodes flanked by sequencing primer targets enables quantitative assessment of the self-renewal and differentiation patterns of these cells in a myeloablative rhesus macaque model. Compared with other approaches to clonal tracking, this approach is highly quantitative and reproducible. We documented stable multipotent long-term hematopoietic clonal output of monocytes, granulocytes, B cells, and T cells from a polyclonal pool of hematopoietic stem and progenitor cells in 4 macaques observed for up to 49 months posttransplantation. A broad range of clonal behaviors characterized by contribution level and biases toward certain cell types were extremely stable over time. Correlations between granulocyte and monocyte clonalities were greatest, followed by correlations between these cell types and B cells. We also detected quantitative expansion of T cell-biased clones consistent with an adaptive immune response. In contrast to recent data from a nonquantitative murine model, there was little evidence for clonal succession after initial hematopoietic reconstitution. These findings have important implications for human hematopoiesis, given the similarities between macaque and human physiologies.
Collapse
|
46
|
One or two umbilical cord blood cell units? Caveat emptor. Bone Marrow Transplant 2017; 52:341-343. [DOI: 10.1038/bmt.2016.277] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 07/21/2016] [Indexed: 12/31/2022]
|
47
|
Abstract
Delineating the behaviour of haematopoietic stem cells (HSCs) in vivo has thus far proven challenging. Two studies in zebrafish and mouse models now track HSCs in vivo using fate mapping with multicolour approaches to provide further insights into clonal events that regulate blood development, HSC function and differentiation during homeostasis and stress conditions.
Collapse
Affiliation(s)
- Trista E North
- Beth Israel Deaconess Medical Center, Department of Pathology, Harvard Medical School, Harvard Stem Cell Institute, CLS 536, 3 Blackfan Circle, Boston, Massachusetts 02115, USA
| | - Wolfram Goessling
- Brigham and Women's Hospital, Genetics and Gastroenterology Division, Harvard Medical School, Harvard Stem Cell Institute, NRB 458, 77 Avenue Louis Pasteur, Boston, Massachusetts 02215, USA
| |
Collapse
|
48
|
Henninger J, Santoso B, Hans S, Durand E, Moore J, Mosimann C, Brand M, Traver D, Zon L. Clonal fate mapping quantifies the number of haematopoietic stem cells that arise during development. Nat Cell Biol 2016; 19:17-27. [PMID: 27870830 DOI: 10.1038/ncb3444] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 10/27/2016] [Indexed: 12/15/2022]
Abstract
Haematopoietic stem cells (HSCs) arise in the developing aorta during embryogenesis. The number of HSC clones born has been estimated through transplantation, but experimental approaches to assess the absolute number of forming HSCs in a native setting have remained challenging. Here, we applied single-cell and clonal analysis of HSCs in zebrafish to quantify developing HSCs. Targeting creERT2 in developing cd41:eGFP+ HSCs enabled long-term assessment of their blood contribution. We also applied the Brainbow-based multicolour Zebrabow system with drl:creERT2 that is active in early haematopoiesis to induce heritable colour barcoding unique to each HSC and its progeny. Our findings reveal that approximately 21 HSC clones exist prior to HSC emergence and 30 clones are present during peak production from aortic endothelium. Our methods further reveal that stress haematopoiesis, including sublethal irradiation and transplantation, reduces clonal diversity. Our findings provide quantitative insights into the early clonal events that regulate haematopoietic development.
Collapse
Affiliation(s)
- Jonathan Henninger
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts 02115, USA.,Biological and Biomedical Sciences, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Buyung Santoso
- Section of Cell and Developmental Biology, University of California at San Diego, La Jolla, California 92093, USA
| | - Stefan Hans
- Biotechnology Center and Center for Regenerative Therapies Dresden, Dresden University of Technology, Tatzberg 47-49, 01307 Dresden, Germany
| | - Ellen Durand
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts 02115, USA.,Biological and Biomedical Sciences, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Jessica Moore
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Christian Mosimann
- Institute of Molecular Life Sciences, University of Zürich, 8057 Zürich, Switzerland
| | - Michael Brand
- Biotechnology Center and Center for Regenerative Therapies Dresden, Dresden University of Technology, Tatzberg 47-49, 01307 Dresden, Germany
| | - David Traver
- Section of Cell and Developmental Biology, University of California at San Diego, La Jolla, California 92093, USA.,Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093-0380, USA
| | - Leonard Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts 02115, USA.,Biological and Biomedical Sciences, Harvard University, Cambridge, Massachusetts 02138, USA
| |
Collapse
|
49
|
Epigenetic Memory Underlies Cell-Autonomous Heterogeneous Behavior of Hematopoietic Stem Cells. Cell 2016; 167:1310-1322.e17. [DOI: 10.1016/j.cell.2016.10.045] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 08/09/2016] [Accepted: 10/25/2016] [Indexed: 12/11/2022]
|
50
|
Pleyer L, Valent P, Greil R. Mesenchymal Stem and Progenitor Cells in Normal and Dysplastic Hematopoiesis-Masters of Survival and Clonality? Int J Mol Sci 2016; 17:ijms17071009. [PMID: 27355944 PMCID: PMC4964385 DOI: 10.3390/ijms17071009] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 05/20/2016] [Accepted: 06/08/2016] [Indexed: 02/07/2023] Open
Abstract
Myelodysplastic syndromes (MDS) are malignant hematopoietic stem cell disorders that have the capacity to progress to acute myeloid leukemia (AML). Accumulating evidence suggests that the altered bone marrow (BM) microenvironment in general, and in particular the components of the stem cell niche, including mesenchymal stem cells (MSCs) and their progeny, play a pivotal role in the evolution and propagation of MDS. We here present an overview of the role of MSCs in the pathogenesis of MDS, with emphasis on cellular interactions in the BM microenvironment and related stem cell niche concepts. MSCs have potent immunomodulatory capacities and communicate with diverse immune cells, but also interact with various other cellular components of the microenvironment as well as with normal and leukemic stem and progenitor cells. Moreover, compared to normal MSCs, MSCs in MDS and AML often exhibit altered gene expression profiles, an aberrant phenotype, and abnormal functional properties. These alterations supposedly contribute to the “reprogramming” of the stem cell niche into a disease-permissive microenvironment where an altered immune system, abnormal stem cell niche interactions, and an impaired growth control lead to disease progression. The current article also reviews molecular targets that play a role in such cellular interactions and possibilities to interfere with abnormal stem cell niche interactions by using specific targeted drugs.
Collapse
Affiliation(s)
- Lisa Pleyer
- 3rd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory for Immunological and Molecular Cancer Research, Oncologic Center, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria.
- Center for Clinical Cancer and Immunology Trials at Salzburg Cancer Research Institute, 5020 Salzburg, Austria.
- 3rd Medical Department, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology & Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, 1090 Vienna, Austria.
| | - Richard Greil
- 3rd Medical Department with Hematology and Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory for Immunological and Molecular Cancer Research, Oncologic Center, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria.
- Center for Clinical Cancer and Immunology Trials at Salzburg Cancer Research Institute, 5020 Salzburg, Austria.
- 3rd Medical Department, Cancer Cluster Salzburg, 5020 Salzburg, Austria.
| |
Collapse
|