1
|
Swann JW, Zhang R, Verovskaya EV, Calero-Nieto FJ, Wang X, Proven MA, Shyu PT, Guo XE, Göttgens B, Passegué E. Inflammation perturbs hematopoiesis by remodeling specific compartments of the bone marrow niche. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.612751. [PMID: 39314376 PMCID: PMC11419052 DOI: 10.1101/2024.09.12.612751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Hematopoietic stem and progenitor cells (HSPC) are regulated by interactions with stromal cells in the bone marrow (BM) cavity, which can be segregated into two spatially defined central marrow (CM) and endosteal (Endo) compartments. However, the importance of this spatial compartmentalization for BM responses to inflammation and neoplasia remains largely unknown. Here, we extensively validate a combination of scRNA-seq profiling and matching flow cytometry isolation that reproducibly identifies 7 key CM and Endo populations across mouse strains and accurately surveys both niche locations. We demonstrate that different perturbations exert specific effects on different compartments, with type I interferon responses causing CM mesenchymal stromal cells to adopt an inflammatory phenotype associated with overproduction of chemokines modulating local monocyte dynamics in the surrounding microenvironment. Our results provide a comprehensive method for molecular and functional stromal characterization and highlight the importance of altered stomal cell activity in regulating hematopoietic responses to inflammatory challenges.
Collapse
|
2
|
Pereira AL, Galli S, Nombela‐Arrieta C. Bone marrow niches for hematopoietic stem cells. Hemasphere 2024; 8:e133. [PMID: 39086665 PMCID: PMC11289431 DOI: 10.1002/hem3.133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/05/2024] [Accepted: 05/06/2024] [Indexed: 08/02/2024] Open
Abstract
Hematopoietic stem cells (HSCs) are the cornerstone of the hematopoietic system. HSCs sustain the continuous generation of mature blood derivatives while self-renewing to preserve a relatively constant pool of progenitors throughout life. Yet, long-term maintenance of functional HSCs exclusively takes place in association with their native tissue microenvironment of the bone marrow (BM). HSCs have been long proposed to reside in fixed and identifiable anatomical units found in the complex BM tissue landscape, which control their identity and fate in a deterministic manner. In the last decades, tremendous progress has been made in the dissection of the cellular and molecular fabric of the BM, the structural organization governing tissue function, and the plethora of interactions established by HSCs. Nonetheless, a holistic model of the mechanisms controlling HSC regulation in their niche is lacking to date. Here, we provide an overview of our current understanding of BM anatomy, HSC localization, and crosstalk within local cellular neighborhoods in murine and human tissues, and highlight fundamental open questions on how HSCs functionally integrate in the BM microenvironment.
Collapse
Affiliation(s)
- Ana Luísa Pereira
- Department of Medical Oncology and HematologyUniversity Hospital and University of ZurichZurichSwitzerland
| | - Serena Galli
- Department of Medical Oncology and HematologyUniversity Hospital and University of ZurichZurichSwitzerland
| | - César Nombela‐Arrieta
- Department of Medical Oncology and HematologyUniversity Hospital and University of ZurichZurichSwitzerland
| |
Collapse
|
3
|
Sukhnanan K, Ross JR, Chao NJ, Chen BJ. Endothelial Cell Derived Extracellular Vesicles and Hematopoiesis. Radiat Res 2024; 202:215-226. [PMID: 38918003 DOI: 10.1667/rade-24-00039.1] [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: 01/31/2024] [Accepted: 04/23/2024] [Indexed: 06/27/2024]
Abstract
Extracellular vesicles (EVs) have been recognized as a novel way of cell-to-cell communication in the last several decades. It is believed that EVs exert their functions on nearby or distant cells through transfer of the cargo that they carry. In this review, we focus on EVs produced by endothelial cells, with emphasis on their role in hematopoiesis. We first describe how endothelial cells interact with hematopoietic stem/progenitor cells during development and in disease conditions. We then discuss EVs, ranging from their subtypes to isolation methods and analysis of EVs. With the above background information, we next review the literature related to endothelial cell derived EVs (ECEVs), including physiological functions and their clinical uses. In the last sections, we summarize the current results about the effect of ECEVs on hematopoiesis under physiological and stress conditions.
Collapse
Affiliation(s)
| | - Joel R Ross
- Department of Medicine, Duke University, Durham, North Carolina
| | - Nelson J Chao
- Department of Medicine, Duke University, Durham, North Carolina
- Department of Pathology, Duke University, Durham, North Carolina
- Department of Immunology, Duke University, Durham, North Carolina
- Duke Cancer Institute, Duke University, Durham, North Carolina
- Duke Global Health Institute, Duke University, Durham, North Carolina
| | - Benny J Chen
- Department of Medicine, Duke University, Durham, North Carolina
- Department of Immunology, Duke University, Durham, North Carolina
- Duke Fitzpatrick Institute for Photonics, Duke University, Durham, North Carolina
- Duke Regeneration Center, Duke University, Durham, North Carolina
| |
Collapse
|
4
|
Huang Y, Liu Z, Li M, Wang D, Ye J, Hu Q, Zhang Q, Lin Y, Chen R, Liang X, Li X, Lin X. Deciphering the impact of aging on splenic endothelial cell heterogeneity and immunosenescence through single-cell RNA sequencing analysis. Immun Ageing 2024; 21:48. [PMID: 39026350 PMCID: PMC11256597 DOI: 10.1186/s12979-024-00452-1] [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: 02/04/2024] [Accepted: 07/01/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUND Aging is associated with significant structural and functional changes in the spleen, leading to immunosenescence, yet the detailed effects on splenic vascular endothelial cells (ECs) and their immunomodulatory roles are not fully understood. In this study, a single-cell RNA (scRNA) atlas of EC transcriptomes from young and aged mouse spleens was constructed to reveal age-related molecular changes, including increased inflammation and reduced vascular development and also the potential interaction between splenic endothelial cells and immune cells. RESULTS Ten clusters of splenic endothelial cells were identified. DEGs analysis across different EC clusters revealed the molecular changes with aging, showing the increase in the overall inflammatory microenvironment and the loss in vascular development function of aged ECs. Notably, four EC clusters with immunological functions were identified, suggesting an Endothelial-to-Immune-like Cell Transition (EndICLT) potentially driven by aging. Pseudotime analysis of the Immunology4 cluster further indicated a possible aging-induced transitional state, potentially initiated by Ctss gene activation. Finally, the effects of aging on cell signaling communication between different EC clusters and immune cells were analyzed. CONCLUSIONS This comprehensive atlas elucidates the complex interplay between ECs and immune cells in the aging spleen, offering new insights into endothelial heterogeneity, reprogramming, and the mechanisms of immunosenescence.
Collapse
Affiliation(s)
- Yanjing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, 510060, China
| | - Zhong Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, 510060, China
| | - Mengke Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, 510060, China
| | - Dongliang Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, 510060, China
| | - Jinguo Ye
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, 510060, China
| | - Qiuling Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, 510060, China
| | - Qikai Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, 510060, China
| | - Yuheng Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, 510060, China
| | - Rongxin Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, 510060, China
| | - Xuanwei Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, 510060, China
| | - Xingyi Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, 510060, China.
| | - Xianchai Lin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-sen University, Guangzhou, 510060, China.
| |
Collapse
|
5
|
Karima G, Kim HD. Unlocking the regenerative key: Targeting stem cell factors for bone renewal. J Tissue Eng 2024; 15:20417314241287491. [PMID: 39479284 PMCID: PMC11523181 DOI: 10.1177/20417314241287491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 09/12/2024] [Indexed: 11/02/2024] Open
Abstract
Stem cell factors (SCFs) are pivotal factors existing in both soluble and membrane-bound forms, expressed by endothelial cells (ECs) and fibroblasts throughout the body. These factors enhance cell growth, viability, and migration in multipotent cell lineages. The preferential expression of SCF by arteriolar ECs indicates that arterioles create a unique microenvironment tailored to hematopoietic stem cells (HSCs). Insufficiency of SCF within bone marrow (BM)-derived adipose tissue results in decreased their overall cellularity, affecting HSCs and their immediate progenitors critical for generating diverse blood cells and maintaining the hematopoietic microenvironment. SCF deficiency disrupts BM function, impacting the production and differentiation of HSCs. Additionally, deleting SCF from adipocytes reduces lipogenesis, highlighting the crucial role of SCF/c-kit signaling in controlling lipid accumulation. This review elucidates the sources, roles, mechanisms, and molecular strategies of SCF in bone renewal, offering a comprehensive overview of recent advancements, challenges, and future directions for leveraging SCF as a key agent in regenerative medicine.
Collapse
Affiliation(s)
- Gul Karima
- Department of Polymer Science and Engineering, Korea National University of Transportation, Chungju, Republic of Korea
| | - Hwan D. Kim
- Department of Polymer Science and Engineering, Korea National University of Transportation, Chungju, Republic of Korea
- Department of IT Convergence (Brain Korea Plus 21), Korea National University of Transportation, Chungju, Republic of Korea
- Department of Biomedical Engineering, Korea National University of Transportation, Chungju, Republic of Korea
| |
Collapse
|
6
|
Adzraku SY, Wang G, Cao C, Bao Y, Wang Y, Smith AO, Du Y, Wang H, Li Y, Xu K, Qiao J, Ju W, Zeng L. Robo4 inhibits gamma radiation-induced permeability of a murine microvascular endothelial cell by regulating the junctions. Cell Mol Biol Lett 2023; 28:2. [PMID: 36647012 PMCID: PMC9843922 DOI: 10.1186/s11658-022-00413-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/19/2022] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Hematopoietic stem cell transplantation involves irradiation preconditioning which causes bone marrow endothelial cell dysfunction. While much emphasis is on the reconstitution of hematopoietic stem cells in the bone marrow microenvironment, endothelial cell preservation is indispensable to overcome the preconditioning damages. This study aims to ascertain the role of Roundabout 4 (Robo4) in regulating irradiation-induced damage to the endothelium. METHODS Microvascular endothelial cells were treated with γ-radiation to establish an endothelial cell injury model. Robo4 expression in the endothelial cells was manipulated employing lentiviral-mediated RNAi and gene overexpression technology before irradiation treatment. The permeability of endothelial cells was measured using qPCR, immunocytochemistry, and immunoblotting to analyze the effect on the expression and distribution of junctional molecules, adherens junctions, tight junctions, and gap junctions. Using Transwell endothelial monolayer staining, FITC-Dextran permeability, and gap junction-mediated intercellular communication (GJIC) assays, we determined the changes in endothelial functions after Robo4 gene manipulation and irradiation. Moreover, we measured the proportion of CD31 expression in endothelial cells by flow cytometry. We analyzed variations between two or multiple groups using Student's t-tests and ANOVA. RESULTS Ionizing radiation upregulates Robo4 expression but disrupts endothelial junctional molecules. Robo4 deletion causes further degradation of endothelial junctions hence increasing the permeability of the endothelial cell monolayer. Robo4 knockdown in microvascular endothelial cells increases the degradation and delocalization of ZO-1, PECAM-1, occludin, and claudin-5 molecules after irradiation. Conversely, connexin 43 expression increases after silencing Robo4 in endothelial cells to induce permeability but are readily destroyed when exposed to 10 Gy of gamma radiation. Also, Robo4 knockdown enhances Y731-VE-cadherin phosphorylation leading to the depletion and destabilization of VE-cadherin at the endothelial junctions following irradiation. However, Robo4 overexpression mitigates irradiation-induced degradation of tight junctional proteins and stabilizes claudin-5 and ZO-1 distribution. Finally, the enhanced expression of Robo4 ameliorates the irradiation-induced depletion of VE-cadherin and connexin 43, improves the integrity of microvascular endothelial cell junctions, and decreases permeability. CONCLUSION This study reveals that Robo4 maintains microvascular integrity after radiation preconditioning treatment by regulating endothelial permeability and protecting endothelial functions. Our results also provided a potential mechanism to repair the bone marrow vascular niche after irradiation by modulating Robo4 expression.
Collapse
Affiliation(s)
- Seyram Yao Adzraku
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China ,Xuzhou Ruihu Health Management Consulting Co., Ltd, Xuzhou, 221002 China
| | - Guozhang Wang
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China ,Xuzhou Ruihu Health Management Consulting Co., Ltd, Xuzhou, 221002 China
| | - Can Cao
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China ,Xuzhou Ruihu Health Management Consulting Co., Ltd, Xuzhou, 221002 China
| | - Yurong Bao
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Yizhou Wang
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Alhaji Osman Smith
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Yuwei Du
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Haiyang Wang
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Yue Li
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Kailin Xu
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Jianlin Qiao
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| | - Wen Ju
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China ,Xuzhou Ruihu Health Management Consulting Co., Ltd, Xuzhou, 221002 China
| | - Lingyu Zeng
- grid.417303.20000 0000 9927 0537Blood Diseases Institute, Xuzhou Medical University, Xuzhou, 221002 China ,Key Laboratory of Bone Marrow Stem Cell, Xuzhou, 221002 Jiangsu China ,grid.413389.40000 0004 1758 1622Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002 China
| |
Collapse
|
7
|
Wang Z, Jiang D, Vergel-Rodriguez M, Nogalska A, Lu R. Lineage tracking to reveal the fate of hematopoietic stem cells influenced by Flk2 - multipotent progenitors after transplantation. Exp Mol Med 2023; 55:205-214. [PMID: 36639717 PMCID: PMC9898540 DOI: 10.1038/s12276-022-00922-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 08/21/2022] [Accepted: 11/16/2022] [Indexed: 01/15/2023] Open
Abstract
After transplantation, hematopoietic stem cells (HSCs) sustain blood cell regeneration throughout the patient's life. Recent studies suggest that several types of mature blood cells provide feedback signals to regulate HSC fate. However, the potential feedback effect of hematopoietic progenitor cells has not been characterized to date. The present investigation demonstrated that multipotent progenitors (MPPs) promoted T cell production of HSCs when both cell types were cotransplanted in mice. Using genetic barcodes to track individual HSCs in mice, we found that the increased T cell production by HSCs was associated with the combined effects of altered lineage bias and clonal expansion during HSC differentiation. We showed that MPP and HSC co-transplantation promoted the multilineage differentiation of HSCs in the short term while preserving lymphoid-specialized HSC differentiation in the long term. Our findings indicate that MPPs derived from HSCs regulate the fate of HSCs after bone marrow transplantation.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Jinfeng Laboratory, Chongqing, 401329, China
- Medical Center of Hematology, Xinqiao Hospital, Army Medical University, Chongqing, 400037, China
| | - Du Jiang
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Mary Vergel-Rodriguez
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Anna Nogalska
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Rong Lu
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| |
Collapse
|
8
|
Akindona FA, Frederico SC, Hancock JC, Gilbert MR. Exploring the origin of the cancer stem cell niche and its role in anti-angiogenic treatment for glioblastoma. Front Oncol 2022; 12:947634. [PMID: 36091174 PMCID: PMC9454306 DOI: 10.3389/fonc.2022.947634] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/08/2022] [Indexed: 11/21/2022] Open
Abstract
Cancer stem cells are thought to be the main drivers of tumorigenesis for malignancies such as glioblastoma (GBM). They are maintained through a close relationship with the tumor vasculature. Previous literature has well-characterized the components and signaling pathways for maintenance of this stem cell niche, but details on how the niche initially forms are limited. This review discusses development of the nonmalignant neural and hematopoietic stem cell niches in order to draw important parallels to the malignant environment. We then discuss what is known about the cancer stem cell niche, its relationship with angiogenesis, and provide a hypothesis for its development in GBM. A better understanding of the mechanisms of development of the tumor stem cell niche may provide new insights to potentially therapeutically exploit.
Collapse
Affiliation(s)
- Funto A. Akindona
- Neuro-Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD, United States
| | - Stephen C. Frederico
- Neuro-Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD, United States
- University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - John C. Hancock
- Neuro-Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD, United States
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Mark R. Gilbert
- Neuro-Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Mark R. Gilbert,
| |
Collapse
|
9
|
Gopal A, Ibrahim R, Fuller M, Umlandt P, Parker J, Tran J, Chang L, Wegrzyn-Woltosz J, Lam J, Li J, Lu M, Karsan A. TIRAP drives myelosuppression through an Ifnγ-Hmgb1 axis that disrupts the endothelial niche in mice. J Exp Med 2022; 219:212987. [PMID: 35089323 PMCID: PMC8932532 DOI: 10.1084/jem.20200731] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 09/15/2021] [Accepted: 12/16/2021] [Indexed: 12/31/2022] Open
Abstract
Inflammation is associated with bone marrow failure syndromes, but how specific molecules impact the bone marrow microenvironment is not well elucidated. We report a novel role for the miR-145 target, Toll/interleukin-1 receptor domain containing adaptor protein (TIRAP), in driving bone marrow failure. We show that TIRAP is overexpressed in various types of myelodysplastic syndromes (MDS) and suppresses all three major hematopoietic lineages. TIRAP expression promotes up-regulation of Ifnγ, leading to myelosuppression through Ifnγ-Ifnγr–mediated release of the alarmin, Hmgb1, which disrupts the bone marrow endothelial niche. Deletion of Ifnγ blocks Hmgb1 release and is sufficient to reverse the endothelial defect and restore myelopoiesis. Contrary to current dogma, TIRAP-activated Ifnγ-driven bone marrow suppression is independent of T cell function or pyroptosis. In the absence of Ifnγ, TIRAP drives myeloproliferation, implicating Ifnγ in suppressing the transformation of MDS to acute leukemia. These findings reveal novel, noncanonical roles of TIRAP, Hmgb1, and Ifnγ in the bone marrow microenvironment and provide insight into the pathophysiology of preleukemic syndromes.
Collapse
Affiliation(s)
- Aparna Gopal
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Rawa Ibrahim
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Megan Fuller
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Patricia Umlandt
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Jeremy Parker
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Jessica Tran
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Linda Chang
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joanna Wegrzyn-Woltosz
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jeffrey Lam
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Jenny Li
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Melody Lu
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada
| | - Aly Karsan
- Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, British Columbia, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
10
|
Abstract
Fracture healing is a complex, multistep process that is highly sensitive to mechanical signaling. To optimize repair, surgeons prescribe immediate weight-bearing as-tolerated within 24 hours after surgical fixation; however, this recommendation is based on anecdotal evidence and assessment of bulk healing outcomes (e.g., callus size, bone volume, etc.). Given challenges in accurately characterizing the mechanical environment and the ever-changing properties of the regenerate, the principles governing mechanical regulation of repair, including their cell and molecular basis, are not yet well defined. However, the use of mechanobiological rodent models, and their relatively large genetic toolbox, combined with recent advances in imaging approaches and single-cell analyses is improving our understanding of the bone microenvironment in response to loading. This review describes the identification and characterization of distinct cell populations involved in bone healing and highlights the most recent findings on mechanical regulation of bone homeostasis and repair with an emphasis on osteo-angio coupling. A discussion on aging and its impact on bone mechanoresponsiveness emphasizes the need for novel mechanotherapeutics that can re-sensitize skeletal stem and progenitor cells to physical rehabilitation protocols.
Collapse
Affiliation(s)
- Tareq Anani
- Department of Orthopedic Surgery, New York University Langone Health, New York, NY 10010, USA
| | - Alesha B Castillo
- Department of Orthopedic Surgery, New York University Langone Health, New York, NY 10010, USA; Department of Biomedical Engineering, Tandon School of Engineering, New York University, New York, NY 11201, USA; Department of Veterans Affairs, New York Harbor Healthcare System, Manhattan Campus, New York, NY 10010, USA.
| |
Collapse
|
11
|
Soukup AA, Matson DR, Liu P, Johnson KD, Bresnick EH. Conditionally pathogenic genetic variants of a hematopoietic disease-suppressing enhancer. SCIENCE ADVANCES 2021; 7:eabk3521. [PMID: 34890222 PMCID: PMC8664263 DOI: 10.1126/sciadv.abk3521] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/22/2021] [Indexed: 05/11/2023]
Abstract
Human genetic variants are classified on the basis of potential pathogenicity to guide clinical decisions. However, mechanistic uncertainties often preclude definitive categorization. Germline coding and enhancer variants within the hematopoietic regulator GATA2 create a bone marrow failure and leukemia predisposition. The conserved murine enhancer promotes hematopoietic stem cell (HSC) genesis, and a single-nucleotide human variant in an Ets motif attenuates chemotherapy-induced hematopoietic regeneration. We describe “conditionally pathogenic” (CP) enhancer motif variants that differentially affect hematopoietic development and regeneration. The Ets motif variant functioned autonomously in hematopoietic cells to disrupt hematopoiesis. Because an epigenetically silenced normal allele can exacerbate phenotypes of a pathogenic heterozygous variant, we engineered a bone marrow failure model harboring the Ets motif variant and a severe enhancer mutation on the second allele. Despite normal developmental hematopoiesis, regeneration in response to chemotherapy, inflammation, and a therapeutic HSC mobilizer was compromised. The CP paradigm informs mechanisms underlying phenotypic plasticity and clinical genetics.
Collapse
Affiliation(s)
- Alexandra A. Soukup
- Wisconsin Blood Cancer Research Institute, Department of Cell and Regenerative Biology, Wisconsin Institutes for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- UW Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Daniel R. Matson
- Wisconsin Blood Cancer Research Institute, Department of Cell and Regenerative Biology, Wisconsin Institutes for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- UW Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Peng Liu
- University of Wisconsin Carbone Cancer Center, Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Kirby D. Johnson
- Wisconsin Blood Cancer Research Institute, Department of Cell and Regenerative Biology, Wisconsin Institutes for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- UW Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Emery H. Bresnick
- Wisconsin Blood Cancer Research Institute, Department of Cell and Regenerative Biology, Wisconsin Institutes for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- UW Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
12
|
Developmental angiocrine diversification of endothelial cells for organotypic regeneration. Dev Cell 2021; 56:3042-3051. [PMID: 34813766 DOI: 10.1016/j.devcel.2021.10.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/17/2021] [Accepted: 10/26/2021] [Indexed: 02/08/2023]
Abstract
Adult organs are vascularized by specialized blood vessels. In addition to inter-organ vascular heterogeneity, each organ is arborized by structurally and functionally diversified populations of endothelial cells (ECs). The molecular pathways that are induced to orchestrate inter- and intra- organ vascular heterogeneity and zonation are shaped during development and fully specified postnatally. Notably, intra-organ specialization of ECs is associated with induction of angiocrine factors that guide cross-talk between ECs and parenchymal cells, establishing co-zonated vascular regions within each organ. In this review, we describe how microenvironmental tissue-specific biophysical, biochemical, immune, and inflammatory cues dictate the specialization of ECs with zonated functions. We delineate how physiological and biophysical stressors in the developing liver, lung, and kidney vasculature induce specialization of capillary beds. Deciphering mechanisms by which vascular microvasculature diversity is attained could set the stage for treating regenerative disorders and promote healing of organs without provoking fibrosis.
Collapse
|
13
|
Abstract
Hematopoietic stem cells (HSCs) are ultimately responsible for the lifelong renewal of all blood cell lineages. In the bone marrow (BM), HSCs reside in specialized microenvironments referred to as the "niche." HSC niche consists of complex components including heterogeneous cell populations, growth factors, and extracellular matrix molecules. The crosstalk between HSCs and their niche is essential to regulate the survival, self-renewal, migration, quiescence, and differentiation of HSCs. The application of mice models with endogenous ablation of specific cell types, advanced imaging technologies, high-throughput single-cell RNA sequencing, and single-cell mass cytometry methods have provided deep insights into communications between HSCs and niche cells. In this chapter, we have focused on three important cell types in the BM niche: mesenchymal stem cells (MSCs), osteoblasts (OBs), and endothelial cells (ECs). In order to address the interaction between HSCs and these three cell populations in BM niche, we have described methodology for (1) collecting total BM from femur and tibia of C57BL/6 mice; (2) analyzing or sorting of MSCs, OBs, and ECs based on the selection of surface markers CD45, Ter119, CD31, Sca1, and CD51 with flow cytometry; and (3) co-culturing the sorted cells with purified HSCs for further functional assays of HSCs.
Collapse
|
14
|
Control of Tumor Progression by Angiocrine Factors. Cancers (Basel) 2021; 13:cancers13112610. [PMID: 34073394 PMCID: PMC8198241 DOI: 10.3390/cancers13112610] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/21/2021] [Accepted: 05/21/2021] [Indexed: 12/24/2022] Open
Abstract
Tumor progression, therapy resistance and metastasis are profoundly controlled by the tumor microenvironment. The contribution of endothelial cells to tumor progression was initially only attributed to the formation of new blood vessels (angiogenesis). Research in the last decade has revealed however that endothelial cells control their microenvironment through the expression of membrane-bound and secreted factors. Such angiocrine functions are frequently hijacked by cancer cells, which deregulate the signaling pathways controlling the expression of angiocrine factors. Here, we review the crosstalk between cancer cells and endothelial cells and how this contributes to the cancer stem cell phenotype, epithelial to mesenchymal transition, immunosuppression, remodeling of the extracellular matrix and intravasation of cancer cells into the bloodstream. We also address the long-distance crosstalk of a primary tumor with endothelial cells at the pre-metastatic niche and how this contributes to metastasis.
Collapse
|
15
|
Crippa S, Santi L, Berti M, De Ponti G, Bernardo ME. Role of ex vivo Expanded Mesenchymal Stromal Cells in Determining Hematopoietic Stem Cell Transplantation Outcome. Front Cell Dev Biol 2021; 9:663316. [PMID: 34017834 PMCID: PMC8129582 DOI: 10.3389/fcell.2021.663316] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/17/2021] [Indexed: 02/06/2023] Open
Abstract
Overall, the human organism requires the production of ∼1 trillion new blood cells per day. Such goal is achieved via hematopoiesis occurring within the bone marrow (BM) under the tight regulation of hematopoietic stem and progenitor cell (HSPC) homeostasis made by the BM microenvironment. The BM niche is defined by the close interactions of HSPCs and non-hematopoietic cells of different origin, which control the maintenance of HSPCs and orchestrate hematopoiesis in response to the body’s requirements. The activity of the BM niche is regulated by specific signaling pathways in physiological conditions and in case of stress, including the one induced by the HSPC transplantation (HSCT) procedures. HSCT is the curative option for several hematological and non-hematological diseases, despite being associated with early and late complications, mainly due to a low level of HSPC engraftment, impaired hematopoietic recovery, immune-mediated graft rejection, and graft-versus-host disease (GvHD) in case of allogenic transplant. Mesenchymal stromal cells (MSCs) are key elements of the BM niche, regulating HSPC homeostasis by direct contact and secreting several paracrine factors. In this review, we will explore the several mechanisms through which MSCs impact on the supportive activity of the BM niche and regulate HSPC homeostasis. We will further discuss how the growing understanding of such mechanisms have impacted, under a clinical point of view, on the transplantation field. In more recent years, these results have instructed the design of clinical trials to ameliorate the outcome of HSCT, especially in the allogenic setting, and when low doses of HSPCs were available for transplantation.
Collapse
Affiliation(s)
- Stefania Crippa
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ludovica Santi
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Margherita Berti
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giada De Ponti
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Centro Ricerca M. Tettamanti, Department of Pediatrics, University of Milano-Bicocca, Monza, Italy
| | - Maria Ester Bernardo
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Pediatric Immunohematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, Milan, Italy.,University Vita-Salute San Raffaele, Faculty of Medicine, Milan, Italy
| |
Collapse
|
16
|
Barcia Durán JG, Lu T, Houghton S, Geng F, Schreiner R, Xiang J, Rafii S, Redmond D, Lis R. Endothelial Jak3 expression enhances pro-hematopoietic angiocrine function in mice. Commun Biol 2021; 4:406. [PMID: 33767339 PMCID: PMC7994450 DOI: 10.1038/s42003-021-01846-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 02/12/2021] [Indexed: 02/08/2023] Open
Abstract
Jak3 is the only non-promiscuous member of the Jak family of secondary messengers. Studies to date have focused on understanding and targeting the cell-autonomous role of Jak3 in immunity, while functional Jak3 expression outside the hematopoietic system remains largely unreported. We show that Jak3 is expressed in endothelial cells across hematopoietic and non-hematopoietic organs, with heightened expression in the bone marrow. The bone marrow niche is understood as a network of different cell types that regulate hematopoietic function. We show that the Jak3-/- bone marrow niche is deleterious for the maintenance of long-term repopulating hematopoietic stem cells (LT-HSCs) and that JAK3-overexpressing endothelial cells have increased potential to expand LT-HSCs in vitro. This work may serve to identify a novel function for a highly specific tyrosine kinase in the bone marrow vascular niche and to further characterize the LT-HSC function of sinusoidal endothelium.
Collapse
Affiliation(s)
- José Gabriel Barcia Durán
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Tyler Lu
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Sean Houghton
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Fuqiang Geng
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Ryan Schreiner
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Department of Ophthalmology, Margaret Dyson Vision Research Institute, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Jenny Xiang
- Genomics Resources Core Facility, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Shahin Rafii
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY, 10065, USA
| | - David Redmond
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA.
| | - Raphaël Lis
- Ansary Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY, 10065, USA.
- Ronald O. Perelman and Claudia Cohen Center for Reproductive Medicine and Infertility, Weill Cornell Medicine, New York, NY, 10065, USA.
| |
Collapse
|
17
|
Mosteo L, Storer J, Batta K, Searle EJ, Duarte D, Wiseman DH. The Dynamic Interface Between the Bone Marrow Vascular Niche and Hematopoietic Stem Cells in Myeloid Malignancy. Front Cell Dev Biol 2021; 9:635189. [PMID: 33777944 PMCID: PMC7991089 DOI: 10.3389/fcell.2021.635189] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/10/2021] [Indexed: 12/19/2022] Open
Abstract
Hematopoietic stem cells interact with bone marrow niches, including highly specialized blood vessels. Recent studies have revealed the phenotypic and functional heterogeneity of bone marrow endothelial cells. This has facilitated the analysis of the vascular microenvironment in steady state and malignant hematopoiesis. In this review, we provide an overview of the bone marrow microenvironment, focusing on refined analyses of the marrow vascular compartment performed in mouse studies. We also discuss the emerging role of the vascular niche in “inflamm-aging” and clonal hematopoiesis, and how the endothelial microenvironment influences, supports and interacts with hematopoietic cells in acute myeloid leukemia and myelodysplastic syndromes, as exemplar states of malignant myelopoiesis. Finally, we provide an overview of strategies for modulating these bidirectional interactions to therapeutic effect in myeloid malignancies.
Collapse
Affiliation(s)
- Laura Mosteo
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Joanna Storer
- Epigenetics of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| | - Kiran Batta
- Epigenetics of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom
| | - Emma J Searle
- Epigenetics of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom.,Department of Haematology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Delfim Duarte
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal.,Department of Biomedicine, Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal.,Department of Onco-Hematology, Instituto Português de Oncologia (IPO)-Porto, Porto, Portugal
| | - Daniel H Wiseman
- Epigenetics of Haematopoiesis Group, Division of Cancer Sciences, The University of Manchester, Manchester, United Kingdom.,Department of Haematology, The Christie NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
18
|
Abstract
Obesity and obesity-related diseases like type 2 diabetes (T2D) are prominent global health issues; therefore, there is a need to better understand the mechanisms underlying these conditions. The onset of obesity is characterized by accumulation of proinflammatory cells, including Ly6chi monocytes (which differentiate into proinflammatory macrophages) and neutrophils, in metabolic tissues. This shift toward chronic, low-grade inflammation is an obese-state hallmark and highly linked to metabolic disorders and other obesity comorbidities. The mechanisms that induce and maintain increased inflammatory myelopoiesis are of great interest, with a recent focus on how obesity affects more primitive hematopoietic cells. The hematopoietic system is constantly replenished by proper regulation of hematopoietic stem and progenitor (HSPC) pools in the BM. While early research suggests that chronic obesity promotes expansion of myeloid-skewed HSPCs, the involvement of the hematopoietic stem cell (HSC) niche in regulating obesity-induced myelopoiesis remains undefined. In this review, we explore the role of the multicellular HSC niche in hematopoiesis and inflammation, and the potential contribution of this niche to the hematopoietic response to obesity. This review further aims to summarize the potential HSC niche involvement as a target of obesity-induced inflammation and a driver of obesity-induced myelopoiesis.
Collapse
|
19
|
Golub R. The Notch signaling pathway involvement in innate lymphoid cell biology. Biomed J 2020; 44:133-143. [PMID: 33863682 PMCID: PMC8178581 DOI: 10.1016/j.bj.2020.12.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/11/2020] [Accepted: 12/16/2020] [Indexed: 12/30/2022] Open
Abstract
The role of Notch in the immune system was first described in the late 90s. Reports revealed that Notch is one of the most conserved developmental pathways involved in diverse biological processes such as the development, differentiation, survival and functions of many immune populations. Here, we provide an extended view of the pleiotropic effects of the Notch signaling on the innate lymphoid cell (ILC) biology. We review the current knowledge on Notch signaling in the regulation of ILC differentiation, plasticity and functions in diverse tissue types and at both the fetal and adult developmental stages. ILCs are early responder cells that secrete a large panel of cytokines after stimulation. By controlling the abundance of ILCs and the specificity of their release, the Notch pathway is also implicated in the regulation of their functions. The Notch pathway is therefore an important player in both ILC cell fate decision and ILC immune response.
Collapse
Affiliation(s)
- Rachel Golub
- Unit of Lymphocytes and Immunity, Department of Immunology, Institut Pasteur, Paris, France.
| |
Collapse
|
20
|
Ju W, Lu W, Ding L, Bao Y, Hong F, Chen Y, Gao H, Xu X, Wang G, Wang W, Zhang X, Fu C, Qi K, Li Z, Xu K, Qiao J, Zeng L. PEDF promotes the repair of bone marrow endothelial cell injury and accelerates hematopoietic reconstruction after bone marrow transplantation. J Biomed Sci 2020; 27:91. [PMID: 32873283 PMCID: PMC7466818 DOI: 10.1186/s12929-020-00685-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 08/26/2020] [Indexed: 12/27/2022] Open
Abstract
Background Preconditioning before bone marrow transplantation such as irradiation causes vascular endothelial cells damage and promoting the repair of damaged endothelial cells is beneficial for hematopoietic reconstitution. Pigment epithelium-derived factor (PEDF) regulates vascular permeability. However, PEDF’s role in the repair of damaged endothelial cells during preconditioning remains unclear. The purpose of our study is to investigate PEDF’s effect on preconditioning-induced damage of endothelial cells and hematopoietic reconstitution. Methods Damaged endothelial cells induced by irradiation was co-cultured with hematopoietic stem cells (HSC) in the absence or presence of PEDF followed by analysis of HSC number, cell cycle, colony formation and differentiation. In addition, PEDF was injected into mice model of bone marrow transplantation followed by analysis of bone marrow injury, HSC number and peripheral hematopoietic reconstitution as well as the secretion of cytokines (SCF, TGF-β, IL-6 and TNF-α). Comparisons between two groups were performed by student t-test and multiple groups by one-way or two-way ANOVA. Results Damaged endothelial cells reduced HSC expansion and colony formation, induced HSC cell cycle arrest and apoptosis and promoted HSC differentiation as well as decreased PEDF expression. Addition of PEDF increased CD144 expression in damaged endothelial cells and inhibited the increase of endothelial permeability, which were abolished after addition of PEDF receptor inhibitor Atglistatin. Additionally, PEDF ameliorated the inhibitory effect of damaged endothelial cells on HSC expansion in vitro. Finally, PEDF accelerated hematopoietic reconstitution after bone marrow transplantation in mice and promoted the secretion of SCF, TGF-β and IL-6. Conclusions PEDF inhibits the increased endothelial permeability induced by irradiation and reverse the inhibitory effect of injured endothelial cells on hematopoietic stem cells and promote hematopoietic reconstruction.
Collapse
Affiliation(s)
- Wen Ju
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wenyi Lu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Lan Ding
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yurong Bao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Fei Hong
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yuting Chen
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Hui Gao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiaoqi Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Guozhang Wang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Weiwei Wang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Chunling Fu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Kunming Qi
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zhenyu Li
- Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China.,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Kailin Xu
- Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China. .,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| | - Jianlin Qiao
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China. .,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China. .,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| | - Lingyu Zeng
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China. .,Key Laboratory of Bone Marrow Stem Cell, Jiangsu Province, Xuzhou, China. .,Department of Hematology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
21
|
Li H, Pei H, Wang S, Zhang B, Fan Z, Liu Y, Xie X, Yang Z, Xu L, Jia Y, Bai Y, Han Y, Chen L, He L, Nan X, Yue W, Pei X. Arterial endothelium creates a permissive niche for expansion of human cord blood hematopoietic stem and progenitor cells. Stem Cell Res Ther 2020; 11:358. [PMID: 32799928 PMCID: PMC7429738 DOI: 10.1186/s13287-020-01880-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/22/2020] [Accepted: 08/06/2020] [Indexed: 12/03/2022] Open
Abstract
Background Although cord blood (CB) offers promise for treatment of patients with high-risk hematological malignancies and immune disorders, the limited numbers of hematopoietic stem cell (HSC)/progenitor cell in a CB unit and straitened circumstances in expanding ex vivo make it quite challenging to develop the successful cell therapies. Methods In this study, a novel strategy has been developed to support ex vivo expansion of hematopoietic stem and progenitor cells (HSPCs) by coculture with engineered human umbilical arterial endothelial cells (HuAECs-E4orf1-GFP), which expresses E4ORF1 stably by using a retroviral system. Results Coculture of CD34+ hCB cells with HuAECs-E4orf1-GFP resulted in generation of considerably more total nucleated cells, CD34+CD38−, and CD34+CD38−CD90+ HSPCs in comparison with that of cytokines alone or that of coculture with human umbilical vein endothelial cells (HuVECs) after 14-day amplification. The in vitro multilineage differentiation potential and in vivo repopulating capacity of the expanded hematopoietic cells cocultured with HuAECs-E4orf1-GFP were also markedly enhanced compared with the other two control groups. DLL4, a major determinant of arterial endothelial cell (EC) identity, was associated with CD34+ hCB cells amplified on HuAECs-E4orf1-GFP. Conclusions Collectively, we demonstrated that HuAECs acted as a permissive niche in facilitating expansion of HSPCs. Our study further implicated that the crucial factors and related pathways presented in HuAECs may give a hint to maintain self-renewal of bona fide HSCs.
Collapse
Affiliation(s)
- Huilin Li
- Stem Cell and Regenerative Medicine Lab, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Haiyun Pei
- Experimental Hematology and Biochemistry Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China. .,South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou, 510005, China.
| | - Sihan Wang
- Stem Cell and Regenerative Medicine Lab, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China.,South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou, 510005, China
| | - Bowen Zhang
- Experimental Hematology and Biochemistry Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China.,South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou, 510005, China
| | - Zeng Fan
- Stem Cell and Regenerative Medicine Lab, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Yiming Liu
- Stem Cell and Regenerative Medicine Lab, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China.,South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou, 510005, China
| | - Xiaoyan Xie
- Stem Cell and Regenerative Medicine Lab, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China.,South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou, 510005, China
| | - Zhou Yang
- Stem Cell and Regenerative Medicine Lab, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Lei Xu
- Stem Cell and Regenerative Medicine Lab, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Yali Jia
- Experimental Hematology and Biochemistry Lab, Beijing Institute of Radiation Medicine, Beijing, 100850, China.,South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou, 510005, China
| | - Yun Bai
- Stem Cell and Regenerative Medicine Lab, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Yi Han
- South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou, 510005, China
| | - Lin Chen
- Stem Cell and Regenerative Medicine Lab, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China.,South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou, 510005, China
| | - Lijuan He
- Stem Cell and Regenerative Medicine Lab, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China.,South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou, 510005, China
| | - Xue Nan
- Stem Cell and Regenerative Medicine Lab, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China.,South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou, 510005, China
| | - Wen Yue
- Stem Cell and Regenerative Medicine Lab, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China. .,South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou, 510005, China.
| | - Xuetao Pei
- Stem Cell and Regenerative Medicine Lab, Institute of Health Service and Transfusion Medicine, Beijing, 100850, China. .,South China Research Center for Stem Cell & Regenerative Medicine, SCIB, Guangzhou, 510005, China.
| |
Collapse
|
22
|
Lin KH, Chiang JC, Ho YH, Yao CL, Lee H. Lysophosphatidic Acid and Hematopoiesis: From Microenvironmental Effects to Intracellular Signaling. Int J Mol Sci 2020; 21:ijms21062015. [PMID: 32188052 PMCID: PMC7139687 DOI: 10.3390/ijms21062015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 03/13/2020] [Accepted: 03/13/2020] [Indexed: 02/06/2023] Open
Abstract
Vertebrate hematopoiesis is a complex physiological process that is tightly regulated by intracellular signaling and extracellular microenvironment. In recent decades, breakthroughs in lineage-tracing technologies and lipidomics have revealed the existence of numerous lipid molecules in hematopoietic microenvironment. Lysophosphatidic acid (LPA), a bioactive phospholipid molecule, is one of the identified lipids that participates in hematopoiesis. LPA exhibits various physiological functions through activation of G-protein-coupled receptors. The functions of these LPARs have been widely studied in stem cells, while the roles of LPARs in hematopoietic stem cells have rarely been examined. Nonetheless, mounting evidence supports the importance of the LPA-LPAR axis in hematopoiesis. In this article, we have reviewed regulation of hematopoiesis in general and focused on the microenvironmental and intracellular effects of the LPA in hematopoiesis. Discoveries in these areas may be beneficial to our understanding of blood-related disorders, especially in the context of prevention and therapy for anemia.
Collapse
Affiliation(s)
- Kuan-Hung Lin
- Department of Life Science, National Taiwan University, Taipei 10617, Taiwan; (K.-H.L.); (J.-C.C.)
| | - Jui-Chung Chiang
- Department of Life Science, National Taiwan University, Taipei 10617, Taiwan; (K.-H.L.); (J.-C.C.)
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ya-Hsuan Ho
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Haematology, University of Cambridge, Cambridge CB2 0AW, UK;
| | - Chao-Ling Yao
- Department of Chemical Engineering and Materials Science, Yuan Ze University, Taoyuan 32003, Taiwan;
| | - Hsinyu Lee
- Department of Life Science, National Taiwan University, Taipei 10617, Taiwan; (K.-H.L.); (J.-C.C.)
- Department of Electrical Engineering, National Taiwan University, Taipei 10617, Taiwan
- Angiogenesis Research Center, National Taiwan University, Taipei 10617, Taiwan
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei 10617, Taiwan
- Center for Biotechnology, National Taiwan University, Taipei 10617, Taiwan
- Correspondence: ; Tel.: +8862-3366-2499; Fax: +8862-2363-6837
| |
Collapse
|
23
|
A Mathematical Model of the Transition from Normal Hematopoiesis to the Chronic and Accelerated-Acute Stages in Myeloid Leukemia. MATHEMATICS 2020. [DOI: 10.3390/math8030376] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
A mathematical model given by a two-dimensional differential system is introduced in order to understand the transition process from the normal hematopoiesis to the chronic and accelerated-acute stages in chronic myeloid leukemia. A previous model of Dingli and Michor is refined by introducing a new parameter in order to differentiate the bone marrow microenvironment sensitivities of normal and mutant stem cells. In the light of the new parameter, the system now has three distinct equilibria corresponding to the normal hematopoietic state, to the chronic state, and to the accelerated-acute phase of the disease. A characterization of the three hematopoietic states is obtained based on the stability analysis. Numerical simulations are included to illustrate the theoretical results.
Collapse
|
24
|
Pinho S, Frenette PS. Haematopoietic stem cell activity and interactions with the niche. Nat Rev Mol Cell Biol 2020; 20:303-320. [PMID: 30745579 DOI: 10.1038/s41580-019-0103-9] [Citation(s) in RCA: 618] [Impact Index Per Article: 123.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The haematopoietic stem cell (HSC) microenvironment in the bone marrow, termed the niche, ensures haematopoietic homeostasis by controlling the proliferation, self-renewal, differentiation and migration of HSCs and progenitor cells at steady state and in response to emergencies and injury. Improved methods for HSC isolation, driven by advances in single-cell and molecular technologies, have led to a better understanding of their behaviour, heterogeneity and lineage fate and of the niche cells and signals that regulate their function. Niche regulatory signals can be in the form of cell-bound or secreted factors and other local physical cues. A combination of technological advances in bone marrow imaging and genetic manipulation of crucial regulatory factors has enabled the identification of several candidate cell types regulating the niche, including both non-haematopoietic (for example, perivascular mesenchymal stem and endothelial cells) and HSC-derived (for example, megakaryocytes, macrophages and regulatory T cells), with better topographical understanding of HSC localization in the bone marrow. Here, we review advances in our understanding of HSC regulation by niches during homeostasis, ageing and cancer, and we discuss their implications for the development of therapies to rejuvenate aged HSCs or niches or to disrupt self-reinforcing malignant niches.
Collapse
Affiliation(s)
- Sandra Pinho
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York, NY, USA.,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY, USA.,Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, NY, USA
| | - Paul S Frenette
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York, NY, USA. .,Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY, USA. .,Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, NY, USA.
| |
Collapse
|
25
|
Kikuchi-Taura A, Okinaka Y, Takeuchi Y, Ogawa Y, Maeda M, Kataoka Y, Yasui T, Kimura T, Gul S, Claussen C, Boltze J, Taguchi A. Bone Marrow Mononuclear Cells Activate Angiogenesis via Gap Junction-Mediated Cell-Cell Interaction. Stroke 2020; 51:1279-1289. [PMID: 32075549 DOI: 10.1161/strokeaha.119.028072] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background and Purpose- Bone marrow mononuclear cells (BM-MNCs) are a rich source of hematopoietic stem cells and have been widely used in experimental therapies for patients with ischemic diseases. Activation of angiogenesis is believed to be one of major BM-MNC mode of actions, but the essential mechanism by which BM-MNCs activate angiogenesis have hitherto been elusive. The objective of this study is to reveal the mechanism how BM-MNCs activate angiogenesis. Methods- We have evaluated the effect of direct cell-cell interaction between BM-MNC and endothelial cell on uptake of VEGF (vascular endothelial growth factor) into endothelial cells in vitro. Cerebral ischemia model was used to evaluate the effects of direct cell-cell interaction with transplanted BM-MNC on endothelial cell at ischemic tissue. Results- The uptake of VEGF into endothelial cells was increased by BM-MNC, while being inhibited by blockading the gap junction. Low-molecular-weight substance was transferred from BM-MNC into endothelial cells via gap junctions in vivo, followed by increased expression of hypoxia-inducible factor-1α and suppression of autophagy in endothelial cells. The concentration of glucose in BM-MNC cytoplasm was significantly higher than in endothelial cells, and transfer of glucose homologue from BM-MNC to endothelial cells was observed. Conclusions- Our findings demonstrated cell-cell interaction via gap junction is the prominent pathway for activation of angiogenesis at endothelial cells after ischemia and provided novel paradigm that energy source supply by stem cell to injured cell is one of the therapeutic mechanisms of cell-based therapy. Visual Overview- An online visual overview is available for this article.
Collapse
Affiliation(s)
- Akie Kikuchi-Taura
- From the Department of Regenerative Medicine Research, Institute of Biomedical Research and Innovation, Kobe, Japan (A.K.-T., Y. Okinaka, Y.T., Y. Ogawa, M.M., J.B., A.T.)
| | - Yuka Okinaka
- From the Department of Regenerative Medicine Research, Institute of Biomedical Research and Innovation, Kobe, Japan (A.K.-T., Y. Okinaka, Y.T., Y. Ogawa, M.M., J.B., A.T.)
| | - Yukiko Takeuchi
- From the Department of Regenerative Medicine Research, Institute of Biomedical Research and Innovation, Kobe, Japan (A.K.-T., Y. Okinaka, Y.T., Y. Ogawa, M.M., J.B., A.T.)
| | - Yuko Ogawa
- From the Department of Regenerative Medicine Research, Institute of Biomedical Research and Innovation, Kobe, Japan (A.K.-T., Y. Okinaka, Y.T., Y. Ogawa, M.M., J.B., A.T.)
| | - Mitsuyo Maeda
- From the Department of Regenerative Medicine Research, Institute of Biomedical Research and Innovation, Kobe, Japan (A.K.-T., Y. Okinaka, Y.T., Y. Ogawa, M.M., J.B., A.T.).,Multi-Modal Microstructure Analysis Unit, RIKEN-JEOL Collaboration Center, RIKEN, Kobe, Japan (M.M., Y.K.).,Laboratory for Cellular Function Imaging, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan (M.M., Y.K.)
| | - Yosky Kataoka
- Multi-Modal Microstructure Analysis Unit, RIKEN-JEOL Collaboration Center, RIKEN, Kobe, Japan (M.M., Y.K.).,Laboratory for Cellular Function Imaging, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan (M.M., Y.K.)
| | - Teruhito Yasui
- National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Japan (T.Y.)
| | - Takafumi Kimura
- Japanese Red Cross Kinki Block Blood Center, Ibaraki, Japan (T.K.)
| | - Sheraz Gul
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME - ScreeningPort, Hamburg, Germany (S.G., C.C.)
| | - Carsten Claussen
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME - ScreeningPort, Hamburg, Germany (S.G., C.C.)
| | - Johannes Boltze
- From the Department of Regenerative Medicine Research, Institute of Biomedical Research and Innovation, Kobe, Japan (A.K.-T., Y. Okinaka, Y.T., Y. Ogawa, M.M., J.B., A.T.).,School of Life Sciences, University of Warwick, Coventry, United Kingdom (J.B.)
| | - Akihiko Taguchi
- From the Department of Regenerative Medicine Research, Institute of Biomedical Research and Innovation, Kobe, Japan (A.K.-T., Y. Okinaka, Y.T., Y. Ogawa, M.M., J.B., A.T.)
| |
Collapse
|
26
|
Abstract
The generation of hematopoietic stem cells (HSCs) from pluripotent stem cell (PSC) sources is a long-standing goal that will require a comprehensive understanding of the molecular and cellular factors that determine HSC fate during embryogenesis. A precise interplay between niche components, such as the vascular, mesenchymal, primitive myeloid cells, and the nervous system provides the unique signaling milieu for the emergence of functional HSCs in the aorta-gonad-mesonephros (AGM) region. Over the last several years, the interrogation of these aspects in the embryo model and in the PSC differentiation system has provided valuable knowledge that will continue educating the design of more efficient protocols to enable the differentiation of PSCs into
bona fide, functionally transplantable HSCs. Herein, we provide a synopsis of early hematopoietic development, with particular focus on the recent discoveries and remaining questions concerning AGM hematopoiesis. Moreover, we acknowledge the recent advances towards the generation of HSCs
in vitro and discuss possible approaches to achieve this goal in light of the current knowledge.
Collapse
Affiliation(s)
- Ana G Freire
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, USA
| | - Jason M Butler
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, USA.,Molecular Oncology Program, Georgetown University, Washington D.C., USA
| |
Collapse
|
27
|
Benova A, Tencerova M. Obesity-Induced Changes in Bone Marrow Homeostasis. Front Endocrinol (Lausanne) 2020; 11:294. [PMID: 32477271 PMCID: PMC7235195 DOI: 10.3389/fendo.2020.00294] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/20/2020] [Indexed: 12/24/2022] Open
Abstract
Obesity is characterized by low-grade inflammation, which is accompanied by increased accumulation of immune cells in peripheral tissues including adipose tissue (AT), skeletal muscle, liver and pancreas, thereby impairing their primary metabolic functions in the regulation of glucose homeostasis. Obesity has also shown to have a detrimental effect on bone homeostasis by altering bone marrow and hematopoietic stem cell differentiation and thus impairing bone integrity and immune cell properties. The origin of immune cells arises in the bone marrow, which has been shown to be affected with the obesogenic condition via increased cellularity and shifting differentiation and function of hematopoietic and bone marrow mesenchymal stem cells in favor of myeloid progenitors and increased bone marrow adiposity. These obesity-induced changes in the bone marrow microenvironment lead to dramatic bone marrow remodeling and compromising immune cell functions, which in turn affect systemic inflammatory conditions and regulation of whole-body metabolism. However, there is limited information on the inflammatory secretory factors creating the bone marrow microenvironment and how these factors changed during metabolic complications. This review summarizes recent findings on inflammatory and cellular changes in the bone marrow in relation to obesity and further discuss whether dietary intervention or physical activity may have beneficial effects on the bone marrow microenvironment and whole-body metabolism.
Collapse
|
28
|
Balistreri CR, Garagnani P, Madonna R, Vaiserman A, Melino G. Developmental programming of adult haematopoiesis system. Ageing Res Rev 2019; 54:100918. [PMID: 31226498 DOI: 10.1016/j.arr.2019.100918] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 05/15/2019] [Accepted: 06/17/2019] [Indexed: 12/15/2022]
Abstract
The Barker hypothesis of 'foetal origin of adult diseases' has led to emphasize the concept of 'developmental programming', based on the crucial role of epigenetic factors. Accordingly, it has been demonstrated that parental adversity (before conception and during pregnancy) and foetal factors (i.e., hypoxia, malnutrition and placental insufficiency) permanently modify the physiological systems of the progeny, predisposing them to premature ageing and chronic disease during adulthood. Thus, an altered functionality of the endocrine, immune, nervous and cardiovascular systems is observed in the progeny. However, it remains to be understood whether the haematopoietic system itself also represents a portrait of foetal programming. Here, we provide evidence, reporting and discussing related theories, and results of studies described in the literature. In addition, we have outlined our opinions and suggest how it is possible to intervene to correct foetal mal-programming. Some pro-health interventions and recommendations are proposed, with the hope of guarantee the health of future generations and trying to combat the continuous increase in age-related diseases in human populations.
Collapse
|
29
|
Feng Y, Zhu R, Shen J, Wu J, Lu W, Zhang J, Zhang J, Liu K. Human Bone Marrow Mesenchymal Stem Cells Rescue Endothelial Cells Experiencing Chemotherapy Stress by Mitochondrial Transfer Via Tunneling Nanotubes. Stem Cells Dev 2019; 28:674-682. [PMID: 30808254 DOI: 10.1089/scd.2018.0248] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Tunneling nanotubes (TNTs) are newly discovered tubular structures between two distant cells that facilitate the intercellular exchange of signals and components. Recent reports show that mesenchymal stem cells (MSCs) can rescue injured target cells and promote recovery from a variety of stresses via TNT-mediated mitochondrial transfer. In this study, we explored how TNTs form between bone marrow MSCs and endothelial cells (ECs) by using a human umbilical cord vein endothelial cell (HUVEC) model. TNT formation between MSCs and HUVECs could be induced by treating HUVECs with cytarabine (Ara-C), and human bone marrow mesenchymal stem cells (hBMMSCs) could transfer mitochondria to injured HUVECs through TNTs. Mitochondrial transfer from hBMMSCs to HUVECs via TNTs rescued the injured HUVECs by reducing apoptosis, promoting proliferation and restoring the transmembrane migration ability as well as the capillary angiogenic capacity of HUVECs. This study provides novel insights into the cell-cell communication between MSCs and ECs and supports the results of prior studies indicating that ECs promote hematopoietic regeneration. An improved understanding of MSC-EC cross-talk will promote the development of MSC-directed strategies for improving EC function and hematopoietic system regeneration following myelosuppressive and myeloablative injuries.
Collapse
Affiliation(s)
- Yonghuai Feng
- 1 Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation, Institute of Haematology, Peking University People's Hospital, Beijing, China.,2 Institute of Haematology, Affiliated Hospital of Zunyi Medical College, Zunyi, China
| | - Rongjia Zhu
- 3 Chinese Academy of Medical Science and Peking Union Medical College, Institute of Basic Medical Sciences and School of Basic Medicine, Beijing, China
| | - Jing Shen
- 4 Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
| | - JiMin Wu
- 4 Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
| | - Wenyi Lu
- 1 Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation, Institute of Haematology, Peking University People's Hospital, Beijing, China
| | - JiaMin Zhang
- 1 Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation, Institute of Haematology, Peking University People's Hospital, Beijing, China
| | - Jing Zhang
- 1 Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation, Institute of Haematology, Peking University People's Hospital, Beijing, China
| | - Kaiyan Liu
- 1 Beijing Key Laboratory of Haematopoietic Stem Cell Transplantation, Institute of Haematology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
30
|
Lazzarini R, Caffarini M, Tang H, Cerqueni G, Pellegrino P, Monsurrò V, Di Primio R, Orciani M. The senescent status of endothelial cells affects proliferation, inflammatory profile and SOX2 expression in bone marrow-derived mesenchymal stem cells. Exp Gerontol 2019; 120:21-27. [PMID: 30822486 DOI: 10.1016/j.exger.2019.02.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 02/05/2019] [Accepted: 02/24/2019] [Indexed: 12/16/2022]
Abstract
Human aging is a physiological process characterized by a chronic low-grade inflammation. Senescence may affect endothelial cells, subsequently involved in the most common age-related diseases (ARDs), as well as mesenchymal stem cells (MSCs) with an impairment of their properties in tissues regeneration. Endothelial cells seem to be able to exert a paracrine effect on BM-MSCs through the secretion of pro-inflammatory factors. This work is aimed to evaluate if the senescent status of human umbilical vein endothelial cells (HUVECs) could affect bone marrow derived MSCs (BM-MSCs) proliferative ability and stemness. HUVECs were cultured until the senescence status. Young (passage 3) and senescent HUVECs (passage 13) were indirectly co-cultured with BM-MSCs for 8 days in order to evaluate the effect of their senescence status on proliferative ability and stemness of MSCs. The co-culture of senescent HUVECs with BM-MSCs was associated with a reduced proliferative ability of BM-MSCs, an enforced pro-inflammatory phenotype of BM-MSCs (increased synthesis of proinflammatory cytokines such as IL-6 and TNF-α) and an increased expression of miR-126a-3p, in association with a significant decrease of SOX2, a stemmness- associated gene, targeted by miR-126a-3p. A more general IPA analysis, revealed as miR-126a-3p also modulates the expression of IRS1, IRS2, IL6ST and PIK3R2, all targets that enforce the hypothesis that senescent endothelial cells may reduce the proliferative ability and the stemness phenotype of bone marrow-derived mesenchymal stem cells.
Collapse
Affiliation(s)
- Raffaella Lazzarini
- Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Miriam Caffarini
- Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Huijuan Tang
- Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Giorgia Cerqueni
- Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Pamela Pellegrino
- Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| | | | - Roberto Di Primio
- Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy.
| | - Monia Orciani
- Department of Clinical and Molecular Sciences, Polytechnic University of Marche, Ancona, Italy
| |
Collapse
|
31
|
Abstract
Purpose of review In this review, we aim to discuss the role of the bone marrow microenvironment in supporting hematopoiesis, with particular focus on the contribution of the endothelial niche in dictating hematopoietic stem cell (HSC) fate. Recent findings Evidence gathered in the past two decades revealed that specific cell types within the bone marrow niche influence the hematopoietic system. Endothelial cells have emerged as a key component of the HSC niche, directly affecting stem cell quiescence, self-renewal, and lineage differentiation. Physiological alterations of the bone marrow niche occurring in aging have been described to be sufficient to promote functional aging of young HSCs. Furthermore, a growing body of evidence suggests that aberrant activation of endothelial-derived signaling pathways can aid or trigger neoplastic transformation. Summary Several groups have contributed to the characterization of the different cell types that comprise the complex bone marrow environment, whose function was long perceived as an undiscernible sum of many parts. Further studies will need to uncover niche cell-type-specific pathways, in order to provide new targets and therapeutic options that aim at withdrawing the microenvironmental support to malignant cells while sparing normal HSCs.
Collapse
|
32
|
Making HSCs in vitro: don't forget the hemogenic endothelium. Blood 2018; 132:1372-1378. [PMID: 30089629 DOI: 10.1182/blood-2018-04-784140] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 08/04/2018] [Indexed: 12/29/2022] Open
Abstract
Generating a hematopoietic stem cell (HSC) in vitro from nonhematopoietic tissue has been a goal of experimental hematologists for decades. Until recently, no in vitro-derived cell has closely demonstrated the full lineage potential and self-renewal capacity of a true HSC. Studies revealing stem cell ontogeny from embryonic mesoderm to hemogenic endothelium to HSC provided the key to inducing HSC-like cells in vitro from a variety of cell types. Here we review the path to this discovery and discuss the future of autologous transplantation with in vitro-derived HSCs as a therapeutic modality.
Collapse
|
33
|
Abstract
Purpose of Review Hematopoietic stem cells (HSC) reside in a specialized microenvironment called the HSC niche. While key components of the niche have been known for several years, recent advances have identified several additional cell types that regulate HSC in the bone marrow (BM). Here we review our current understanding of the components and dynamics of the HSC niche. Recent Findings While the niche has been considered a stable structure, recent advances clearly show that the niche is regulated in a dynamic manner to control HSC traffic and function. Moreover the niche can rapidly remodel in response to insults to the BM in a process controlled by positive and negative regulators. Summary Multiple niche cells have been shown to be dynamically regulated by systemic and local signals to influence how the niche controls HSC function. Elucidating how different components of the niche coordinate to orchestrate HSC behavior is essential to understand how the hematopoietic system adjusts blood cell production to the demands of the body.
Collapse
|
34
|
Xu R, Yallowitz A, Qin A, Wu Z, Shin DY, Kim JM, Debnath S, Ji G, Bostrom MP, Yang X, Zhang C, Dong H, Kermani P, Lalani S, Li N, Liu Y, Poulos MG, Wach A, Zhang Y, Inoue K, Di Lorenzo A, Zhao B, Butler JM, Shim JH, Glimcher LH, Greenblatt MB. Targeting skeletal endothelium to ameliorate bone loss. Nat Med 2018; 24:823-833. [PMID: 29785024 PMCID: PMC5992080 DOI: 10.1038/s41591-018-0020-z] [Citation(s) in RCA: 235] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 03/22/2018] [Indexed: 11/08/2022]
Abstract
Recent studies have identified a specialized subset of CD31hiendomucinhi (CD31hiEMCNhi) vascular endothelium that positively regulates bone formation. However, it remains unclear how CD31hiEMCNhi endothelium levels are coupled to anabolic bone formation. Mice with an osteoblast-specific deletion of Shn3, which have markedly elevated bone formation, demonstrated an increase in CD31hiEMCNhi endothelium. Transcriptomic analysis identified SLIT3 as an osteoblast-derived, SHN3-regulated proangiogenic factor. Genetic deletion of Slit3 reduced skeletal CD31hiEMCNhi endothelium, resulted in low bone mass because of impaired bone formation and partially reversed the high bone mass phenotype of Shn3-/- mice. This coupling between osteoblasts and CD31hiEMCNhi endothelium is essential for bone healing, as shown by defective fracture repair in SLIT3-mutant mice and enhanced fracture repair in SHN3-mutant mice. Finally, administration of recombinant SLIT3 both enhanced bone fracture healing and counteracted bone loss in a mouse model of postmenopausal osteoporosis. Thus, drugs that target the SLIT3 pathway may represent a new approach for vascular-targeted osteoanabolic therapy to treat bone loss.
Collapse
Affiliation(s)
- Ren Xu
- Department of Pathology and Laboratory Medicine, Cornell University, New York, NY, USA
| | - Alisha Yallowitz
- Department of Pathology and Laboratory Medicine, Cornell University, New York, NY, USA
| | - An Qin
- Department of Orthopaedics, Shanghai Key Laboratory of Orthopaedic Implant, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhuhao Wu
- Laboratory of Brain Development and Repair, The Rockefeller University, New York, NY, USA
| | - Dong Yeon Shin
- Department of Pathology and Laboratory Medicine, Cornell University, New York, NY, USA
| | - Jung-Min Kim
- Division of Rheumatology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Shawon Debnath
- Department of Pathology and Laboratory Medicine, Cornell University, New York, NY, USA
| | - Gang Ji
- Research Division, Hospital for Special Surgery, New York, NY, USA
- Department of Joint Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Mathias P Bostrom
- Research Division, Hospital for Special Surgery, New York, NY, USA
- Division of Adult Reconstruction and Joint Replacement, Department of Orthopaedic Surgery, Hospital for Special Surgery, New York, NY, USA
| | - Xu Yang
- Research Division, Hospital for Special Surgery, New York, NY, USA
| | - Chao Zhang
- Institute for Computational Biomedicine, Cornell University, New York, NY, USA
| | - Han Dong
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute and Harvard University Medical School, Boston, MA, USA
- Department of Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Pouneh Kermani
- Division of Regenerative Medicine, Department of Medicine, Ansary Stem Cell Institute, Cornell University, New York, NY, USA
| | - Sarfaraz Lalani
- Department of Pathology and Laboratory Medicine, Cornell University, New York, NY, USA
| | - Na Li
- Department of Pathology and Laboratory Medicine, Cornell University, New York, NY, USA
| | - Yifang Liu
- Department of Pathology and Laboratory Medicine, Cornell University, New York, NY, USA
| | - Michael G Poulos
- Division of Regenerative Medicine, Department of Medicine, Ansary Stem Cell Institute, Cornell University, New York, NY, USA
| | - Amanda Wach
- Department of Biomechanics, Hospital for Special Surgery, New York, NY, USA
| | - Yi Zhang
- Department of Pathology and Laboratory Medicine, Cornell University, New York, NY, USA
| | - Kazuki Inoue
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Annarita Di Lorenzo
- Department of Pathology and Laboratory Medicine, Cornell University, New York, NY, USA
| | - Baohong Zhao
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Jason M Butler
- Division of Regenerative Medicine, Department of Medicine, Ansary Stem Cell Institute, Cornell University, New York, NY, USA
| | - Jae-Hyuck Shim
- Division of Rheumatology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Laurie H Glimcher
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute and Harvard University Medical School, Boston, MA, USA.
- Department of Medicine, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA.
| | - Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
35
|
Lucas D. The Bone Marrow Microenvironment for Hematopoietic Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1041:5-18. [PMID: 29204826 DOI: 10.1007/978-3-319-69194-7_2] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The main function of the microenvironment in the bone marrow (BM) is to provide signals that regulate and support the production of the billions of blood cells necessary to maintain homeostasis. The best characterized BM microenvironment is the niche that regulates hematopoietic stem cells. Efforts from many different laboratories have revealed that the niche is mainly perivascular and that blood vessels and perivascular stromal cells are the key components. In addition numerous cell types have been shown to be components of the niche. Here we discuss our current understanding of the niche and the evidence supporting the role of different types of cells in regulating hematopoietic stem cell numbers and function in vivo.
Collapse
Affiliation(s)
- Daniel Lucas
- Department of Cell and Developmental Biology, University of Michigan School of Medicine, Ann Arbor, MI, USA. .,Center for Organogenesis, University of Michigan School of Medicine, Ann Arbor, MI, USA. .,The University of Michigan Comprehensive Cancer Center, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
36
|
Guo P, Poulos MG, Palikuqi B, Badwe CR, Lis R, Kunar B, Ding BS, Rabbany SY, Shido K, Butler JM, Rafii S. Endothelial jagged-2 sustains hematopoietic stem and progenitor reconstitution after myelosuppression. J Clin Invest 2017; 127:4242-4256. [PMID: 29058691 PMCID: PMC5707154 DOI: 10.1172/jci92309] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 08/29/2017] [Indexed: 12/19/2022] Open
Abstract
Angiocrine factors, such as Notch ligands, supplied by the specialized endothelial cells (ECs) within the bone marrow and splenic vascular niche play an essential role in modulating the physiology of adult hematopoietic stem and progenitor cells (HSPCs). However, the relative contribution of various Notch ligands, specifically jagged-2, to the homeostasis of HSPCs is unknown. Here, we show that under steady state, jagged-2 is differentially expressed in tissue-specific vascular beds, but its expression is induced in hematopoietic vascular niches after myelosuppressive injury. We used mice with EC-specific deletion of the gene encoding jagged-2 (Jag2) to demonstrate that while EC-derived jagged-2 was dispensable for maintaining the capacity of HSPCs to repopulate under steady-state conditions, by activating Notch2 it did contribute to the recovery of HSPCs in response to myelosuppressive conditions. Engraftment and/or expansion of HSPCs was dependent on the expression of endothelial-derived jagged-2 following myeloablation. Additionally, jagged-2 expressed in bone marrow ECs regulated HSPC cell cycle and quiescence during regeneration. Endothelial-deployed jagged-2 triggered Notch2/Hey1, while tempering Notch2/Hes1 signaling in HSPCs. Collectively, these data demonstrate that EC-derived jagged-2 activates Notch2 signaling in HSPCs to promote hematopoietic recovery and has potential as a therapeutic target to accelerate balanced hematopoietic reconstitution after myelosuppression.
Collapse
Affiliation(s)
- Peipei Guo
- Department of Medicine, Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, New York, USA.,Department of Physiology, Biophysics, and Systems Biology, Weill Cornell Medicine, New York, New York, USA
| | - Michael G Poulos
- Department of Medicine, Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, New York, USA
| | - Brisa Palikuqi
- Department of Medicine, Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, New York, USA
| | - Chaitanya R Badwe
- Department of Medicine, Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, New York, USA
| | - Raphael Lis
- Department of Medicine, Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, New York, USA
| | - Balvir Kunar
- Department of Medicine, Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, New York, USA.,Department of Physiology, Biophysics, and Systems Biology, Weill Cornell Medicine, New York, New York, USA
| | - Bi-Sen Ding
- Department of Medicine, Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, New York, USA
| | - Sina Y Rabbany
- Department of Medicine, Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, New York, USA.,Bioengineering Program, DeMatteis School of Engineering and Applied Science, Hofstra University, Long Island, New York, USA
| | - Koji Shido
- Department of Medicine, Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, New York, USA
| | - Jason M Butler
- Department of Medicine, Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, New York, USA
| | - Shahin Rafii
- Department of Medicine, Division of Regenerative Medicine, Ansary Stem Cell Institute, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|