51
|
Mendes MS, Majewska AK. An overview of microglia ontogeny and maturation in the homeostatic and pathological brain. Eur J Neurosci 2021; 53:3525-3547. [PMID: 33835613 PMCID: PMC8225243 DOI: 10.1111/ejn.15225] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 03/17/2021] [Accepted: 04/01/2021] [Indexed: 12/21/2022]
Abstract
Microglia are the resident immune cells of the central nervous system (CNS) and are increasingly recognized as critical players in development, brain homeostasis, and disease pathogenesis. The lifespan, maintenance, proliferation, and turnover of microglia are important factors that regulate microglial behavior and affect their roles in the CNS. However, emerging evidence suggests that microglia are morphologically and phenotypically distinct in different brain areas, at different ages, and during disease. Ongoing research focuses on understanding how microglia acquire specific phenotypes in response to extrinsic cues in the environment and how phenotypes are specified by intrinsic properties of different populations of microglia. With the development of pharmacological and genetic tools that allow the investigation of microglia in vivo, there have been considerable advances in understanding molecular signatures of both homeostatic microglia and those reacting to injury and disease. Here, we review the master gene regulators that define microglia as well as discuss the evidence that microglia are heterogeneous and fall into distinct clusters that display specific intrinsic properties and perform unique tasks in different settings. Taken together, the information presented supports the idea that microglia morphology and transcriptional heterogeneity should be considered when studying the complex nature of microglia and their roles in brain health and disease.
Collapse
Affiliation(s)
- Monique S Mendes
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
| | - Ania K Majewska
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
- Center for Visual Science, University of Rochester, Rochester, NY, USA
| |
Collapse
|
52
|
Sirkis DW, Bonham LW, Yokoyama JS. The Role of Microglia in Inherited White-Matter Disorders and Connections to Frontotemporal Dementia. Appl Clin Genet 2021; 14:195-207. [PMID: 33833548 PMCID: PMC8020808 DOI: 10.2147/tacg.s245029] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/17/2021] [Indexed: 12/13/2022] Open
Abstract
Microglia play a critical but poorly understood role in promoting white-matter homeostasis. In this review, we leverage advances in human genetics and mouse models of leukodystrophies to delineate our current knowledge and identify outstanding questions regarding the impact of microglia on central nervous system white matter. We first focus on the role of pathogenic mutations in genes, such as TREM2, TYROBP, and CSF1R, that cause leukodystrophies in which the primary deficit is thought to originate in microglia. We next discuss recent advances in disorders such as adrenoleukodystrophy and Krabbe disease, in which microglia play an increasingly recognized role. We conclude by reviewing the roles of GRN and related genes, such as TMEM106B, PSAP, and SORT1, that affect microglial biology and associate with several types of disease, including multiple leukodystrophies as well as forms of frontotemporal dementia (FTD) presenting with white-matter abnormalities. Taken together, mouse and human data support the notion that loss of microglia-facilitated white-matter homeostasis plays an important role in the development of leukodystrophies and suggest novel mechanisms contributing to FTD.
Collapse
Affiliation(s)
- Daniel W Sirkis
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Luke W Bonham
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, 94158, USA.,Department of Radiology & Biomedical Imaging, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Jennifer S Yokoyama
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, 94158, USA.,Department of Radiology & Biomedical Imaging, University of California San Francisco, San Francisco, CA, 94158, USA
| |
Collapse
|
53
|
Estrada-Capetillo L, Aragoneses-Fenoll L, Domínguez-Soto Á, Fuentelsaz-Romero S, Nieto C, Simón-Fuentes M, Alonso B, Portolés P, Corbí AL, Rojo JM, Puig-Kröger A. CD28 is expressed by macrophages with anti-inflammatory potential and limits their T-cell activating capacity. Eur J Immunol 2021; 51:824-834. [PMID: 33169838 DOI: 10.1002/eji.202048806] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 10/18/2020] [Accepted: 10/22/2020] [Indexed: 11/10/2022]
Abstract
CD28 expression is generally considered to be T lymphocyte specific. We have previously shown CD28 mRNA expression in M-CSF-dependent anti-inflammatory monocyte-derived macrophages (M-MØ), and now demonstrate that CD28 cell surface expression is higher in M-MØ than in GM-CSF-dependent macrophages, and that macrophage CD28 expression is regulated by MAFB and activin A. In vivo, CD28 was found in tumor-associated macrophages and, to a lower extent, in pro-inflammatory synovial fluid macrophages from rheumatoid arthritis patients. Analysis of mouse macrophages confirmed Cd28 expression in bone-marrow derived M-MØ. Indeed, anti-CD28 antibodies triggered ERK1/2 phosphorylation in mouse M-MØ. At the functional level, Cd28KO M-MØ exhibited a significantly higher capacity to activate the OVA-specific proliferation of OT-II CD4+ T cells than WT M-MØ, as well as enhanced LPS-induced IL-6 production. Besides, the Cd28KO M-MØ transcriptome was significantly different from WT M-MØ regarding the expression IFN response, inflammatory response, and TGF-β signaling related gene sets. Therefore, defective CD28 expression in mouse macrophages associates to changes in gene expression profile, what might contribute to the altered functionality displayed by Cd28KO M-MØ. Thus, CD28 expression appears as a hallmark of anti-inflammatory macrophages and might be a target for immunotherapy.
Collapse
Affiliation(s)
- Lizbeth Estrada-Capetillo
- Unidad de InmunoMetabolismo e Inflamación, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Laura Aragoneses-Fenoll
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Sara Fuentelsaz-Romero
- Unidad de InmunoMetabolismo e Inflamación, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Concha Nieto
- Centro de Investigaciones Biológicas Margarita Salas, Madrid, Spain
| | | | - Bárbara Alonso
- Centro de Investigaciones Biológicas Margarita Salas, Madrid, Spain
| | - Pilar Portolés
- Unidad de Inmunología Celular, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - Angel L Corbí
- Centro de Investigaciones Biológicas Margarita Salas, Madrid, Spain
| | - Jose M Rojo
- Centro de Investigaciones Biológicas Margarita Salas, Madrid, Spain
| | - Amaya Puig-Kröger
- Unidad de InmunoMetabolismo e Inflamación, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| |
Collapse
|
54
|
Freuchet A, Salama A, Remy S, Guillonneau C, Anegon I. IL-34 and CSF-1, deciphering similarities and differences at steady state and in diseases. J Leukoc Biol 2021; 110:771-796. [PMID: 33600012 DOI: 10.1002/jlb.3ru1120-773r] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/04/2021] [Accepted: 01/04/2021] [Indexed: 12/11/2022] Open
Abstract
Although IL-34 and CSF-1 share actions as key mediators of monocytes/macrophages survival and differentiation, they also display differences that should be identified to better define their respective roles in health and diseases. IL-34 displays low sequence homology with CSF-1 but has a similar general structure and they both bind to a common receptor CSF-1R, although binding and subsequent intracellular signaling shows differences. CSF-1R expression has been until now mainly described at a steady state in monocytes/macrophages and myeloid dendritic cells, as well as in some cancers. IL-34 has also 2 other receptors, protein-tyrosine phosphatase zeta (PTPζ) and CD138 (Syndecan-1), expressed in some epithelium, cells of the central nervous system (CNS), as well as in numerous cancers. While most, if not all, of CSF-1 actions are mediated through monocyte/macrophages, IL-34 has also other potential actions through PTPζ and CD138. Additionally, IL-34 and CSF-1 are produced by different cells in different tissues. This review describes and discusses similarities and differences between IL-34 and CSF-1 at steady state and in pathological situations and identifies possible ways to target IL-34, CSF-1, and its receptors.
Collapse
Affiliation(s)
- Antoine Freuchet
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Apolline Salama
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Séverine Remy
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Carole Guillonneau
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Ignacio Anegon
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France.,LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| |
Collapse
|
55
|
Delaney C, Farrell M, Doherty CP, Brennan K, O’Keeffe E, Greene C, Byrne K, Kelly E, Birmingham N, Hickey P, Cronin S, Savvides SN, Doyle SL, Campbell M. Attenuated CSF-1R signalling drives cerebrovascular pathology. EMBO Mol Med 2021; 13:e12889. [PMID: 33350588 PMCID: PMC7863388 DOI: 10.15252/emmm.202012889] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 11/19/2020] [Accepted: 11/23/2020] [Indexed: 12/21/2022] Open
Abstract
Cerebrovascular pathologies occur in up to 80% of cases of Alzheimer's disease; however, the underlying mechanisms that lead to perivascular pathology and accompanying blood-brain barrier (BBB) disruption are still not fully understood. We have identified previously unreported mutations in colony stimulating factor-1 receptor (CSF-1R) in an ultra-rare autosomal dominant condition termed adult-onset leucoencephalopathy with axonal spheroids and pigmented glia (ALSP). Cerebrovascular pathologies such as cerebral amyloid angiopathy (CAA) and perivascular p-Tau were some of the primary neuropathological features of this condition. We have identified two families with different dominant acting alleles with variants located in the kinase region of the CSF-1R gene, which confer a lack of kinase activity and signalling. The protein product of this gene acts as the receptor for 2 cognate ligands, namely colony stimulating factor-1 (CSF-1) and interleukin-34 (IL-34). Here, we show that depletion in CSF-1R signalling induces BBB disruption and decreases the phagocytic capacity of peripheral macrophages but not microglia. CSF-1R signalling appears to be critical for macrophage and microglial activation, and macrophage localisation to amyloid appears reduced following the induction of Csf-1r heterozygosity in macrophages. Finally, we show that endothelial/microglial crosstalk and concomitant attenuation of CSF-1R signalling causes re-modelling of BBB-associated tight junctions and suggest that regulating BBB integrity and systemic macrophage recruitment to the brain may be therapeutically relevant in ALSP and other Alzheimer's-like dementias.
Collapse
Affiliation(s)
- Conor Delaney
- Smurfit Institute of GeneticsTrinity College DublinDublin 2Ireland
| | - Michael Farrell
- Department of NeuropathologyBeaumont HospitalDublin 9Ireland
| | - Colin P Doherty
- Department of NeurologyHealth Care CentreSt James's HospitalDublin 8Ireland
- Academic Unit of NeurologyBiomedical Sciences InstituteTrinity College DublinDublin 2Ireland
- FutureNeuro SFI Research CentreRoyal College of Surgeons in IrelandDublinIreland
| | - Kiva Brennan
- Trinity College Institute of NeuroscienceTrinity College Dublin 2Dublin 2Ireland
| | - Eoin O’Keeffe
- Smurfit Institute of GeneticsTrinity College DublinDublin 2Ireland
| | - Chris Greene
- Smurfit Institute of GeneticsTrinity College DublinDublin 2Ireland
| | - Kieva Byrne
- Smurfit Institute of GeneticsTrinity College DublinDublin 2Ireland
| | - Eoin Kelly
- Department of NeurologyHealth Care CentreSt James's HospitalDublin 8Ireland
| | | | | | - Simon Cronin
- Department of MedicineUniversity College CorkCorkIreland
| | - Savvas N Savvides
- Unit for Structural BiologyDepartment of Biochemistry and MicrobiologyGhent UniversityGhentBelgium
- VIB‐UGent Center for Inflammation ResearchGhentBelgium
| | - Sarah L Doyle
- Trinity College Institute of NeuroscienceTrinity College Dublin 2Dublin 2Ireland
| | - Matthew Campbell
- Smurfit Institute of GeneticsTrinity College DublinDublin 2Ireland
- FutureNeuro SFI Research CentreRoyal College of Surgeons in IrelandDublinIreland
| |
Collapse
|
56
|
Ferrero G, Miserocchi M, Di Ruggiero E, Wittamer V. A c sf1rb mutation uncouples two waves of microglia development in zebrafish. Development 2021; 148:dev.194241. [PMID: 33298459 DOI: 10.1242/dev.194241] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 12/02/2020] [Indexed: 12/15/2022]
Abstract
In vertebrates, the ontogeny of microglia, the resident macrophages of the central nervous system, initiates early during development from primitive macrophages. Although murine embryonic microglia then persist through life, in zebrafish these cells are transient, as they are fully replaced by an adult population originating from larval hematopoietic stem cell (HSC)-derived progenitors. Colony-stimulating factor 1 receptor (Csf1r) is a fundamental regulator of microglia ontogeny in vertebrates, including zebrafish, which possess two paralogous genes: csf1ra and csf1rb Although previous work has shown that mutation in both genes completely abrogates microglia development, the specific contribution of each paralog remains largely unknown. Here, using a fate-mapping strategy to discriminate between the two microglial waves, we uncover non-overlapping roles for csf1ra and csf1rb in hematopoiesis, and identified csf1rb as an essential regulator of adult microglia development. Notably, we demonstrate that csf1rb positively regulates HSC-derived myelopoiesis, resulting in macrophage deficiency, including microglia, in adult mutant animals. Overall, this study contributes to new insights into evolutionary aspects of Csf1r signaling and provides an unprecedented framework for the functional dissection of embryonic versus adult microglia in vivo.
Collapse
Affiliation(s)
- Giuliano Ferrero
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), Brussels 1070, Belgium.,ULB Institute of Neuroscience (UNI), Université Libre de Bruxelles (ULB), Brussels 1070, Belgium
| | - Magali Miserocchi
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), Brussels 1070, Belgium.,ULB Institute of Neuroscience (UNI), Université Libre de Bruxelles (ULB), Brussels 1070, Belgium
| | - Elodie Di Ruggiero
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), Brussels 1070, Belgium
| | - Valérie Wittamer
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB), Brussels 1070, Belgium .,ULB Institute of Neuroscience (UNI), Université Libre de Bruxelles (ULB), Brussels 1070, Belgium.,WELBIO, Université Libre de Bruxelles (ULB), Brussels 1070, Belgium
| |
Collapse
|
57
|
Muñoz-Garcia J, Cochonneau D, Télétchéa S, Moranton E, Lanoe D, Brion R, Lézot F, Heymann MF, Heymann D. The twin cytokines interleukin-34 and CSF-1: masterful conductors of macrophage homeostasis. Theranostics 2021; 11:1568-1593. [PMID: 33408768 PMCID: PMC7778581 DOI: 10.7150/thno.50683] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/03/2020] [Indexed: 12/19/2022] Open
Abstract
Macrophages are specialized cells that control tissue homeostasis. They include non-resident and tissue-resident macrophage populations which are characterized by the expression of particular cell surface markers and the secretion of molecules with a wide range of biological functions. The differentiation and polarization of macrophages relies on specific growth factors and their receptors. Macrophage-colony stimulating factor (CSF-1) and interleukine-34 (IL-34), also known as "twin" cytokines, are part of this regluatory landscape. CSF-1 and IL-34 share a common receptor, the macrophage-colony stimulating factor receptor (CSF-1R), which is activated in a similar way by both factors and turns on identical signaling pathways. However, there is some discrete differential activation leading to specific activities. In this review, we disscuss recent progress in understanding of the role of the twin cytokines in macrophage differentiation, from their interaction with CSF-1R and the activation of signaling pathways, to their implication in macrophage polarization of non-resident and tissue-resident macrophages. A special focus on IL-34, its involvement in pathophsyiological contexts, and its potential as a theranostic target for macrophage therapy will be proposed.
Collapse
Affiliation(s)
- Javier Muñoz-Garcia
- Université de Nantes, Institut de Cancérologie de l'Ouest, Saint-Herblain, F-44805, France
- SATT Ouest Valorisation, Nantes, France
| | - Denis Cochonneau
- Université de Nantes, Institut de Cancérologie de l'Ouest, Saint-Herblain, F-44805, France
| | | | - Emilie Moranton
- Université de Nantes, Institut de Cancérologie de l'Ouest, Saint-Herblain, F-44805, France
| | - Didier Lanoe
- Université de Nantes, Institut de Cancérologie de l'Ouest, Saint-Herblain, F-44805, France
| | - Régis Brion
- Université de Nantes, INSERM, U1238, Nantes, France
| | | | | | - Dominique Heymann
- Université de Nantes, Institut de Cancérologie de l'Ouest, Saint-Herblain, F-44805, France
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, UK
| |
Collapse
|
58
|
Abstract
Tissue or organ regeneration is a complex process with successful outcomes depending on the type of tissue and organism. Upon damage, mammals can only efficiently restore a few tissues including the liver, skin, epithelia of the lung, kidney, and gut. In contrast, lower vertebrates such as zebrafish possess an extraordinary regeneration ability, which restores the normal function of a broad spectrum of tissues including heart, fin, brain, spinal cord, and retina. This regeneration process is either mediated by the proliferation of resident stem cells, or cells that dedifferentiate into a stem cell-like. In recent years, evidence has suggested that the innate immune system can modulate stem cell activity to initiate the regenerative response to damage. This review will explore some of the newer concepts of inflammation in zebrafish regeneration in different tissues. Understanding how inflammation regulates regeneration in zebrafish would provide important clues to improve the therapeutic strategies for repairing injured mammalian tissues that do not have an inherent regenerative capacity.
Collapse
Affiliation(s)
- Maria Iribarne
- Center for Zebrafish Research, Department of Biological Sciences; Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, USA
| |
Collapse
|
59
|
Wu S, Nguyen LTM, Pan H, Hassan S, Dai Y, Xu J, Wen Z. Two phenotypically and functionally distinct microglial populations in adult zebrafish. SCIENCE ADVANCES 2020; 6:6/47/eabd1160. [PMID: 33208372 PMCID: PMC7673811 DOI: 10.1126/sciadv.abd1160] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 10/02/2020] [Indexed: 05/23/2023]
Abstract
Microglia are the tissue-resident macrophages in the central nervous system and are critically involved in immune defense, neural development and function, and neuroinflammation. The versatility of microglia has long been attributed to heterogeneity. Recent studies have revealed possible heterogeneity in human but not in murine microglia, yet a firm demonstration linking microglial heterogeneity to functional phenotypes remains scarce. Here, we identified two distinct microglial populations in adult zebrafish that differ in morphology, distribution, development, and function. The predominant population, phagocytotic microglia, which expresses ccl34b.1, is broadly distributed, amoeboid in shape, highly mobile, and phagocytotic. The other white matter-enriched ccl34b.1- population, regulatory microglia, has ramified protrusions but has limited mobility and phagocytosis capability. These functional differences are further supported by distinct transcriptomes and responses to bacterial infection, where ccl34b.1+ microglia function in tissue clearance and ccl34b.1- microglia release immune regulators. Our study sheds light on the heterogeneity and functional diversification of microglia.
Collapse
Affiliation(s)
- Shuting Wu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Linh T M Nguyen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Hongru Pan
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Shaoli Hassan
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Yimei Dai
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Jin Xu
- Division of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangdong, Guangzhou 510630, China
| | - Zilong Wen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.
- Center of Systems Biology and Human Health, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen 518055, China
| |
Collapse
|
60
|
Lelios I, Cansever D, Utz SG, Mildenberger W, Stifter SA, Greter M. Emerging roles of IL-34 in health and disease. J Exp Med 2020; 217:133604. [PMID: 31940023 PMCID: PMC7062519 DOI: 10.1084/jem.20190290] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 09/11/2019] [Accepted: 11/26/2019] [Indexed: 12/14/2022] Open
Abstract
Macrophages are part of the innate immune system and are present in every organ of the body. They fulfill critical roles in tissue homeostasis and development and are involved in various pathologies. An essential factor for the development, homeostasis, and function of mononuclear phagocytes is the colony stimulating factor-1 receptor (CSF-1R), which has two known ligands: CSF-1 and interleukin-34 (IL-34). While CSF-1 has been extensively studied, the biology and functions of IL-34 are only now beginning to be uncovered. In this review, we discuss recent advances of IL-34 biology in health and disease with a specific focus on mononuclear phagocytes.
Collapse
Affiliation(s)
- Iva Lelios
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Dilay Cansever
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Sebastian G Utz
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Wiebke Mildenberger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Sebastian A Stifter
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
61
|
Demy DL, Carrère M, Noche R, Tauzin M, Le Bris M, Baek C, Leshchiner I, Goessling W, Herbomel P. The cationic amino acid exporter Slc7a7 is induced and vital in zebrafish tissue macrophages with sustained efferocytic activity. J Cell Sci 2020; 133:jcs249037. [PMID: 32973110 DOI: 10.1242/jcs.249037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/09/2020] [Indexed: 01/02/2023] Open
Abstract
Most tissues harbor a substantial population of resident macrophages. Here, we elucidate a functional link between the Slc7a7 cationic amino acid transporter and tissue macrophages. We identified a mutant zebrafish devoid of microglia due to a mutation in the slc7a7 gene. We found that in Slc7a7-deficient larvae, macrophages do enter the retina and brain to become microglia, but then die during the developmental wave of neuronal apoptosis, which triggers intense efferocytic work from them. A similar macrophage demise occurs in other tissues, at stages where macrophages have to engulf many cell corpses, whether due to developmental or experimentally triggered cell death. We found that Slc7a7 is the main cationic amino acid transporter expressed in macrophages of zebrafish larvae, and that its expression is induced in tissue macrophages within 1-2 h upon efferocytosis. Our data indicate that Slc7a7 is vital not only for microglia but also for any steadily efferocytic tissue macrophages, and that slc7a7 gene induction is one of the adaptive responses that allow them to cope with the catabolism of numerous dead cells without compromising their own viability.
Collapse
Affiliation(s)
- Doris Lou Demy
- Institut Pasteur, Department of Developmental & Stem Cell Biology, 75015 Paris, France
- CNRS, UMR3738, 75015 Paris, France
| | - Mireille Carrère
- Institut Pasteur, Department of Developmental & Stem Cell Biology, 75015 Paris, France
- CNRS, UMR3738, 75015 Paris, France
| | - Ramil Noche
- Institut Pasteur, Department of Developmental & Stem Cell Biology, 75015 Paris, France
- CNRS, UMR3738, 75015 Paris, France
| | - Muriel Tauzin
- Institut Pasteur, Department of Developmental & Stem Cell Biology, 75015 Paris, France
- CNRS, UMR3738, 75015 Paris, France
| | - Marion Le Bris
- Institut Pasteur, Department of Developmental & Stem Cell Biology, 75015 Paris, France
- CNRS, UMR3738, 75015 Paris, France
| | - Chooyoung Baek
- Institut Pasteur, Department of Developmental & Stem Cell Biology, 75015 Paris, France
- CNRS, UMR3738, 75015 Paris, France
| | | | - Wolfram Goessling
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Philippe Herbomel
- Institut Pasteur, Department of Developmental & Stem Cell Biology, 75015 Paris, France
- CNRS, UMR3738, 75015 Paris, France
| |
Collapse
|
62
|
The M-CSF receptor in osteoclasts and beyond. Exp Mol Med 2020; 52:1239-1254. [PMID: 32801364 PMCID: PMC8080670 DOI: 10.1038/s12276-020-0484-z] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 12/18/2022] Open
Abstract
Colony-stimulating factor 1 receptor (CSF1R, also known as c-FMS) is a receptor tyrosine kinase. Macrophage colony-stimulating factor (M-CSF) and IL-34 are ligands of CSF1R. CSF1R-mediated signaling is crucial for the survival, function, proliferation, and differentiation of myeloid lineage cells, including osteoclasts, monocytes/macrophages, microglia, Langerhans cells in the skin, and Paneth cells in the intestine. CSF1R also plays an important role in oocytes and trophoblastic cells in the female reproductive tract and in the maintenance and maturation of neural progenitor cells. Given that CSF1R is expressed in a wide range of myeloid cells, altered CSF1R signaling is implicated in inflammatory, neoplastic, and neurodegenerative diseases. Inhibiting CSF1R signaling through an inhibitory anti-CSF1R antibody or small molecule inhibitors that target the kinase activity of CSF1R has thus been a promising therapeutic strategy for those diseases. In this review, we cover the recent progress in our understanding of the various roles of CSF1R in osteoclasts and other myeloid cells, highlighting the therapeutic applications of CSF1R inhibitors in disease conditions. Drugs directed at a key signaling receptor involved in breaking down bone tissue could help treat diseases marked by pathological bone loss and destruction. In a review article, Kyung-Hyun Park-Min and colleagues from the Hospital for Special Surgery in New York, USA, discuss the essential roles played by the colony-stimulating factor 1 receptor (CSF1R) protein in the survival, function, proliferation and differentiation of myeloid lineage stem cells in the bone marrow, including bone-resorbing osteoclasts. They explore the links between the CSF1R-mediated signaling pathway and diseases such as cancer and neurodegeneration. The authors largely focus on bone conditions, highlighting mouse studies in which CSF1R-blocking drugs were shown to ameliorate bone loss and inflammatory symptoms in models of arthritis, osteoporosis and metastatic cancer. Clinical trials are ongoing to test therapeutic applications.
Collapse
|
63
|
Yang L, Jiménez JA, Earley AM, Hamlin V, Kwon V, Dixon CT, Shiau CE. Drainage of inflammatory macromolecules from the brain to periphery targets the liver for macrophage infiltration. eLife 2020; 9:58191. [PMID: 32735214 PMCID: PMC7434444 DOI: 10.7554/elife.58191] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/27/2020] [Indexed: 12/16/2022] Open
Abstract
Many brain pathologies are associated with liver damage, but a direct link has long remained elusive. Here, we establish a new paradigm for interrogating brain-periphery interactions by leveraging zebrafish for its unparalleled access to the intact whole animal for in vivo analysis in real time after triggering focal brain inflammation. Using traceable lipopolysaccharides (LPS), we reveal that drainage of these inflammatory macromolecules from the brain led to a strikingly robust peripheral infiltration of macrophages into the liver independent of Kupffer cells. We further demonstrate that this macrophage recruitment requires signaling from the cytokine IL-34 and Toll-like receptor adaptor MyD88, and occurs in coordination with neutrophils. These results highlight the possibility for circulation of brain-derived substances to serve as a rapid mode of communication from brain to the liver. Understanding how the brain engages the periphery at times of danger may offer new perspectives for detecting and treating brain pathologies.
Collapse
Affiliation(s)
- Linlin Yang
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Jessica A Jiménez
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Alison M Earley
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Victoria Hamlin
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Victoria Kwon
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Cameron T Dixon
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Celia E Shiau
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, United States.,Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, United States
| |
Collapse
|
64
|
Yolk sac-derived Pdcd11-positive cells modulate zebrafish microglia differentiation through the NF-κB-Tgfβ1 pathway. Cell Death Differ 2020; 28:170-183. [PMID: 32709934 PMCID: PMC7853042 DOI: 10.1038/s41418-020-0591-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/07/2020] [Accepted: 07/09/2020] [Indexed: 12/22/2022] Open
Abstract
Microglia are the primary immune cells in the central nervous system, which plays a vital role in neuron development and neurodegenerative diseases. Microglial precursors in peripheral hematopoietic tissues colonize the central nervous system during early embryogenesis. However, how intrinsic and extrinsic signals integrate to regulate microglia’s differentiation remains undefined. In this study, we identified the cerebral white matter hyperintensities susceptibility gene, programmed cell death protein 11 (PDCD11), as an essential factor regulating microglia differentiation. In zebrafish, pdcd11 deficiency prevents the differentiation of the precursors to mature brain microglia. Although, the inflammatory featured macrophage brain colonization is augmented. At 22 h post fertilization, the Pdcd11-positive cells on the yolk sac are distinct from macrophages and neutrophils. Mechanistically, PDCD11 exerts its physiological role by differentially regulating the functions of nuclear factor-kappa B family members, P65 and c-Rel, suppressing P65-mediated expression of inflammatory cytokines, such as tnfα, and enhancing the c-Rel-dependent appearance of tgfβ1. The present study provides novel insights in understanding microglia differentiation during zebrafish development.
Collapse
|
65
|
Yu T, Kuang H, Chen J, Lin X, Wu Y, Chen K, Zhang M, Zhang W, Wen Z. Tripartite-motif family protein 35-28 regulates microglia development by preventing necrotic death of microglial precursors in zebrafish. J Biol Chem 2020; 295:8846-8856. [PMID: 32398256 DOI: 10.1074/jbc.ra119.012043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 05/04/2020] [Indexed: 02/02/2023] Open
Abstract
Microglia are tissue-resident macrophages in the central nervous system (CNS) that play essential roles in the regulation of CNS development and homeostasis. Yet, the genetic networks governing microglia development remain incompletely defined. Here, we report the identification and characterization of a microglia-defective zebrafish mutant wulonghkz12 (wulhkz12 ) isolated from an ethylnitrosourea (ENU)-based genetic screen. We show that wulhkz12 mutants harbors a missense point mutation in the gene region encoding the PRY/SPRY domain of the tripartite-motif family protein 35-28 (trim35-28) gene. Time-lapse imaging revealed that the loss of Trim35-28 function causes lytic necrosis of microglial precursors/peripheral macrophages, as indicated by cytoplasmic swelling and membrane rupture of these precursors and accompanied by neutrophil infiltration and systemic inflammation. Intriguingly, the lytic necrosis of microglial precursors in trim35-28-deficient mutants appeared to depend neither on the canonical pyroptotic nor necroptotic pathways, as inhibition of the key component in each pathway could not rescue the microglia phenotype in trim35-28-deficient mutants. Finally, results from tissue-specific rescue experiments suggested that Trim35-28 acts cell-autonomously in the survival of microglial precursors. Taken together, the findings of our study reveal Trim35-28 as a regulatory protein essential for microglia development.
Collapse
Affiliation(s)
- Tao Yu
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University- The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Haoyue Kuang
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University- The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Jiahao Chen
- Department of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangdong, Guangzhou, China
| | - Xi Lin
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Center of Systems Biology and Human Health, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Yi Wu
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University- The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Keyu Chen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Center of Systems Biology and Human Health, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, China
| | - Mingjie Zhang
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University- The Hong Kong University of Science and Technology Medical Center, Shenzhen, China; Division of Life Science, State Key Laboratory of Molecular Neuroscience, Center of Systems Biology and Human Health, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, China
| | - Wenqing Zhang
- Department of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangdong, Guangzhou, China.
| | - Zilong Wen
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University- The Hong Kong University of Science and Technology Medical Center, Shenzhen, China; Division of Life Science, State Key Laboratory of Molecular Neuroscience, Center of Systems Biology and Human Health, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Greater Bay Biomedical Innocenter, Shenzhen Bay Laboratory, Shenzhen, China.
| |
Collapse
|
66
|
Kuil LE, Oosterhof N, Ferrero G, Mikulášová T, Hason M, Dekker J, Rovira M, van der Linde HC, van Strien PMH, de Pater E, Schaaf G, Bindels EMJ, Wittamer V, van Ham TJ. Zebrafish macrophage developmental arrest underlies depletion of microglia and reveals Csf1r-independent metaphocytes. eLife 2020; 9:e53403. [PMID: 32367800 PMCID: PMC7237208 DOI: 10.7554/elife.53403] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 04/24/2020] [Indexed: 12/18/2022] Open
Abstract
Macrophages derive from multiple sources of hematopoietic progenitors. Most macrophages require colony-stimulating factor 1 receptor (CSF1R), but some macrophages persist in the absence of CSF1R. Here, we analyzed mpeg1:GFP-expressing macrophages in csf1r-deficient zebrafish and report that embryonic macrophages emerge followed by their developmental arrest. In larvae, mpeg1+ cell numbers then increased showing two distinct types in the skin: branched, putative Langerhans cells, and amoeboid cells. In contrast, although numbers also increased in csf1r-mutants, exclusively amoeboid mpeg1+ cells were present, which we showed by genetic lineage tracing to have a non-hematopoietic origin. They expressed macrophage-associated genes, but also showed decreased phagocytic gene expression and increased epithelial-associated gene expression, characteristic of metaphocytes, recently discovered ectoderm-derived cells. We further demonstrated that juvenile csf1r-deficient zebrafish exhibit systemic macrophage depletion. Thus, csf1r deficiency disrupts embryonic to adult macrophage development. Zebrafish deficient for csf1r are viable and permit analyzing the consequences of macrophage loss throughout life.
Collapse
Affiliation(s)
- Laura E Kuil
- Department of Clinical Genetics, Erasmus University Medical Center RotterdamRotterdamNetherlands
| | - Nynke Oosterhof
- Department of Clinical Genetics, Erasmus University Medical Center RotterdamRotterdamNetherlands
| | - Giuliano Ferrero
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB)BrusselsBelgium
| | - Tereza Mikulášová
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Martina Hason
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Jordy Dekker
- Department of Clinical Genetics, Erasmus University Medical Center RotterdamRotterdamNetherlands
| | - Mireia Rovira
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB)BrusselsBelgium
| | - Herma C van der Linde
- Department of Clinical Genetics, Erasmus University Medical Center RotterdamRotterdamNetherlands
| | | | - Emma de Pater
- Department of Hematology, Erasmus University Medical CenterRotterdamNetherlands
| | - Gerben Schaaf
- Department of Clinical Genetics, Erasmus University Medical Center RotterdamRotterdamNetherlands
| | - Erik MJ Bindels
- Department of Hematology, Erasmus University Medical CenterRotterdamNetherlands
| | - Valerie Wittamer
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles (ULB)BrusselsBelgium
- WELBIO, ULBBrusselsBelgium
| | - Tjakko J van Ham
- Department of Clinical Genetics, Erasmus University Medical Center RotterdamRotterdamNetherlands
| |
Collapse
|
67
|
Rosowski EE. Determining macrophage versus neutrophil contributions to innate immunity using larval zebrafish. Dis Model Mech 2020; 13:13/1/dmm041889. [PMID: 31932292 PMCID: PMC6994940 DOI: 10.1242/dmm.041889] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The specific roles of the two major innate immune cell types – neutrophils and macrophages – in response to infection and sterile inflammation are areas of great interest. The larval zebrafish model of innate immunity, and the imaging capabilities it provides, is a source of new research and discoveries in this field. Multiple methods have been developed in larval zebrafish to specifically deplete functional macrophages or neutrophils. Each of these has pros and cons, as well as caveats, that often make it difficult to directly compare results from different studies. The purpose of this Review is to (1) explore the pros, cons and caveats of each of these immune cell-depleted models; (2) highlight and place into a broader context recent key findings on the specific functions of innate immune cells using these models; and (3) explore future directions in which immune cell depletion methods are being expanded. Summary: Macrophages and neutrophils are distinct innate immune cells with diverse roles in diverse inflammatory contexts. Recent research in larval zebrafish using cell-specific depletion methods has revealed new insights into these cells' functions.
Collapse
Affiliation(s)
- Emily E Rosowski
- Department of Biological Sciences, Clemson University, Clemson, SC 29634, USA
| |
Collapse
|
68
|
Shwartz A, Goessling W, Yin C. Macrophages in Zebrafish Models of Liver Diseases. Front Immunol 2019; 10:2840. [PMID: 31867007 PMCID: PMC6904306 DOI: 10.3389/fimmu.2019.02840] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 11/19/2019] [Indexed: 12/16/2022] Open
Abstract
Hepatic macrophages are key components of the liver immunity and consist of two main populations. Liver resident macrophages, known as Kupffer cells in mammals, are crucial for maintaining normal liver homeostasis. Upon injury, they become activated to release proinflammatory cytokines and chemokines and recruit a large population of inflammatory monocyte-derived macrophages to the liver. During the progression of liver diseases, macrophages are highly plastic and have opposing functions depending on the signaling cues that they receive from the microenvironment. A comprehensive understanding of liver macrophages is essential for developing therapeutic interventions that target these cells in acute and chronic liver diseases. Mouse studies have provided the bulk of our current knowledge of liver macrophages. The emergence of various liver disease models and availability of transgenic tools to visualize and manipulate macrophages have made the teleost zebrafish (Danio rerio) an attractive new vertebrate model to study liver macrophages. In this review, we summarize the origin and behaviors of macrophages in healthy and injured livers in zebrafish. We highlight the roles of macrophages in zebrafish models of alcoholic and non-alcoholic liver diseases, hepatocellular carcinoma, and liver regeneration, and how they compare with the roles that have been described in mammals. We also discuss the advantages and challenges of using zebrafish to study liver macrophages.
Collapse
Affiliation(s)
- Arkadi Shwartz
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Wolfram Goessling
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
- Harvard Stem Cell Institute, Cambridge, MA, United States
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
- Broad Institute, Massachusetts Institute of Technology and Harvard, Cambridge, MA, United States
- Division of Health Sciences and Technology, Harvard and Massachusetts Institute of Technology, Boston, MA, United States
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Chunyue Yin
- Division of Gastroenterology, Hepatology and Nutrition and Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| |
Collapse
|
69
|
Abstract
Tissue-resident macrophages (TRMs), generally found in tissues under normal physiological conditions, play crucial roles not only in immunity but also in tissue development and homeostasis. Because of their diverse functions, dysregulation of their development and function has been implicated in many human disorders. In the past decade, a great deal of extensive studies have been conducted in various model organisms with cutting-edge technologies to explore the origin and function of TRMs. In this review, we summarize the recent findings on TRMs in mouse and zebrafish and compare the similarity/differences between these two species.
Collapse
|
70
|
Anderson SR, Roberts JM, Zhang J, Steele MR, Romero CO, Bosco A, Vetter ML. Developmental Apoptosis Promotes a Disease-Related Gene Signature and Independence from CSF1R Signaling in Retinal Microglia. Cell Rep 2019; 27:2002-2013.e5. [PMID: 31091440 PMCID: PMC6544177 DOI: 10.1016/j.celrep.2019.04.062] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/21/2019] [Accepted: 04/11/2019] [Indexed: 12/19/2022] Open
Abstract
Microglia have important remodeling functions in neurodevelopment, aging, and disease, with evidence for molecular diversity. However, the signaling pathways and environmental cues that drive diverse states of microglia are incompletely understood. We profiled microglia of a discrete developing CNS region, the murine retina. We found distinct transcriptional signatures for retinal microglia across development and peak postnatal density of a population that resembles aging and disease-associated microglia (DAM) and CD11c+ microglia of developing white matter. While TREM2 signaling modulates the expression of select genes, the DAM-related signature is significantly reduced in retinas lacking Bax, a proapoptotic factor required for neuronal death. Furthermore, we found postnatal retinal microglia highly expressing CD11c are resistant to loss or inhibition of colony stimulating factor 1 receptor (CSF1R), while most microglia can be eliminated in Bax knockout retina. Thus, developmental apoptosis promotes a microglia gene signature linked to CSF1R independence that shares features with microglia in developing white matter and in disease.
Collapse
Affiliation(s)
- Sarah R Anderson
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Jacqueline M Roberts
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Jianmin Zhang
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Michael R Steele
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Cesar O Romero
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Alejandra Bosco
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Monica L Vetter
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
71
|
Smolders SMT, Kessels S, Vangansewinkel T, Rigo JM, Legendre P, Brône B. Microglia: Brain cells on the move. Prog Neurobiol 2019; 178:101612. [PMID: 30954517 DOI: 10.1016/j.pneurobio.2019.04.001] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 02/13/2019] [Accepted: 04/01/2019] [Indexed: 02/08/2023]
Abstract
In the last decade, tremendous progress has been made in understanding the biology of microglia - i.e. the fascinating immigrated resident immune cell population of the central nervous system (CNS). Recent literature reviews have largely dealt with the plentiful functions of microglia in CNS homeostasis, development and pathology, and the influences of sex and the microbiome. In this review, the intriguing aspect of their physical plasticity during CNS development will get specific attention. Microglia move around (mobility) and reshape their processes (motility). Microglial migration into and inside the CNS is most prominent throughout development and consequently most of the data described in this review concern mobility and motility in the changing environment of the developing brain. Here, we first define microglia based on their highly specialized age- and region-dependent gene expression signature and associated functional heterogeneity. Next, we describe their origin, the migration route of immature microglial cells towards the CNS, the mechanisms underlying their invasion of the CNS, and their spatiotemporal localization and surveying behaviour inside the developing CNS. These processes are dependent on microglial mobility and motility which are determined by the microenvironment of the CNS. Therefore, we further zoom in on the changing environment during CNS development. We elaborate on the extracellular matrix and the respective integrin receptors on microglia and we discuss the purinergic and molecular signalling in microglial mobility. In the last section, we discuss the physiological and pathological functions of microglia in which mobility and motility are involved to stress the importance of microglial 'movement'.
Collapse
Affiliation(s)
- Sophie Marie-Thérèse Smolders
- UHasselt, BIOMED, Diepenbeek, Belgium; INSERM, UMR-S 1130, CNRS, UMR 8246, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France; Sorbonne Universités, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | | | | | | | - Pascal Legendre
- INSERM, UMR-S 1130, CNRS, UMR 8246, Neuroscience Paris Seine, Institute of Biology Paris Seine, Paris, France; Sorbonne Universités, UPMC Université Paris 06, UM CR18, Neuroscience Paris Seine, Paris, France
| | | |
Collapse
|
72
|
Kuil LE, Oosterhof N, Geurts SN, van der Linde HC, Meijering E, van Ham TJ. Reverse genetic screen reveals that Il34 facilitates yolk sac macrophage distribution and seeding of the brain. Dis Model Mech 2019; 12:dmm037762. [PMID: 30765415 PMCID: PMC6451432 DOI: 10.1242/dmm.037762] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 02/06/2019] [Indexed: 12/30/2022] Open
Abstract
Microglia are brain-resident macrophages, which have specialized functions important in brain development and in disease. They colonize the brain in early embryonic stages, but few factors that drive the migration of yolk sac macrophages (YSMs) into the embryonic brain, or regulate their acquisition of specialized properties, are currently known. Here, we present a CRISPR/Cas9-based in vivo reverse genetic screening pipeline to identify new microglia regulators using zebrafish. Zebrafish larvae are particularly suitable due to their external development, transparency and conserved microglia features. We targeted putative microglia regulators, by Cas9/gRNA complex injections, followed by Neutral-Red-based visualization of microglia. Microglia were quantified automatically in 3-day-old larvae using a software tool we called SpotNGlia. We identified that loss of zebrafish colony-stimulating factor 1 receptor (Csf1r) ligand, Il34, caused reduced microglia numbers. Previous studies on the role of IL34 in microglia development in vivo were ambiguous. Our data, and a concurrent paper, show that, in zebrafish, il34 is required during the earliest seeding of the brain by microglia. Our data also indicate that Il34 is required for YSM distribution to other organs. Disruption of the other Csf1r ligand, Csf1, did not reduce microglia numbers in mutants, whereas overexpression increased the number of microglia. This shows that Csf1 can influence microglia numbers, but might not be essential for the early seeding of the brain. In all, we identified il34 as a modifier of microglia colonization, by affecting distribution of YSMs to target organs, validating our reverse genetic screening pipeline in zebrafish.This article has an associated First Person interview with the joint first authors of the paper.
Collapse
Affiliation(s)
- Laura E Kuil
- Department of Clinical Genetics, Erasmus University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Nynke Oosterhof
- Department of Clinical Genetics, Erasmus University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
- European Research Institute for the Biology of Ageing, University Medical Center Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Samuël N Geurts
- Biomedical Imaging Group Rotterdam, Departments of Medical Informatics and Radiology, Erasmus University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
- Quantitative Imaging, Faculty of Applied Sciences, Delft University of Technology, Lorentzweg 1, 2628 CJ Delft, The Netherlands
| | - Herma C van der Linde
- Department of Clinical Genetics, Erasmus University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Erik Meijering
- Biomedical Imaging Group Rotterdam, Departments of Medical Informatics and Radiology, Erasmus University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Tjakko J van Ham
- Department of Clinical Genetics, Erasmus University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| |
Collapse
|
73
|
Jin W, Dai Y, Li F, Zhu L, Huang Z, Liu W, Li J, Zhang M, Du J, Zhang W, Wen Z. Dysregulation of Microglial Function Contributes to Neuronal Impairment in Mcoln1a-Deficient Zebrafish. iScience 2019; 13:391-401. [PMID: 30897512 PMCID: PMC6426713 DOI: 10.1016/j.isci.2019.02.031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 10/28/2018] [Accepted: 02/26/2019] [Indexed: 01/21/2023] Open
Abstract
Type IV mucolipidosis (ML-IV) is a neurodegenerative lysosome storage disorder caused by mutations in the MCOLN1 gene. However, the cellular and molecular bases underlying the neuronal phenotypes of ML-IV disease remain elusive. Using a forward genetic screening, we identified a zebrafish mutant, biluo, that harbors a hypomorphic mutation in mcoln1a, one of the two zebrafish homologs of mammalian MCOLN1. The mcoln1a-deficient mutants display phenotypes partially recapitulating the key features of ML-IV disorder, including the accumulation of enlarged late endosomes in microglia and aberrant neuronal activities in both spontaneous and visual-evoking conditions in optic tectal neurons. We further show that the accumulation of enlarged late endosomes in microglia is caused by the impairment of late endosome and lysosome fusion and the aberrant neuronal activities can be partially rescued by the reconstitution of Mcoln1a function in microglia. Our findings suggest that dysregulation of microglial function may contribute to the development and progression of ML-IV disease. mcoln1a-deficient fish display microglia impairment and aberrant neuronal activity The aberrant neuronal activity can be rescued by expressing WT mcoln1a in microglia Impairment of microglia-neuron contact contributes to the aberrant neuronal activity
Collapse
Affiliation(s)
- Wan Jin
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Center of Systems Biology and Human Health, the Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, PR. China
| | - Yimei Dai
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Center of Systems Biology and Human Health, the Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, PR. China
| | - Funing Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, PR. China
| | - Lu Zhu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Center of Systems Biology and Human Health, the Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, PR. China
| | - Zhibin Huang
- Department of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006, PR. China
| | - Wei Liu
- Department of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006, PR. China
| | - Jianchao Li
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Center of Systems Biology and Human Health, the Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, PR. China
| | - Mingjie Zhang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Center of Systems Biology and Human Health, the Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, PR. China
| | - Jiulin Du
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, PR. China
| | - Wenqing Zhang
- Department of Cell, Developmental and Integrative Biology, School of Medicine, South China University of Technology, Guangzhou 510006, PR. China.
| | - Zilong Wen
- Division of Life Science, State Key Laboratory of Molecular Neuroscience and Center of Systems Biology and Human Health, the Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, PR. China; Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, PR. China.
| |
Collapse
|
74
|
Morales RA, Allende ML. Peripheral Macrophages Promote Tissue Regeneration in Zebrafish by Fine-Tuning the Inflammatory Response. Front Immunol 2019; 10:253. [PMID: 30891030 PMCID: PMC6413720 DOI: 10.3389/fimmu.2019.00253] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 01/29/2019] [Indexed: 12/14/2022] Open
Abstract
The role of macrophages during regeneration in zebrafish has been well-documented. Nevertheless, new evidence indicates that zebrafish macrophages are a heterogeneous population of cells, and that they can play different roles during immune responses and in tissue restoration after damage and infection. In this work, we first aimed to classify zebrafish macrophages according to their distribution in the larva during homeostasis and after tissue damage, distinguishing peripheral, and hematopoietic tissue resident macrophages. We discovered differences between the migratory behavior of these two macrophage populations both before and after tissue damage, triggered by the amputation of the tail fin. Further, we found a specific role for peripheral tissue-resident macrophages, and we propose that these cells contribute to tail fin regeneration by down-regulating inflammatory mediators such as interleukin-1b (il1b) and by diminishing reactive oxygen species (ROS) in the damage site. Our work suggests that specific macrophage populations recruited after tissue damage in zebrafish larvae can display different functions during both inflammation and tissue regeneration.
Collapse
Affiliation(s)
- Rodrigo A Morales
- FONDAP Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Miguel L Allende
- FONDAP Center for Genome Regulation, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| |
Collapse
|
75
|
Oltova J, Svoboda O, Bartunek P. Hematopoietic Cytokine Gene Duplication in Zebrafish Erythroid and Myeloid Lineages. Front Cell Dev Biol 2018; 6:174. [PMID: 30619854 PMCID: PMC6306437 DOI: 10.3389/fcell.2018.00174] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 12/06/2018] [Indexed: 12/11/2022] Open
Abstract
Hematopoiesis is a precisely orchestrated process regulated by the activity of hematopoietic cytokines and their respective receptors. Due to an extra round of whole genome duplication during vertebrate evolution in teleost fish, zebrafish have two paralogs of many important genes, including genes involved in hematopoiesis. Importantly, these duplication events brought increased level of complexity in such cases, where both ligands and receptors have been duplicated in parallel. Therefore, precise understanding of binding specificities between duplicated ligand-receptor signalosomes as well as understanding of their differential expression provide an important basis for future studies to better understand the role of duplication of these genes. However, although many recent studies in the field have partly addressed functional redundancy or sub-specialization of some of those duplicated paralogs, this information remains to be scattered over many publications and unpublished data. Therefore, the focus of this review is to provide an overview of recent findings in the zebrafish hematopoietic field regarding activity, role and specificity of some of the hematopoietic cytokines with emphasis on crucial regulators of the erythro-myeloid lineages.
Collapse
Affiliation(s)
- Jana Oltova
- Department of Cell Differentiation, Institute of Molecular Genetics of the ASCR, v.v.i., Prague, Czechia
| | - Ondrej Svoboda
- Department of Cell Differentiation, Institute of Molecular Genetics of the ASCR, v.v.i., Prague, Czechia
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Petr Bartunek
- Department of Cell Differentiation, Institute of Molecular Genetics of the ASCR, v.v.i., Prague, Czechia
| |
Collapse
|
76
|
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
|
77
|
Soyer N, Celik F, Tombuloglu M, Sahin F, Saydam G, Vural F. Role of Allogeneic Stem Cell Transplant in the Treatment of Primary Myelofibrosis. EXP CLIN TRANSPLANT 2016; 17:93-96. [PMID: 27855592 DOI: 10.6002/ect.2016.0007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES The only known curative therapy for primary myelofibrosis is allogeneic hematopoietic stem cell transplant. MATERIALS AND METHODS We retrospectively evaluated 11 transplant procedures involving 10 patients (5 men and 5 women) diagnosed with primary myelofibrosis between 2005 and 2014. RESULTS The median age at the time of transplant was 60.5 years (range, 22-62 years). Stem cell sources were unrelated (n=1) and related (n=11) peripheral blood stem cells. Conditioning regimen was myeloablative for 8 and reduced intensity for 3 transplants. The median number of infused CD34+ cells was 6.8 × 106 cells/kg (range, 3.2-10.4 × 106 cells/kg). Neutrophil and platelet engraftment occurred at median of 22 days (range, 12-31 days) and 19.5 days (range, 13-56 days). Acute and chronic graft-versus-host disease was seen in 4 of 11 allografts. Relapse and nonrelapse mortality rates were 20%. Six patients (60%) were still alive without disease after median follow-up of 68.5 months (range, 17-120 months). Median progression-free survival and overall survival were 61 months (range, 2-120 months) and 65 months (range, 2-120 months). CONCLUSIONS Our results suggest that allogeneic hematopoietic stem cell transplant may provide a curative treatment for primary myelofibrosis patients. A myeloablative regimen seems to be effective and safe, especially for younger primary myelofibrosis patients.
Collapse
Affiliation(s)
- Nur Soyer
- Department of Hematology, Ege University Medical Faculty, Izmir, Turkey
| | - Ferit Celik
- Department of Internal Medicine, Ege University Medical Faculty, Izmir, Turkey
| | - Murat Tombuloglu
- Department of Hematology, Ege University Medical Faculty, Izmir, Turkey
| | - Fahri Sahin
- Department of Hematology, Ege University Medical Faculty, Izmir, Turkey
| | - Guray Saydam
- Department of Hematology, Ege University Medical Faculty, Izmir, Turkey
| | - Filiz Vural
- Department of Hematology, Ege University Medical Faculty, Izmir, Turkey
| |
Collapse
|