1
|
Ide AD, Carpenter KA, Elaswad M, Opria K, Marcellin K, Gilliland C, Grainger S. Secreted Frizzled-Related Protein 1a regulates hematopoietic development in a dose-dependent manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.10.632371. [PMID: 39829913 PMCID: PMC11741364 DOI: 10.1101/2025.01.10.632371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Hematopoietic stem and progenitor cells (HSPCs) arise only during embryonic development, and their identity specification, emergence from the floor of the dorsal aorta, and proliferation are all tightly regulated by molecular mechanisms such as signaling cues. Among these, Wnt signaling plays an important role in HSPC specification, differentiation, and self-renewal, requiring precise modulation for proper development and homeostasis. Wnt signaling is initiated when a Wnt ligand binds to cell surface receptors such as those encoded by the frizzled gene family, activating intracellular signaling pathways that regulate gene expression. Secreted frizzled-related proteins (Sfrps) are known modulators of Wnt signaling, acting as both agonists and antagonists of this pathway. Yet, in vivo functions of Sfrps in HSPC development remain incompletely understood. Here, we demonstrate that Sfrp1a regulates zebrafish HSPC development and differentiation in a dose-dependent manner. In Sfrp1a loss of function animals, we observe an increase in HSPCs, an upregulation of canonical Wnt signaling, and a decrease in differentiation into both lymphoid and myeloid lineages. Conversely, at low-dose sfrp1a overexpression, there is a decrease in HSPCs and an increase in lymphoid differentiation. High-dose sfrp1a overexpression phenocopies the loss of function animals, with an increase in HSPCs, increased canonical Wnt signaling, and decreased lymphoid and myeloid differentiation. These findings highlight the importance of dose-dependent modulation of Sfrps, paralleling what is observed in hematopoietic cancers where SFRP1 loss-of-function and gain-of-function variants can drive tumorigenesis.
Collapse
Affiliation(s)
- Amber D. Ide
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA RRID: SCR_021956
| | - Kelsey A. Carpenter
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA RRID: SCR_021956
| | - Mohamed Elaswad
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA RRID: SCR_021956
| | - Katherine Opria
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA RRID: SCR_021956
| | - Kendersley Marcellin
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA RRID: SCR_021956
| | - Carla Gilliland
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA RRID: SCR_021956
| | - Stephanie Grainger
- Department of Cell Biology, Van Andel Institute, Grand Rapids, Michigan, 49503, USA RRID: SCR_021956
| |
Collapse
|
2
|
Cain TL, Derecka M, McKinney-Freeman S. The role of the haematopoietic stem cell niche in development and ageing. Nat Rev Mol Cell Biol 2025; 26:32-50. [PMID: 39256623 DOI: 10.1038/s41580-024-00770-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2024] [Indexed: 09/12/2024]
Abstract
Blood production depends on rare haematopoietic stem cells (HSCs) and haematopoietic stem and progenitor cells (HSPCs) that ultimately take up residence in the bone marrow during development. HSPCs and HSCs are subject to extrinsic regulation by the bone marrow microenvironment, or niche. Studying the interactions between HSCs and their niche is critical for improving ex vivo culturing conditions and genetic manipulation of HSCs, which is pivotal for improving autologous HSC therapies and transplantations. Additionally, understanding how the complex molecular network in the bone marrow is altered during ageing is paramount for developing novel therapeutics for ageing-related haematopoietic disorders. HSCs are unique amongst stem and progenitor cell pools in that they engage with multiple physically distinct niches during their ontogeny. HSCs are specified from haemogenic endothelium in the aorta, migrate to the fetal liver and, ultimately, colonize their final niche in the bone marrow. Recent studies employing single-cell transcriptomics and microscopy have identified novel cellular interactions that govern HSC specification and engagement with their niches throughout ontogeny. New lineage-tracing models and microscopy tools have raised questions about the numbers of HSCs specified, as well as the functional consequences of HSCs interacting with each developmental niche. Advances have also been made in understanding how these niches are modified and perturbed during ageing, and the role of these altered interactions in haematopoietic diseases. In this Review, we discuss these new findings and highlight the questions that remain to be explored.
Collapse
Affiliation(s)
- Terri L Cain
- Department of Haematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Marta Derecka
- Department of Haematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | |
Collapse
|
3
|
Liu Y, Huang J, Li L, Duan Y, Chong BH, Li L, Yang M. Regulatory Effect of PDGF/PDGFR on Hematopoiesis. Semin Thromb Hemost 2024. [PMID: 39608410 DOI: 10.1055/s-0044-1796630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Platelet-derived growth factor (PDGF) is a critical cytokine with substantial regulatory effects on hematopoiesis. Recent research highlights the essential role of PDGF in the modulation of hematopoietic stem/progenitor cells (HSPCs), megakaryocytes/platelets, and thrombopoietin (TPO) synthesis within the bone marrow microenvironment. PDGF directly stimulates the proliferation and differentiation of HSPCs while also inhibiting apoptosis. In addition, PDGF indirectly enhances the production of other growth factors, including granulocyte-macrophage colony-stimulating factors. Further, PDGF regulates TPO production and influences the bone marrow milieu, thus impacting hematopoiesis and platelet formation. Mechanistically, PDGF binds to its receptor, PDGF receptor (PDGFR), thus activating the PDGF/PDGFR signaling pathway. This pathway subsequently activates phosphoinositide 3-kinase/protein kinase B, leading to the activation of downstream cytokines, including c-Fos and NF-E2, while inhibiting caspase-3 activation. Collectively, these actions have prodifferentiation and antiapoptotic effects on megakaryocytes, thereby regulating platelet production. This review provides a comprehensive analysis of the regulatory role of the PDGF/PDGFR axis in hematopoiesis, with a particular focus on platelet production, by summarizing all studies on PDGF/PDGFR from our group and globally.
Collapse
Affiliation(s)
- Yong Liu
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Junbin Huang
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Lindi Li
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Yifei Duan
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Beng H Chong
- Haematology Research Unit, School of Clinical Medicine, St George and Sutherland Campus, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
- New South Wales Health Pathology, St George Hospital, Kogarah, New South Wales, Australia
| | - Liang Li
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Mo Yang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
- Pediatric Hematology Laboratory, Division of Hematology/Oncology, Department of Pediatrics, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
- Department of Hematology, Affiliated Hospital of Guangdong Medical University (GDMU), Zhanjiang, Guangdong, China
| |
Collapse
|
4
|
Sánchez-Lanzas R, Jiménez-Pompa A, Ganuza M. The evolving hematopoietic niche during development. Front Mol Biosci 2024; 11:1488199. [PMID: 39417006 PMCID: PMC11480086 DOI: 10.3389/fmolb.2024.1488199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
Mammalian hematopoietic stem cells (HSCs) emerge from the hemogenic endothelium in the major embryonic arteries. HSCs undergo a complex journey first migrating to the fetal liver (FL) and from there to the fetal bone marrow (FBM), where they mostly remain during adult life. In this process, a pool of adult HSCs is produced, which sustains lifelong hematopoiesis. Multiple cellular components support HSC maturation and expansion and modulate their response to environmental and developmental cues. While the adult HSC niche has been extensively studied over the last two decades, the niches present in the major embryonic arteries, FL, FBM and perinatal bone marrow (BM) are poorly described. Recent investigations highlight important differences among FL, FBM and adult BM niches and emphasize the important role that inflammation, microbiota and hormonal factors play regulating HSCs and their niches. We provide a review on our current understanding of these important cellular microenvironments across ontogeny. We mainly focused on mice, as the most widely used research model, and, when possible, include relevant insights from other vertebrates including birds, zebrafish, and human. Developing a comprehensive picture on these processes is critical to understand the earliest origins of childhood leukemia and to achieve multiple goals in regenerative medicine, such as mimicking HSC development in vitro to produce HSCs for broad transplantation purposes in leukemia, following chemotherapy, bone marrow failure, and in HSC-based gene therapy.
Collapse
Affiliation(s)
| | | | - Miguel Ganuza
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
5
|
Ren H, Feng J, Hong M, Liu Z, Muyey DM, Zhang Y, Xu Z, Tan Y, Ren F, Chang J, Chen X, Wang H. Baicalein attenuates oxidative damage in mice haematopoietic cells through regulation of PDGFRβ. Mol Cell Probes 2024; 76:101966. [PMID: 38866345 DOI: 10.1016/j.mcp.2024.101966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 05/26/2024] [Accepted: 06/06/2024] [Indexed: 06/14/2024]
Abstract
Platelet-derived growth factor receptor β (PDGFRβ) plays a crucial role in murine haematopoiesis. Baicalein (BAI), a naturally occurring flavonoid, can alleviate disease damage through anti-oxidative, anti-apoptotic, and anti-inflammatory mechanisms. However, whether BAI attenuates oxidative damage in murine haematopoietic cells by PDGFRβ remains unexplored. In this study, we utilized a tert-butyl hydroperoxide (TBHP)-induced BaF3 cell injury model and an ionising radiation (IR)-induced mice injury model to investigate the impact of the presence or absence of PDGFRβ on the pharmacological effects of BAI. In addition, the BAI-PDGFRβ interaction was characterized by molecular docking and dynamics simulations. The results show that a specific concentration of BAI led to increased cell viability, reduced reactive oxygen species (ROS) content, upregulated nuclear factor erythroid 2-related factor 2 (NRF2) expression, and its downstream target genes heme oxygenase 1 (HO-1) and NAD(P)H Quinone Dehydrogenase 1 (NQO1), and activated protein kinase B (AKT) pathway in cells expressing PDGFRβ plasmid and experiencing damage. Similarly, BAI elevated lineage-Sca1+cKIT+ (LSK) cell proportion, promoted haematopoietic restoration, enhanced NRF2-mediated antioxidant response in PDGFRβ+/+ mice. However, despite BAI usage, PDGFRβ knockout mice (PDGFRβ-/-) showed lower LSK proportion and less antioxidant capacity than the total body irradiation (TBI) group. Furthermore, we demonstrated an interaction between BAI and PDGFRβ at the molecular level. Collectively, our results indicate that BAI attenuates oxidative stress injury and helps promote haematopoietic cell recovery through regulation of PDGFRβ.
Collapse
Affiliation(s)
- Huanying Ren
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Jingyi Feng
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Minglin Hong
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Zhuang Liu
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Daniel Muteb Muyey
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Yaofang Zhang
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Zhifang Xu
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Yanhong Tan
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Fanggang Ren
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Jianmei Chang
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China
| | - Xiuhua Chen
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| | - Hongwei Wang
- Institute of Hematology, The Second Hospital of Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
6
|
Clements WK, Khoury H. The molecular and cellular hematopoietic stem cell specification niche. Exp Hematol 2024; 136:104280. [PMID: 39009276 PMCID: PMC11338702 DOI: 10.1016/j.exphem.2024.104280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024]
Abstract
Hematopoietic stem cells (HSCs) are a population of tissue-specific stem cells that reside in the bone marrow of adult mammals, where they self-renew and continuously regenerate the adult hematopoietic lineages over the life of the individual. Prominence as a stem cell model and clinical usefulness have driven interest in understanding the physiologic processes that lead to the specification of HSCs during embryonic development. High-efficiency directed differentiation of HSCs by the instruction of defined progenitor cells using sequentially defined instructive molecules and conditions remains impossible, indicating that comprehensive knowledge of the complete set of precursor intermediate identities and required inductive inputs remains incompletely understood. Recently, interest in the molecular and cellular microenvironment where HSCs are specified from endothelial precursors-the "specification niche"-has increased. Here we review recent progress in understanding these niche spaces across vertebrate phyla, as well as how a better characterization of the origin and molecular phenotypes of the niche cell populations has helped inform and complicate previous understanding of signaling required for HSC emergence and maturation.
Collapse
Affiliation(s)
- Wilson K Clements
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN.
| | - Hanane Khoury
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN
| |
Collapse
|
7
|
Belt AJ, Grant S, Tombes RM, Rothschild SC. Myeloid Targeted Human MLL-ENL and MLL-AF9 Induces cdk9 and bcl2 Expression in Zebrafish Embryos. PLoS Genet 2024; 20:e1011308. [PMID: 38829886 PMCID: PMC11175583 DOI: 10.1371/journal.pgen.1011308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 06/13/2024] [Accepted: 05/19/2024] [Indexed: 06/05/2024] Open
Abstract
Acute myeloid leukemia (AML) accounts for greater than twenty thousand new cases of leukemia annually in the United States. The average five-year survival rate is approximately 30%, pointing to the need for developing novel model systems for drug discovery. In particular, patients with chromosomal rearrangements in the mixed lineage leukemia (MLL) gene have higher relapse rates with poor outcomes. In this study we investigated the expression of human MLL-ENL and MLL-AF9 in the myeloid lineage of zebrafish embryos. We observed an expansion of MLL positive cells and determined these cells colocalized with the myeloid markers spi1b, mpx, and mpeg. In addition, expression of MLL-ENL and MLL-AF9 induced the expression of endogenous bcl2 and cdk9, genes that are often dysregulated in MLL-r-AML. Co-treatment of lyz: MLL-ENL or lyz:MLL-AF9 expressing embryos with the BCL2 inhibitor, Venetoclax, and the CDK9 inhibitor, Flavopiridol, significantly reduced the number of MLL positive cells compared to embryos treated with vehicle or either drug alone. In addition, cotreatment with Venetoclax and Flavopiridol significantly reduced the expression of endogenous mcl1a compared to vehicle, consistent with AML. This new model of MLL-r-AML provides a novel tool to understand the molecular mechanisms underlying disease progression and a platform for drug discovery.
Collapse
MESH Headings
- Zebrafish/genetics
- Zebrafish/embryology
- Animals
- Cyclin-Dependent Kinase 9/genetics
- Cyclin-Dependent Kinase 9/metabolism
- Cyclin-Dependent Kinase 9/antagonists & inhibitors
- Myeloid-Lymphoid Leukemia Protein/genetics
- Myeloid-Lymphoid Leukemia Protein/metabolism
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Humans
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/metabolism
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Sulfonamides/pharmacology
- Piperidines/pharmacology
- Embryo, Nonmammalian
- Flavonoids/pharmacology
- Myeloid Cells/metabolism
- Myeloid Cells/drug effects
- Histone-Lysine N-Methyltransferase/genetics
- Histone-Lysine N-Methyltransferase/metabolism
- Zebrafish Proteins/genetics
- Zebrafish Proteins/metabolism
Collapse
Affiliation(s)
- Alex J. Belt
- Life Sciences, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Steven Grant
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Robert M. Tombes
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Biology, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Sarah C. Rothschild
- Life Sciences, Virginia Commonwealth University, Richmond, Virginia, United States of America
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia, United States of America
| |
Collapse
|
8
|
Miladinovic O, Canto PY, Pouget C, Piau O, Radic N, Freschu P, Megherbi A, Brujas Prats C, Jacques S, Hirsinger E, Geeverding A, Dufour S, Petit L, Souyri M, North T, Isambert H, Traver D, Jaffredo T, Charbord P, Durand C. A multistep computational approach reveals a neuro-mesenchymal cell population in the embryonic hematopoietic stem cell niche. Development 2024; 151:dev202614. [PMID: 38451068 PMCID: PMC11057820 DOI: 10.1242/dev.202614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 02/23/2024] [Indexed: 03/08/2024]
Abstract
The first hematopoietic stem and progenitor cells (HSPCs) emerge in the Aorta-Gonad-Mesonephros (AGM) region of the mid-gestation mouse embryo. However, the precise nature of their supportive mesenchymal microenvironment remains largely unexplored. Here, we profiled transcriptomes of laser micro-dissected aortic tissues at three developmental stages and individual AGM cells. Computational analyses allowed the identification of several cell subpopulations within the E11.5 AGM mesenchyme, with the presence of a yet unidentified subpopulation characterized by the dual expression of genes implicated in adhesive or neuronal functions. We confirmed the identity of this cell subset as a neuro-mesenchymal population, through morphological and lineage tracing assays. Loss of function in the zebrafish confirmed that Decorin, a characteristic extracellular matrix component of the neuro-mesenchyme, is essential for HSPC development. We further demonstrated that this cell population is not merely derived from the neural crest, and hence, is a bona fide novel subpopulation of the AGM mesenchyme.
Collapse
Affiliation(s)
- Olivera Miladinovic
- Laboratoire de Biologie du Développement/UMR7622, Institut de Biologie Paris Seine, Sorbonne Université, CNRS, Inserm U1156,9 Quai St-Bernard, 75005 Paris, France
| | - Pierre-Yves Canto
- Laboratoire de Biologie du Développement/UMR7622, Institut de Biologie Paris Seine, Sorbonne Université, CNRS, Inserm U1156,9 Quai St-Bernard, 75005 Paris, France
| | - Claire Pouget
- Department of Cell and Developmental Biology, University of California San Diego, La Jolla, CA 92093-0380, USA
| | - Olivier Piau
- Laboratoire de Biologie du Développement/UMR7622, Institut de Biologie Paris Seine, Sorbonne Université, CNRS, Inserm U1156,9 Quai St-Bernard, 75005 Paris, France
- Centre de Recherche Saint-Antoine-Team Proliferation and Differentiation of Stem Cells, Institut Universitaire de Cancérologie, Sorbonne Université, Inserm, UMR-S 938,F-75012 Paris, France
| | - Nevenka Radic
- Laboratoire de Biologie du Développement/UMR7622, Institut de Biologie Paris Seine, Sorbonne Université, CNRS, Inserm U1156,9 Quai St-Bernard, 75005 Paris, France
| | - Priscilla Freschu
- Laboratoire de Biologie du Développement/UMR7622, Institut de Biologie Paris Seine, Sorbonne Université, CNRS, Inserm U1156,9 Quai St-Bernard, 75005 Paris, France
| | - Alexandre Megherbi
- Laboratoire de Biologie du Développement/UMR7622, Institut de Biologie Paris Seine, Sorbonne Université, CNRS, Inserm U1156,9 Quai St-Bernard, 75005 Paris, France
| | - Carla Brujas Prats
- Laboratoire de Biologie du Développement/UMR7622, Institut de Biologie Paris Seine, Sorbonne Université, CNRS, Inserm U1156,9 Quai St-Bernard, 75005 Paris, France
| | - Sebastien Jacques
- Plateforme de génomique, Université de Paris, Institut Cochin, Inserm, CNRS, F-75014 Paris, France
| | - Estelle Hirsinger
- Laboratoire de Biologie du Développement/UMR7622, Institut de Biologie Paris Seine, Sorbonne Université, CNRS, Inserm U1156,9 Quai St-Bernard, 75005 Paris, France
| | - Audrey Geeverding
- Service de microscopie électronique, Fr3631 Institut de Biologie Paris Seine, Sorbonne Université, CNRS, 7-9Quai St-Bernard, 75005 Paris, France
| | - Sylvie Dufour
- Université Paris-Est Créteil, Inserm, IMRB, F94010 Créteil, France
| | - Laurence Petit
- Laboratoire de Biologie du Développement/UMR7622, Institut de Biologie Paris Seine, Sorbonne Université, CNRS, Inserm U1156,9 Quai St-Bernard, 75005 Paris, France
| | - Michele Souyri
- Université de Paris, Inserm UMR 1131, Institut de Recherche Saint Louis, Hôpital Saint Louis, 1 Avenue Claude Vellefaux, 75010 Paris, France
| | - Trista North
- Stem Cell Program, Department of Hematology/Oncology, Boston Children's Hospital, Boston, MA 02115, USA
- Developmental and Regenerative Biology Program, Harvard Medical School, Boston, MA 02115, USA
| | - Hervé Isambert
- Institut Curie, PSL Research University, CNRS UMR168, Paris, France
| | - David Traver
- Department of Cell and Developmental Biology, University of California San Diego, La Jolla, CA 92093-0380, USA
| | - Thierry Jaffredo
- Laboratoire de Biologie du Développement/UMR7622, Institut de Biologie Paris Seine, Sorbonne Université, CNRS, Inserm U1156,9 Quai St-Bernard, 75005 Paris, France
| | - Pierre Charbord
- Laboratoire de Biologie du Développement/UMR7622, Institut de Biologie Paris Seine, Sorbonne Université, CNRS, Inserm U1156,9 Quai St-Bernard, 75005 Paris, France
| | - Charles Durand
- Laboratoire de Biologie du Développement/UMR7622, Institut de Biologie Paris Seine, Sorbonne Université, CNRS, Inserm U1156,9 Quai St-Bernard, 75005 Paris, France
| |
Collapse
|
9
|
Gonzalez Galofre ZN, Kilpatrick AM, Marques M, Sá da Bandeira D, Ventura T, Gomez Salazar M, Bouilleau L, Marc Y, Barbosa AB, Rossi F, Beltran M, van de Werken HJG, van IJcken WFJ, Henderson NC, Forbes SJ, Crisan M. Runx1+ vascular smooth muscle cells are essential for hematopoietic stem and progenitor cell development in vivo. Nat Commun 2024; 15:1653. [PMID: 38395882 PMCID: PMC10891074 DOI: 10.1038/s41467-024-44913-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 01/09/2024] [Indexed: 02/25/2024] Open
Abstract
Hematopoietic stem cells (HSCs) produce all essential cellular components of the blood. Stromal cell lines supporting HSCs follow a vascular smooth muscle cell (vSMC) differentiation pathway, suggesting that some hematopoiesis-supporting cells originate from vSMC precursors. These pericyte-like precursors were recently identified in the aorta-gonad-mesonephros (AGM) region; however, their role in the hematopoietic development in vivo remains unknown. Here, we identify a subpopulation of NG2+Runx1+ perivascular cells that display a sclerotome-derived vSMC transcriptomic profile. We show that deleting Runx1 in NG2+ cells impairs the hematopoietic development in vivo and causes transcriptional changes in pericytes/vSMCs, endothelial cells and hematopoietic cells in the murine AGM. Importantly, this deletion leads also to a significant reduction of HSC reconstitution potential in the bone marrow in vivo. This defect is developmental, as NG2+Runx1+ cells were not detected in the adult bone marrow, demonstrating the existence of a specialised pericyte population in the HSC-generating niche, unique to the embryo.
Collapse
Affiliation(s)
- Zaniah N Gonzalez Galofre
- Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Alastair M Kilpatrick
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Madalena Marques
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Diana Sá da Bandeira
- Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Telma Ventura
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Mario Gomez Salazar
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Léa Bouilleau
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Yvan Marc
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Ana B Barbosa
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Fiona Rossi
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Mariana Beltran
- Centre for Inflammation Research/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Harmen J G van de Werken
- Cancer Computational Biology Center, Erasmus MC Cancer Institute, University Medical Center, 3000 CA, Rotterdam, The Netherlands
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center, 3000 CA, Rotterdam, The Netherlands
- Department of Immunology, Erasmus MC Cancer Institute, University Medical Center, 3000 CA, Rotterdam, The Netherlands
| | - Wilfred F J van IJcken
- Center for Biomics, Department of Cell Biology, Erasmus MC University Medical Centre, 3015 GE, Rotterdam, The Netherlands
| | - Neil C Henderson
- Centre for Inflammation Research/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Stuart J Forbes
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK
| | - Mihaela Crisan
- Centre for Cardiovascular Science, The University of Edinburgh, Edinburgh, UK.
- Centre for Regenerative Medicine/Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
10
|
Di X, Chen J, Li Y, Wang M, Wei J, Li T, Liao B, Luo D. Crosstalk between fibroblasts and immunocytes in fibrosis: From molecular mechanisms to clinical trials. Clin Transl Med 2024; 14:e1545. [PMID: 38264932 PMCID: PMC10807359 DOI: 10.1002/ctm2.1545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 12/25/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024] Open
Abstract
BACKGROUND The impact of fibroblasts on the immune system provides insight into the function of fibroblasts. In various tissue microenvironments, multiple fibroblast subtypes interact with immunocytes by secreting growth factors, cytokines, and chemokines, leading to wound healing, fibrosis, and escape of cancer immune surveillance. However, the specific mechanisms involved in the fibroblast-immunocyte interaction network have not yet been fully elucidated. MAIN BODY AND CONCLUSION Therefore, we systematically reviewed the molecular mechanisms of fibroblast-immunocyte interactions in fibrosis, from the history of cellular evolution and cell subtype divisions to the regulatory networks between fibroblasts and immunocytes. We also discuss how these communications function in different tissue and organ statuses, as well as potential therapies targeting the reciprocal fibroblast-immunocyte interplay in fibrosis. A comprehensive understanding of these functional cells under pathophysiological conditions and the mechanisms by which they communicate may lead to the development of effective and specific therapies targeting fibrosis.
Collapse
Affiliation(s)
- Xingpeng Di
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Jiawei Chen
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Ya Li
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Menghua Wang
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Jingwen Wei
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Tianyue Li
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Banghua Liao
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| | - Deyi Luo
- Department of Urology and Institute of UrologyWest China HospitalSichuan UniversityChengduP.R. China
| |
Collapse
|
11
|
Mao A, Li Z, Ning G, Zhou Z, Wei C, Li J, He X, Wang Q. Sclerotome-derived PDGF signaling functions as a niche cue responsible for primitive erythropoiesis. Development 2023; 150:dev201807. [PMID: 37882745 PMCID: PMC10690055 DOI: 10.1242/dev.201807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 10/19/2023] [Indexed: 10/27/2023]
Abstract
Primitive erythropoiesis serves a vital role in embryonic development, generating primitive red blood cells responsible for transportation of oxygen throughout the body. Although diverse niche factors are known to function in definitive hematopoiesis, the microenvironment contributing to primitive hematopoiesis remains largely elusive. Here, we report that platelet-derived growth factor (PDGF) signaling is required for erythroid progenitor differentiation in zebrafish. Ablating pdgfαa (also known as pdgfaa) and pdgfαb (also known as pdgfab) or blocking PDGF signaling with an inhibitor impairs erythroid progenitor differentiation, thus resulting in a significant decrease in the number of erythrocytes. We reveal that pdgfαb is expressed in sclerotomal cells, and that its receptor genes, pdgfra and pdgfrb, are expressed in the adjacent erythroid progenitor cells. Sclerotome-specific overexpression of pdgfαb effectively restores primitive erythropoiesis in pdgfαa-/-;pdgfαb-/- mutant embryos. In addition, we have defined ERK1/2 signaling as a downstream pathway of PDGF signaling during embryonic erythropoiesis. Taken together, our findings indicate that PDGF signaling derived from sclerotome functions as a niche cue for primitive erythropoiesis.
Collapse
Affiliation(s)
- Aihua Mao
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou, Guangdong 515063, China
| | - Zhuyun Li
- Innovation Centre of Ministry of Education for Development and Diseases, Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China
- School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Guozhu Ning
- Affiliated Hospital of Guangdong Medical University and Key Laboratory of Zebrafish Model for Development and Disease, Guangdong Medical University, Zhanjiang 524001, China
| | - Zhengrong Zhou
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Chiju Wei
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, Shantou, Guangdong 515063, China
| | - Jianchao Li
- Innovation Centre of Ministry of Education for Development and Diseases, Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Xinyu He
- Innovation Centre of Ministry of Education for Development and Diseases, Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China
| | - Qiang Wang
- Innovation Centre of Ministry of Education for Development and Diseases, Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
12
|
Yvernogeau L, Dainese G, Jaffredo T. Dorsal aorta polarization and haematopoietic stem cell emergence. Development 2023; 150:286251. [PMID: 36602140 DOI: 10.1242/dev.201173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Recent studies have highlighted the crucial role of the aorta microenvironment in the generation of the first haematopoietic stem cells (HSCs) from specialized haemogenic endothelial cells (HECs). Despite more than two decades of investigations, we require a better understanding of the cellular and molecular events driving aorta formation and polarization, which will be pivotal to establish the mechanisms that operate during HEC specification and HSC competency. Here, we outline the early mechanisms involved in vertebrate aorta formation by comparing four different species: zebrafish, chicken, mouse and human. We highlight how this process, which is tightly controlled in time and space, requires a coordinated specification of several cell types, in particular endothelial cells originating from distinct mesodermal tissues. We also discuss how molecular signals originating from the aorta environment result in its polarization, creating a unique entity for HSC generation.
Collapse
Affiliation(s)
- Laurent Yvernogeau
- Sorbonne Université, IBPS, CNRS UMR7622, Inserm U1156, Laboratoire de Biologie du Développement, 75005 Paris, France
| | - Giovanna Dainese
- Sorbonne Université, IBPS, CNRS UMR7622, Inserm U1156, Laboratoire de Biologie du Développement, 75005 Paris, France
| | - Thierry Jaffredo
- Sorbonne Université, IBPS, CNRS UMR7622, Inserm U1156, Laboratoire de Biologie du Développement, 75005 Paris, France
| |
Collapse
|
13
|
Kapeni C, Nitsche L, Kilpatrick AM, Wilson NK, Xia K, Mirshekar-Syahkal B, Chandrakanthan V, Malouf C, Pimanda JE, Göttgens B, Kirschner K, Tomlinson SR, Ottersbach K. p57Kip2 regulates embryonic blood stem cells by controlling sympathoadrenal progenitor expansion. Blood 2022; 140:464-477. [PMID: 35653588 PMCID: PMC9353151 DOI: 10.1182/blood.2021014853] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 05/13/2022] [Indexed: 11/20/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are of major clinical importance, and finding methods for their in vitro generation is a prime research focus. We show here that the cell cycle inhibitor p57Kip2/Cdkn1c limits the number of emerging HSCs by restricting the size of the sympathetic nervous system (SNS) and the amount of HSC-supportive catecholamines secreted by these cells. This regulation occurs at the SNS progenitor level and is in contrast to the cell-intrinsic function of p57Kip2 in maintaining adult HSCs, highlighting profound differences in cell cycle requirements of adult HSCs compared with their embryonic counterparts. Furthermore, this effect is specific to the aorta-gonad-mesonephros (AGM) region and shows that the AGM is the main contributor to early fetal liver colonization, as early fetal liver HSC numbers are equally affected. Using a range of antagonists in vivo, we show a requirement for intact β2-adrenergic signaling for SNS-dependent HSC expansion. To gain further molecular insights, we have generated a single-cell RNA-sequencing data set of all Ngfr+ sympathoadrenal cells around the dorsal aorta to dissect their differentiation pathway. Importantly, this not only defined the relevant p57Kip2-expressing SNS progenitor stage but also revealed that some neural crest cells, upon arrival at the aorta, are able to take an alternative differentiation pathway, giving rise to a subset of ventrally restricted mesenchymal cells that express important HSC-supportive factors. Neural crest cells thus appear to contribute to the AGM HSC niche via 2 different mechanisms: SNS-mediated catecholamine secretion and HSC-supportive mesenchymal cell production.
Collapse
Affiliation(s)
- Chrysa Kapeni
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Leslie Nitsche
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Alastair M Kilpatrick
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Nicola K Wilson
- Department of Haematology, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Kankan Xia
- Department of Haematology, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Bahar Mirshekar-Syahkal
- Department of Haematology, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Vashe Chandrakanthan
- School of Medical Sciences, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
| | - Camille Malouf
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - John E Pimanda
- School of Medical Sciences, Lowy Cancer Research Centre, UNSW, Sydney, NSW, Australia
- Department of Haematology, The Prince of Wales Hospital, Sydney, NSW, Australia
| | - Berthold Göttgens
- Department of Haematology, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Kristina Kirschner
- Institute of Cancer Sciences and
- CRUK Beatson Institute for Cancer Research, University of Glasgow, Glasgow, United Kingdom
| | - Simon R Tomlinson
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| | - Katrin Ottersbach
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
14
|
Hariom SK, Nelson EJR. Effects of short-term hypergravity on hematopoiesis and vasculogenesis in embryonic zebrafish. LIFE SCIENCES IN SPACE RESEARCH 2022; 34:21-29. [PMID: 35940686 DOI: 10.1016/j.lssr.2022.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 05/12/2022] [Accepted: 05/16/2022] [Indexed: 06/15/2023]
Abstract
Microgravity and hypergravity-induced changes affect both molecular and organismal responses as demonstrated in various animal models. In addition to its inherent advantages, zebrafish have been shown to be incredibly resilient to altered gravity conditions. To understand the effects of altered gravity on animal physiology, especially the cardiovascular system, we used 2 h centrifugations to simulate short-term hypergravity and investigated its effects on zebrafish development. Morphological and in situ hybridization observations show a comparable overall development in both control and treated embryos. Spatiotemporal analysis revealed varied gene expression patterns across different developmental times. Genes driving primitive hematopoiesis (tal1, gata1) and vascular specificity (vegf, etv2) displayed an early onset of expression following hypergravity exposure. Upregulated expression of hematopoiesis-linked genes, such as runx1, cmyb, nos, and pdgf family demonstrate short-term hypergravity to be a factor inducing definitive hematopoiesis through a combinatorial mechanism. We speculate that these short-term hypergravity-induced physiological changes in the developing zebrafish embryos constitute a rescue mechanism to regain homeostasis.
Collapse
Affiliation(s)
- Senthil Kumar Hariom
- SMV124A, Gene Therapy Laboratory, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, TN 632 014, India
| | - Everette Jacob Remington Nelson
- SMV124A, Gene Therapy Laboratory, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, TN 632 014, India.
| |
Collapse
|
15
|
Sá da Bandeira D, Kilpatrick AM, Marques M, Gomez-Salazar M, Ventura T, Gonzalez ZN, Stefancova D, Rossi F, Vermeren M, Vink CS, Beltran M, Henderson NC, Jung B, van der Linden R, van de Werken HJG, van Ijcken WFJ, Betsholtz C, Forbes SJ, Cuervo H, Crisan M. PDGFRβ + cells play a dual role as hematopoietic precursors and niche cells during mouse ontogeny. Cell Rep 2022; 40:111114. [PMID: 35858557 PMCID: PMC9638014 DOI: 10.1016/j.celrep.2022.111114] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/18/2022] [Accepted: 06/28/2022] [Indexed: 11/27/2022] Open
Abstract
Hematopoietic stem cell (HSC) generation in the aorta-gonad-mesonephros region requires HSC specification signals from the surrounding microenvironment. In zebrafish, PDGF-B/PDGFRβ signaling controls hematopoietic stem/progenitor cell (HSPC) generation and is required in the HSC specification niche. Little is known about murine HSPC specification in vivo and whether PDGF-B/PDGFRβ is involved. Here, we show that PDGFRβ is expressed in distinct perivascular stromal cell layers surrounding the mid-gestation dorsal aorta, and its deletion impairs hematopoiesis. We demonstrate that PDGFRβ+ cells play a dual role in murine hematopoiesis. They act in the aortic niche to support HSPCs, and in addition, PDGFRβ+ embryonic precursors give rise to a subset of HSPCs that persist into adulthood. These findings provide crucial information for the controlled production of HSPCs in vitro. PDGFRβ deletion affects hematopoietic development in the AGM in vivo The transcriptome and hematopoietic support of the PDGFRβ-KO niche are altered The osteogenic gene profile and differentiation of KO AGM MSCs are affected PDGFRβ+ early embryonic precursors contribute to EC and HSPC lineages in vivo
Collapse
Affiliation(s)
- Diana Sá da Bandeira
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, UK; Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, EH16 4UU Edinburgh, UK
| | - Alastair Morris Kilpatrick
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, EH16 4UU Edinburgh, UK
| | - Madalena Marques
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, EH16 4UU Edinburgh, UK
| | - Mario Gomez-Salazar
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, EH16 4UU Edinburgh, UK
| | - Telma Ventura
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, EH16 4UU Edinburgh, UK
| | - Zaniah Nashira Gonzalez
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, UK; Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, EH16 4UU Edinburgh, UK
| | - Dorota Stefancova
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, EH16 4UU Edinburgh, UK
| | - Fiona Rossi
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, EH16 4UU Edinburgh, UK
| | - Matthieu Vermeren
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, EH16 4UU Edinburgh, UK
| | - Chris Sebastiaan Vink
- Centre for Inflammation Research, Institute for Regeneration and Repair, The Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, UK
| | - Mariana Beltran
- Centre for Inflammation Research, Institute for Regeneration and Repair, The Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, UK
| | - Neil Cowan Henderson
- Centre for Inflammation Research, Institute for Regeneration and Repair, The Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, UK; MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, EH4 2XU Edinburgh, UK
| | - Bongnam Jung
- Department of Immunology, Genetics, and Pathology, Uppsala University, 751 85 Uppsala, Sweden; Harvard Medical School, Department of Surgery, Boston Children's Hospital, Boston, MA 02115, USA
| | - Reinier van der Linden
- Hubrecht Institute, Department van Oudenaarden Quantitative Biology, 3584 Utrecht, the Netherlands
| | - Harmen Jan George van de Werken
- Erasmus MC Cancer Institute, University Medical Center, Cancer Computational Biology Center, and Departments of Urology and Immunology, 3000 Rotterdam, the Netherlands
| | - Wilfred F J van Ijcken
- Center for Biomics, Department of Cell Biology, Erasmus MC University Medical Centre, 3015 Rotterdam, the Netherlands
| | - Christer Betsholtz
- Department of Immunology, Genetics, and Pathology, Uppsala University, 751 85 Uppsala, Sweden; Department of Medicine Huddinge, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Stuart John Forbes
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, EH16 4UU Edinburgh, UK
| | - Henar Cuervo
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Mihaela Crisan
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, EH16 4TJ Edinburgh, UK; Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 5 Little France Drive, EH16 4UU Edinburgh, UK.
| |
Collapse
|
16
|
Li D, Huang LT, Zhang CP, Li Q, Wang JH. Insights Into the Role of Platelet-Derived Growth Factors: Implications for Parkinson’s Disease Pathogenesis and Treatment. Front Aging Neurosci 2022; 14:890509. [PMID: 35847662 PMCID: PMC9283766 DOI: 10.3389/fnagi.2022.890509] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Parkinson’s disease (PD), the second most common neurodegenerative disease after Alzheimer’s disease, commonly occurs in the elderly population, causing a significant medical and economic burden to the aging society worldwide. At present, there are few effective methods that achieve satisfactory clinical results in the treatment of PD. Platelet-derived growth factors (PDGFs) and platelet-derived growth factor receptors (PDGFRs) are important neurotrophic factors that are expressed in various cell types. Their unique structures allow for specific binding that can effectively regulate vital functions in the nervous system. In this review, we summarized the possible mechanisms by which PDGFs/PDGFRs regulate the occurrence and development of PD by affecting oxidative stress, mitochondrial function, protein folding and aggregation, Ca2+ homeostasis, and cell neuroinflammation. These modes of action mainly depend on the type and distribution of PDGFs in different nerve cells. We also summarized the possible clinical applications and prospects for PDGF in the treatment of PD, especially in genetic treatment. Recent advances have shown that PDGFs have contradictory roles within the central nervous system (CNS). Although they exert neuroprotective effects through multiple pathways, they are also associated with the disruption of the blood–brain barrier (BBB). Our recommendations based on our findings include further investigation of the contradictory neurotrophic and neurotoxic effects of the PDGFs acting on the CNS.
Collapse
Affiliation(s)
- Dan Li
- Department of Family Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Le-Tian Huang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Cheng-pu Zhang
- Department of Family Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Qiang Li
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Qiang Li,
| | - Jia-He Wang
- Department of Family Medicine, Shengjing Hospital of China Medical University, Shenyang, China
- Jia-He Wang,
| |
Collapse
|
17
|
Yang S, Ning G, Hou Y, Cao Y, Xu J, Wu J, Zhang T, Wang Q. Myoneurin regulates BMP signaling by competing with Ppm1a for Smad binding. iScience 2022; 25:104495. [PMID: 35712083 PMCID: PMC9194458 DOI: 10.1016/j.isci.2022.104495] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 04/07/2022] [Accepted: 05/23/2022] [Indexed: 11/25/2022] Open
Abstract
A delicate balance of BMP activity is critical for tissue formation and organogenesis. However, the mechanical molecular details in ensuring the proper duration and intensity of BMP signaling have yet to be fully elucidated. Here, we identified a zebrafish mutant with a disrupted gene encoding for the BTB/POZ and zinc finger protein myoneurin (Mynn). mynn−/− mutants exhibited severe loss of pharyngeal cartilage elements, owing to poor proliferation, blocked differentiation, and low viability of cranial neural crest cells. Depletion of mynn in both zebrafish embryos and mammalian cells led to a reduction of the BMP signal activity. Mechanistically, Mynn interacts with Smad proteins in the nucleus, thereby disrupting the association between Smad protein and the phosphatase Ppm1a. Ultimately, this interaction prevents Smad dephosphorylation. More broadly, our findings may provide a new strategy to balance BMP signal activity via competitive binding of Mynn and Ppm1a to Smad proteins during pharyngeal cartilage formation. mynn gene is essential for pharyngeal cartilage development mynn is required for the proliferation, differentiation, and survival of the CNCCs Mynn has an evolutionarily conserved function in supporting BMP signal Mynn maintains BMP signal activity by competing with Ppm1a for Smad binding
Collapse
|
18
|
Ando K, Ishii T, Fukuhara S. Zebrafish Vascular Mural Cell Biology: Recent Advances, Development, and Functions. Life (Basel) 2021; 11:1041. [PMID: 34685412 PMCID: PMC8537713 DOI: 10.3390/life11101041] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/29/2021] [Accepted: 09/29/2021] [Indexed: 12/14/2022] Open
Abstract
Recruitment of mural cells to the vascular wall is essential for forming the vasculature as well as maintaining proper vascular functions. In recent years, zebrafish genetic tools for mural cell biology have improved substantially. Fluorescently labeled zebrafish mural cell reporter lines enable us to study, with higher spatiotemporal resolution than ever, the processes of mural cell development from their progenitors. Furthermore, recent phenotypic analysis of platelet-derived growth factor beta mutant zebrafish revealed well-conserved organotypic mural cell development and functions in vertebrates with the unique features of zebrafish. However, comprehensive reviews of zebrafish mural cells are lacking. Therefore, herein, we highlight recent advances in zebrafish mural cell tools. We also summarize the fundamental features of zebrafish mural cell development, especially at early stages, and functions.
Collapse
Affiliation(s)
- Koji Ando
- Department of Molecular Pathophysiology, Institute of Advanced Medical Sciences, Nippon Medical School, Tokyo 113 8602, Japan; (T.I.); (S.F.)
| | | | | |
Collapse
|
19
|
Sugden WW, North TE. Making Blood from the Vessel: Extrinsic and Environmental Cues Guiding the Endothelial-to-Hematopoietic Transition. Life (Basel) 2021; 11:life11101027. [PMID: 34685398 PMCID: PMC8539454 DOI: 10.3390/life11101027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/25/2021] [Accepted: 09/27/2021] [Indexed: 01/10/2023] Open
Abstract
It is increasingly recognized that specialized subsets of endothelial cells carry out unique functions in specific organs and regions of the vascular tree. Perhaps the most striking example of this specialization is the ability to contribute to the generation of the blood system, in which a distinct population of “hemogenic” endothelial cells in the embryo transforms irreversibly into hematopoietic stem and progenitor cells that produce circulating erythroid, myeloid and lymphoid cells for the lifetime of an animal. This review will focus on recent advances made in the zebrafish model organism uncovering the extrinsic and environmental factors that facilitate hemogenic commitment and the process of endothelial-to-hematopoietic transition that produces blood stem cells. We highlight in particular biomechanical influences of hemodynamic forces and the extracellular matrix, metabolic and sterile inflammatory cues present during this developmental stage, and outline new avenues opened by transcriptomic-based approaches to decipher cell–cell communication mechanisms as examples of key signals in the embryonic niche that regulate hematopoiesis.
Collapse
Affiliation(s)
- Wade W. Sugden
- Stem Cell Program, Department of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA;
- Developmental and Regenerative Biology Program, Harvard Medical School, Boston, MA 02115, USA
| | - Trista E. North
- Stem Cell Program, Department of Hematology/Oncology, Boston Children’s Hospital, Boston, MA 02115, USA;
- Developmental and Regenerative Biology Program, Harvard Medical School, Boston, MA 02115, USA
- Correspondence:
| |
Collapse
|
20
|
Choe CP, Choi SY, Kee Y, Kim MJ, Kim SH, Lee Y, Park HC, Ro H. Transgenic fluorescent zebrafish lines that have revolutionized biomedical research. Lab Anim Res 2021; 37:26. [PMID: 34496973 PMCID: PMC8424172 DOI: 10.1186/s42826-021-00103-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/26/2021] [Indexed: 12/22/2022] Open
Abstract
Since its debut in the biomedical research fields in 1981, zebrafish have been used as a vertebrate model organism in more than 40,000 biomedical research studies. Especially useful are zebrafish lines expressing fluorescent proteins in a molecule, intracellular organelle, cell or tissue specific manner because they allow the visualization and tracking of molecules, intracellular organelles, cells or tissues of interest in real time and in vivo. In this review, we summarize representative transgenic fluorescent zebrafish lines that have revolutionized biomedical research on signal transduction, the craniofacial skeletal system, the hematopoietic system, the nervous system, the urogenital system, the digestive system and intracellular organelles.
Collapse
Affiliation(s)
- Chong Pyo Choe
- Division of Life Science, Gyeongsang National University, Jinju, 52828, Republic of Korea.,Division of Applied Life Science, Plant Molecular Biology and Biotechnology Research Center, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Seok-Yong Choi
- Department of Biomedical Sciences, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Yun Kee
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| | - Min Jung Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul, 04310, Republic of Korea
| | - Seok-Hyung Kim
- Department of Marine Life Sciences and Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea
| | - Yoonsung Lee
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan, 44919, Republic of Korea
| | - Hae-Chul Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan, 15355, Republic of Korea
| | - Hyunju Ro
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon, 34134, Republic of Korea
| |
Collapse
|
21
|
Franklin RA. Fibroblasts and macrophages: Collaborators in tissue homeostasis. Immunol Rev 2021; 302:86-103. [PMID: 34101202 DOI: 10.1111/imr.12989] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/12/2021] [Accepted: 05/15/2021] [Indexed: 12/19/2022]
Abstract
Fibroblasts and macrophages are universal cell types across all mammalian tissues. These cells differ in many ways including their cellular origins; dynamics of renewal, recruitment, and motility within tissues; roles in tissue structure and secretion of signaling molecules; and contributions to the activation and progression of immune responses. However, many of the features that make these two cell types unique are not opposing, but instead complementary. This review will present cell-cell communication in this context and discuss how complementarity makes fibroblasts and macrophages highly compatible partners in the maintenance of tissue homeostasis.
Collapse
Affiliation(s)
- Ruth A Franklin
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.,Department of Immunology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
22
|
Girard D, Torossian F, Oberlin E, Alexander KA, Gueguen J, Tseng HW, Genêt F, Lataillade JJ, Salga M, Levesque JP, Le Bousse-Kerdilès MC, Banzet S. Neurogenic Heterotopic Ossifications Recapitulate Hematopoietic Stem Cell Niche Development Within an Adult Osteogenic Muscle Environment. Front Cell Dev Biol 2021; 9:611842. [PMID: 33748104 PMCID: PMC7973025 DOI: 10.3389/fcell.2021.611842] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/17/2021] [Indexed: 12/11/2022] Open
Abstract
Hematopoiesis and bone interact in various developmental and pathological processes. Neurogenic heterotopic ossifications (NHO) are the formation of ectopic hematopoietic bones in peri-articular muscles that develop following severe lesions of the central nervous system such as traumatic cerebral or spinal injuries or strokes. This review will focus on the hematopoietic facet of NHO. The characterization of NHO demonstrates the presence of hematopoietic marrow in which quiescent hematopoietic stem cells (HSC) are maintained by a functional stromal microenvironment, thus documenting that NHOs are neo-formed ectopic HSC niches. Similarly to adult bone marrow, the NHO permissive environment supports HSC maintenance, proliferation and differentiation through bidirectional signaling with mesenchymal stromal cells and endothelial cells, involving cell adhesion molecules, membrane-bound growth factors, hormones, and secreted matrix proteins. The participation of the nervous system, macrophages and inflammatory cytokines including oncostatin M and transforming growth factor (TGF)-β in this process, reveals how neural circuitry fine-tunes the inflammatory response to generate hematopoietic bones in injured muscles. The localization of NHOs in the peri-articular muscle environment also suggests a role of muscle mesenchymal cells and bone metabolism in development of hematopoiesis in adults. Little is known about the establishment of bone marrow niches and the regulation of HSC cycling during fetal development. Similarities between NHO and development of fetal bones make NHOs an interesting model to study the establishment of bone marrow hematopoiesis during development. Conversely, identification of stage-specific factors that specify HSC developmental state during fetal bone development will give more mechanistic insights into NHO.
Collapse
Affiliation(s)
- Dorothée Girard
- INSERM UMRS-MD 1197, Institut de Recherche Biomédicale des Armées (IRBA), Clamart, France
| | - Frédéric Torossian
- INSERM UMRS-MD 1197, Université Paris-Saclay, Hôpital Paul Brousse, Villejuif, France
| | - Estelle Oberlin
- INSERM UMRS-MD 1197, Université Paris-Saclay, Hôpital Paul Brousse, Villejuif, France
| | - Kylie A. Alexander
- Mater Research Institute—The University of Queensland, Woolloongabba, QLD, Australia
| | - Jules Gueguen
- INSERM UMRS-MD 1197, Institut de Recherche Biomédicale des Armées (IRBA), Clamart, France
| | - Hsu-Wen Tseng
- Mater Research Institute—The University of Queensland, Woolloongabba, QLD, Australia
| | - François Genêt
- INSERM U1179, Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Versailles, France
| | | | - Marjorie Salga
- INSERM U1179, Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), Versailles, France
| | - Jean-Pierre Levesque
- Mater Research Institute—The University of Queensland, Woolloongabba, QLD, Australia
| | | | - Sébastien Banzet
- INSERM UMRS-MD 1197, Institut de Recherche Biomédicale des Armées (IRBA), Clamart, France
| |
Collapse
|
23
|
Belmonte RL, Engbretson IL, Kim JH, Cajias I, Ahn EYE, Stachura DL. son is necessary for proper vertebrate blood development. PLoS One 2021; 16:e0247489. [PMID: 33630943 PMCID: PMC7906411 DOI: 10.1371/journal.pone.0247489] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 02/08/2021] [Indexed: 12/15/2022] Open
Abstract
The gene SON is on human chromosome 21 (21q22.11) and is thought to be associated with hematopoietic disorders that accompany Down syndrome. Additionally, SON is an RNA splicing factor that plays a role in the transcription of leukemia-associated genes. Previously, we showed that mutations in SON cause malformations in human and zebrafish spines and brains during early embryonic development. To examine the role of SON in normal hematopoiesis, we reduced expression of the zebrafish homolog of SON in zebrafish at the single-cell developmental stage with specific morpholinos. In addition to the brain and spinal malformations we also observed abnormal blood cell levels upon son knockdown. We then investigated how blood production was altered when levels of son were reduced. Decreased levels of son resulted in lower amounts of red blood cells when visualized with lcr:GFP transgenic fish. There were also reduced thrombocytes seen with cd41:GFP fish, and myeloid cells when mpx:GFP fish were examined. We also observed a significant decrease in the quantity of T cells, visualized with lck:GFP fish. However, when we examined their hematopoietic stem and progenitor cells (HSPCs), we saw no difference in colony-forming capability. These studies indicate that son is essential for the proper differentiation of the innate and adaptive immune system, and further investigation determining the molecular pathways involved during blood development should elucidate important information about vertebrate HSPC generation, proliferation, and differentiation.
Collapse
Affiliation(s)
- Rebecca L. Belmonte
- Department of Biological Sciences, California State University Chico, Chico, California, United States of America
| | - Isabella L. Engbretson
- Department of Biological Sciences, California State University Chico, Chico, California, United States of America
| | - Jung-Hyun Kim
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, United States of America
| | - Illiana Cajias
- Department of Biological Sciences, California State University Chico, Chico, California, United States of America
| | - Eun-Young Erin Ahn
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - David L. Stachura
- Department of Biological Sciences, California State University Chico, Chico, California, United States of America
- * E-mail:
| |
Collapse
|
24
|
Grainger S, Nguyen N, Richter J, Setayesh J, Lonquich B, Oon CH, Wozniak JM, Barahona R, Kamei CN, Houston J, Carrillo-Terrazas M, Drummond IA, Gonzalez D, Willert K, Traver D. EGFR is required for Wnt9a-Fzd9b signalling specificity in haematopoietic stem cells. Nat Cell Biol 2019; 21:721-730. [PMID: 31110287 PMCID: PMC6559346 DOI: 10.1038/s41556-019-0330-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 04/11/2019] [Indexed: 12/31/2022]
Abstract
Wnt signalling drives a plethora of processes in development, homeostasis, and disease; however, the role and mechanism of individual ligand/receptor (Wnt/Frizzled, Fzd) interactions in specific biological processes remain poorly understood. Wnt9a is specifically required for the amplification of blood progenitor cells during development. Using genetic studies in zebrafish and human embryonic stem cells, paired with in vitro cell biology and biochemistry, we have determined that Wnt9a signals specifically through Fzd9b to elicit β-catenin-dependent Wnt signalling that regulates haematopoietic stem and progenitor cell emergence. We demonstrate that the epidermal growth factor receptor (EGFR) is required as a co-factor for Wnt9a/Fzd9b signalling. EGFR-mediated phosphorylation of one tyrosine residue on the Fzd9b intracellular tail in response to Wnt9a promotes internalization of the Wnt9a/Fzd9b/LRP signalosome and subsequent signal transduction. These findings provide mechanistic insights for specific Wnt/Fzd signals, which will be crucial for specific therapeutic targeting and regenerative medicine.
Collapse
Affiliation(s)
- Stephanie Grainger
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Nicole Nguyen
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jenna Richter
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA.,Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Jordan Setayesh
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Brianna Lonquich
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Chet Huan Oon
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jacob M Wozniak
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA, USA.,Skaggs School of Pharmacy and Pharmaceutical Science, University of California, San Diego, La Jolla, CA, USA.,Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Rocio Barahona
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Caramai N Kamei
- Massachusetts General Hospital Nephrology Division, Charlestown, MA, USA
| | - Jack Houston
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA.,Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Marvic Carrillo-Terrazas
- Skaggs School of Pharmacy and Pharmaceutical Science, University of California, San Diego, La Jolla, CA, USA.,Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Iain A Drummond
- Massachusetts General Hospital Nephrology Division, Charlestown, MA, USA.,Harvard Medical School, Department of Genetics, Boston, MA, USA
| | - David Gonzalez
- Skaggs School of Pharmacy and Pharmaceutical Science, University of California, San Diego, La Jolla, CA, USA.,Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Karl Willert
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA.
| | - David Traver
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA. .,Section of Cell and Developmental Biology, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
25
|
Bajanca F, Gouignard N, Colle C, Parsons M, Mayor R, Theveneau E. In vivo topology converts competition for cell-matrix adhesion into directional migration. Nat Commun 2019; 10:1518. [PMID: 30944331 PMCID: PMC6447549 DOI: 10.1038/s41467-019-09548-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 03/11/2019] [Indexed: 12/14/2022] Open
Abstract
When migrating in vivo, cells are exposed to numerous conflicting signals: chemokines, repellents, extracellular matrix, growth factors. The roles of several of these molecules have been studied individually in vitro or in vivo, but we have yet to understand how cells integrate them. To start addressing this question, we used the cephalic neural crest as a model system and looked at the roles of its best examples of positive and negative signals: stromal-cell derived factor 1 (Sdf1/Cxcl12) and class3-Semaphorins. Here we show that Sdf1 and Sema3A antagonistically control cell-matrix adhesion via opposite effects on Rac1 activity at the single cell level. Directional migration at the population level emerges as a result of global Semaphorin-dependent confinement and broad activation of adhesion by Sdf1 in the context of a biased Fibronectin distribution. These results indicate that uneven in vivo topology renders the need for precise distribution of secreted signals mostly dispensable.
Collapse
Affiliation(s)
- Fernanda Bajanca
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 118 route de Narbonne, 31062, Toulouse, Cedex 09, France
| | - Nadège Gouignard
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 118 route de Narbonne, 31062, Toulouse, Cedex 09, France
| | - Charlotte Colle
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Maddy Parsons
- Kings College London, Randall Centre for Cell and Molecular Biophysics Room 3.22B, New Hunts House, Guys Campus, London, SE1 1UL, UK
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Eric Theveneau
- Centre de Biologie du Développement (CBD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 118 route de Narbonne, 31062, Toulouse, Cedex 09, France.
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
26
|
Zhang Y, Liu F. Multidimensional Single-Cell Analyses in Organ Development and Maintenance. Trends Cell Biol 2019; 29:477-486. [PMID: 30928527 DOI: 10.1016/j.tcb.2019.02.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 02/12/2019] [Accepted: 02/19/2019] [Indexed: 12/15/2022]
Abstract
The revolution of single-cell analysis tools in epigenomics, transcriptomics, lineage tracing, and transcriptome-scale RNA imaging, has boosted our understanding of the underlying molecular mechanisms during organ development and maintenance. Application of these tools enables the multidimensional study of organs, from cell atlas profiling, spatial organization, to cell-cell interaction. Here, we discuss recent progress in employing multidimensional single-cell analyses to address fundamental questions related to the development and maintenance of hematopoietic organs, brain and lung, which will also help provide insights into a better understanding of relevant diseases.
Collapse
Affiliation(s)
- Yifan Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Feng Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
27
|
Delloye-Bourgeois C, Castellani V. Hijacking of Embryonic Programs by Neural Crest-Derived Neuroblastoma: From Physiological Migration to Metastatic Dissemination. Front Mol Neurosci 2019; 12:52. [PMID: 30881286 PMCID: PMC6405627 DOI: 10.3389/fnmol.2019.00052] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 02/12/2019] [Indexed: 12/12/2022] Open
Abstract
In the developing organism, complex molecular programs orchestrate the generation of cells in adequate numbers, drive them to migrate along the correct pathways towards appropriate territories, eliminate superfluous cells, and induce terminal differentiation of survivors into the appropriate cell-types. Despite strict controls constraining developmental processes, malignancies can emerge in still immature organisms. This is the case of neuroblastoma (NB), a highly heterogeneous disease, predominantly affecting children before the age of 5 years. Highly metastatic forms represent half of the cases and are diagnosed when disseminated foci are detectable. NB arise from a transient population of embryonic cells, the neural crest (NC), and especially NC committed to the establishment of the sympatho-adrenal tissues. The NC is generated at the dorsal edge of the neural tube (NT) of the vertebrate embryo, under the action of NC specifier gene programs. NC cells (NCCs) undergo an epithelial to mesenchymal transition, and engage on a remarkable journey in the developing embryo, contributing to a plethora of cell-types and tissues. Various NCC sub-populations and derived lineages adopt specific migratory behaviors, moving individually as well as collectively, exploiting the different embryonic substrates they encounter along their path. Here we discuss how the specific features of NCC in development are re-iterated during NB metastatic behaviors.
Collapse
Affiliation(s)
- Céline Delloye-Bourgeois
- University of Lyon, University of Lyon 1 Claude Bernard Lyon 1, NeuroMyoGene Institute, CNRS UMR5310, INSERM U1217, Lyon, France
| | - Valérie Castellani
- University of Lyon, University of Lyon 1 Claude Bernard Lyon 1, NeuroMyoGene Institute, CNRS UMR5310, INSERM U1217, Lyon, France
| |
Collapse
|
28
|
Abstract
Humoral regulation by ligand/receptor interactions is a fundamental feature of vertebrate hematopoiesis. Zebrafish are an established vertebrate animal model of hematopoiesis, sharing with mammals conserved genetic, molecular and cell biological regulatory mechanisms. This comprehensive review considers zebrafish hematopoiesis from the perspective of the hematopoietic growth factors (HGFs), their receptors and their actions. Zebrafish possess multiple HGFs: CSF1 (M-CSF) and CSF3 (G-CSF), kit ligand (KL, SCF), erythropoietin (EPO), thrombopoietin (THPO/TPO), and the interleukins IL6, IL11, and IL34. Some ligands and/or receptor components have been duplicated by various mechanisms including the teleost whole genome duplication, adding complexity to the ligand/receptor interactions possible, but also providing examples of several different outcomes of ligand and receptor subfunctionalization or neofunctionalization. CSF2 (GM-CSF), IL3 and IL5 and their receptors are absent from zebrafish. Overall the humoral regulation of hematopoiesis in zebrafish displays considerable similarity with mammals, which can be applied in biological and disease modelling research.
Collapse
Affiliation(s)
- Vahid Pazhakh
- a Australian Regenerative Medicine Institute, Monash University , Clayton , Australia
| | - Graham J Lieschke
- a Australian Regenerative Medicine Institute, Monash University , Clayton , Australia
| |
Collapse
|
29
|
Agarwala S, Tamplin OJ. Neural Crossroads in the Hematopoietic Stem Cell Niche. Trends Cell Biol 2018; 28:987-998. [PMID: 29857963 DOI: 10.1016/j.tcb.2018.05.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 05/05/2018] [Accepted: 05/10/2018] [Indexed: 01/25/2023]
Abstract
The hematopoietic stem cell (HSC) niche supports steady-state hematopoiesis and responds to changing needs during stress and disease. The nervous system is an important regulator of the niche, and its influence is established early in development when stem cells are specified. Most research has focused on direct innervation of the niche, however recent findings show there are different modes of neural control, including globally by the central nervous system (CNS) and hormone release, locally by neural crest-derived mesenchymal stem cells, and intrinsically by hematopoietic cells that express neural receptors and neurotransmitters. Dysregulation between neural and hematopoietic systems can contribute to disease, however new therapeutic opportunities may be found among neuroregulator drugs repurposed to support hematopoiesis.
Collapse
Affiliation(s)
- Sobhika Agarwala
- Center for Stem Cell and Regenerative Medicine, Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Owen J Tamplin
- Center for Stem Cell and Regenerative Medicine, Department of Pharmacology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
30
|
Gore AV, Pillay LM, Venero Galanternik M, Weinstein BM. The zebrafish: A fintastic model for hematopoietic development and disease. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2018; 7:e312. [PMID: 29436122 DOI: 10.1002/wdev.312] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 11/30/2017] [Accepted: 12/03/2017] [Indexed: 12/19/2022]
Abstract
Hematopoiesis is a complex process with a variety of different signaling pathways influencing every step of blood cell formation from the earliest precursors to final differentiated blood cell types. Formation of blood cells is crucial for survival. Blood cells carry oxygen, promote organ development and protect organs in different pathological conditions. Hematopoietic stem and progenitor cells (HSPCs) are responsible for generating all adult differentiated blood cells. Defects in HSPCs or their downstream lineages can lead to anemia and other hematological disorders including leukemia. The zebrafish has recently emerged as a powerful vertebrate model system to study hematopoiesis. The developmental processes and molecular mechanisms involved in zebrafish hematopoiesis are conserved with higher vertebrates, and the genetic and experimental accessibility of the fish and the optical transparency of its embryos and larvae make it ideal for in vivo analysis of hematopoietic development. Defects in zebrafish hematopoiesis reliably phenocopy human blood disorders, making it a highly attractive model system to screen small molecules to design therapeutic strategies. In this review, we summarize the key developmental processes and molecular mechanisms of zebrafish hematopoiesis. We also discuss recent findings highlighting the strengths of zebrafish as a model system for drug discovery against hematopoietic disorders. This article is categorized under: Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cell Differentiation and Reversion Vertebrate Organogenesis > Musculoskeletal and Vascular Nervous System Development > Vertebrates: Regional Development Comparative Development and Evolution > Organ System Comparisons Between Species.
Collapse
Affiliation(s)
- Aniket V Gore
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland
| | - Laura M Pillay
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland
| | - Marina Venero Galanternik
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland
| | - Brant M Weinstein
- Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland
| |
Collapse
|
31
|
Zhou X, Franklin RA, Adler M, Jacox JB, Bailis W, Shyer JA, Flavell RA, Mayo A, Alon U, Medzhitov R. Circuit Design Features of a Stable Two-Cell System. Cell 2018; 172:744-757.e17. [PMID: 29398113 PMCID: PMC7377352 DOI: 10.1016/j.cell.2018.01.015] [Citation(s) in RCA: 264] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 11/06/2017] [Accepted: 01/08/2018] [Indexed: 12/14/2022]
Abstract
Cell communication within tissues is mediated by multiple paracrine signals including growth factors, which control cell survival and proliferation. Cells and the growth factors they produce and receive constitute a circuit with specific properties that ensure homeostasis. Here, we used computational and experimental approaches to characterize the features of cell circuits based on growth factor exchange between macrophages and fibroblasts, two cell types found in most mammalian tissues. We found that the macrophage-fibroblast cell circuit is stable and robust to perturbations. Analytical screening of all possible two-cell circuit topologies revealed the circuit features sufficient for stability, including environmental constraint and negative-feedback regulation. Moreover, we found that cell-cell contact is essential for the stability of the macrophage-fibroblast circuit. These findings illustrate principles of cell circuit design and provide a quantitative perspective on cell interactions.
Collapse
Affiliation(s)
- Xu Zhou
- Howard Hughes Medical Institute, Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Ruth A Franklin
- Howard Hughes Medical Institute, Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Miri Adler
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Jeremy B Jacox
- Howard Hughes Medical Institute, Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Will Bailis
- Howard Hughes Medical Institute, Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Justin A Shyer
- Howard Hughes Medical Institute, Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Richard A Flavell
- Howard Hughes Medical Institute, Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Avi Mayo
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Uri Alon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, 7610001, Israel.
| | - Ruslan Medzhitov
- Howard Hughes Medical Institute, Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
32
|
Ganuza M, Hall T, Finkelstein D, Chabot A, Kang G, McKinney-Freeman S. Lifelong haematopoiesis is established by hundreds of precursors throughout mammalian ontogeny. Nat Cell Biol 2017; 19:1153-1163. [PMID: 28920953 PMCID: PMC5705075 DOI: 10.1038/ncb3607] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 08/09/2017] [Indexed: 12/17/2022]
Abstract
Current dogma asserts that mammalian lifelong blood production is established by a small number of blood progenitors. However, this model is based on assays that require the disruption, transplantation and/or culture of embryonic tissues. Here, we used the sample-to-sample variance of a multicoloured lineage trace reporter to assess the frequency of emerging lifelong blood progenitors while avoiding the disruption, culture or transplantation of embryos. We find that approximately 719 Flk1+ mesodermal precursors, 633 VE-cadherin+ endothelial precursors and 545 Vav1+ nascent blood stem and progenitor cells emerge to establish the haematopoietic system at embryonic days (E)7-E8.5, E8.5-E11.5 and E11.5-E14.5, respectively. We also determined that the spatio-temporal recruitment of endothelial blood precursors begins at E8.5 and ends by E10.5, and that many c-Kit+ clusters of newly specified blood progenitors in the aorta are polyclonal in origin. Our work illuminates the dynamics of the developing mammalian blood system during homeostasis.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Cadherins/genetics
- Cadherins/metabolism
- Cell Differentiation
- Cell Lineage
- Cell Tracking/methods
- Cells, Cultured
- Coculture Techniques
- Endothelial Cells/metabolism
- Endothelial Cells/transplantation
- Gene Expression Regulation, Developmental
- Genotype
- Gestational Age
- Hematopoiesis
- Hematopoietic Stem Cell Transplantation
- Hematopoietic Stem Cells/metabolism
- Integrases/genetics
- Integrases/metabolism
- Linear Models
- Luminescent Proteins/genetics
- Luminescent Proteins/metabolism
- Mice, Inbred C57BL
- Mice, Transgenic
- Microscopy, Fluorescence
- Models, Biological
- Phenotype
- Proto-Oncogene Proteins c-kit/genetics
- Proto-Oncogene Proteins c-kit/metabolism
- Proto-Oncogene Proteins c-vav/genetics
- Proto-Oncogene Proteins c-vav/metabolism
- RNA, Untranslated/genetics
- RNA, Untranslated/metabolism
- Signal Transduction
- Time Factors
- Vascular Endothelial Growth Factor Receptor-2/genetics
- Vascular Endothelial Growth Factor Receptor-2/metabolism
Collapse
Affiliation(s)
- Miguel Ganuza
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, 38105
| | - Trent Hall
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, 38105
| | - David Finkelstein
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105
| | - Ashley Chabot
- Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, 38105
| | - Guolian Kang
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN, 38105
| | | |
Collapse
|
33
|
Nik S, Weinreb JT, Bowman TV. Developmental HSC Microenvironments: Lessons from Zebrafish. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1041:33-53. [PMID: 29204828 DOI: 10.1007/978-3-319-69194-7_4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hematopoietic stem cells (HSCs) posses the ability to maintain the blood system of an organism from birth to adulthood. The behavior of HSCs is modulated by its microenvironment. During development, HSCs acquire the instructions to self-renew and differentiate into all blood cell fates by passing through several developmental microenvironments. In this chapter, we discuss the signals and cell types that inform HSC decisions throughout ontogeny with a focus on HSC specification, mobilization, migration, and engraftment.
Collapse
Affiliation(s)
- Sara Nik
- Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Joshua T Weinreb
- Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.,Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Teresa V Bowman
- Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA. .,Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA. .,Departments of Molecular Biology and Medicine (Oncology), Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|