51
|
Swann JW, Olson OC, Passegué E. Made to order: emergency myelopoiesis and demand-adapted innate immune cell production. Nat Rev Immunol 2024:10.1038/s41577-024-00998-7. [PMID: 38467802 DOI: 10.1038/s41577-024-00998-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2024] [Indexed: 03/13/2024]
Abstract
Definitive haematopoiesis is the process by which haematopoietic stem cells, located in the bone marrow, generate all haematopoietic cell lineages in healthy adults. Although highly regulated to maintain a stable output of blood cells in health, the haematopoietic system is capable of extensive remodelling in response to external challenges, prioritizing the production of certain cell types at the expense of others. In this Review, we consider how acute insults, such as infections and cytotoxic drug-induced myeloablation, cause molecular, cellular and metabolic changes in haematopoietic stem and progenitor cells at multiple levels of the haematopoietic hierarchy to drive accelerated production of the mature myeloid cells needed to resolve the initiating insult. Moreover, we discuss how dysregulation or subversion of these emergency myelopoiesis mechanisms contributes to the progression of chronic inflammatory diseases and cancer.
Collapse
Affiliation(s)
- James W Swann
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University, New York, NY, USA
| | - Oakley C Olson
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University, New York, NY, USA
| | - Emmanuelle Passegué
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University, New York, NY, USA.
| |
Collapse
|
52
|
Zhang Q, Olofzon R, Konturek-Ciesla A, Yuan O, Bryder D. Ex vivo expansion potential of murine hematopoietic stem cells is a rare property only partially predicted by phenotype. eLife 2024; 12:RP91826. [PMID: 38446538 PMCID: PMC10942641 DOI: 10.7554/elife.91826] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024] Open
Abstract
The scarcity of hematopoietic stem cells (HSCs) restricts their use in both clinical settings and experimental research. Here, we examined a recently developed method for expanding rigorously purified murine HSCs ex vivo. After 3 weeks of culture, only 0.1% of cells exhibited the input HSC phenotype, but these accounted for almost all functional long-term HSC activity. Input HSCs displayed varying potential for ex vivo self-renewal, with alternative outcomes revealed by single-cell multimodal RNA and ATAC sequencing profiling. While most HSC progeny offered only transient in vivo reconstitution, these cells efficiently rescued mice from lethal myeloablation. The amplification of functional HSC activity allowed for long-term multilineage engraftment in unconditioned hosts that associated with a return of HSCs to quiescence. Thereby, our findings identify several key considerations for ex vivo HSC expansion, with major implications also for assessment of normal HSC activity.
Collapse
Affiliation(s)
- Qinyu Zhang
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Faculty of Medical, Lund UniversityLundSweden
| | - Rasmus Olofzon
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Faculty of Medical, Lund UniversityLundSweden
| | - Anna Konturek-Ciesla
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Faculty of Medical, Lund UniversityLundSweden
| | - Ouyang Yuan
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Faculty of Medical, Lund UniversityLundSweden
| | - David Bryder
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Faculty of Medical, Lund UniversityLundSweden
| |
Collapse
|
53
|
Kreger J, Mooney JA, Shibata D, MacLean AL. Developmental hematopoietic stem cell variation explains clonal hematopoiesis later in life. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.02.583106. [PMID: 38496542 PMCID: PMC10942294 DOI: 10.1101/2024.03.02.583106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Clonal hematopoiesis becomes increasingly common with age, but its cause is enigmatic because driver mutations are often absent. Serial observations infer weak selection indicating variants are acquired much earlier in life with unexplained initial growth spurts. Here we use fluctuating CpG methylation as a lineage marker to track stem cell clonal dynamics of hematopoiesis. We show, via the shared prenatal circulation of monozygotic twins, that weak selection conferred by stem cell variation created before birth can reliably yield clonal hematopoiesis later in life. Theory indicates weak selection will lead to dominance given enough time and large enough population sizes. Human hematopoiesis satisfies both these conditions. Stochastic loss of weakly selected variants is naturally prevented by the expansion of stem cell lineages during development. The dominance of stem cell clones created before birth is supported by blood fluctuating CpG methylation patterns that exhibit low correlation between unrelated individuals but are highly correlated between many elderly monozygotic twins. Therefore, clonal hematopoiesis driven by weak selection in later life appears to reflect variation created before birth.
Collapse
Affiliation(s)
- Jesse Kreger
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA
| | - Jazlyn A. Mooney
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA
| | - Darryl Shibata
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Adam L. MacLean
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
54
|
Nechanitzky R, Ramachandran P, Nechanitzky D, Li WY, Wakeham AC, Haight J, Saunders ME, Epelman S, Mak TW. CaSSiDI: novel single-cell "Cluster Similarity Scoring and Distinction Index" reveals critical functions for PirB and context-dependent Cebpb repression. Cell Death Differ 2024; 31:265-279. [PMID: 38383888 PMCID: PMC10923835 DOI: 10.1038/s41418-024-01268-8] [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] [Received: 05/16/2023] [Revised: 01/15/2024] [Accepted: 02/07/2024] [Indexed: 02/23/2024] Open
Abstract
PirB is an inhibitory cell surface receptor particularly prominent on myeloid cells. PirB curtails the phenotypes of activated macrophages during inflammation or tumorigenesis, but its functions in macrophage homeostasis are obscure. To elucidate PirB-related functions in macrophages at steady-state, we generated and compared single-cell RNA-sequencing (scRNAseq) datasets obtained from myeloid cell subsets of wild type (WT) and PirB-deficient knockout (PirB KO) mice. To facilitate this analysis, we developed a novel approach to clustering parameter optimization called "Cluster Similarity Scoring and Distinction Index" (CaSSiDI). We demonstrate that CaSSiDI is an adaptable computational framework that facilitates tandem analysis of two scRNAseq datasets by optimizing clustering parameters. We further show that CaSSiDI offers more advantages than a standard Seurat analysis because it allows direct comparison of two or more independently clustered datasets, thereby alleviating the need for batch-correction while identifying the most similar and different clusters. Using CaSSiDI, we found that PirB is a novel regulator of Cebpb expression that controls the generation of Ly6Clo patrolling monocytes and the expansion properties of peritoneal macrophages. PirB's effect on Cebpb is tissue-specific since it was not observed in splenic red pulp macrophages (RPMs). However, CaSSiDI revealed a segregation of the WT RPM population into a CD68loIrf8+ "neuronal-primed" subset and an CD68hiFtl1+ "iron-loaded" subset. Our results establish the utility of CaSSiDI for single-cell assay analyses and the determination of optimal clustering parameters. Our application of CaSSiDI in this study has revealed previously unknown roles for PirB in myeloid cell populations. In particular, we have discovered homeostatic functions for PirB that are related to Cebpb expression in distinct macrophage subsets.
Collapse
Affiliation(s)
- Robert Nechanitzky
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, ON, Canada.
- Providence Therapeutics Holdings Inc., Calgary, AB, Canada.
| | - Parameswaran Ramachandran
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, ON, Canada
| | - Duygu Nechanitzky
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, ON, Canada
| | - Wanda Y Li
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR, China
| | - Andrew C Wakeham
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, ON, Canada
| | - Jillian Haight
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, ON, Canada
| | - Mary E Saunders
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, ON, Canada
| | - Slava Epelman
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ted Rogers Centre for Heart Research, Translational Biology and Engineering Program, Toronto, ON, Canada
- Peter Munk Cardiac Centre, UHN, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Departments of Immunology and Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Tak W Mak
- Princess Margaret Cancer Centre, Ontario Cancer Institute, University Health Network, Toronto, ON, Canada.
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR, China.
- Department of Pathology Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
55
|
Weng C, Yu F, Yang D, Poeschla M, Liggett LA, Jones MG, Qiu X, Wahlster L, Caulier A, Hussmann JA, Schnell A, Yost KE, Koblan LW, Martin-Rufino JD, Min J, Hammond A, Ssozi D, Bueno R, Mallidi H, Kreso A, Escabi J, Rideout WM, Jacks T, Hormoz S, van Galen P, Weissman JS, Sankaran VG. Deciphering cell states and genealogies of human haematopoiesis. Nature 2024; 627:389-398. [PMID: 38253266 PMCID: PMC10937407 DOI: 10.1038/s41586-024-07066-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/12/2024] [Indexed: 01/24/2024]
Abstract
The human blood system is maintained through the differentiation and massive amplification of a limited number of long-lived haematopoietic stem cells (HSCs)1. Perturbations to this process underlie diverse diseases, but the clonal contributions to human haematopoiesis and how this changes with age remain incompletely understood. Although recent insights have emerged from barcoding studies in model systems2-5, simultaneous detection of cell states and phylogenies from natural barcodes in humans remains challenging. Here we introduce an improved, single-cell lineage-tracing system based on deep detection of naturally occurring mitochondrial DNA mutations with simultaneous readout of transcriptional states and chromatin accessibility. We use this system to define the clonal architecture of HSCs and map the physiological state and output of clones. We uncover functional heterogeneity in HSC clones, which is stable over months and manifests as both differences in total HSC output and biases towards the production of different mature cell types. We also find that the diversity of HSC clones decreases markedly with age, leading to an oligoclonal structure with multiple distinct clonal expansions. Our study thus provides a clonally resolved and cell-state-aware atlas of human haematopoiesis at single-cell resolution, showing an unappreciated functional diversity of human HSC clones and, more broadly, paving the way for refined studies of clonal dynamics across a range of tissues in human health and disease.
Collapse
Affiliation(s)
- Chen Weng
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biology and Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Fulong Yu
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, P.R. China
| | - Dian Yang
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology and Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Molecular Pharmacology and Therapeutics, Department of Systems Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Michael Poeschla
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - L Alexander Liggett
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Matthew G Jones
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology and Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Dermatology, Stanford University, Stanford, CA, USA
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA
| | - Xiaojie Qiu
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology and Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Genetics and Computer Science, BASE Research Initiative, Betty Irene Moore Children's Heart Center, Stanford University, Stanford, CA, USA
| | - Lara Wahlster
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alexis Caulier
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jeffrey A Hussmann
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology and Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alexandra Schnell
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology and Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kathryn E Yost
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology and Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Luke W Koblan
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology and Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jorge D Martin-Rufino
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joseph Min
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology and Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alessandro Hammond
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Daniel Ssozi
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Hematology, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Raphael Bueno
- Division of Thoracic and Cardiac Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Hari Mallidi
- Division of Thoracic and Cardiac Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Antonia Kreso
- Division of Cardiac Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Javier Escabi
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - William M Rideout
- Koch Institute For Integrative Cancer Research at MIT, MIT, Cambridge, MA, USA
| | - Tyler Jacks
- Koch Institute For Integrative Cancer Research at MIT, MIT, Cambridge, MA, USA
| | - Sahand Hormoz
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Peter van Galen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Hematology, Brigham and Women's Hospital, Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA, USA
| | - Jonathan S Weissman
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.
- Department of Biology and Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Koch Institute For Integrative Cancer Research at MIT, MIT, Cambridge, MA, USA.
| | - Vijay G Sankaran
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
56
|
Lee S, Zhan H. Deciphering the differential impact of thrombopoietin/MPL signaling on hematopoietic stem/progenitor cell function in bone marrow and spleen. Stem Cell Reports 2024; 19:211-223. [PMID: 38215758 PMCID: PMC10874852 DOI: 10.1016/j.stemcr.2023.12.004] [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: 04/27/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 01/14/2024] Open
Abstract
Thrombopoietin (TPO) and its receptor MPL play crucial roles in hematopoietic stem cell (HSC) function and platelet production. However, the precise effects of TPO/MPL signaling on HSC regulation in different hematopoietic niches remain unclear. Here, we investigated the effects of TPO/MPL ablation on marrow and splenic hematopoiesis in TPO-/- and MPL-/- mice during aging. Despite severe thrombocytopenia, TPO-/- and MPL-/- mice did not develop marrow failure during a 2-year follow-up. Marrow and splenic HSCs exhibited different responses to TPO/MPL ablation and exogenous TPO treatment. Splenic niche cells compensated for marrow HSC loss in TPO-/- and MPL-/- mice by upregulating CXCL12 levels. These findings provide new insights into the complex regulation of HSCs by TPO/MPL and reveal a previously unknown link between TPO and CXCL12, two key growth factors for HSC maintenance. Understanding the distinct regulatory mechanisms between marrow and spleen hematopoiesis will help to develop novel therapeutic approaches for hematopoietic disorders.
Collapse
Affiliation(s)
- Sandy Lee
- Graduate Program in Molecular & Cellular Pharmacology, Stony Brook University, Stony Brook, NY, USA
| | - Huichun Zhan
- Department of Medicine, Stony Brook School of Medicine, Stony Brook, NY, USA; Medical Service, Northport VA Medical Center, Northport, NY, USA.
| |
Collapse
|
57
|
Wang H, Yan H, Chen W, Tang H, Pei Y, Shan Q, Cang J, Miao C, Tan L, Tan L. Association of clonal haematopoiesis with severe postoperative complications in patients undergoing radical oesophagectomy. Br J Anaesth 2024; 132:277-284. [PMID: 38044238 DOI: 10.1016/j.bja.2023.10.035] [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: 01/02/2023] [Revised: 10/11/2023] [Accepted: 10/15/2023] [Indexed: 12/05/2023] Open
Abstract
BACKGROUND Clonal haematopoiesis (CH) is an age-associated clonal expansion of blood cells driven by leukaemia-associated somatic mutations. Although CH has been reported to be a risk factor for leukaemia and a number of non-haematopoietic diseases, its role in perioperative medicine remains unexplored. METHODS This was a single-centre, prospective, observational study. Patients undergoing radical oesophagectomy were enrolled, and peripheral blood samples were collected for DNA sequencing. Patients with haematopoietic somatic mutations (variant allele frequencies ≥1%) in the DNMT3A gene, TET2 gene, or both were defined as CH carriers. The primary outcome was the incidence of severe postoperative complications (Clavien-Dindo classification ≥3). The secondary outcomes included the major types of postoperative complications, mortality, and other common perioperative variables. RESULTS Clonal haematopoiesis was found in 21.2% (33/156) of the patients (mean age: 66 yr [range: 26-79 yr]; 83% males). Some 14/33 (42.4%) patients with CH had severe postoperative complications, compared with patients without CH carriers (28/123 [22.8%]; P=0.024). Multivariable logistic regression analysis showed that CH was associated with an increased risk of developing severe postoperative complications (odds ratio, 3.63; 95% confidence interval, 1.37-9.66; P=0.010). Among the major postoperative complications, the incidence of pulmonary complications was significantly higher in the patients with CH than in those without CH (15 in 33 [45.5%] vs 30 in 123 [24.4%], P=0.018). CONCLUSIONS Clonal haematopoiesis was associated with a higher incidence of severe postoperative complications in patients undergoing radical oesophagectomy, suggesting that clonal haematopoiesis can play an important role in perioperative medicine. CLINICAL TRIAL REGISTRATION ChiCTR2100044175 (Chinese Clinical Trial Registry, http://www.chictr.org.cn/showproj.aspx?proj=123193).
Collapse
Affiliation(s)
- Hao Wang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Huan Yan
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wannan Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Han Tang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanzi Pei
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Centre, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Qi Shan
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Cang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lijie Tan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Li Tan
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Centre, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
58
|
Stanger BZ, Wahl GM. Cancer as a Disease of Development Gone Awry. ANNUAL REVIEW OF PATHOLOGY 2024; 19:397-421. [PMID: 37832945 PMCID: PMC11486542 DOI: 10.1146/annurev-pathmechdis-031621-025610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2023]
Abstract
In the 160 years since Rudolf Virchow first postulated that neoplasia arises by the same law that regulates embryonic development, scientists have come to recognize the striking overlap between the molecular and cellular programs used by cancers and embryos. Advances in cancer biology and molecular techniques have further highlighted the similarities between carcinogenesis and embryogenesis, where cellular growth, differentiation, motility, and intercellular cross talk are mediated by common drivers and regulatory networks. This review highlights the many connections linking cancer biology and developmental biology to provide a deeper understanding of how a tissue's developmental history may both enable and constrain cancer cell evolution.
Collapse
Affiliation(s)
- Ben Z Stanger
- Division of Gastroenterology, Department of Medicine, Abramson Family Cancer Research Institute, and Department of Cell and Developmental Biology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA;
| | - Geoffrey M Wahl
- Gene Expression Laboratory, The Salk Institute for Biological Studies, La Jolla, California, USA;
| |
Collapse
|
59
|
Feng J, Jang G, Esteva E, Adams NM, Jin H, Reizis B. Clonal barcoding of endogenous adult hematopoietic stem cells reveals a spectrum of lineage contributions. Proc Natl Acad Sci U S A 2024; 121:e2317929121. [PMID: 38227649 PMCID: PMC10823160 DOI: 10.1073/pnas.2317929121] [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: 11/01/2023] [Accepted: 12/11/2023] [Indexed: 01/18/2024] Open
Abstract
The hierarchical model of hematopoiesis posits that self-renewing, multipotent hematopoietic stem cells (HSCs) give rise to all blood cell lineages. While this model accounts for hematopoiesis in transplant settings, its applicability to steady-state hematopoiesis remains to be clarified. Here, we used inducible clonal DNA barcoding of endogenous adult HSCs to trace their contribution to major hematopoietic cell lineages in unmanipulated animals. While the majority of barcodes were unique to a single lineage, we also observed frequent barcode sharing between multiple lineages, specifically between lymphocytes and myeloid cells. These results suggest that both single-lineage and multilineage contributions by HSCs collectively drive continuous hematopoiesis, and highlight a close relationship of myeloid and lymphoid development.
Collapse
Affiliation(s)
- Jue Feng
- Department of Pathology, New York University Grossman School of Medicine, New York, NY10016
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY10029
| | - Geunhyo Jang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY10016
| | - Eduardo Esteva
- Department of Pathology, New York University Grossman School of Medicine, New York, NY10016
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, NY10016
| | - Nicholas M. Adams
- Department of Pathology, New York University Grossman School of Medicine, New York, NY10016
| | - Hua Jin
- Department of Pathology, New York University Grossman School of Medicine, New York, NY10016
| | - Boris Reizis
- Department of Pathology, New York University Grossman School of Medicine, New York, NY10016
| |
Collapse
|
60
|
Tseng YJ, Kageyama Y, Murdaugh RL, Kitano A, Kim JH, Hoegenauer KA, Tiessen J, Smith MH, Uryu H, Takahashi K, Martin JF, Samee MAH, Nakada D. Increased iron uptake by splenic hematopoietic stem cells promotes TET2-dependent erythroid regeneration. Nat Commun 2024; 15:538. [PMID: 38225226 PMCID: PMC10789814 DOI: 10.1038/s41467-024-44718-0] [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: 10/01/2021] [Accepted: 01/02/2024] [Indexed: 01/17/2024] Open
Abstract
Hematopoietic stem cells (HSCs) are capable of regenerating the blood system, but the instructive cues that direct HSCs to regenerate particular lineages lost to the injury remain elusive. Here, we show that iron is increasingly taken up by HSCs during anemia and induces erythroid gene expression and regeneration in a Tet2-dependent manner. Lineage tracing of HSCs reveals that HSCs respond to hemolytic anemia by increasing erythroid output. The number of HSCs in the spleen, but not bone marrow, increases upon anemia and these HSCs exhibit enhanced proliferation, erythroid differentiation, iron uptake, and TET2 protein expression. Increased iron in HSCs promotes DNA demethylation and expression of erythroid genes. Suppressing iron uptake or TET2 expression impairs erythroid genes expression and erythroid differentiation of HSCs; iron supplementation, however, augments these processes. These results establish that the physiological level of iron taken up by HSCs has an instructive role in promoting erythroid-biased differentiation of HSCs.
Collapse
Affiliation(s)
- Yu-Jung Tseng
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yuki Kageyama
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Rebecca L Murdaugh
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ayumi Kitano
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jong Hwan Kim
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kevin A Hoegenauer
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jonathan Tiessen
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Mackenzie H Smith
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Hidetaka Uryu
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Koichi Takahashi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - James F Martin
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
- Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, TX, 77030, USA
| | - Md Abul Hassan Samee
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Daisuke Nakada
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
61
|
Solomon M, Song B, Govindarajah V, Good S, Arasu A, Hinton EB, Thakkar K, Bartram J, Filippi MD, Cancelas JA, Salomonis N, Grimes HL, Reynaud D. Slow cycling and durable Flt3+ progenitors contribute to hematopoiesis under native conditions. J Exp Med 2024; 221:e20231035. [PMID: 37910046 PMCID: PMC10620607 DOI: 10.1084/jem.20231035] [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: 06/16/2023] [Revised: 09/18/2023] [Accepted: 10/13/2023] [Indexed: 11/03/2023] Open
Abstract
The dynamics of the hematopoietic flux responsible for blood cell production in native conditions remains a matter of debate. Using CITE-seq analyses, we uncovered a distinct progenitor population that displays a cell cycle gene signature similar to the one found in quiescent hematopoietic stem cells. We further determined that the CD62L marker can be used to phenotypically enrich this population in the Flt3+ multipotent progenitor (MPP4) compartment. Functional in vitro and in vivo analyses validated the heterogeneity of the MPP4 compartment and established the quiescent/slow-cycling properties of the CD62L- MPP4 cells. Furthermore, studies under native conditions revealed a novel hierarchical organization of the MPP compartments in which quiescent/slow-cycling MPP4 cells sustain a prolonged hematopoietic activity at steady-state while giving rise to other lineage-biased MPP populations. Altogether, our data characterize a durable and productive quiescent/slow-cycling hematopoietic intermediary within the MPP4 compartment and highlight early paths of progenitor differentiation during unperturbed hematopoiesis.
Collapse
Affiliation(s)
- Michael Solomon
- Division of Experimental Hematology and Cancer Biology, Stem Cell Program, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Baobao Song
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Immunology Graduate Program, University of Cincinnati, Cincinnati, OH, USA
| | - Vinothini Govindarajah
- Division of Experimental Hematology and Cancer Biology, Stem Cell Program, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Samantha Good
- Division of Experimental Hematology and Cancer Biology, Stem Cell Program, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Ashok Arasu
- Division of Experimental Hematology and Cancer Biology, Stem Cell Program, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - E. Broderick Hinton
- Division of Experimental Hematology and Cancer Biology, Stem Cell Program, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Kairavee Thakkar
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - James Bartram
- Division of Experimental Hematology and Cancer Biology, Stem Cell Program, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Marie-Dominique Filippi
- Division of Experimental Hematology and Cancer Biology, Stem Cell Program, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jose A. Cancelas
- Division of Experimental Hematology and Cancer Biology, Stem Cell Program, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Nathan Salomonis
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - H. Leighton Grimes
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Damien Reynaud
- Division of Experimental Hematology and Cancer Biology, Stem Cell Program, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
62
|
Zhang Y, Liu F. The evolving views of hematopoiesis: from embryo to adulthood and from in vivo to in vitro. J Genet Genomics 2024; 51:3-15. [PMID: 37734711 DOI: 10.1016/j.jgg.2023.09.005] [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: 08/23/2023] [Revised: 09/13/2023] [Accepted: 09/13/2023] [Indexed: 09/23/2023]
Abstract
The hematopoietic system composed of hematopoietic stem and progenitor cells (HSPCs) and their differentiated lineages serves as an ideal model to uncover generic principles of cell fate transitions. From gastrulation onwards, there successively emerge primitive hematopoiesis (that produces specialized hematopoietic cells), pro-definitive hematopoiesis (that produces lineage-restricted progenitor cells), and definitive hematopoiesis (that produces multipotent HSPCs). These nascent lineages develop in several transient hematopoietic sites and finally colonize into lifelong hematopoietic sites. The development and maintenance of hematopoietic lineages are orchestrated by cell-intrinsic gene regulatory networks and cell-extrinsic microenvironmental cues. Owing to the progressive methodology (e.g., high-throughput lineage tracing and single-cell functional and omics analyses), our understanding of the developmental origin of hematopoietic lineages and functional properties of certain hematopoietic organs has been updated; meanwhile, new paradigms to characterize rare cell types, cell heterogeneity and its causes, and comprehensive regulatory landscapes have been provided. Here, we review the evolving views of HSPC biology during developmental and postnatal hematopoiesis. Moreover, we discuss recent advances in the in vitro induction and expansion of HSPCs, with a focus on the implications for clinical applications.
Collapse
Affiliation(s)
- Yifan Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China
| | - Feng Liu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, Shandong 266237, China; State Key Laboratory of Membrane Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
63
|
Fath MA, Liu D, Ewald JT, Robles-Planells C, Tomanek-Chalkley AM, Graves SA, Howe JR, O’Dorisio TM, Rastogi P, Bellizzi AM, Sue O’Dorisio M, Menda Y, Spitz DR. Chemokine Receptor CXCR4 Radioligand Targeted Therapy Using 177Lutetium-pentixather for Pulmonary Neuroendocrine Cancers. Radiat Res 2024; 201:35-47. [PMID: 37989124 PMCID: PMC10896455 DOI: 10.1667/rade-23-00064.1] [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: 04/07/2023] [Accepted: 10/23/2023] [Indexed: 11/23/2023]
Abstract
Intermediate to high-grade lung neuroendocrine tumors (NETs; i.e., atypical carcinoid tumors) and neuroendocrine carcinomas (NECs) are currently difficult to cure. These tumors were found to express the CXCR4 G-protein coupled receptor that can be targeted with radioligands. PCR and flow cytometric analysis of lung NET and NEC cell lines using an anti-CXCR4 antibody demonstrated that all cell lines tested expressed CXCR4. PET/CT imaging with 68Galium-pentixafor in mouse xenografts of NETs and NECs verified tumor targeting that was blocked by a CXCR4 agonist. Clonogenic survival analysis demonstrated a more than additive enhancement of killing when 1 μM auranofin (a thioredoxin reductase inhibitor) was used as a radiosensitizer in combination with 177Lu-pentixather (10 μCi). DMS273 small cell lung cancer xenografts in female nude mice treated with 25 μCi/g 177Lu-pentixather induced inhibition of tumor growth and resulted in an increase in overall survival without causing unacceptable normal tissue toxicities. Immunohistochemical staining of 95 retrospective human samples (containing 90 small cell lung carcinomas) demonstrated 84% CXCR4 positivity. In a multivariable analysis of this cohort that included age, gender, stage, primary site, SSTR2 status, and CXCR4 status, Cox regression models determined that only distant metastasis at presentation (P < 0.01) and a CXCR4 H-score >30 (P = 0.04) were significantly associated with reduced survival. Prospective clinical testing of patient tumors identified CXCR4-positivity in 76% of 21 NECs, 67% of 15 lung NETs (including 8 of 10 atypical carcinoids), and 0% of 25 non-lung NETs (including 5 NETS G3s). These data support the hypothesis that CXCR4-targeted theranostics can be utilized effectively for select NETs and NECs.
Collapse
Affiliation(s)
- Melissa A. Fath
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - Dijie Liu
- Department of Pediatrics, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - Jordan T. Ewald
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - Claudia Robles-Planells
- Department of Pediatrics, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - Ann M. Tomanek-Chalkley
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - Stephen A. Graves
- Department of Radiology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - James R. Howe
- Department of Surgery, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - Thomas M. O’Dorisio
- Department of Internal Medicine, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - Prerna Rastogi
- Department of Pathology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - Andrew M. Bellizzi
- Department of Pathology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - M Sue O’Dorisio
- Department of Pediatrics, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - Yusuf Menda
- Department of Radiology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| | - Douglas R. Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52240
| |
Collapse
|
64
|
Zhang YW, Schönberger K, Cabezas‐Wallscheid N. Bidirectional interplay between metabolism and epigenetics in hematopoietic stem cells and leukemia. EMBO J 2023; 42:e112348. [PMID: 38010205 PMCID: PMC10711668 DOI: 10.15252/embj.2022112348] [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: 08/11/2022] [Revised: 08/24/2023] [Accepted: 08/28/2023] [Indexed: 11/29/2023] Open
Abstract
During the last decades, remarkable progress has been made in further understanding the complex molecular regulatory networks that maintain hematopoietic stem cell (HSC) function. Cellular and organismal metabolisms have been shown to directly instruct epigenetic alterations, and thereby dictate stem cell fate, in the bone marrow. Epigenetic regulatory enzymes are dependent on the availability of metabolites to facilitate DNA- and histone-modifying reactions. The metabolic and epigenetic features of HSCs and their downstream progenitors can be significantly altered by environmental perturbations, dietary habits, and hematological diseases. Therefore, understanding metabolic and epigenetic mechanisms that regulate healthy HSCs can contribute to the discovery of novel metabolic therapeutic targets that specifically eliminate leukemia stem cells while sparing healthy HSCs. Here, we provide an in-depth review of the metabolic and epigenetic interplay regulating hematopoietic stem cell fate. We discuss the influence of metabolic stress stimuli, as well as alterations occurring during leukemic development. Additionally, we highlight recent therapeutic advancements toward eradicating acute myeloid leukemia cells by intervening in metabolic and epigenetic pathways.
Collapse
Affiliation(s)
- Yu Wei Zhang
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | | | | |
Collapse
|
65
|
Schönberger K, Cabezas-Wallscheid N. How nutrition regulates hematopoietic stem cell features. Exp Hematol 2023; 128:10-18. [PMID: 37816445 DOI: 10.1016/j.exphem.2023.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/12/2023]
Abstract
Our dietary choices significantly impact all the cells in our body. Increasing evidence suggests that diet-derived metabolites influence hematopoietic stem cell (HSC) metabolism and function, thereby actively modulating blood homeostasis. This is of particular relevance because regulating the metabolic activity of HSCs is crucial for maintaining stem cell fitness and mitigating the risk of hematologic disorders. In this review, we examine the current scientific knowledge of the impact of diet on stemness features, and we specifically highlight the established mechanisms by which dietary components modulate metabolic and transcriptional programs in adult HSCs. Gaining a deeper understanding of how nutrition influences our HSC compartment may pave the way for targeted dietary interventions with the potential to decelerate aging and improve the effectiveness of transplantation and cancer therapies.
Collapse
|
66
|
Cunningham KT, Mills KHG. Modulation of haematopoiesis by protozoal and helminth parasites. Parasite Immunol 2023; 45:e12975. [PMID: 36797216 PMCID: PMC10909493 DOI: 10.1111/pim.12975] [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: 12/13/2022] [Revised: 02/07/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023]
Abstract
During inflammation, haematopoietic stem cells (HSCs) in the bone marrow (BM) and periphery rapidly expand and preferentially differentiate into myeloid cells that mediate innate immune responses. HSCs can be directed into quiescence or differentiation by sensing alterations to the haematopoietic niche, including cytokines, chemokines, and pathogen-derived products. Most studies attempting to identify the mechanisms of haematopoiesis have focused on bacterial and viral infections. From intracellular protozoan infections to large multicellular worms, parasites are a global health burden and represent major immunological challenges that remain poorly defined in the context of haematopoiesis. Immune responses to parasites vary drastically, and parasites have developed sophisticated immunomodulatory mechanisms that allow development of chronic infections. Recent advances in imaging, genomic sequencing, and mouse models have shed new light on how parasites induce unique forms of emergency haematopoiesis. In addition, parasites can modify the haematopoiesis in the BM and periphery to improve their survival in the host. Parasites can also induce long-lasting modifications to HSCs, altering future immune responses to infection, inflammation or transplantation, a term sometimes referred to as central trained immunity. In this review, we highlight the current understanding of parasite-induced haematopoiesis and how parasites target this process to promote chronic infections.
Collapse
Affiliation(s)
- Kyle T. Cunningham
- Wellcome Centre for Integrative ParasitologyInstitute of Infection and Immunity, University of GlasgowGlasgowUK
| | - Kingston H. G. Mills
- Immune Regulation Research GroupTrinity Biomedical Sciences Institute, Trinity College DublinDublinIreland
| |
Collapse
|
67
|
Shetty S, Duesman SJ, Patel S, Huyhn P, Shroff S, Das A, Chowhan D, Sebra R, Beaumont K, McAlpine CS, Rajbhandari P, Rajbhandari AK. Sexually dimorphic role of diet and stress on behavior, energy metabolism, and the ventromedial hypothalamus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.17.567534. [PMID: 38014350 PMCID: PMC10680837 DOI: 10.1101/2023.11.17.567534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Scientific evidence underscores the influence of biological sex on the interplay between stress and metabolic dysfunctions. However, there is limited understanding of how diet and stress jointly contribute to metabolic dysregulation in both males and females. To address this gap, our study aimed to investigate the combined effects of a high-fat diet (HFD) and repeated footshock stress on fear-related behaviors and metabolic outcomes in male and female mice. Using a robust rodent model that recapitulates key aspects of post-traumatic stress disorder (PTSD), we subjected mice to footshock stressor followed by weekly reminder footshock stressor or no stressor for 14 weeks while on either an HFD or chow diet. Our findings revealed that HFD impaired fear memory extinction in male mice that received initial stressor but not in female mice. Blood glucose levels were influenced by both diet and sex, with HFD-fed female mice displaying elevated levels that returned to baseline in the absence of stress, a pattern not observed in male mice. Male mice on HFD exhibited higher energy expenditure, while HFD-fed female mice showed a decreased respiratory exchange ratio (RER). Sex-specific alterations in pro-inflammatory markers and abundance of hematopoietic stem cells were observed in chronically stressed mice on an HFD in different peripheral tissues, indicating the manifestation of distinct comorbid disorders. Single-nuclei RNA sequencing of the ventromedial hypothalamus from stressed mice on an HFD provided insights into sex-specific glial cell activation and cell-type-specific transcriptomic changes. In conclusion, our study offers a comprehensive understanding of the intricate interactions between stress, diet, sex, and various physiological and behavioral outcomes, shedding light on a potential brain region coordinating these interactions.
Collapse
Affiliation(s)
- Sanutha Shetty
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Samuel J. Duesman
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Sanil Patel
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Pacific Huyhn
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Sanjana Shroff
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anika Das
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, NY, New York 10029
- Center for Excellence in Youth Education, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Disha Chowhan
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Sebra
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristin Beaumont
- Center for Advanced Genomic Technology, Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Cameron S. McAlpine
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, NY, New York 10029
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, NY, New York 10029
| | - Prashant Rajbhandari
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, NY, New York 10029
- Disease Mechanism and Therapeutics Program, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Senior authors
| | - Abha K. Rajbhandari
- Department of Neuroscience and Psychiatry, Icahn School of Medicine at Mount Sinai, NY, New York 10029
- Senior authors
| |
Collapse
|
68
|
Li L, Bowling S, McGeary SE, Yu Q, Lemke B, Alcedo K, Jia Y, Liu X, Ferreira M, Klein AM, Wang SW, Camargo FD. A mouse model with high clonal barcode diversity for joint lineage, transcriptomic, and epigenomic profiling in single cells. Cell 2023; 186:5183-5199.e22. [PMID: 37852258 DOI: 10.1016/j.cell.2023.09.019] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 07/11/2023] [Accepted: 09/19/2023] [Indexed: 10/20/2023]
Abstract
Cellular lineage histories and their molecular states encode fundamental principles of tissue development and homeostasis. Current lineage-recording mouse models have insufficient barcode diversity and single-cell lineage coverage for profiling tissues composed of millions of cells. Here, we developed DARLIN, an inducible Cas9 barcoding mouse line that utilizes terminal deoxynucleotidyl transferase (TdT) and 30 CRISPR target sites. DARLIN is inducible, generates massive lineage barcodes across tissues, and enables the detection of edited barcodes in ∼70% of profiled single cells. Using DARLIN, we examined fate bias within developing hematopoietic stem cells (HSCs) and revealed unique features of HSC migration. Additionally, we established a protocol for joint transcriptomic and epigenomic single-cell measurements with DARLIN and found that cellular clonal memory is associated with genome-wide DNA methylation rather than gene expression or chromatin accessibility. DARLIN will enable the high-resolution study of lineage relationships and their molecular signatures in diverse tissues and physiological contexts.
Collapse
Affiliation(s)
- Li Li
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Sarah Bowling
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Sean E McGeary
- Department of Systems Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Qi Yu
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Bianca Lemke
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Karel Alcedo
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Yuemeng Jia
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Xugeng Liu
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Mark Ferreira
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Allon M Klein
- Department of Systems Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Shou-Wen Wang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; School of Science, Westlake University, Hangzhou, Zhejiang 310024, China.
| | - Fernando D Camargo
- Stem Cell Program, Boston Children's Hospital, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
69
|
Rydström A, Grahn THM, Niroula A, Mansell E, van der Garde M, Pertesi M, Subramaniam A, Soneji S, Zubarev R, Enver T, Nilsson B, Miharada K, Larsson J, Karlsson S. Functional and molecular profiling of hematopoietic stem cells during regeneration. Exp Hematol 2023; 127:40-51. [PMID: 37666355 DOI: 10.1016/j.exphem.2023.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/06/2023]
Abstract
Hematopoietic stem cells (HSCs) enable hematopoietic stem cell transplantation (HCT) through their ability to replenish the entire blood system. Proliferation of HSCs is linked to decreased reconstitution potential, and a precise regulation of actively dividing HSCs is thus essential to ensure long-term functionality. This regulation becomes important in the transplantation setting where HSCs undergo proliferation followed by a gradual transition to quiescence and homeostasis. Although mouse HSCs have been well studied under homeostatic conditions, the mechanisms regulating HSC activation under stress remain unclear. Here, we analyzed the different phases of regeneration after transplantation. We isolated bone marrow from mice at 8 time points after transplantation and examined the reconstitution dynamics and transcriptional profiles of stem and progenitor populations. We found that regenerating HSCs initially produced rapidly expanding progenitors and displayed distinct changes in fatty acid metabolism and glycolysis. Moreover, we observed molecular changes in cell cycle, MYC and mTOR signaling in both HSCs, and progenitor subsets. We used a decay rate model to fit the temporal transcription profiles of regenerating HSCs and identified genes with progressively decreased or increased expression after transplantation. These genes overlapped to a large extent with published gene sets associated with key aspects of HSC function, demonstrating the potential of this data set as a resource for identification of novel HSC regulators. Taken together, our study provides a detailed functional and molecular characterization of HSCs at different phases of regeneration and identifies a gene set associated with the transition from proliferation to quiescence.
Collapse
Affiliation(s)
- Anna Rydström
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Tan H M Grahn
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Abhishek Niroula
- Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Els Mansell
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Mark van der Garde
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Maroulio Pertesi
- Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | | | - Shamit Soneji
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Roman Zubarev
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solna, Sweden
| | - Tariq Enver
- Stem Cell Group, Cancer Institute, University College London, United Kingdom
| | - Björn Nilsson
- Hematology and Transfusion Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Kenichi Miharada
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Jonas Larsson
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Stefan Karlsson
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden.
| |
Collapse
|
70
|
Takeishi S, Marchand T, Koba WR, Borger DK, Xu C, Guha C, Bergman A, Frenette PS, Gritsman K, Steidl U. Haematopoietic stem cell numbers are not solely determined by niche availability. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.28.564559. [PMID: 37961493 PMCID: PMC10634881 DOI: 10.1101/2023.10.28.564559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Haematopoietic stem cells (HSCs) reside in specialized microenvironments, also referred to as niches, and it has been widely believed that HSC numbers are determined by the niche size alone 1-5 . However, the vast excess of the number of niche cells over that of HSCs raises questions about this model. We initially established a mathematical model of niche availability and occupancy, which predicted that HSC numbers are restricted at both systemic and local levels. To address this question experimentally, we developed a femoral bone transplantation system, enabling us to increase the number of available HSC niches. We found that the addition of niches does not alter total HSC numbers in the body, regardless of whether the endogenous (host) niche is intact or defective, suggesting that HSC numbers are limited at the systemic level. Additionally, HSC numbers in transplanted wild-type femurs did not increase beyond physiological levels when HSCs were mobilized from defective endogenous niches to the periphery, indicating that HSC numbers are also constrained at the local level. Our study demonstrates that HSC numbers are not solely determined by niche availability, thereby rewriting the long-standing model for the regulation of HSC numbers.
Collapse
|
71
|
Cochran J, Yura Y, Thel MC, Doviak H, Polizio AH, Arai Y, Arai Y, Horitani K, Park E, Chavkin NW, Kour A, Sano S, Mahajan N, Evans M, Huba M, Naya NM, Sun H, Ban Y, Hirschi KK, Toldo S, Abbate A, Druley TE, Ruberg FL, Maurer MS, Ezekowitz JA, Dyck JR, Walsh K. Clonal Hematopoiesis in Clinical and Experimental Heart Failure With Preserved Ejection Fraction. Circulation 2023; 148:1165-1178. [PMID: 37681311 PMCID: PMC10575571 DOI: 10.1161/circulationaha.123.064170] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 08/07/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Clonal hematopoiesis (CH), which results from an array of nonmalignant driver gene mutations, can lead to altered immune cell function and chronic disease, and has been associated with worse outcomes in patients with heart failure (HF) with reduced ejection fraction. However, the role of CH in the prognosis of HF with preserved ejection fraction (HFpEF) has been understudied. This study aimed to characterize CH in patients with HFpEF and elucidate its causal role in a murine model. METHODS Using a panel of 20 candidate CH driver genes and a variant allele fraction cutoff of 0.5%, ultradeep error-corrected sequencing identified CH in a cohort of 81 patients with HFpEF (mean age, 71±6 years; ejection fraction, 63±5%) and 36 controls without a diagnosis of HFpEF (mean age, 74±7 years; ejection fraction, 61.5±8%). CH was also evaluated in a replication cohort of 59 individuals with HFpEF. RESULTS Compared with controls, there was an enrichment of TET2-mediated CH in the HFpEF patient cohort (12% versus 0%, respectively; P=0.02). In the HFpEF cohort, patients with CH exhibited exacerbated diastolic dysfunction in terms of E/e' (14.9 versus 11.7, respectively; P=0.0096) and E/A (1.69 versus 0.89, respectively; P=0.0206) compared with those without CH. The association of CH with exacerbated diastolic dysfunction was corroborated in a validation cohort of individuals with HFpEF. In accordance, patients with HFpEF, an age ≥70 years, and CH exhibited worse prognosis in terms of 5-year cardiovascular-related hospitalization rate (hazard ratio, 5.06; P=0.042) compared with patients with HFpEF and an age ≥70 years without CH. To investigate the causal role of CH in HFpEF, nonconditioned mice underwent adoptive transfer with Tet2-wild-type or Tet2-deficient bone marrow and were subsequently subjected to a high-fat diet/L-NAME (Nω-nitro-l-arginine methyl ester) combination treatment to induce features of HFpEF. This model of Tet2-CH exacerbated cardiac hypertrophy by heart weight/tibia length and cardiomyocyte size, diastolic dysfunction by E/e' and left ventricular end-diastolic pressure, and cardiac fibrosis compared with the Tet2-wild-type condition. CONCLUSIONS CH is associated with worse heart function and prognosis in patients with HFpEF, and a murine experimental model of Tet2-mediated CH displays greater features of HFpEF.
Collapse
Affiliation(s)
- Jesse Cochran
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
- Medical Scientist Training Program, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Yoshimitsu Yura
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
- Current address: Department of Cardiovascular Medicine, Nagoya University School of Medicine, Nagoya 466-8550, Japan
| | - Mark C. Thel
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Heather Doviak
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Ariel H. Polizio
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Yuka Arai
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Yohei Arai
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Keita Horitani
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
- Current address: Department of Internal Medicine II, Kansai Medical University, Osaka 573-1010, Japan
| | - Eunbee Park
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Nicholas W. Chavkin
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Anupreet Kour
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Soichi Sano
- Laboratory of Cardiovascular Mosaicism, National Cerebral and Cardiovascular Center, Osaka 564-8565, Japan
| | | | - Megan Evans
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Mahalia Huba
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | | | - Hanna Sun
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Youngho Ban
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Karen K. Hirschi
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Stefano Toldo
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | | | - Frederick L. Ruberg
- Section of Cardiovascular Medicine, Department of Medicine and Amyloidosis Center, Boston University Chobanian & Avedisian School of Medicine/Boston Medical Center, Boston, MA 02118, USA
| | - Mathew S. Maurer
- Seymour, Paul, and Gloria Milstein Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Justin A. Ezekowitz
- Alberta Heart Failure Etiology and Analysis Research Team (HEART) project
- Department of Medicine, Division of Cardiology, University of Alberta, Edmonton, Alberta, T6G 2R3, Canada
| | - Jason R.B. Dyck
- Alberta Heart Failure Etiology and Analysis Research Team (HEART) project
- Cardiovascular Research Centre, Department of Pediatrics, University of Alberta, Edmonton, Alberta, T6G 2S2, Canada
| | - Kenneth Walsh
- Robert M. Berne Cardiovascular Research Center, Division of Cardiovascular Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| |
Collapse
|
72
|
Huang AH, Galloway JL. Current and emerging technologies for defining and validating tendon cell fate. J Orthop Res 2023; 41:2082-2092. [PMID: 37211925 DOI: 10.1002/jor.25632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 05/09/2023] [Accepted: 05/18/2023] [Indexed: 05/23/2023]
Abstract
The tendon field has been flourishing in recent years with the advent of new tools and model systems. The recent ORS 2022 Tendon Section Conference brought together researchers from diverse disciplines and backgrounds, showcasing studies in biomechanics and tissue engineering to cell and developmental biology and using models from zebrafish and mouse to humans. This perspective aims to summarize progress in tendon research as it pertains to understanding and studying tendon cell fate. The successful integration of new technologies and approaches have the potential to further propel tendon research into a new renaissance of discovery. However, there are also limitations with the current methodologies that are important to consider when tackling research questions. Altogether, we will highlight recent advances and technologies and propose new avenues to explore tendon biology.
Collapse
Affiliation(s)
- Alice H Huang
- Department of Orthopedic Surgery, Columbia University, New York, New York, USA
| | - Jenna L Galloway
- Department of Orthopaedic Surgery, Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
73
|
Almotiri A, Boyd AS, Rodrigues NP. Zeb1 Regulates the Function of Lympho-Myeloid Primed Progenitors after Transplantation. Biomolecules 2023; 13:1386. [PMID: 37759786 PMCID: PMC10526482 DOI: 10.3390/biom13091386] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/07/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
Zeb1, a zinc finger E-box binding homeobox epithelial-mesenchymal (EMT) transcription factor, acts as a critical regulator of hematopoietic stem cell (HSC) self-renewal and multi-lineage differentiation. Whether Zeb1 directly regulates the function of multi-potent progenitors primed for hematopoietic lineage commitment remains ill defined. By using an inducible Mx-1 Cre conditional mouse model where Zeb1 was genetically engineered to be deficient in the adult hematopoietic system (hereafter Zeb1-/-), we found that the absolute cell number of immunophenotypically defined lympho-myeloid primed progenitors (LMPPs) from Zeb1-/- mice was reduced. Myeloid- and lymphoid-biased HSCs in Zeb1-/- mice were unchanged, implying that defective LMPP generation from Zeb1-/- mice was not directly caused by an imbalance of lineage-biased HSCs. Functional analysis of LMPP from Zeb1-/- mice, as judged by competitive transplantation, revealed an overall reduction in engraftment to hematopoietic organs over 4 weeks, which correlated with minimal T-cell engraftment, reduced B-cell and monocyte/macrophage engraftment, and unperturbed granulocyte engraftment. Thus, Zeb1 regulates LMPP differentiation potential to select lympho-myeloid lineages in the context of transplantation.
Collapse
Affiliation(s)
- Alhomidi Almotiri
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences-Dawadmi, Shaqra University, Dawadmi 17464, Saudi Arabia;
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK
| | - Ashleigh S. Boyd
- Department of Surgical Biotechnology, Division of Surgery and Interventional Science, Royal Free Hospital, University College London, London NW3 2PS, UK;
- Institute of Immunity and Transplantation, University College London, London NW3 2PP, UK
| | - Neil P. Rodrigues
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Hadyn Ellis Building, Cardiff CF24 4HQ, UK
| |
Collapse
|
74
|
Pizzato HA, Wang Y, Wolfgang MJ, Finck BN, Patti GJ, Bhattacharya D. Mitochondrial pyruvate metabolism and glutaminolysis toggle steady-state and emergency myelopoiesis. J Exp Med 2023; 220:e20221373. [PMID: 37249600 PMCID: PMC10227646 DOI: 10.1084/jem.20221373] [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: 08/10/2022] [Revised: 03/23/2023] [Accepted: 05/10/2023] [Indexed: 05/31/2023] Open
Abstract
To define the metabolic requirements of hematopoiesis, we examined blood lineages in mice conditionally deficient in genes required for long-chain fatty acid oxidation (Cpt2), glutaminolysis (Gls), or mitochondrial pyruvate import (Mpc2). Genetic ablation of Cpt2 or Gls minimally impacted most blood lineages. In contrast, deletion of Mpc2 led to a sharp decline in mature myeloid cells and a slower reduction in T cells, whereas other hematopoietic lineages were unaffected. Yet MPC2-deficient monocytes and neutrophils rapidly recovered due to a transient and specific increase in myeloid progenitor proliferation. Competitive bone marrow chimera and stable isotope tracing experiments demonstrated that this proliferative burst was progenitor intrinsic and accompanied by a metabolic switch to glutaminolysis. Myeloid recovery after loss of MPC2 or cyclophosphamide treatment was delayed in the absence of GLS. Reciprocally, MPC2 was not required for myeloid recovery after cyclophosphamide treatment. Thus, mitochondrial pyruvate metabolism maintains myelopoiesis under steady-state conditions, while glutaminolysis in progenitors promotes emergency myelopoiesis.
Collapse
Affiliation(s)
- Hannah A. Pizzato
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Immunobiology, University of Arizona, Tucson, AZ, USA
| | - Yahui Wang
- Department of Chemistry, Washington University, Saint Louis, MO, USA
| | - Michael J. Wolfgang
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Brian N. Finck
- Division of Geriatrics and Nutritional Sciences, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Gary J. Patti
- Department of Chemistry, Washington University, Saint Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
- Siteman Cancer Center, Washington University, Saint Louis, MO, USA
| | - Deepta Bhattacharya
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Immunobiology, University of Arizona, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
75
|
Nizharadze T, Busch K, Fanti AK, Rodewald HR, Höfer T. Differentiation tracing identifies hematopoietic regeneration from multipotent progenitors but not stem cells. Cells Dev 2023; 175:203861. [PMID: 37286105 DOI: 10.1016/j.cdev.2023.203861] [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: 04/12/2023] [Revised: 05/26/2023] [Accepted: 05/31/2023] [Indexed: 06/09/2023]
Abstract
Hematopoietic stem cells (HSCs) and multipotent progenitors (MPPs) generate the immune system in development, and contribute to its maintenance under steady-state conditions. How stem and progenitor cells respond to increased demand for mature cells upon injury is a fundamental question of stem cell biology. Several studies of murine hematopoiesis have reported increased proliferation of HSCs in situ when exposed to inflammatory stimuli, which has been taken as a proxy for increased HSC differentiation. Such surplus generation of HSC may fuel enhanced HSC differentiation or, alternatively, maintain HSC cellularity in the face of increased cell death without enhanced HSC differentiation. This key question calls for direct measurements of HSC differentiation in their natural niches in vivo. Here, we review work that quantifies native HSC differentiation by fate mapping and mathematical inference. Recent differentiation tracing studies show that HSC do not increase their differentiation rate upon a wide range of challenges, including systemic bacterial infection (sepsis), blood loss, and transient or persistent ablation of specific mature immune cells. By contrast, MPPs differentiate more rapidly in response to systemic infection to accelerate the production of myeloid cells. These new in vivo data identify MPPs as a major source of hematopoietic regeneration; HSCs might not contribute to regeneration while remaining protected.
Collapse
Affiliation(s)
- Tamar Nizharadze
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Katrin Busch
- Division of Cellular Immunology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; Faculty of Medicine, Heidelberg University, 69120 Heidelberg, Germany
| | - Ann-Kathrin Fanti
- Division of Cellular Immunology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Thomas Höfer
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
76
|
Kang YA, Paik H, Zhang SY, Chen JJ, Olson OC, Mitchell CA, Collins A, Swann JW, Warr MR, Fan R, Passegué E. Secretory MPP3 reinforce myeloid differentiation trajectory and amplify myeloid cell production. J Exp Med 2023; 220:214059. [PMID: 37115584 PMCID: PMC10140385 DOI: 10.1084/jem.20230088] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/25/2023] [Accepted: 03/30/2023] [Indexed: 04/29/2023] Open
Abstract
Hematopoietic stem cells (HSC) and downstream lineage-biased multipotent progenitors (MPP) tailor blood production and control myelopoiesis on demand. Recent lineage tracing analyses revealed MPPs to be major functional contributors to steady-state hematopoiesis. However, we still lack a precise resolution of myeloid differentiation trajectories and cellular heterogeneity in the MPP compartment. Here, we found that myeloid-biased MPP3 are functionally and molecularly heterogeneous, with a distinct subset of myeloid-primed secretory cells with high endoplasmic reticulum (ER) volume and FcγR expression. We show that FcγR+/ERhigh MPP3 are a transitional population serving as a reservoir for rapid production of granulocyte/macrophage progenitors (GMP), which directly amplify myelopoiesis through inflammation-triggered secretion of cytokines in the local bone marrow (BM) microenvironment. Our results identify a novel regulatory function for a secretory MPP3 subset that controls myeloid differentiation through lineage-priming and cytokine production and acts as a self-reinforcing amplification compartment in inflammatory stress and disease conditions.
Collapse
Affiliation(s)
- Yoon-A Kang
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University , New York, NY, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Department of Medicine, Hem/Onc Division, University of California, San Francisco , San Francisco, CA, USA
| | - Hyojung Paik
- Center for Applied Scientific Computing, Korea Institute of Science and Technology Information, and University of Science and Technology , Daejeon, South Korea
| | - Si Yi Zhang
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Department of Medicine, Hem/Onc Division, University of California, San Francisco , San Francisco, CA, USA
| | - Jonathan J Chen
- Department of Biomedical Engineering, Yale University , New Haven, CT, USA
| | - Oakley C Olson
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University , New York, NY, USA
| | - Carl A Mitchell
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University , New York, NY, USA
| | - Amelie Collins
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University , New York, NY, USA
| | - James W Swann
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University , New York, NY, USA
| | - Matthew R Warr
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Department of Medicine, Hem/Onc Division, University of California, San Francisco , San Francisco, CA, USA
| | - Rong Fan
- Department of Biomedical Engineering, Yale University , New Haven, CT, USA
| | - Emmanuelle Passegué
- Columbia Stem Cell Initiative, Department of Genetics and Development, Columbia University , New York, NY, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Department of Medicine, Hem/Onc Division, University of California, San Francisco , San Francisco, CA, USA
| |
Collapse
|
77
|
Ghersi JJ, Baldissera G, Hintzen J, Luff SA, Cheng S, Xia IF, Sturgeon CM, Nicoli S. Haematopoietic stem and progenitor cell heterogeneity is inherited from the embryonic endothelium. Nat Cell Biol 2023; 25:1135-1145. [PMID: 37460694 PMCID: PMC10415179 DOI: 10.1038/s41556-023-01187-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 06/09/2023] [Indexed: 08/12/2023]
Abstract
Definitive haematopoietic stem and progenitor cells (HSPCs) generate erythroid, lymphoid and myeloid lineages. HSPCs are produced in the embryo via transdifferentiation of haemogenic endothelial cells in the aorta-gonad-mesonephros (AGM). HSPCs in the AGM are heterogeneous in differentiation and proliferative output, but how these intrinsic differences are acquired remains unanswered. Here we discovered that loss of microRNA (miR)-128 in zebrafish leads to an expansion of HSPCs in the AGM with different cell cycle states and a skew towards erythroid and lymphoid progenitors. Manipulating miR-128 in differentiating haemogenic endothelial cells, before their transition to HSPCs, recapitulated the lineage skewing in both zebrafish and human pluripotent stem cells. miR-128 promotes Wnt and Notch signalling in the AGM via post-transcriptional repression of the Wnt inhibitor csnk1a1 and the Notch ligand jag1b. De-repression of cskn1a1 resulted in replicative and erythroid-biased HSPCs, whereas de-repression of jag1b resulted in G2/M and lymphoid-biased HSPCs with long-term consequence on the respective blood lineages. We propose that HSPC heterogeneity arises in the AGM endothelium and is programmed in part by Wnt and Notch signalling.
Collapse
Affiliation(s)
- Joey J Ghersi
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology & Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Gabriel Baldissera
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology & Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Jared Hintzen
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology & Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Stephanie A Luff
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Siyuan Cheng
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology & Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Ivan Fan Xia
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
- Vascular Biology & Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Christopher M Sturgeon
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stefania Nicoli
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT, USA.
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.
- Vascular Biology & Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
78
|
Wang Y, Zhang X, Wang Z. Cellular barcoding: From developmental tracing to anti-tumor drug discovery. Cancer Lett 2023; 567:216281. [PMID: 37336285 DOI: 10.1016/j.canlet.2023.216281] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/31/2023] [Accepted: 06/10/2023] [Indexed: 06/21/2023]
Abstract
Clonal evolution has gained immense attention in explaining cancer cell status, history, and fate during cancer progression. Current single-cell or spatial transcriptome technologies have broadened our understanding of various mechanisms underlying cancer initiation, relapse, and drug resistance. However, technical challenges still hinder a better understanding of the dynamics of distinctive phenotypic states and abnormal trajectories from normal physiological transition to malignant stages. Cellular barcoding enabled lineage tracing on parallelly massive cells at single-cell resolution through different mechanisms lately, enabling new insights into exploring developmental trajectories, cancer progression, and targeted therapies. This review summarizes the latest noteworthy and robust strategies for different types of cellular barcodes. To introduce the major characteristics, advantages and limitations of these different strategies, this review will further guide in choosing or improving cellular barcoding technologies and their applications in cancer research.
Collapse
Affiliation(s)
- Yuqing Wang
- Medical Center of Hematology, The Second Affiliated Hospital, Army Medical University, Chongqing, 40037, China; State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 40037, China
| | - Xi Zhang
- Medical Center of Hematology, The Second Affiliated Hospital, Army Medical University, Chongqing, 40037, China; State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 40037, China; Jinfeng Laboratory, Chongqing, 401329, China.
| | - Zheng Wang
- Medical Center of Hematology, The Second Affiliated Hospital, Army Medical University, Chongqing, 40037, China; State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 40037, China; Bio-Med Informatics Research Center & Clinical Research Center, The Second Affiliated Hospital, Army Medical University, Chongqing, 400037, China; Jinfeng Laboratory, Chongqing, 401329, China.
| |
Collapse
|
79
|
Radtke S, Enstrom M, Pande D, Duke ER, Cardozo-Ojeda EF, Madhu R, Owen S, Kanestrom G, Cui M, Perez AM, Schiffer JT, Kiem HP. Stochastic fate decisions of HSCs after transplantation: early contribution, symmetric expansion, and pool formation. Blood 2023; 142:33-43. [PMID: 36821766 PMCID: PMC10935507 DOI: 10.1182/blood.2022018564] [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: 09/29/2022] [Revised: 02/16/2023] [Accepted: 02/16/2023] [Indexed: 02/25/2023] Open
Abstract
Hematopoietic stem cells (HSCs) are assumed to be rare, infrequently dividing, long-lived cells not involved in immediate recovery after transplantation. Here, we performed unprecedented high-density clonal tracking in nonhuman primates and found long-term persisting HSC clones to actively contribute during early neutrophil recovery, and to be the main source of blood production as early as 50 days after transplantation. Most surprisingly, we observed a rapid decline in the number of unique HSC clones, while persisting HSCs expanded, undergoing symmetric divisions to create identical siblings and formed clonal pools ex vivo as well as in vivo. In contrast to the currently assumed model of hematopoietic reconstitution, we provide evidence for contribution of HSCs in short-term recovery as well as symmetric expansion of individual clones into pools. These findings provide novel insights into HSC biology, informing the design of HSC transplantation and gene therapy studies.
Collapse
Affiliation(s)
- Stefan Radtke
- Stem Cell and Gene Therapy Program, Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Mark Enstrom
- Stem Cell and Gene Therapy Program, Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Dnyanada Pande
- Stem Cell and Gene Therapy Program, Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Elizabeth R. Duke
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | | | - Ravishankar Madhu
- Stem Cell and Gene Therapy Program, Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Staci Owen
- Stem Cell and Gene Therapy Program, Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Greta Kanestrom
- Stem Cell and Gene Therapy Program, Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Margaret Cui
- Stem Cell and Gene Therapy Program, Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Anai M. Perez
- Stem Cell and Gene Therapy Program, Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Joshua T. Schiffer
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Hans-Peter Kiem
- Stem Cell and Gene Therapy Program, Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
- Department of Pathology, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
80
|
Boyd AL, Lu J, Hollands CG, Alsostovar L, Murali S, Reid JC, Ye W, Vandersluis S, Johnson P, ElRafie A, Porras DP, Xenocostas D, Leber A, Leber B, Foley R, Trus M, Berg T, Kawata E, Xenocostas A, Bhatia M. Leukemic progenitor compartment serves as a prognostic measure of cancer stemness in patients with acute myeloid leukemia. Cell Rep Med 2023:101108. [PMID: 37433297 PMCID: PMC10394166 DOI: 10.1016/j.xcrm.2023.101108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 01/29/2023] [Accepted: 06/16/2023] [Indexed: 07/13/2023]
Abstract
We systematically investigate functional and molecular measures of stemness in patients with acute myeloid leukemia (AML) using a cohort of 121 individuals. We confirm that the presence of leukemic stem cells (LSCs) detected through in vivo xenograft transplantation is associated with poor survival. However, the measurement of leukemic progenitor cells (LPCs) through in vitro colony-forming assays provides an even stronger predictor of overall and event-free survival. LPCs not only capture patient-specific mutations but also retain serial re-plating ability, demonstrating their biological relevance. Notably, LPC content represents an independent prognostic factor in multivariate analyses including clinical guidelines of risk stratification. Our findings suggest that LPCs provide a robust functional measure of AML, enabling quantitative and rapid assessment of a wide range of patients. This highlights the potential of LPCs as a valuable prognostic factor in AML management.
Collapse
Affiliation(s)
- Allison L Boyd
- Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Justin Lu
- Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Cameron G Hollands
- Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Lili Alsostovar
- Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Shiva Murali
- Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Jennifer C Reid
- Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Wendy Ye
- Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Sean Vandersluis
- Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Paige Johnson
- Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Amro ElRafie
- Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Deanna P Porras
- Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Dimetri Xenocostas
- Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Andrew Leber
- Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Brian Leber
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Ronan Foley
- Department of Oncology, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Michael Trus
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Tobias Berg
- Department of Oncology, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Eri Kawata
- Department of Medicine, Division of Hematology, Schulich School of Medicine, University of Western Ontario, London, Ontario N6A 3K7, Canada
| | - Anargyros Xenocostas
- Department of Medicine, Division of Hematology, Schulich School of Medicine, University of Western Ontario, London, Ontario N6A 3K7, Canada
| | - Mickie Bhatia
- Michael G. DeGroote School of Medicine, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada.
| |
Collapse
|
81
|
Zhao HG, Deininger M. Always stressed but never exhausted: how stem cells in myeloid neoplasms avoid extinction in inflammatory conditions. Blood 2023; 141:2797-2812. [PMID: 36947811 PMCID: PMC10315634 DOI: 10.1182/blood.2022017152] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 02/27/2023] [Accepted: 03/13/2023] [Indexed: 03/24/2023] Open
Abstract
Chronic or recurrent episodes of acute inflammation cause attrition of normal hematopoietic stem cells (HSCs) that can lead to hematopoietic failure but they drive progression in myeloid malignancies and their precursor clonal hematopoiesis. Mechanistic parallels exist between hematopoiesis in chronic inflammation and the continuously increased proliferation of myeloid malignancies, particularly myeloproliferative neoplasms (MPNs). The ability to enter dormancy, a state of deep quiescence characterized by low oxidative phosphorylation, low glycolysis, reduced protein synthesis, and increased autophagy is central to the preservation of long-term HSCs and likely MPN SCs. The metabolic features of dormancy resemble those of diapause, a state of arrested embryonic development triggered by adverse environmental conditions. To outcompete their normal counterparts in the inflammatory MPN environment, MPN SCs co-opt mechanisms used by HSCs to avoid exhaustion, including signal attenuation by negative regulators, insulation from activating cytokine signals, anti-inflammatory signaling, and epigenetic reprogramming. We propose that new therapeutic strategies may be derived from conceptualizing myeloid malignancies as an ecosystem out of balance, in which residual normal and malignant hematopoietic cells interact in multiple ways, only few of which have been characterized in detail. Disrupting MPN SC insulation to overcome dormancy, interfering with aberrant cytokine circuits that favor MPN cells, and directly boosting residual normal HSCs are potential strategies to tip the balance in favor of normal hematopoiesis. Although eradicating the malignant cell clones remains the goal of therapy, rebalancing the ecosystem may be a more attainable objective in the short term.
Collapse
Affiliation(s)
- Helong Gary Zhao
- Versiti Blood Research Institute and Medical College of Wisconsin, Milwaukee, WI
| | - Michael Deininger
- Versiti Blood Research Institute and Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
82
|
Diamond B, Ziccheddu B, Maclachlan K, Taylor J, Boyle E, Ossa JA, Jahn J, Affer M, Totiger TM, Coffey D, Chandhok N, Watts J, Cimmino L, Lu SX, Bolli N, Bolton K, Landau H, Park JH, Ganesh K, McPherson A, Sekeres MA, Lesokhin A, Chung DJ, Zhang Y, Ho C, Roshal M, Tyner J, Nimer S, Papaemmanuil E, Usmani S, Morgan G, Landgren O, Maura F. Tracking the evolution of therapy-related myeloid neoplasms using chemotherapy signatures. Blood 2023; 141:2359-2371. [PMID: 36626250 PMCID: PMC10273163 DOI: 10.1182/blood.2022018244] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/22/2022] [Accepted: 12/19/2022] [Indexed: 01/11/2023] Open
Abstract
Patients treated with cytotoxic therapies, including autologous stem cell transplantation, are at risk for developing therapy-related myeloid neoplasms (tMN). Preleukemic clones (ie, clonal hematopoiesis [CH]) are detectable years before the development of these aggressive malignancies, although the genomic events leading to transformation and expansion are not well defined. Here, by leveraging distinctive chemotherapy-associated mutational signatures from whole-genome sequencing data and targeted sequencing of prechemotherapy samples, we reconstructed the evolutionary life-history of 39 therapy-related myeloid malignancies. A dichotomy was revealed, in which neoplasms with evidence of chemotherapy-induced mutagenesis from platinum and melphalan were hypermutated and enriched for complex structural variants (ie, chromothripsis), whereas neoplasms with nonmutagenic chemotherapy exposures were genomically similar to de novo acute myeloid leukemia. Using chemotherapy-associated mutational signatures as temporal barcodes linked to discrete clinical exposure in each patient's life, we estimated that several complex events and genomic drivers were acquired after chemotherapy was administered. For patients with prior multiple myeloma who were treated with high-dose melphalan and autologous stem cell transplantation, we demonstrate that tMN can develop from either a reinfused CH clone that escapes melphalan exposure and is selected after reinfusion, or from TP53-mutant CH that survives direct myeloablative conditioning and acquires melphalan-induced DNA damage. Overall, we revealed a novel mode of tMN progression that is not reliant on direct mutagenesis or even exposure to chemotherapy. Conversely, for tMN that evolve under the influence of chemotherapy-induced mutagenesis, distinct chemotherapies not only select preexisting CH but also promote the acquisition of recurrent genomic drivers.
Collapse
Affiliation(s)
- Benjamin Diamond
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | | | - Kylee Maclachlan
- Division of Myeloma, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Justin Taylor
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Eileen Boyle
- Myeloma Research Program, New York University Langone, Perlmutter Cancer Center, New York, NY
| | - Juan Arango Ossa
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jacob Jahn
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Maurizio Affer
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | | | - David Coffey
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Namrata Chandhok
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Justin Watts
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Luisa Cimmino
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Sydney X. Lu
- Division of Hematology, Stanford Hospital and Clinics, Stanford University, Stanford, CA
| | - Niccolò Bolli
- Department of Oncology and Onco-Hematology, Università degli Studi di Milano, Milan, Italy
- Hematology Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Kelly Bolton
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Heather Landau
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jae H. Park
- Department of Medicine, Memorial Hospital, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Karuna Ganesh
- Department of Medicine, Memorial Hospital, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andrew McPherson
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Alexander Lesokhin
- Division of Myeloma, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - David J. Chung
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Yanming Zhang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Caleb Ho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Mikhail Roshal
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jeffrey Tyner
- Division of Hematology and Medical Oncology, Oregon Health & Science University, Portland, OR
| | - Stephen Nimer
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Elli Papaemmanuil
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Saad Usmani
- Division of Myeloma, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Gareth Morgan
- Myeloma Research Program, New York University Langone, Perlmutter Cancer Center, New York, NY
| | - Ola Landgren
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Francesco Maura
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| |
Collapse
|
83
|
Katiyar S, Shah A, Rahman K, Tripathy NK, Kashyap R, Nityanand S, Chaturvedi CP. Analysis of Immunophenotypic Changes during Ex Vivo Human Erythropoiesis and Its Application in the Study of Normal and Defective Erythropoiesis. Cells 2023; 12:cells12091303. [PMID: 37174702 PMCID: PMC10177526 DOI: 10.3390/cells12091303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Erythropoiesis is a highly regulated process and undergoes several genotypic and phenotypic changes during differentiation. The phenotypic changes can be evaluated using a combination of cell surface markers expressed at different cellular stages of erythropoiesis using FACS. However, limited studies are available on the in-depth phenotypic characterization of progenitors from human adult hematopoietic stem and progenitor cells (HSPCs) to red blood cells. Therefore, using a set of designed marker panels, in the current study we have kinetically characterized the hematopoietic, erythroid progenitors, and terminally differentiated erythroblasts ex vivo. Furthermore, the progenitor stages were explored for expression of CD117, CD31, CD41a, CD133, and CD45, along with known key markers CD36, CD71, CD105, and GPA. Additionally, we used these marker panels to study the stage-specific phenotypic changes regulated by the epigenetic regulator; Nuclear receptor binding SET Domain protein 1 (NSD1) during erythropoiesis and to study ineffective erythropoiesis in myelodysplastic syndrome (MDS) and pure red cell aplasia (PRCA) patients. Our immunophenotyping strategy can be used to sort and study erythroid-primed hematopoietic and erythroid precursors at specified time points and to study diseases resulting from erythroid dyspoiesis. Overall, the current study explores the in-depth kinetics of phenotypic changes occurring during human erythropoiesis and applies this strategy to study normal and defective erythropoiesis.
Collapse
Affiliation(s)
- Shobhita Katiyar
- Stem Cell Research Center, Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Arunim Shah
- Stem Cell Research Center, Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Khaliqur Rahman
- Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Naresh Kumar Tripathy
- Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Rajesh Kashyap
- Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Soniya Nityanand
- Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| | - Chandra Prakash Chaturvedi
- Stem Cell Research Center, Department of Hematology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, India
| |
Collapse
|
84
|
Salavaty A, Azadian E, Naik SH, Currie PD. Clonal selection parallels between normal and cancer tissues. Trends Genet 2023; 39:358-380. [PMID: 36842901 DOI: 10.1016/j.tig.2023.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 01/12/2023] [Accepted: 01/26/2023] [Indexed: 02/28/2023]
Abstract
Clonal selection and drift drive both normal tissue and cancer development. However, the biological mechanisms and environmental conditions underpinning these processes remain to be elucidated. Clonal selection models are centered in Darwinian evolutionary theory, where some clones with the fittest features are selected and populate the tissue or tumor. We suggest that different subclasses of stem cells, each of which is responsible for a distinct feature of the selection process, share common features between normal and cancer conditions. While active stem cells populate the tissue, dormant cells account for tissue replenishment/regeneration in both normal and cancerous tissues. We also discuss potential mechanisms that drive clonal drift, their interactions with clonal selection, and their similarities during normal and cancer tissue development.
Collapse
Affiliation(s)
- Adrian Salavaty
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; Systems Biology Institute Australia, Monash University, Clayton, VIC 3800, Australia.
| | - Esmaeel Azadian
- Immunology Division, Walter and Eliza Hall Institute, Parkville, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Shalin H Naik
- Immunology Division, Walter and Eliza Hall Institute, Parkville, VIC, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia; Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Peter D Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; EMBL Australia, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
85
|
Kobayashi M, Yoshimoto M. Multiple waves of fetal-derived immune cells constitute adult immune system. Immunol Rev 2023; 315:11-30. [PMID: 36929134 PMCID: PMC10754384 DOI: 10.1111/imr.13192] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
It has been over three decades since Drs. Herzenberg and Herzenberg proposed the layered immune system hypothesis, suggesting that different types of stem cells with distinct hematopoietic potential produce specific immune cells. This layering of immune system development is now supported by recent studies showing the presence of fetal-derived immune cells that function in adults. It has been shown that various immune cells arise at different embryonic ages via multiple waves of hematopoiesis from special endothelial cells (ECs), referred to as hemogenic ECs. However, it remains unknown whether these fetal-derived immune cells are produced by hematopoietic stem cells (HSCs) during the fetal to neonatal period. To address this question, many advanced tools have been used, including lineage-tracing mouse models, cellular barcoding techniques, clonal assays, and transplantation assays at the single-cell level. In this review, we will review the history of the search for the origins of HSCs, B-1a progenitors, and mast cells in the mouse embryo. HSCs can produce both B-1a and mast cells within a very limited time window, and this ability declines after embryonic day (E) 14.5. Furthermore, the latest data have revealed that HSC-independent adaptive immune cells exist in adult mice, which implies more complicated developmental pathways of immune cells. We propose revised road maps of immune cell development.
Collapse
Affiliation(s)
- Michihiro Kobayashi
- Center for Stem Cell and Regenerative Medicine, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Momoko Yoshimoto
- Center for Stem Cell and Regenerative Medicine, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
86
|
Ng LG, Liu Z, Kwok I, Ginhoux F. Origin and Heterogeneity of Tissue Myeloid Cells: A Focus on GMP-Derived Monocytes and Neutrophils. Annu Rev Immunol 2023; 41:375-404. [PMID: 37126421 DOI: 10.1146/annurev-immunol-081022-113627] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Myeloid cells are a significant proportion of leukocytes within tissues, comprising granulocytes, monocytes, dendritic cells, and macrophages. With the identification of various myeloid cells that perform separate but complementary functions during homeostasis and disease, our understanding of tissue myeloid cells has evolved significantly. Exciting findings from transcriptomics profiling and fate-mapping mouse models have facilitated the identification of their developmental origins, maturation, and tissue-specific specializations. This review highlights the current understanding of tissue myeloid cells and the contributing factors of functional heterogeneity to better comprehend the complex and dynamic immune interactions within the healthy or inflamed tissue. Specifically, we discuss the new understanding of the contributions of granulocyte-monocyte progenitor-derived phagocytes to tissue myeloid cell heterogeneity as well as the impact of niche-specific factors on monocyte and neutrophil phenotype and function. Lastly, we explore the developing paradigm of myeloid cell heterogeneity during inflammation and disease.
Collapse
Affiliation(s)
- Lai Guan Ng
- Shanghai Immune Therapy Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China;
- Singapore Immunology Network (SIgN), A*STAR (Agency for Science, Technology and Research), Biopolis, Singapore; ,
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Zhaoyuan Liu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Immanuel Kwok
- Singapore Immunology Network (SIgN), ASTAR (Agency for Science, Technology and Research), Biopolis, Singapore; ,
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), ASTAR (Agency for Science, Technology and Research), Biopolis, Singapore; ,
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institut Gustave Roussy, INSERM U1015, Villejuif, France
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore
| |
Collapse
|
87
|
Loeffler D. HSC aging: a sleeping beauty fairy tale? Blood 2023; 141:1902-1904. [PMID: 37079334 DOI: 10.1182/blood.2023019780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023] Open
Affiliation(s)
- Dirk Loeffler
- St. Jude Children's Research Hospital
- University of Tennessee
| |
Collapse
|
88
|
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
|
89
|
Anginot A, Nguyen J, Abou Nader Z, Rondeau V, Bonaud A, Kalogeraki M, Boutin A, Lemos JP, Bisio V, Koenen J, Hanna Doumit Sakr L, Picart A, Coudert A, Provot S, Dulphy N, Aurrand-Lions M, Mancini SJC, Lazennec G, McDermott DH, Guidez F, Blin-Wakkach C, Murphy PM, Cohen-Solal M, Espéli M, Rouleau M, Balabanian K. WHIM Syndrome-linked CXCR4 mutations drive osteoporosis. Nat Commun 2023; 14:2058. [PMID: 37045841 PMCID: PMC10097661 DOI: 10.1038/s41467-023-37791-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 03/07/2023] [Indexed: 04/14/2023] Open
Abstract
WHIM Syndrome is a rare immunodeficiency caused by gain-of-function CXCR4 mutations. Here we report a decrease in bone mineral density in 25% of WHIM patients and bone defects leading to osteoporosis in a WHIM mouse model. Imbalanced bone tissue is observed in mutant mice combining reduced osteoprogenitor cells and increased osteoclast numbers. Mechanistically, impaired CXCR4 desensitization disrupts cell cycle progression and osteogenic commitment of skeletal stromal/stem cells, while increasing their pro-osteoclastogenic capacities. Impaired osteogenic differentiation is evidenced in primary bone marrow stromal cells from WHIM patients. In mice, chronic treatment with the CXCR4 antagonist AMD3100 normalizes in vitro osteogenic fate of mutant skeletal stromal/stem cells and reverses in vivo the loss of skeletal cells, demonstrating that proper CXCR4 desensitization is required for the osteogenic specification of skeletal stromal/stem cells. Our study provides mechanistic insights into how CXCR4 signaling regulates the osteogenic fate of skeletal cells and the balance between bone formation and resorption.
Collapse
Affiliation(s)
- Adrienne Anginot
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Julie Nguyen
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- Inflammation, Microbiome and Immunosurveillance, INSERM, Université Paris-Saclay, Orsay, France
| | - Zeina Abou Nader
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Vincent Rondeau
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Amélie Bonaud
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Maria Kalogeraki
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | | | - Julia P Lemos
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Valeria Bisio
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Joyce Koenen
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- Inflammation, Microbiome and Immunosurveillance, INSERM, Université Paris-Saclay, Orsay, France
| | - Lea Hanna Doumit Sakr
- Université Paris Cité, BIOSCAR Inserm U1132, Department of Rheumatology and Reference Center for Rare Bone Diseases, AP-HP Hospital Lariboisière, Paris, France
| | - Amandine Picart
- Université Paris Cité, BIOSCAR Inserm U1132, Department of Rheumatology and Reference Center for Rare Bone Diseases, AP-HP Hospital Lariboisière, Paris, France
| | - Amélie Coudert
- Université Paris Cité, BIOSCAR Inserm U1132, Department of Rheumatology and Reference Center for Rare Bone Diseases, AP-HP Hospital Lariboisière, Paris, France
| | - Sylvain Provot
- Université Paris Cité, BIOSCAR Inserm U1132, Department of Rheumatology and Reference Center for Rare Bone Diseases, AP-HP Hospital Lariboisière, Paris, France
| | - Nicolas Dulphy
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Michel Aurrand-Lions
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Stéphane J C Mancini
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Gwendal Lazennec
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- CNRS, SYS2DIAG-ALCEDIAG, Cap Delta, Montpellier, France
| | - David H McDermott
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Fabien Guidez
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1131, Paris, France
| | | | - Philip M Murphy
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Martine Cohen-Solal
- Université Paris Cité, BIOSCAR Inserm U1132, Department of Rheumatology and Reference Center for Rare Bone Diseases, AP-HP Hospital Lariboisière, Paris, France
| | - Marion Espéli
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | | | - Karl Balabanian
- Université Paris Cité, Institut de Recherche Saint-Louis, INSERM U1160, Paris, France.
- CNRS, GDR3697 "Microenvironment of tumor niches", Micronit, France.
- OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France.
| |
Collapse
|
90
|
Banjac I, Maimets M, Jensen KB. Maintenance of high-turnover tissues during and beyond homeostasis. Cell Stem Cell 2023; 30:348-361. [PMID: 37028402 DOI: 10.1016/j.stem.2023.03.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/23/2023] [Accepted: 03/15/2023] [Indexed: 04/09/2023]
Abstract
Tissues with a high turnover rate produce millions of cells daily and have abundant regenerative capacity. At the core of their maintenance are populations of stem cells that balance self-renewal and differentiation to produce the adequate numbers of specialized cells required for carrying out essential tissue functions. Here, we compare and contrast the intricate mechanisms and elements of homeostasis and injury-driven regeneration in the epidermis, hematopoietic system, and intestinal epithelium-the fastest renewing tissues in mammals. We highlight the functional relevance of the main mechanisms and identify open questions in the field of tissue maintenance.
Collapse
Affiliation(s)
- Isidora Banjac
- The Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Martti Maimets
- The Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark.
| | - Kim B Jensen
- The Novo Nordisk Foundation Center for Stem Cell Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark.
| |
Collapse
|
91
|
Li F, Sun J. Differentiation Kinetics of Hematopoietic Stem and Progenitor Cells In Vivo Are Not Affected by β-Glucan Treatment in Trained Immunity. Inflammation 2023; 46:718-729. [PMID: 36414879 DOI: 10.1007/s10753-022-01767-1] [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: 10/12/2022] [Revised: 11/08/2022] [Accepted: 11/13/2022] [Indexed: 11/24/2022]
Abstract
Agonists of trained immunity induce epigenetic changes in hematopoietic stem and progenitor cells (HSPCs) to generate long-lasting immune protection. Although trained HSPCs generate myeloid cells with increased responsiveness to secondary challenges, whether their differentiation kinetics is affected by prior exposure to inducers of trained immunity remains elusive. Here, we used lineage tracing to examine the cell fates of endothelial protein C receptor-positive hematopoietic stem cells (EPCR+ HSCs) and fms-like tyrosine kinase 3-positive multipotent progenitor cells (Flt3+ MPPs) in β-glucan-induced trained immunity. We found that although β-glucan triggered the expected expansion of myeloid progenitors, the differentiation behaviors of EPCR+ HSCs and Flt3+ MPPs in multiple cycles of hematopoietic regeneration were hardly affected. Thus, our results rule out changed kinetics in cell differentiation by EPCR+ HSC and Flt3+ MPP as the cause of enhanced myelopoiesis upon secondary immune challenges.
Collapse
Affiliation(s)
- Feiyang Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Jianlong Sun
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
92
|
Huo Y, Wu L, Pang A, Li Q, Hong F, Zhu C, Yang Z, Dai W, Zheng Y, Meng Q, Sun J, Ma S, Hu L, Zhu P, Dong F, Gao X, Jiang E, Hao S, Cheng T. Single-cell dissection of human hematopoietic reconstitution after allogeneic hematopoietic stem cell transplantation. Sci Immunol 2023; 8:eabn6429. [PMID: 36930730 DOI: 10.1126/sciimmunol.abn6429] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
Abstract
Hematopoietic stem cell transplantation is an effective regenerative therapy for many malignant, inherited, or autoimmune diseases. However, our understanding of reconstituted hematopoiesis in transplant patients remains limited. Here, we uncover the reconstitution dynamics of human allogeneic hematopoietic stem and progenitor cells (HSPCs) at single-cell resolution after transplantation. Transplanted HSPCs underwent rapid and measurable changes during the first 30 days after transplantation, characterized by a strong proliferative response on the first day. Transcriptomic analysis of HSPCs enabled us to observe that immunoregulatory neutrophil progenitors expressing high levels of the S100A gene family were enriched in granulocyte colony-stimulating factor-mobilized peripheral blood stem cells. Transplant recipients who developed acute graft-versus-host disease (aGVHD) infused fewer S100Ahigh immunoregulatory neutrophil progenitors, immunophenotyped as Lin-CD34+CD66b+CD177+, than those who did not develop aGVHD. Therefore, our study provides insights into the regenerative process of transplanted HSPCs in human patients and identifies a potential criterion for identifying patients at high risk for developing aGVHD early after transplant.
Collapse
Affiliation(s)
- Yingying Huo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Linjie Wu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Aiming Pang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Qing Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Fang Hong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Caiying Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Zining Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Weiqian Dai
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Yawei Zheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Qianqian Meng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Jiali Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Shihui Ma
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Linping Hu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Ping Zhu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Fang Dong
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Xin Gao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Erlie Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Sha Hao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
| |
Collapse
|
93
|
Yao L, Li F, Yu C, Wang H, Wang Y, Ye L, Yu F. Chronological and Replicative Aging of CD51 +/PDGFR-α + Pulp Stromal Cells. J Dent Res 2023:220345231158038. [PMID: 36919905 DOI: 10.1177/00220345231158038] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
As a crucial source of mesenchymal stromal cells, CD51+/PDGFR-α+ human dental pulp stromal cells (hDPSCs) are promising seeding cells for regenerative medicine. Cellular senescence hinders the translational application of hDPSCs. However, it remains unclear whether chronological and replicative senescence results in distinct outcomes for hDPSCs. To investigate the influence of senescence on DPSCs, we used transgenic lineage tracking, immunofluorescence, flow cytometry, and various molecular experiments to depict the dynamic pattern of hDPSCs in mice and humans during chronological and replicative senescence. The data demonstrated that CD51+/PDGFR-α+ cells were decreased in chronological senescence. Impaired self-renewal and higher ossificatory differentiation were observed in chronologically senescent hDPSCs. Regarding replicative senescence, a decreased CD51+ but upregulated PDGFR-α+ population was observed in culture. Furthermore, weakened self-renewal and osteogenic differentiation were observed in replicatively senescent hDPSCs. In summary, CD51+/PDGFR-α+ hDPSCs decrease in chronologically aged pulp, with self-renewal that is impaired without impaired osteogenic differentiation. However, replicative senescence has a different impact: self-renewal and ossific differentiation are impaired and CD51 expression is reduced, but PDGFR-α expression remains. These findings demonstrate the different outcomes of chronological and replicative senescence in CD51+/PDGFR-α+ hDPSCs. Furthermore, we revealed that impaired self-renewal is the core dysfunction for both types of cellular aging and that osteogenic differentiation capability differs between them. This study provides insights into the influence of chronological and replicative senescence on the characteristics and capabilities of hDPSCs. These advances provide fundamental knowledge to alleviate cellular aging of CD51+/PDGFR-α+ hDPSCs and promote their translational applications.
Collapse
Affiliation(s)
- L Yao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - F Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - C Yu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - H Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - L Ye
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - F Yu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
94
|
Kobayashi M, Wei H, Yamanashi T, Azevedo Portilho N, Cornelius S, Valiente N, Nishida C, Cheng H, Latorre A, Zheng WJ, Kang J, Seita J, Shih DJ, Wu JQ, Yoshimoto M. HSC-independent definitive hematopoiesis persists into adult life. Cell Rep 2023; 42:112239. [PMID: 36906851 PMCID: PMC10122268 DOI: 10.1016/j.celrep.2023.112239] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/18/2023] [Accepted: 02/24/2023] [Indexed: 03/13/2023] Open
Abstract
It is widely believed that hematopoiesis after birth is established by hematopoietic stem cells (HSCs) in the bone marrow and that HSC-independent hematopoiesis is limited only to primitive erythro-myeloid cells and tissue-resident innate immune cells arising in the embryo. Here, surprisingly, we find that significant percentages of lymphocytes are not derived from HSCs, even in 1-year-old mice. Instead, multiple waves of hematopoiesis occur from embryonic day 7.5 (E7.5) to E11.5 endothelial cells, which simultaneously produce HSCs and lymphoid progenitors that constitute many layers of adaptive T and B lymphocytes in adult mice. Additionally, HSC lineage tracing reveals that the contribution of fetal liver HSCs to peritoneal B-1a cells is minimal and that the majority of B-1a cells are HSC independent. Our discovery of extensive HSC-independent lymphocytes in adult mice attests to the complex blood developmental dynamics spanning the embryo-to-adult transition and challenges the paradigm of HSCs exclusively underpinning the postnatal immune system.
Collapse
Affiliation(s)
- Michihiro Kobayashi
- Center for Stem Cell and Regenerative Medicine, Brown Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Haichao Wei
- Center for Stem Cell and Regenerative Medicine, Brown Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Takashi Yamanashi
- Advanced Data Science Project, RIKEN Information R&D and Strategy Headquarters, Tokyo 103-0027, Japan; Center for Integrative Medical Sciences, RIKEN, Yokohama, Kanagawa 230-0045, Japan
| | - Nathalia Azevedo Portilho
- Center for Stem Cell and Regenerative Medicine, Brown Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Samuel Cornelius
- Center for Stem Cell and Regenerative Medicine, Brown Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Noemi Valiente
- Center for Stem Cell and Regenerative Medicine, Brown Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Chika Nishida
- Center for Stem Cell and Regenerative Medicine, Brown Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Haizi Cheng
- Center for Stem Cell and Regenerative Medicine, Brown Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Augusto Latorre
- Center for Stem Cell and Regenerative Medicine, Brown Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - W Jim Zheng
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Joonsoo Kang
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Jun Seita
- Advanced Data Science Project, RIKEN Information R&D and Strategy Headquarters, Tokyo 103-0027, Japan; Center for Integrative Medical Sciences, RIKEN, Yokohama, Kanagawa 230-0045, Japan
| | - David J Shih
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Jia Qian Wu
- Center for Stem Cell and Regenerative Medicine, Brown Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Momoko Yoshimoto
- Center for Stem Cell and Regenerative Medicine, Brown Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
95
|
Jahn J, Diamond B, Hsu J, Montoya S, Totiger TM, Landgren O, Maura F, Taylor J. Therapy-selected clonal hematopoiesis and its role in myeloid neoplasms. Leuk Res 2023; 126:107020. [PMID: 36696829 PMCID: PMC11305114 DOI: 10.1016/j.leukres.2023.107020] [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: 11/22/2022] [Revised: 01/06/2023] [Accepted: 01/19/2023] [Indexed: 01/21/2023]
Abstract
Therapy-related myeloid neoplasms (t-MN) account for approximately 10-15% of all myeloid neoplasms and are associated with poor prognosis. Genomic characterization of t-MN to date has been limited in comparison to the considerable sequencing efforts performed for de novo myeloid neoplasms. Until recently, targeted deep sequencing (TDS) or whole exome sequencing (WES) have been the primary technologies utilized and thus limited the ability to explore the landscape of structural variants and mutational signatures. In the past decade, population-level studies have identified clonal hematopoiesis as a risk factor for the development of myeloid neoplasms. However, emerging research on clonal hematopoiesis as a risk factor for developing t-MN is evolving, and much is unknown about the progression of CH to t-MN. In this work, we will review the current knowledge of the genomic landscape of t-MN, discuss background knowledge of clonal hematopoiesis gained from studies of de novo myeloid neoplasms, and examine the recent literature studying the role of therapeutic selection of CH and its evolution under the effects of antineoplastic therapy. Finally, we will discuss the potential implications on current clinical practice and the areas of focus needed for future research into therapy-selected clonal hematopoiesis in myeloid neoplasms.
Collapse
Affiliation(s)
- Jacob Jahn
- Sylvester Comprehensive Cancer Center at the University of Miami Miller School of Medicine, United States
| | - Benjamin Diamond
- Myeloma Division, Department of Medicine, University of Miami Miller School of Medicine, United States
| | - Jeffrey Hsu
- Sylvester Comprehensive Cancer Center at the University of Miami Miller School of Medicine, United States
| | - Skye Montoya
- Sylvester Comprehensive Cancer Center at the University of Miami Miller School of Medicine, United States
| | - Tulasigeri M Totiger
- Sylvester Comprehensive Cancer Center at the University of Miami Miller School of Medicine, United States
| | - Ola Landgren
- Myeloma Division, Department of Medicine, University of Miami Miller School of Medicine, United States
| | - Francesco Maura
- Myeloma Division, Department of Medicine, University of Miami Miller School of Medicine, United States
| | - Justin Taylor
- Sylvester Comprehensive Cancer Center at the University of Miami Miller School of Medicine, United States; Leukemia Program, Department of Medicine, University of Miami Miller School of Medicine, United States.
| |
Collapse
|
96
|
Fanti AK, Busch K, Greco A, Wang X, Cirovic B, Shang F, Nizharadze T, Frank L, Barile M, Feyerabend TB, Höfer T, Rodewald HR. Flt3- and Tie2-Cre tracing identifies regeneration in sepsis from multipotent progenitors but not hematopoietic stem cells. Cell Stem Cell 2023; 30:207-218.e7. [PMID: 36652946 DOI: 10.1016/j.stem.2022.12.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 10/04/2022] [Accepted: 12/20/2022] [Indexed: 01/19/2023]
Abstract
In response to infections and stress, hematopoiesis rapidly enhances blood and immune cell production. The stage within the hematopoietic hierarchy that accounts for this regeneration is unclear under natural conditions in vivo. We analyzed by differentiation tracing, using inducible Tie2- or Flt3-driven Cre recombinase, the roles of mouse hematopoietic stem cells (HSCs) and multipotent progenitors (MPPs). During polymicrobial sepsis, HSCs responded transcriptionally and increased their proliferation and cell death, yet HSC differentiation rates remained at steady-state levels. HSC differentiation was also independent from the ablation of various cellular compartments-bleeding, the antibody-mediated ablation of granulocytes or B lymphocytes, and genetic lymphocyte deficiency. By marked contrast, the fate mapping of MPPs in polymicrobial sepsis identified these cells as a major source for accelerated myeloid cell production. The regulation of blood and immune cell homeostasis by progenitors rather than stem cells may ensure a rapid response while preserving the integrity of the HSC population.
Collapse
Affiliation(s)
- Ann-Kathrin Fanti
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Katrin Busch
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany; Faculty of Medicine, Heidelberg University, 69120 Heidelberg, Germany
| | - Alessandro Greco
- Division of Theoretical Systems Biology, German Cancer Research Center, 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Xi Wang
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany; State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Avenue, Nanjing 211166, China
| | - Branko Cirovic
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Fuwei Shang
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany; Faculty of Medicine, Heidelberg University, 69120 Heidelberg, Germany
| | - Tamar Nizharadze
- Division of Theoretical Systems Biology, German Cancer Research Center, 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Larissa Frank
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Melania Barile
- Division of Theoretical Systems Biology, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Thorsten B Feyerabend
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Thomas Höfer
- Division of Theoretical Systems Biology, German Cancer Research Center, 69120 Heidelberg, Germany.
| | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany.
| |
Collapse
|
97
|
Vink CS, Dzierzak E. The (intra-aortic) hematopoietic cluster cocktail: what is in the mix? Exp Hematol 2023; 118:1-11. [PMID: 36529317 DOI: 10.1016/j.exphem.2022.12.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
The adult-definitive hematopoietic hierarchy and hematopoietic stem cells (HSCs) residing in the bone marrow are established during embryonic development. In mouse, human, and many other mammals, it is the sudden formation of so-called intra-aortic/arterial hematopoietic clusters (IAHCs) that best signifies and visualizes this de novo generation of HSCs and hematopoietic progenitor cells (HPCs). Cluster cells arise through an endothelial-to-hematopoietic transition and, for some time, express markers/genes of both tissue types, whilst acquiring more hematopoietic features and losing endothelial ones. Among several hundreds of IAHC cells, the midgestation mouse embryo contains only very few bona fide adult-repopulating HSCs, suggestive of a challenging cell fate to achieve. Most others are HPCs of various types, some of which have the potential to mature into HSCs in vitro. Based on the number of cells that reveal hematopoietic function, a fraction of IAHC cells is uncharacterized. This review aims to explore the current state of knowledge on IAHC cells. We will describe markers useful for isolation and characterization of these fleetingly produced, yet vitally important, cells and for the refined enrichment of the HSCs they contain, and speculate on the role of some IAHC cells that are as-yet functionally uncharacterized.
Collapse
Affiliation(s)
- Chris S Vink
- The University of Edinburgh, Centre for Inflammation Research, Edinburgh, Midlothian, Scotland, UK
| | - Elaine Dzierzak
- The University of Edinburgh, Centre for Inflammation Research, Edinburgh, Midlothian, Scotland, UK.
| |
Collapse
|
98
|
Haghverdi L, Ludwig LS. Single-cell multi-omics and lineage tracing to dissect cell fate decision-making. Stem Cell Reports 2023; 18:13-25. [PMID: 36630900 PMCID: PMC9860164 DOI: 10.1016/j.stemcr.2022.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 01/12/2023] Open
Abstract
The concept of cell fate relates to the future identity of a cell, and its daughters, which is obtained via cell differentiation and division. Understanding, predicting, and manipulating cell fate has been a long-sought goal of developmental and regenerative biology. Recent insights obtained from single-cell genomic and integrative lineage-tracing approaches have further aided to identify molecular features predictive of cell fate. In this perspective, we discuss these approaches with a focus on theoretical concepts and future directions of the field to dissect molecular mechanisms underlying cell fate.
Collapse
Affiliation(s)
- Laleh Haghverdi
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany.
| | - Leif S Ludwig
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
99
|
In vivo clonal tracking reveals evidence of haemangioblast and haematomesoblast contribution to yolk sac haematopoiesis. Nat Commun 2023; 14:41. [PMID: 36596806 PMCID: PMC9810727 DOI: 10.1038/s41467-022-35744-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 12/22/2022] [Indexed: 01/05/2023] Open
Abstract
During embryogenesis, haematopoietic and endothelial lineages emerge closely in time and space. It is thought that the first blood and endothelium derive from a common clonal ancestor, the haemangioblast. However, investigation of candidate haemangioblasts in vitro revealed the capacity for mesenchymal differentiation, a feature more compatible with an earlier mesodermal precursor. To date, no evidence for an in vivo haemangioblast has been discovered. Using single cell RNA-Sequencing and in vivo cellular barcoding, we have unravelled the ancestral relationships that give rise to the haematopoietic lineages of the yolk sac, the endothelium, and the mesenchyme. We show that the mesodermal derivatives of the yolk sac are produced by three distinct precursors with dual-lineage outcomes: the haemangioblast, the mesenchymoangioblast, and a previously undescribed cell type: the haematomesoblast. Between E5.5 and E7.5, this trio of precursors seeds haematopoietic, endothelial, and mesenchymal trajectories.
Collapse
|
100
|
Umeki Y, Ogawa N, Uegaki Y, Saga K, Kaneda Y, Nimura K. DNA barcoding and gene expression recording reveal the presence of cancer cells with unique properties during tumor progression. Cell Mol Life Sci 2023; 80:17. [PMID: 36564568 PMCID: PMC9789022 DOI: 10.1007/s00018-022-04640-4] [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: 08/22/2022] [Revised: 11/02/2022] [Accepted: 11/19/2022] [Indexed: 12/25/2022]
Abstract
Tumors comprise diverse cancer cell populations with specific capabilities for adaptation to the tumor microenvironment, resistance to anticancer treatments, and metastatic dissemination. However, whether these populations are pre-existing in cancer cells or stochastically appear during tumor growth remains unclear. Here, we show the heterogeneous behaviors of cancer cells regarding response to anticancer drug treatments, formation of lung metastases, and expression of transcription factors related to cancer stem-like cells using a DNA barcoding and gene expression recording system. B16F10 cells maintained clonal diversity after treatment with HVJ-E, a UV-irradiated Sendai virus, and the anticancer drug dacarbazine. PBS treatment of the primary tumor and intravenous injection of B16F10 cells resulted in metastases formed from clones of multiple cell lineages. Conversely, BL6 and 4T1 cells developed spontaneous lung metastases by a small number of clones. Notably, an identical clone of 4T1 cells developed lung metastases in different mice, suggesting the existence of cells with high metastatic potential. Cas9-based transcription recording analysis in a human prostate cancer cell line revealed that specific cells express POU5F1 in response to an anticancer drug and sphere formation. Our findings provide insights into the diversity of cancer cells during tumor progression.
Collapse
Affiliation(s)
- Yuka Umeki
- Division of Gene Therapy Science, Department of Genome Biology, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita, Osaka 565-0871 Japan
| | - Noriaki Ogawa
- Division of Gene Therapy Science, Department of Genome Biology, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita, Osaka 565-0871 Japan
| | - Yuko Uegaki
- Division of Gene Therapy Science, Department of Genome Biology, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita, Osaka 565-0871 Japan
| | - Kotaro Saga
- Division of Gene Therapy Science, Department of Genome Biology, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita, Osaka 565-0871 Japan
| | - Yasufumi Kaneda
- Division of Gene Therapy Science, Department of Genome Biology, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita, Osaka 565-0871 Japan
| | - Keisuke Nimura
- Division of Gene Therapy Science, Department of Genome Biology, Graduate School of Medicine, Osaka University, 2-2 Yamada-Oka, Suita, Osaka 565-0871 Japan
| |
Collapse
|