1
|
MacAldaz ME, Shu J, Edin G, Hale M, Eaves CJ. Hematopoietic Stem Cells in Human Fetal Liver Selectively Express CD49f. Exp Hematol 2025:104788. [PMID: 40311859 DOI: 10.1016/j.exphem.2025.104788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/29/2025] [Accepted: 04/10/2025] [Indexed: 05/03/2025]
Abstract
Identification of phenotypes of human hematopoietic cells that display long-term mature cell outputs in vitro and repopulating capability in immunodeficient mice has been important to anticipating the therapeutic potential of fresh harvests of bone marrow or cord blood before or after their physical or genetic manipulation. However, characterizing their key properties and strategies for their isolation from multiple sources at increasing cell purities and elucidating the mechanisms that regulate their ability to sustain mature blood cell production continues to be of major interest. Previous studies have shown that fetal and adult human cells with long-term blood cell output potential are highly enriched in their respective GPI80+ and CD49f+ subsets of a developmentally preserved CD45+CD34+CD38-CD45RA-CD90+ population. The so-called "GPI80" hematopoietic cells found in first trimester human fetal liver are of particular interest because of their very high regenerative capability compared to their adult or even neonatal (cord blood) "CD49f" counterparts. Here it was hypothesized that high regenerative activity of the GPI80+ cells could be further enriched within a CD49f+ subset. We now demonstrate that co-expression of CD49f within the GPI80+ population identifies a subset with reduced short-term myeloid colony-forming activity in semi-solid medium, and greater progeny outputs in both 12-week growth factor-supplemented stromal co-cultures, and in transplanted immunodeficient mice. These findings demonstrate CD49f is a pervasive marker of human HSCs throughout ontogeny and aging.
Collapse
Affiliation(s)
| | - Jeremy Shu
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Glenn Edin
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Margaret Hale
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Connie J Eaves
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
2
|
Watt SM, Roubelakis MG. Deciphering the Complexities of Adult Human Steady State and Stress-Induced Hematopoiesis: Progress and Challenges. Int J Mol Sci 2025; 26:671. [PMID: 39859383 PMCID: PMC11766050 DOI: 10.3390/ijms26020671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/05/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Human hematopoietic stem cells (HSCs) have traditionally been viewed as self-renewing, multipotent cells with enormous potential in sustaining essential steady state blood and immune cell production throughout life. Indeed, around 86% (1011-1012) of new cells generated daily in a healthy young human adult are of hematopoietic origin. Therapeutically, human HSCs have contributed to over 1.5 million hematopoietic cell transplants (HCTs) globally, making this the most successful regenerative therapy to date. We will commence this review by briefly highlighting selected key achievements (from 1868 to the end of the 20th century) that have contributed to this accomplishment. Much of our knowledge of hematopoiesis is based on small animal models that, despite their enormous importance, do not always recapitulate human hematopoiesis. Given this, we will critically review the progress and challenges faced in identifying adult human HSCs and tracing their lineage differentiation trajectories, referring to murine studies as needed. Moving forward and given that human hematopoiesis is dynamic and can readily adjust to a variety of stressors, we will then discuss recent research advances contributing to understanding (i) which HSPCs maintain daily steady state human hematopoiesis, (ii) where these are located, and (iii) which mechanisms come into play when homeostatic hematopoiesis switches to stress-induced or emergency hematopoiesis.
Collapse
Affiliation(s)
- Suzanne M. Watt
- Stem Cell Research, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9BQ, UK
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, North Terrace, Adelaide 5005, Australia
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide 5001, Australia
| | - Maria G. Roubelakis
- Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens (NKUA), 11527 Athens, Greece;
- Cell and Gene Therapy Laboratory, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece
| |
Collapse
|
3
|
Wong TN, Mychalowych A, Feldpausch ER, Carson A, Karpova D, Link DC. The Clonal Hematopoiesis-associated Gene Srcap Plays an Essential Role in Hematopoiesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.16.607812. [PMID: 39229096 PMCID: PMC11370474 DOI: 10.1101/2024.08.16.607812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Somatic mutations arising in hematopoietic stem cells (HSCs) may provide the latter with a fitness advantage, allowing the mutant HSC to clonally expand. Such mutations have been recurrently identified in the chromatin modifier, SRCAP, in both non-malignant and leukemic clones, suggesting that this gene plays a significant role in hematopoiesis. We generated a conditional Srcap loss of function murine model and determined the consequences of hematopoietic-specific loss of this gene. We show that Srcap is essential for normal fetal liver erythropoiesis and monocytopoiesis. In Srcap deficient fetal livers, the number of phenotypic HSCs is similar to that of controls, but these HSCs exhibit a profound repopulating defect. Likewise, conditional deletion of Srcap during adult hematopoiesis results in a rapid loss of HSCs. Loss of Srcap is associated with evidence of increased DNA damage in HSCs and lineage-restricted progenitors as assessed by y-H2AX expression. Consistent with this finding, we observed strong transcriptional upregulation of the p53 pathway in Srcap deficient erythroid precursors. Collectively our data highlight the importance of Srcap in maintaining HSC function and supporting hematopoietic differentiation and suggests that it plays an essential role in maintaining genomic integrity.
Collapse
Affiliation(s)
- Terrence N. Wong
- Division of Hematology-Oncology, University of Michigan, Ann Arbor, Michigan
| | - Anna Mychalowych
- Division of Hematology-Oncology, University of Michigan, Ann Arbor, Michigan
| | - Ellie R. Feldpausch
- Division of Hematology-Oncology, University of Michigan, Ann Arbor, Michigan
| | - Alexander Carson
- Division of Hematology-Oncology, University of Michigan, Ann Arbor, Michigan
| | - Darja Karpova
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Daniel C. Link
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
4
|
Kushida Y, Oguma Y, Abe K, Deguchi T, Barbera FG, Nishimura N, Fujioka K, Iwatani S, Dezawa M. Human post-implantation blastocyst-like characteristics of Muse cells isolated from human umbilical cord. Cell Mol Life Sci 2024; 81:297. [PMID: 38992309 PMCID: PMC11335221 DOI: 10.1007/s00018-024-05339-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/13/2024]
Abstract
Muse cells, identified as cells positive for the pluripotent surface marker SSEA-3, are pluripotent-like endogenous stem cells located in the bone marrow (BM), peripheral blood, and organ connective tissues. The detailed characteristics of SSEA-3(+) cells in extraembryonic tissue, however, are unknown. Here, we demonstrated that similar to human-adult tissue-Muse cells collected from the BM, adipose tissue, and dermis as SSEA-3(+), human-umbilical cord (UC)-SSEA-3(+) cells express pluripotency markers, differentiate into triploblastic-lineage cells at a single cell level, migrate to damaged tissue, and exhibit low telomerase activity and non-tumorigenicity. Notably, ~ 20% of human-UC-SSEA-3(+) cells were negative for X-inactive specific transcript (XIST), a naïve pluripotent stem cell characteristic, whereas all human adult tissue-Muse cells are XIST-positive. Single-cell RNA sequencing revealed that the gene expression profile of human-UC-SSEA-3(+) cells was more similar to that of human post-implantation blastocysts than human-adult tissue-Muse cells. The DNA methylation level showed the same trend, and notably, the methylation levels in genes particularly related to differentiation were lower in human-UC-SSEA-3(+) cells than in human-adult tissue-Muse cells. Furthermore, human-UC-SSEA-3(+) cells newly express markers specific to extraembryonic-, germline-, and hematopoietic-lineages after differentiation induction in vitro whereas human-adult tissue-Muse cells respond only partially to the induction. Among various stem/progenitor cells in living bodies, those that exhibit properties similar to post-implantation blastocysts in a naïve state have not yet been found in humans. Easily accessible human-UC-SSEA-3(+) cells may be a valuable tool for studying early-stage human development and human reproductive medicine.
Collapse
Affiliation(s)
- Yoshihiro Kushida
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan.
| | - Yo Oguma
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| | - Kana Abe
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| | - Taichi Deguchi
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| | - Federico Girolamo Barbera
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| | - Noriyuki Nishimura
- Department of Public Health, Kobe University Graduate School of Health Science, Kobe, Japan
| | - Kazumichi Fujioka
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Sota Iwatani
- Department of Neonatology, Hyogo Prefectural Kobe Children's Hospital, Kobe, Hyogo, Japan
| | - Mari Dezawa
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-Machi, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan.
| |
Collapse
|
5
|
O’Hehir ZD, Lynch T, O’Neill S, March L, Xue M. Endothelial Protein C Receptor and Its Impact on Rheumatic Disease. J Clin Med 2024; 13:2030. [PMID: 38610795 PMCID: PMC11012567 DOI: 10.3390/jcm13072030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/14/2024] Open
Abstract
Endothelial Protein C Receptor (EPCR) is a key regulator of the activated protein C anti-coagulation pathway due to its role in the binding and activation of this protein. EPCR also binds to other ligands such as Factor VII and X, γδ T-cells, plasmodium falciparum erythrocyte membrane protein 1, and Secretory group V Phospholipases A2, facilitating ligand-specific functions. The functions of EPCR can also be regulated by soluble (s)EPCR that competes for the binding sites of membrane-bound (m)EPCR. sEPCR is created when mEPCR is shed from the cell surface. The propensity of shedding alters depending on the genetic haplotype of the EPCR gene that an individual may possess. EPCR plays an active role in normal homeostasis, anti-coagulation pathways, inflammation, and cell stemness. Due to these properties, EPCR is considered a potential effector/mediator of inflammatory diseases. Rheumatic diseases such as rheumatoid arthritis and systemic lupus erythematosus are autoimmune/inflammatory conditions that are associated with elevated EPCR levels and disease activity, potentially driven by EPCR. This review highlights the functions of EPCR and its contribution to rheumatic diseases.
Collapse
Affiliation(s)
- Zachary Daniel O’Hehir
- Sutton Arthritis Research Laboratory, Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney at Royal North Shore Hospital, Sydney, NSW 2065, Australia;
| | - Tom Lynch
- The Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, University of Sydney at Royal North Shore Hospital, St Leonards, NSW 2065, Australia; (T.L.); (L.M.)
| | - Sean O’Neill
- Department of Rheumatology, Royal North Shore Hospital, Syndey, NSW 2065, Australia;
| | - Lyn March
- The Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, University of Sydney at Royal North Shore Hospital, St Leonards, NSW 2065, Australia; (T.L.); (L.M.)
- Department of Rheumatology, Royal North Shore Hospital, Syndey, NSW 2065, Australia;
| | - Meilang Xue
- Sutton Arthritis Research Laboratory, Sydney Musculoskeletal Health, Kolling Institute, Faculty of Medicine and Health, The University of Sydney at Royal North Shore Hospital, Sydney, NSW 2065, Australia;
- The Australian Arthritis and Autoimmune Biobank Collaborative (A3BC), Institute of Bone and Joint Research, Kolling Institute, Faculty of Medicine and Health, University of Sydney at Royal North Shore Hospital, St Leonards, NSW 2065, Australia; (T.L.); (L.M.)
| |
Collapse
|
6
|
Talkhoncheh MS, Baudet A, Ek F, Subramaniam A, Kao YR, Miharada N, Karlsson C, Oburoglu L, Rydström A, Zemaitis K, Alattar AG, Rak J, Pietras K, Olsson R, Will B, Larsson J. Ciclopirox ethanolamine preserves the immature state of human HSCs by mediating intracellular iron content. Blood Adv 2023; 7:7407-7417. [PMID: 37487020 PMCID: PMC10758717 DOI: 10.1182/bloodadvances.2023009844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 06/14/2023] [Accepted: 07/05/2023] [Indexed: 07/26/2023] Open
Abstract
Culture conditions in which hematopoietic stem cells (HSCs) can be expanded for clinical benefit are highly sought after. To elucidate regulatory mechanisms governing the maintenance and propagation of human HSCs ex vivo, we screened libraries of annotated small molecules in human cord blood cells using an optimized assay for detection of functional HSCs during culture. We found that the antifungal agent ciclopirox ethanolamine (CPX) selectively supported immature CD34+CD90+ cells during culture and enhanced their long-term in vivo repopulation capacity. Purified HSCs treated with CPX showed a reduced cell division rate and an enrichment of HSC-specific gene expression patterns. Mechanistically, we found that the HSC stimulating effect of CPX was directly mediated by chelation of the intracellular iron pool, which in turn affected iron-dependent proteins and enzymes mediating cellular metabolism and respiration. Our findings unveil a significant impact of iron homeostasis in regulation of human HSCs, with important implications for both basic HSC biology and clinical hematology.
Collapse
Affiliation(s)
| | - Aurélie Baudet
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Fredrik Ek
- Chemical Biology and Therapeutics, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Yun-Ruei Kao
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
| | - Natsumi Miharada
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Christine Karlsson
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Leal Oburoglu
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Anna Rydström
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Kristijonas Zemaitis
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Abdul Ghani Alattar
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Justyna Rak
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Kristian Pietras
- Division of Translational Cancer Research, Medicon Village, Lund University, Lund, Sweden
| | - Roger Olsson
- Chemical Biology and Therapeutics, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Britta Will
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY
- Department of Medicine (Oncology), Albert Einstein College of Medicine, Bronx, NY
| | - Jonas Larsson
- Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
7
|
Crosse EI, Binagui-Casas A, Gordon-Keylock S, Rybtsov S, Tamagno S, Olofsson D, Anderson RA, Medvinsky A. An interactive resource of molecular signalling in the developing human haematopoietic stem cell niche. Development 2023; 150:dev201972. [PMID: 37840454 PMCID: PMC10730088 DOI: 10.1242/dev.201972] [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: 05/09/2023] [Accepted: 10/03/2023] [Indexed: 10/17/2023]
Abstract
The emergence of definitive human haematopoietic stem cells (HSCs) from Carnegie Stage (CS) 14 to CS17 in the aorta-gonad-mesonephros (AGM) region is a tightly regulated process. Previously, we conducted spatial transcriptomic analysis of the human AGM region at the end of this period (CS16/CS17) and identified secreted factors involved in HSC development. Here, we extend our analysis to investigate the progression of dorso-ventral polarised signalling around the dorsal aorta over the entire period of HSC emergence. Our results reveal a dramatic increase in ventral signalling complexity from the CS13-CS14 transition, coinciding with the first appearance of definitive HSCs. We further observe stage-specific changes in signalling up to CS17, which may underpin the step-wise maturation of HSCs described in the mouse model. The data-rich resource is also presented in an online interface enabling in silico analysis of molecular interactions between spatially defined domains of the AGM region. This resource will be of particular interest for researchers studying mechanisms underlying human HSC development as well as those developing in vitro methods for the generation of clinically relevant HSCs from pluripotent stem cells.
Collapse
Affiliation(s)
- Edie I. Crosse
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Anahi Binagui-Casas
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | | | - Stanislav Rybtsov
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Sara Tamagno
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Didrik Olofsson
- Omiqa Bioinformatics GmbH, Altensteinstraße 40, 14195 Berlin, Germany
| | - Richard A. Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Alexander Medvinsky
- Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| |
Collapse
|
8
|
Yeung AK, Villacorta-Martin C, Lindstrom-Vautrin J, Belkina AC, Vanuytsel K, Dowrey TW, Ysasi AB, Bawa P, Wang F, Vrbanac V, Mostoslavsky G, Balazs AB, Murphy GJ. De novo hematopoiesis from the fetal lung. Blood Adv 2023; 7:6898-6912. [PMID: 37729429 PMCID: PMC10685174 DOI: 10.1182/bloodadvances.2022008347] [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: 06/15/2022] [Revised: 08/08/2023] [Accepted: 08/13/2023] [Indexed: 09/22/2023] Open
Abstract
Hemogenic endothelial cells (HECs) are specialized cells that undergo endothelial-to-hematopoietic transition (EHT) to give rise to the earliest precursors of hematopoietic progenitors that will eventually sustain hematopoiesis throughout the lifetime of an organism. Although HECs are thought to be primarily limited to the aorta-gonad-mesonephros (AGM) during early development, EHT has been described in various other hematopoietic organs and embryonic vessels. Though not defined as a hematopoietic organ, the lung houses many resident hematopoietic cells, aids in platelet biogenesis, and is a reservoir for hematopoietic stem and progenitor cells (HSPCs). However, lung HECs have never been described. Here, we demonstrate that the fetal lung is a potential source of HECs that have the functional capacity to undergo EHT to produce de novo HSPCs and their resultant progeny. Explant cultures of murine and human fetal lungs display adherent endothelial cells transitioning into floating hematopoietic cells, accompanied by the gradual loss of an endothelial signature. Flow cytometric and functional assessment of fetal-lung explants showed the production of multipotent HSPCs that expressed the EHT and pre-HSPC markers EPCR, CD41, CD43, and CD44. scRNA-seq and small molecule modulation demonstrated that fetal lung HECs rely on canonical signaling pathways to undergo EHT, including TGFβ/BMP, Notch, and YAP. Collectively, these data support the possibility that post-AGM development, functional HECs are present in the fetal lung, establishing this location as a potential extramedullary site of de novo hematopoiesis.
Collapse
Affiliation(s)
- Anthony K. Yeung
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA
- Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA
| | | | | | - Anna C. Belkina
- Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA
| | - Kim Vanuytsel
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA
- Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA
| | - Todd W. Dowrey
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA
- Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA
| | - Alexandra B. Ysasi
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA
- Pulmonary Center and Department of Medicine, Boston University School of Medicine, Boston, MA
| | - Pushpinder Bawa
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA
| | - Feiya Wang
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA
| | | | - Gustavo Mostoslavsky
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA
- Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA
| | | | - George J. Murphy
- Center for Regenerative Medicine of Boston University and Boston Medical Center, Boston, MA
- Section of Hematology and Medical Oncology, Boston University School of Medicine, Boston, MA
| |
Collapse
|
9
|
Sommarin MNE, Olofzon R, Palo S, Dhapola P, Soneji S, Karlsson G, Böiers C. Single-cell multiomics of human fetal hematopoiesis define a developmental-specific population and a fetal signature. Blood Adv 2023; 7:5325-5340. [PMID: 37379274 PMCID: PMC10506049 DOI: 10.1182/bloodadvances.2023009808] [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: 01/24/2023] [Revised: 06/05/2023] [Accepted: 06/16/2023] [Indexed: 06/30/2023] Open
Abstract
Knowledge of human fetal blood development and how it differs from adult blood is highly relevant to our understanding of congenital blood and immune disorders and childhood leukemia, of which the latter can originate in utero. Blood formation occurs in waves that overlap in time and space, adding to heterogeneity, which necessitates single-cell approaches. Here, a combined single-cell immunophenotypic and transcriptional map of first trimester primitive blood development is presented. Using CITE-seq (cellular indexing of transcriptomes and epitopes by sequencing), the molecular profile of established immunophenotype-gated progenitors was analyzed in the fetal liver (FL). Classical markers for hematopoietic stem cells (HSCs), such as CD90 and CD49F, were largely preserved, whereas CD135 (FLT3) and CD123 (IL3R) had a ubiquitous expression pattern capturing heterogenous populations. Direct molecular comparison with an adult bone marrow data set revealed that the HSC state was less frequent in FL, whereas cells with a lymphomyeloid signature were more abundant. An erythromyeloid-primed multipotent progenitor cluster was identified, potentially representing a transient, fetal-specific population. Furthermore, differentially expressed genes between fetal and adult counterparts were specifically analyzed, and a fetal core signature was identified. The core gene set could separate subgroups of acute lymphoblastic leukemia by age, suggesting that a fetal program may be partially retained in specific subgroups of pediatric leukemia. Our detailed single-cell map presented herein emphasizes molecular and immunophenotypic differences between fetal and adult blood cells, which are of significance for future studies of pediatric leukemia and blood development in general.
Collapse
Affiliation(s)
- Mikael N. E. Sommarin
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
| | - Rasmus Olofzon
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
| | - Sara Palo
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
| | - Parashar Dhapola
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
| | - Shamit Soneji
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
| | - Göran Karlsson
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
| | - Charlotta Böiers
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Faculty of Medicine, Lund University, Lund, Sweden
| |
Collapse
|
10
|
Calvanese V, Mikkola HKA. The genesis of human hematopoietic stem cells. Blood 2023; 142:519-532. [PMID: 37339578 PMCID: PMC10447622 DOI: 10.1182/blood.2022017934] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/27/2023] [Accepted: 05/13/2023] [Indexed: 06/22/2023] Open
Abstract
Developmental hematopoiesis consists of multiple, partially overlapping hematopoietic waves that generate the differentiated blood cells required for embryonic development while establishing a pool of undifferentiated hematopoietic stem cells (HSCs) for postnatal life. This multilayered design in which active hematopoiesis migrates through diverse extra and intraembryonic tissues has made it difficult to define a roadmap for generating HSCs vs non-self-renewing progenitors, especially in humans. Recent single-cell studies have helped in identifying the rare human HSCs at stages when functional assays are unsuitable for distinguishing them from progenitors. This approach has made it possible to track the origin of human HSCs to the unique type of arterial endothelium in the aorta-gonad-mesonephros region and document novel benchmarks for HSC migration and maturation in the conceptus. These studies have delivered new insights into the intricate process of HSC generation and provided tools to inform the in vitro efforts to replicate the physiological developmental journey from pluripotent stem cells via distinct mesodermal and endothelial intermediates to HSCs.
Collapse
Affiliation(s)
- Vincenzo Calvanese
- Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA
| | - Hanna K. A. Mikkola
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, CA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA
| |
Collapse
|
11
|
Li H, Luo Q, Cai S, Tie R, Meng Y, Shan W, Xu Y, Zeng X, Qian P, Huang H. Glia maturation factor-γ is required for initiation and maintenance of hematopoietic stem and progenitor cells. Stem Cell Res Ther 2023; 14:117. [PMID: 37122014 PMCID: PMC10150485 DOI: 10.1186/s13287-023-03328-1] [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: 04/24/2022] [Accepted: 04/05/2023] [Indexed: 05/02/2023] Open
Abstract
BACKGROUND In vertebrates, hematopoietic stem and progenitor cells (HSPCs) emerge from hemogenic endothelium in the floor of the dorsal aorta and subsequently migrate to secondary niches where they expand and differentiate into committed lineages. Glia maturation factor γ (gmfg) is a key regulator of actin dynamics that was shown to be highly expressed in hematopoietic tissue. Our goal is to investigate the role and mechanism of gmfg in embryonic HSPC development. METHODS In-depth bioinformatics analysis of our published RNA-seq data identified gmfg as a cogent candidate gene implicated in HSPC development. Loss and gain-of-function strategies were applied to study the biological function of gmfg. Whole-mount in situ hybridization, confocal microscopy, flow cytometry, and western blotting were used to evaluate changes in the number of various hematopoietic cells and expression levels of cell proliferation, cell apoptosis and hematopoietic-related markers. RNA-seq was performed to screen signaling pathways responsible for gmfg deficiency-induced defects in HSPC initiation. The effect of gmfg on YAP sublocalization was assessed in vitro by utilizing HUVEC cell line. RESULTS We took advantage of zebrafish embryos to illustrate that loss of gmfg impaired HSPC initiation and maintenance. In gmfg-deficient embryos, the number of hemogenic endothelium and HSPCs was significantly reduced, with the accompanying decreased number of erythrocytes, myelocytes and lymphocytes. We found that blood flow modulates gmfg expression and gmfg overexpression could partially rescue the reduction of HSPCs in the absence of blood flow. Assays in zebrafish and HUVEC showed that gmfg deficiency suppressed the activity of YAP, a well-established blood flow mediator, by preventing its shuttling from cytoplasm to nucleus. During HSPC initiation, loss of gmfg resulted in Notch inactivation and the induction of Notch intracellular domain could partially restore the HSPC loss in gmfg-deficient embryos. CONCLUSIONS We conclude that gmfg mediates blood flow-induced HSPC maintenance via regulation of YAP, and contributes to HSPC initiation through the modulation of Notch signaling. Our findings reveal a brand-new aspect of gmfg function and highlight a novel mechanism for embryonic HSPC development.
Collapse
Affiliation(s)
- Honghu Li
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Qian Luo
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Shuyang Cai
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Ruxiu Tie
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Ye Meng
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Wei Shan
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Yulin Xu
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Xiangjun Zeng
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China
| | - Pengxu Qian
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China.
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China.
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China.
- Center of Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, 310012, People's Republic of China.
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310012, Zhejiang, People's Republic of China.
- School of Medicine, Zhejiang University, No. 866 Yuhangtang Road, Hangzhou, People's Republic of China.
| | - He Huang
- Bone Marrow Transplantation Center, School of Medicine, The First Affiliated Hospital, Zhejiang University, No. 79 Qingchun Road, Hangzhou, 310012, Zhejiang, People's Republic of China.
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, People's Republic of China.
- Institute of Hematology, Zhejiang University, Hangzhou, People's Republic of China.
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Hangzhou, People's Republic of China.
- Center of Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, 310012, People's Republic of China.
| |
Collapse
|
12
|
Lin DS, Trumpp A. Differential expression of endothelial protein C receptor (EPCR) in hematopoietic stem and multipotent progenitor cells in young and old mice. Cells Dev 2023; 174:203843. [PMID: 37080459 DOI: 10.1016/j.cdev.2023.203843] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/26/2023] [Accepted: 04/13/2023] [Indexed: 04/22/2023]
Abstract
Endothelial protein C receptor (EPCR) has emerged as one of the most conserved and reliable surface markers for the prospective identification and isolation of hematopoietic stem cells (HSCs). Prior studies have consistently demonstrated that EPCR expression enriches HSCs capable of long-term multilineage repopulation in both mouse and human across different hematopoietic tissues, including bone marrow (BM), fetal liver and ex vivo HSC expansion cultures. However, little is known about the expression profiles of EPCR in multipotent progenitor (MPP) populations located immediately downstream of HSCs in the hematopoietic hierarchy and which play a major role in sustaining lifelong blood cell production. Here, we incorporate EPCR antibody detection into a multi-parameter flow cytometric panel, which allows accurate identification of HSCs and five MPP subsets (MPP1-5) in mouse BM. Our data reveal that all MPP populations contain EPCR-expressing cells. Multipotent MPP1 and MPP5 contain higher proportion of EPCR+ cells compared to the more lineage-biased MPP2-4. Notably, high expression of EPCR enriches phenotypic HSC and MPP5, but not MPP1. Comparison of EPCR expression profiles between young and old BM reveals ageing mediated expansion of EPCR-expressing cells only in HSCs, but not in any of the MPP populations. Collectively, our study provides a comprehensive characterization of the surface expression pattern of EPCR in mouse HSC and MPP1-5 cells during normal and aged hematopoiesis.
Collapse
Affiliation(s)
- Dawn S Lin
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany.
| |
Collapse
|
13
|
Hou S, Liu C, Yao Y, Bai Z, Gong Y, Wang C, He J, You G, Zhang G, Liu B, Lan Y. Hematopoietic Stem Cell Development in Mammalian Embryos. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1442:1-16. [PMID: 38228955 DOI: 10.1007/978-981-99-7471-9_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Hematopoietic stem cells (HSCs) are situated at the top of the adult hematopoietic hierarchy in mammals and give rise to the majority of blood cells throughout life. Recently, with the advance of multiple single-cell technologies, researchers have unprecedentedly deciphered the cellular and molecular evolution, the lineage relationships, and the regulatory mechanisms underlying HSC emergence in mammals. In this review, we describe the precise vascular origin of HSCs in mouse and human embryos, emphasizing the conservation in the unambiguous arterial characteristics of the HSC-primed hemogenic endothelial cells (HECs). Serving as the immediate progeny of some HECs, functional pre-HSCs of mouse embryos can now be isolated at single-cell level using defined surface marker combinations. Heterogeneity regrading cell cycle status or lineage differentiation bias within HECs, pre-HSCs, or emerging HSCs in mouse embryos has been figured out. Several epigenetic regulatory mechanisms of HSC generation, including long noncoding RNA, DNA methylation modification, RNA splicing, and layered epigenetic modifications, have also been recently uncovered. In addition to that of HSCs, the cellular and molecular events underlying the development of multiple hematopoietic progenitors in human embryos/fetus have been unraveled with the use of series of single-cell technologies. Specifically, yolk sac-derived myeloid-biased progenitors have been identified as the earliest multipotent hematopoietic progenitors in human embryo, serving as an important origin of fetal liver monocyte-derived macrophages. Moreover, the development of multiple hematopoietic lineages in human embryos such as T and B lymphocytes, innate lymphoid cells, as well as myeloid cells like monocytes, macrophages, erythrocytes, and megakaryocytes has also been depicted and reviewed here.
Collapse
Affiliation(s)
- Siyuan Hou
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Chen Liu
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yingpeng Yao
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Zhijie Bai
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yandong Gong
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Chaojie Wang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Jian He
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Guoju You
- State Key Laboratory of Primate Biomedical Research, State Key Laboratory of Experimental Hematology, School of Medicine, Tsinghua University, Beijing, China
| | - Guangyu Zhang
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Bing Liu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yu Lan
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
14
|
Vanuytsel K, Yeung AK, Dowrey TW, Murphy GJ, Belkina AC. CPHEN-013: Comprehensive phenotyping of hematopoietic stem and progenitor cells in the human fetal liver. Cytometry A 2022; 101:903-908. [PMID: 35253987 DOI: 10.1002/cyto.a.24540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/21/2021] [Accepted: 01/11/2022] [Indexed: 01/27/2023]
Abstract
Hematopoietic stem cells (HSCs) reside at the top of the hematopoietic hierarchy and can give rise to all the mature blood cell types in our body, while at the same time maintaining a pool of HSCs through self-renewing divisions. This potential is reflected in their functional definition as cells that are capable of long-term multi-lineage engraftment upon transplantation. While all HSCs meet these criteria, subtle differences exist between developmentally different populations of these cells. Here we present a comprehensive overview of traditional and more recently described markers for phenotyping HSCs and their downstream progeny. To address the need to assess the growing number of surface molecules expressed in various HSC-enriched fractions at different developmental stages, we have developed an extensive multi-parameter spectral flow cytometry panel to phenotype hematopoietic stem and multipotent progenitor cells (HSC/MPPs) throughout development. In this study we then employ this panel to comprehensively profile the HSC compartment in the human fetal liver (FL), which is endowed with superior engraftment potential compared to postnatal sources. Spectral cytometry lends an improved resolution of marker expression to our comprehensive approach, allowing to extract combinatorial expression signatures of several relevant HSC/MPP markers to precisely characterize the HSC/MPP fraction in a variety of tissues.
Collapse
Affiliation(s)
- Kim Vanuytsel
- Section of Hematology and Medical Oncology, School of Medicine, Boston University, Boston, Massachusetts, USA.,Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, Massachusetts, USA
| | - Anthony K Yeung
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, Massachusetts, USA
| | - Todd W Dowrey
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, Massachusetts, USA
| | - George J Murphy
- Section of Hematology and Medical Oncology, School of Medicine, Boston University, Boston, Massachusetts, USA.,Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, Massachusetts, USA
| | - Anna C Belkina
- Flow Cytometry Core Facility, Boston University School of Medicine, Boston, Massachusetts, USA.,Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
15
|
RNAi screen identifies MTA1 as an epigenetic modifier of differentiation commitment in human HSPCs. Exp Hematol 2022; 115:20-29. [PMID: 36041657 DOI: 10.1016/j.exphem.2022.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/13/2022] [Accepted: 08/18/2022] [Indexed: 11/22/2022]
Abstract
The molecular mechanisms regulating key fate decisions of hematopoietic stem cells (HSCs) remain incompletely understood. Here, we targeted global shRNA libraries to primary human hematopoietic stem and progenitor cells (HSPCs) to screen for modifiers of self-renewal and differentiation, and identified metastasis-associated 1 (MTA1) as a negative regulator of human HSPC propagation in vitro. Knockdown of MTA1 by independent shRNAs in primary human cord blood (CB) HSPCs led to a cell expansion during culture and a relative accumulation of immature CD34+CD90+ cells with perturbed in vitro differentiation potential. Transplantation experiments in immunodeficient mice showed a significant reduction of human chimerism in both blood and bone marrow from HSPCs with knockdown of MTA1, possibly due to reduced maturation of blood cells. We further show that MTA1 associates with the nucleosome remodeling deacetylase (NuRD) complex in human HSPCs, and upon knockdown of MTA1 we observed an increase in H3K27Ac marks coupled with a downregulation of genes linked to differentiation towards the erythroid lineage. Altogether, our findings identify MTA1 as a novel regulator of human HSPCs in vitro and in vivo with critical functions for differentiation commitment.
Collapse
|
16
|
Vanuytsel K, Villacorta-Martin C, Lindstrom-Vautrin J, Wang Z, Garcia-Beltran WF, Vrbanac V, Parsons D, Lam EC, Matte TM, Dowrey TW, Kumar SS, Li M, Wang F, Yeung AK, Mostoslavsky G, Dries R, Campbell JD, Belkina AC, Balazs AB, Murphy GJ. Multi-modal profiling of human fetal liver hematopoietic stem cells reveals the molecular signature of engraftment. Nat Commun 2022; 13:1103. [PMID: 35232959 PMCID: PMC8888592 DOI: 10.1038/s41467-022-28616-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 01/21/2022] [Indexed: 12/18/2022] Open
Abstract
The human hematopoietic stem cell harbors remarkable regenerative potential that can be harnessed therapeutically. During early development, hematopoietic stem cells in the fetal liver undergo active expansion while simultaneously retaining robust engraftment capacity, yet the underlying molecular program responsible for their efficient engraftment remains unclear. Here, we profile 26,407 fetal liver cells at both the transcriptional and protein level including ~7,000 highly enriched and functional fetal liver hematopoietic stem cells to establish a detailed molecular signature of engraftment potential. Integration of transcript and linked cell surface marker expression reveals a generalizable signature defining functional fetal liver hematopoietic stem cells and allows for the stratification of enrichment strategies with high translational potential. More precisely, our integrated analysis identifies CD201 (endothelial protein C receptor (EPCR), encoded by PROCR) as a marker that can specifically enrich for engraftment potential. This comprehensive, multi-modal profiling of engraftment capacity connects a critical biological function at a key developmental timepoint with its underlying molecular drivers. As such, it serves as a useful resource for the field and forms the basis for further biological exploration of strategies to retain the engraftment potential of hematopoietic stem cells ex vivo or induce this potential during in vitro hematopoietic stem cell generation.
Collapse
Affiliation(s)
- Kim Vanuytsel
- Section of Hematology and Medical Oncology, School of Medicine, Boston University, Boston, MA, USA.
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA, USA.
| | - Carlos Villacorta-Martin
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA, USA
| | | | - Zhe Wang
- Division of Computational Biomedicine, School of Medicine, Boston University, Boston, MA, USA
| | | | | | - Dylan Parsons
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Evan C Lam
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Taylor M Matte
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA, USA
| | - Todd W Dowrey
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA, USA
| | - Sara S Kumar
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA, USA
| | - Mengze Li
- Division of Computational Biomedicine, School of Medicine, Boston University, Boston, MA, USA
| | - Feiya Wang
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA, USA
| | - Anthony K Yeung
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA, USA
| | - Gustavo Mostoslavsky
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA, USA
| | - Ruben Dries
- Section of Hematology and Medical Oncology, School of Medicine, Boston University, Boston, MA, USA
- Division of Computational Biomedicine, School of Medicine, Boston University, Boston, MA, USA
| | - Joshua D Campbell
- Division of Computational Biomedicine, School of Medicine, Boston University, Boston, MA, USA
| | - Anna C Belkina
- Department of Pathology and Laboratory Medicine, School of Medicine, Boston University, Boston, MA, USA
- Flow Cytometry Core Facility, School of Medicine, Boston University, Boston, MA, USA
| | | | - George J Murphy
- Section of Hematology and Medical Oncology, School of Medicine, Boston University, Boston, MA, USA.
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA, USA.
| |
Collapse
|
17
|
Amiri F, Kiani AA, Bahadori M, Roudkenar MH. Co-culture of mesenchymal stem cell spheres with hematopoietic stem cells under hypoxia: a cost-effective method to maintain self-renewal and homing marker expression. Mol Biol Rep 2021; 49:931-941. [PMID: 34741711 DOI: 10.1007/s11033-021-06912-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/29/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Hematopoietic stem cell (HSC) transplantation is considered a possible treatment option capable of curing various diseases. The aim of this study was the co-culturing of mesenchymal stem cell (MSC) spheres with HSCs under hypoxic condition to enhance the proliferation, self-renewal, stemness, and homing capacities of HSCs. METHODS AND RESULTS HSCs were expanded after being subjected to different conditions including cytokines without feeder (Cyto), co-culturing with adherent MSCs (MSC), co-culturing with adherent MSCs + hypoxia (MSC + Hyp), co-culturing with MSCs spheres (Sph-MSC), co-culturing with MSCs spheres + hypoxia (Sph-MSC + Hyp), co-culturing with MSC spheres + cytokines (Sph-MSC + Cyto). After 10 days, total nucleated cell (TNC) and CD34+/CD38- cell counts, colony-forming unit assay (CFU), long-term culture initiating cell (LTC-IC), the expression of endothelial protein C receptor (EPCR), nucleostemin (NS), nuclear factor I/X (Nfix) CXCR4, and VLA-4 were evaluated. The TNC, CD34+/CD38- cell count, CFU, and LTC-IC were higher in the Sph-MSC + Hyp and Sph-MSC + Cyto groups as compared with those of the MSC + Hyp group (P < 0.001). The expanded HSCs co-cultured with MSC spheres in combination with hypoxia expressed more EPCR, CXCR4, VLA-4, NS, and Nfix mRNA. The protein expression was also more up-regulated in the Sph-MSC + Cyto and Sph-MSC + Hyp groups. CONCLUSION Co-culturing HSCs with MSC spheres under hypoxic condition not only leads to higher cellular yield but also increases the expression of self-renewal and homing genes. Therefore, we suggest this approach as a simple and non-expensive strategy that might improve the transplantation efficiency of HSCs.
Collapse
Affiliation(s)
- Fatemeh Amiri
- Department of Medical Laboratory Sciences, School of Para Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Asghar Kiani
- Department of Hematology and Blood Transfusion, Lorestan University of Medical Sciences, Khorramabad, Lorestan, Iran
| | - Marzie Bahadori
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mehryar Habibi Roudkenar
- Cardiovascular Diseases Research Center, Department of Cardiology, Heshmat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran. .,Burn and Regenerative Research Center, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
18
|
Christopher AC, Venkatesan V, Karuppusamy KV, Srinivasan S, Babu P, Azhagiri MKK, C K, Bagchi A, Rajendiran V, Ravi NS, Kumar S, Marepally SK, Mohankumar KM, Srivastava A, Velayudhan SR, Thangavel S. Preferential expansion of human CD34+CD133+CD90+ hematopoietic stem cells enhances gene-modified cell frequency for gene therapy. Hum Gene Ther 2021; 33:188-201. [PMID: 34486377 DOI: 10.1089/hum.2021.089] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
CD34+CD133+CD90+ hematopoietic stem cells (HSCs) are responsible for long-term multi-lineage hematopoiesis and the high frequency of gene-modified HSCs is crucial for the success of hematopoietic stem and progenitor cell (HSPC) gene therapy. However, the ex vivo culture and gene manipulation steps of HSPC graft preparation significantly reduce the frequency of HSCs, thus necessitating large doses of HSPCs and reagents for the manipulation. Here, we identified a combination of small molecules, Resveratrol, UM729, and SR1 that preferentially expands CD34+CD133+CD90+ HSCs over other subpopulations of adult HSPCs in ex vivo culture. The preferential expansion enriches the HSCs in ex vivo culture, enhances the adhesion and results in a 6-fold increase in the long-term engraftment in NSG mice. Further, the culture enriched HSCs are more responsive to gene modification by lentiviral transduction and gene editing, increasing the frequency of gene-modified HSCs up to 10-fold in vivo. The yield of gene-modified HSCs obtained by the culture enrichment is similar to the sort-purification of HSCs and superior to Cyclosporin-H treatment. Our study addresses a critical challenge of low frequency of gene-modified HSCs in HSPC graft by developing and demonstrating a facile HSPC culture condition that increases the frequency of gene-modified cells in vivo. This strategy will improve the outcome of HSPC gene therapy and also simplify the gene manipulation process.
Collapse
Affiliation(s)
| | - Vigneshwaran Venkatesan
- Center for Stem Cell Research, 302927, Vellore, Tamil nadu, India.,Manipal Academy of Higher Education, 76793, Manipal, Karnataka, India;
| | - Karthik V Karuppusamy
- Center for Stem Cell Research, 302927, Vellore, Tamil nadu, India.,Manipal Academy of Higher Education, 76793, Manipal, Karnataka, India;
| | | | - Prathibha Babu
- Center for Stem Cell Research, 302927, Vellore, Tamil nadu, India.,Manipal Academy of Higher Education, 76793, Manipal, Karnataka, India;
| | - Manoj Kumar K Azhagiri
- Center for Stem Cell Research, 302927, Vellore, Tamil nadu, India.,Manipal Academy of Higher Education, 76793, Manipal, Karnataka, India;
| | - Karthik C
- Center for Stem Cell Research, 302927, Vellore, Tamil nadu, India;
| | - Abhirup Bagchi
- Center for Stem Cell Research, 302927, Vellore, Tamil nadu, India;
| | | | - Nithin Sam Ravi
- Center for Stem Cell Research, 302927, Vellore, Tamil Nadu, India;
| | - Sanjay Kumar
- Christian Medical College and Hospital Vellore, 30025, Center for Stem Cell Research, Vellore, Tamil Nadu, India;
| | | | | | - Alok Srivastava
- Christian Medical College, Centre for Stem Cell Research, CMC Campus, Bagayam, Vellore, Tamilnadu, India, 632002.,Christian Medical College, Haematology, Ida Scudder Road, Vellore, Tamil Nadu, India, 632004;
| | | | - Saravanabhavan Thangavel
- Center for Stem Cell Research, 302927, Christian Medical College Campus Bagayam,, Vellore, Tamil nadu, India, 632002;
| |
Collapse
|
19
|
Gan Y, He J, Zhu J, Xu Z, Wang Z, Yan J, Hu O, Bai Z, Chen L, Xie Y, Jin M, Huang S, Liu B, Liu P. Spatially defined single-cell transcriptional profiling characterizes diverse chondrocyte subtypes and nucleus pulposus progenitors in human intervertebral discs. Bone Res 2021; 9:37. [PMID: 34400611 PMCID: PMC8368097 DOI: 10.1038/s41413-021-00163-z] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 04/30/2021] [Accepted: 06/10/2021] [Indexed: 02/07/2023] Open
Abstract
A comprehensive understanding of the cellular heterogeneity and molecular mechanisms underlying the development, homeostasis, and disease of human intervertebral disks (IVDs) remains challenging. Here, the transcriptomic landscape of 108 108 IVD cells was mapped using single-cell RNA sequencing of three main compartments from young and adult healthy IVDs, including the nucleus pulposus (NP), annulus fibrosus, and cartilage endplate (CEP). The chondrocyte subclusters were classified based on their potential regulatory, homeostatic, and effector functions in extracellular matrix (ECM) homeostasis. Notably, in the NP, a PROCR+ resident progenitor population showed enriched colony-forming unit-fibroblast (CFU-F) activity and trilineage differentiation capacity. Finally, intercellular crosstalk based on signaling network analysis uncovered that the PDGF and TGF-β cascades are important cues in the NP microenvironment. In conclusion, a single-cell transcriptomic atlas that resolves spatially regulated cellular heterogeneity together with the critical signaling that underlies homeostasis will help to establish new therapeutic strategies for IVD degeneration in the clinic.
Collapse
Affiliation(s)
- Yibo Gan
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China ,grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jian He
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Jun Zhu
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhengyang Xu
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Zhong Wang
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jing Yan
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Ou Hu
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhijie Bai
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Lin Chen
- grid.410570.70000 0004 1760 6682Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yangli Xie
- grid.410570.70000 0004 1760 6682Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Min Jin
- grid.410570.70000 0004 1760 6682Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shuo Huang
- grid.410570.70000 0004 1760 6682Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Bing Liu
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China ,grid.11135.370000 0001 2256 9319State Key Laboratory of Experimental Hematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China ,grid.258164.c0000 0004 1790 3548Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Peng Liu
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China ,grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
20
|
Gan Y, He J, Zhu J, Xu Z, Wang Z, Yan J, Hu O, Bai Z, Chen L, Xie Y, Jin M, Huang S, Liu B, Liu P. Spatially defined single-cell transcriptional profiling characterizes diverse chondrocyte subtypes and nucleus pulposus progenitors in human intervertebral discs. Bone Res 2021; 9:37. [PMID: 34400611 PMCID: PMC8368097 DOI: 10.1038/s41413-021-00163-z+10.1038/s41413-021-00163-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 04/30/2021] [Accepted: 06/10/2021] [Indexed: 01/21/2024] Open
Abstract
A comprehensive understanding of the cellular heterogeneity and molecular mechanisms underlying the development, homeostasis, and disease of human intervertebral disks (IVDs) remains challenging. Here, the transcriptomic landscape of 108 108 IVD cells was mapped using single-cell RNA sequencing of three main compartments from young and adult healthy IVDs, including the nucleus pulposus (NP), annulus fibrosus, and cartilage endplate (CEP). The chondrocyte subclusters were classified based on their potential regulatory, homeostatic, and effector functions in extracellular matrix (ECM) homeostasis. Notably, in the NP, a PROCR+ resident progenitor population showed enriched colony-forming unit-fibroblast (CFU-F) activity and trilineage differentiation capacity. Finally, intercellular crosstalk based on signaling network analysis uncovered that the PDGF and TGF-β cascades are important cues in the NP microenvironment. In conclusion, a single-cell transcriptomic atlas that resolves spatially regulated cellular heterogeneity together with the critical signaling that underlies homeostasis will help to establish new therapeutic strategies for IVD degeneration in the clinic.
Collapse
Affiliation(s)
- Yibo Gan
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jian He
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Jun Zhu
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhengyang Xu
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Zhong Wang
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jing Yan
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Ou Hu
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhijie Bai
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Lin Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yangli Xie
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Min Jin
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shuo Huang
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Bing Liu
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China.
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China.
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China.
| | - Peng Liu
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China.
- State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University (Third Military Medical University), Chongqing, China.
| |
Collapse
|
21
|
Spatially defined single-cell transcriptional profiling characterizes diverse chondrocyte subtypes and nucleus pulposus progenitors in human intervertebral discs. Bone Res 2021; 9:37. [PMID: 34400611 PMCID: PMC8368097 DOI: 10.1038/s41413-021-00163-z 10.1038/s41413-021-00163-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023] Open
Abstract
A comprehensive understanding of the cellular heterogeneity and molecular mechanisms underlying the development, homeostasis, and disease of human intervertebral disks (IVDs) remains challenging. Here, the transcriptomic landscape of 108 108 IVD cells was mapped using single-cell RNA sequencing of three main compartments from young and adult healthy IVDs, including the nucleus pulposus (NP), annulus fibrosus, and cartilage endplate (CEP). The chondrocyte subclusters were classified based on their potential regulatory, homeostatic, and effector functions in extracellular matrix (ECM) homeostasis. Notably, in the NP, a PROCR+ resident progenitor population showed enriched colony-forming unit-fibroblast (CFU-F) activity and trilineage differentiation capacity. Finally, intercellular crosstalk based on signaling network analysis uncovered that the PDGF and TGF-β cascades are important cues in the NP microenvironment. In conclusion, a single-cell transcriptomic atlas that resolves spatially regulated cellular heterogeneity together with the critical signaling that underlies homeostasis will help to establish new therapeutic strategies for IVD degeneration in the clinic.
Collapse
|
22
|
Grzywa TM, Nowis D, Golab J. The role of CD71 + erythroid cells in the regulation of the immune response. Pharmacol Ther 2021; 228:107927. [PMID: 34171326 DOI: 10.1016/j.pharmthera.2021.107927] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/13/2021] [Accepted: 05/18/2021] [Indexed: 02/07/2023]
Abstract
Complex regulation of the immune response is necessary to support effective defense of an organism against hostile invaders and to maintain tolerance to harmless microorganisms and autoantigens. Recent studies revealed previously unappreciated roles of CD71+ erythroid cells (CECs) in regulation of the immune response. CECs physiologically reside in the bone marrow where erythropoiesis takes place. Under stress conditions, CECs are enriched in some organs outside of the bone marrow as a result of extramedullary erythropoiesis. However, the role of CECs goes well beyond the production of erythrocytes. In neonates, increased numbers of CECs contribute to their vulnerability to infectious diseases. On the other side, neonatal CECs suppress activation of immune cells in response to abrupt colonization with commensal microorganisms after delivery. CECs are also enriched in the peripheral blood of pregnant women as well as in the placenta and are responsible for the regulation of feto-maternal tolerance. In patients with cancer, anemia leads to increased frequency of CECs in the peripheral blood contributing to diminished antiviral and antibacterial immunity, as well as to accelerated cancer progression. Moreover, recent studies revealed the role of CECs in HIV and SARS-CoV-2 infections. CECs use a full arsenal of mechanisms to regulate immune response. These cells suppress proinflammatory responses of myeloid cells and T-cell proliferation by the depletion of ʟ-arginine by arginase. Moreover, CECs produce reactive oxygen species to decrease T-cell proliferation. CECs also secrete cytokines, including transforming growth factor β (TGF-β), which promotes T-cell differentiation into regulatory T-cells. Here, we comprehensively describe the role of CECs in orchestrating immune response and indicate some therapeutic approaches that might be used to regulate their effector functions in the treatment of human conditions.
Collapse
Affiliation(s)
- Tomasz M Grzywa
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5 Street, 02-097 Warsaw, Poland; Doctoral School, Medical University of Warsaw, Zwirki and Wigury 61 Street, 02-091 Warsaw, Poland; Laboratory of Experimental Medicine, Medical University of Warsaw, Nielubowicza 5 Street, 02-097 Warsaw, Poland.
| | - Dominika Nowis
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5 Street, 02-097 Warsaw, Poland; Laboratory of Experimental Medicine, Medical University of Warsaw, Nielubowicza 5 Street, 02-097 Warsaw, Poland.
| | - Jakub Golab
- Department of Immunology, Medical University of Warsaw, Nielubowicza 5 Street, 02-097 Warsaw, Poland; Centre of Preclinical Research, Medical University of Warsaw, Banacha 1b Street, 02-097 Warsaw, Poland.
| |
Collapse
|
23
|
Shan W, Yu Q, Long Y, Luo Q, Li H, Han Y, Xu Y, Fu S, Zeng X, Wei C, Gao Y, Li X, Li X, Zhang L, Liu L, Chen M, Qian P, Huang H. Enhanced HSC-like cell generation from mouse pluripotent stem cells in a 3D induction system cocultured with stromal cells. Stem Cell Res Ther 2021; 12:353. [PMID: 34147128 PMCID: PMC8214308 DOI: 10.1186/s13287-021-02434-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/06/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Decades of efforts have attempted to differentiate the pluripotent stem cells (PSCs) into truly functional hematopoietic stem cells (HSCs), yet the problems of low differentiation efficiency in vitro and poor hematopoiesis reconstitution in vivo still exist, mainly attributing to the lack of solid, reproduced, or pursued differentiation system. METHODS In this study, we established an in vitro differentiation system yielding in vivo hematopoietic reconstitution hematopoietic cells from mouse PSCs through a 3D induction system followed by coculture with OP9 stromal cells. The in vivo hematopoietic reconstitution potential of c-kit+ cells derived from the mouse PSCs was evaluated via m-NSG transplantation assay. Flow cytometry analysis, RNA-seq, and cell cycle analysis were used to detect the in vitro hematopoietic ability of endothelial protein C receptor (EPCR, CD201) cells generated in our induction system. RESULTS The c-kit+ cells from 3D self-assembling peptide induction system followed by the OP9 coculture system possessed apparently superiority in terms of in vivo repopulating activity than that of 3D induction system followed by the 0.1% gelatin culture. We interestingly found that our 3D+OP9 system enriched a higher percentage of CD201+c-kit+cells that showed more similar HSC-like features such as transcriptome level and CFU formation ability than CD201-c-kit+cells, which have not been reported in the field of mouse PSCs hematopoietic differentiation. Moreover, CD201+ hematopoietic cells remained in a relatively slow cycling state, consistent with high expression levels of P57 and Ccng2. Further, we innovatively demonstrated that notch signaling pathway is responsible for in vitro CD201+ hematopoietic cell induction from mouse PSCs. CONCLUSIONS Altogether, our findings lay a foundation for improving the efficiency of hematopoietic differentiation and generating in vivo functional HSC-like cells from mouse PSCs for clinical application.
Collapse
Affiliation(s)
- Wei Shan
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, No.79 Qingchun Road, Hangzhou, Zhejiang, PR China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, PR China.,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, PR China.,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, PR China
| | - Qin Yu
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Yan Long
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, No.79 Qingchun Road, Hangzhou, Zhejiang, PR China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, PR China.,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, PR China.,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, PR China
| | - Qian Luo
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, No.79 Qingchun Road, Hangzhou, Zhejiang, PR China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, PR China.,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, PR China.,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, PR China
| | - Honghu Li
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, No.79 Qingchun Road, Hangzhou, Zhejiang, PR China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, PR China.,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, PR China.,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, PR China
| | - Yingli Han
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, No.79 Qingchun Road, Hangzhou, Zhejiang, PR China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, PR China.,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, PR China.,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, PR China
| | - Yulin Xu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, No.79 Qingchun Road, Hangzhou, Zhejiang, PR China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, PR China.,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, PR China.,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, PR China
| | - Shan Fu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, No.79 Qingchun Road, Hangzhou, Zhejiang, PR China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, PR China.,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, PR China.,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, PR China
| | - Xiangjun Zeng
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, No.79 Qingchun Road, Hangzhou, Zhejiang, PR China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, PR China.,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, PR China.,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, PR China
| | - Cong Wei
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, No.79 Qingchun Road, Hangzhou, Zhejiang, PR China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, PR China.,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, PR China.,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, PR China
| | - Yang Gao
- Department of Hematology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 Qingchun East Rd., Hangzhou, 310016, Zhejiang, PR China
| | - Xiaoqing Li
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, No.79 Qingchun Road, Hangzhou, Zhejiang, PR China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, PR China.,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, PR China.,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, PR China
| | - Xia Li
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, No.79 Qingchun Road, Hangzhou, Zhejiang, PR China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, PR China.,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, PR China.,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, PR China
| | - Lifei Zhang
- Department of Hematology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, No. 3 Qingchun East Rd., Hangzhou, 310016, Zhejiang, PR China
| | - Lizhen Liu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, No.79 Qingchun Road, Hangzhou, Zhejiang, PR China.,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, PR China.,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, PR China.,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, PR China
| | - Ming Chen
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, PR China.,Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Pengxu Qian
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, PR China. .,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, PR China. .,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, PR China. .,Center of Stem Cell and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, 310012, PR China.
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, School of Medicine, Zhejiang University, No.79 Qingchun Road, Hangzhou, Zhejiang, PR China. .,Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, PR China. .,Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, Zhejiang, PR China. .,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, PR China.
| |
Collapse
|
24
|
Lysine-specific demethylase 1A restricts ex vivo propagation of human HSCs and is a target of UM171. Blood 2021; 136:2151-2161. [PMID: 32582923 DOI: 10.1182/blood.2020005827] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/07/2020] [Indexed: 12/23/2022] Open
Abstract
Culture conditions in which hematopoietic stem cells (HSCs) can be expanded for clinical benefit are highly sought after. Here, we report that inhibition of the epigenetic regulator lysine-specific histone demethylase 1A (LSD1) induces a rapid expansion of human cord blood-derived CD34+ cells and promotes in vitro propagation of long-term repopulating HSCs by preventing differentiation. The phenotype and molecular characteristics of cells treated with LSD1 inhibitors were highly similar to cells treated with UM171, an agent promoting expansion of HSCs through undefined mechanisms and currently being tested in clinical trials. Strikingly, we found that LSD1, as well as other members of the LSD1-containing chromatin remodeling complex CoREST, is rapidly polyubiquitinated and degraded upon UM171 treatment. CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 depletion of the CoREST core member, RCOR1, resulted in expansion of CD34+ cells similar to LSD1 inhibition and UM171. Taken together, LSD1 and CoREST restrict HSC expansion and are principal targets of UM171, forming a mechanistic basis for the HSC-promoting activity of UM171.
Collapse
|
25
|
Continuous NF-κB pathway inhibition promotes expansion of human phenotypical hematopoietic stem/progenitor cells through metabolism regulation. Exp Cell Res 2021; 399:112468. [PMID: 33428903 DOI: 10.1016/j.yexcr.2020.112468] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 12/24/2020] [Accepted: 12/27/2020] [Indexed: 01/24/2023]
Abstract
Hematopoietic stem/progenitor cells (HSPCs) ex vivo expansion is critical in facilitating their widespread clinical application. NF-κB pathway is implicated in the energy homeostasis and metabolic adaptation. To explore the effect of NF-κB pathway on the ex vivo HSPC expansion and metabolism, the 50 nM-1 μM inhibitor of NF-κB pathway TPCA-1 was used to expand cord blood derived CD34+ cells in serum-free culture. The expansion folds, function, mitochondrial profile and metabolism of HSPCs were determined. After 10 days of culture with 100 nM TPCA-1, the expansion of total cells, CD34+CD38- cells, and CD34+CD38-CD45RA-CD90+CD49f+ cells were significantly increased compared to the cytokine priming alone. Notably, TPCA-1 treatment generated ~ 2-fold greater percentage of CD34+EPCR+ and CD34+CD38-CD45RA-CD90+CD49f+ cells compared to cytokine only conditions. Moreover, TPCA-1 expanded CD34+ cells displayed enhanced serial colonies forming potential and secondary expansion capability. NF-κB inhibition increased the expression of self-renewal related genes, while downregulated the expression of mitochondrial biogenesis regulator (Pgc1α) and mitochondrial chaperones and proteases (ClpP, Hsp10, Hsp60). Mitochondrial mass and membrane potential were markedly decreased with TPCA-1 treatment, leading to the reduced mitochondrial reactive oxygen species (ROS) level in HSPCs. NF-κB inhibition displayed augmented glycolysis rate with compromising mitochondrial metabolism. This study demonstrated that NF-κB pathway inhibition improved glycolysis and limited ROS production that promoted the ex vivo expansion and maintenance of functional HSPCs.
Collapse
|
26
|
Tracing the first hematopoietic stem cell generation in human embryo by single-cell RNA sequencing. Cell Res 2019; 29:881-894. [PMID: 31501518 PMCID: PMC6888893 DOI: 10.1038/s41422-019-0228-6] [Citation(s) in RCA: 145] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 08/13/2019] [Indexed: 12/20/2022] Open
Abstract
Tracing the emergence of the first hematopoietic stem cells (HSCs) in human embryos, particularly the scarce and transient precursors thereof, is so far challenging, largely due to the technical limitations and the material rarity. Here, using single-cell RNA sequencing, we constructed the first genome-scale gene expression landscape covering the entire course of endothelial-to-HSC transition during human embryogenesis. The transcriptomically defined HSC-primed hemogenic endothelial cells (HECs) were captured at Carnegie stage (CS) 12–14 in an unbiased way, showing an unambiguous feature of arterial endothelial cells (ECs) with the up-regulation of RUNX1, MYB and ANGPT1. Importantly, subcategorizing CD34+CD45− ECs into a CD44+ population strikingly enriched HECs by over 10-fold. We further mapped the developmental path from arterial ECs via HSC-primed HECs to hematopoietic stem progenitor cells, and revealed a distinct expression pattern of genes that were transiently over-represented upon the hemogenic fate choice of arterial ECs, including EMCN, PROCR and RUNX1T1. We also uncovered another temporally and molecularly distinct intra-embryonic HEC population, which was detected mainly at earlier CS 10 and lacked the arterial feature. Finally, we revealed the cellular components of the putative aortic niche and potential cellular interactions acting on the HSC-primed HECs. The cellular and molecular programs that underlie the generation of the first HSCs from HECs in human embryos, together with the ability to distinguish the HSC-primed HECs from others, will shed light on the strategies for the production of clinically useful HSCs from pluripotent stem cells.
Collapse
|
27
|
Endothelial protein C receptor supports hematopoietic stem cell engraftment and expansion in Mpl-deficient mice. Blood 2019; 133:1465-1478. [DOI: 10.1182/blood-2018-03-837344] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 01/15/2019] [Indexed: 11/20/2022] Open
Abstract
Abstract
Thrombopoietin (Thpo)/myeloproliferative leukemia virus oncogene (Mpl) signaling controls hematopoietic stem cell (HSC) self-renewal and quiescence; however, how these 2 seemingly opposing functions are controlled is not well understood. By transplantation of lentiviral-transduced hematopoietic cells in the Mpl-deficient mouse model, we addressed whether known or predicted Thpo target genes were able to rescue the Mpl-deficient phenotype of the mice. Among the tested genes, we identified endothelial protein C receptor (Epcr) to expand HSCs with the long-term (LT)-HSC surface phenotype in Mpl−/− mice and to enable secondary transplantation of Mpl-deficient bone marrow (BM). Epcr-transduced Mpl−/− HSCs enter quiescence earlier after transplantation than control-transduced Mpl−/− cells, and upregulated expression of the anti-apoptotic gene Bcl-xL. Also, in the wild-type background, Epcr expression marked the engrafting population in the BM. Furthermore, Epcr expression in Mpl−/− hematopoiesis increased the number of megakaryocytes in the BM. In vitro Thpo supported the surface expression of Epcr on primary murine hematopoietic stem and progenitor cells. With these data, we add new insights into Thpo-dependent influence on HSC engraftment after transplantation. This may be of use for the in vitro manipulation of HSCs, also in the context of gene therapy.
Collapse
|
28
|
Mahony CB, Bertrand JY. How HSCs Colonize and Expand in the Fetal Niche of the Vertebrate Embryo: An Evolutionary Perspective. Front Cell Dev Biol 2019; 7:34. [PMID: 30915333 PMCID: PMC6422921 DOI: 10.3389/fcell.2019.00034] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 02/25/2019] [Indexed: 12/18/2022] Open
Abstract
Rare hematopoietic stem cells (HSCs) can self-renew, establish the entire blood system and represent the basis of regenerative medicine applied to hematological disorders. Clinical use of HSCs is however limited by their inefficient expansion ex vivo, creating a need to further understand HSC expansion in vivo. After embryonic HSCs are born from the hemogenic endothelium, they migrate to the embryonic/fetal niche, where the future adult HSC pool is established by considerable expansion. This takes place at different anatomical sites and is controlled by numerous signals. HSCs then migrate to their adult niche, where they are maintained throughout adulthood. Exactly how HSC expansion is controlled during embryogenesis remains to be characterized and is an important step to improve the therapeutic use of HSCs. We will review the current knowledge of HSC expansion in the different fetal niches across several model organisms and highlight possible clinical applications.
Collapse
Affiliation(s)
- Christopher B Mahony
- Department of Pathology and Immunology, Faculty of Medicine, CMU, University of Geneva, Geneva, Switzerland
| | - Julien Y Bertrand
- Department of Pathology and Immunology, Faculty of Medicine, CMU, University of Geneva, Geneva, Switzerland
| |
Collapse
|