1
|
Hidayat AS, Pitt JA, Fredricks H, Hahn ME, Aluru N. Domoic acid induces developmental stage-specific effects on microglia in zebrafish. HARMFUL ALGAE 2025; 146:102862. [PMID: 40409865 PMCID: PMC12102529 DOI: 10.1016/j.hal.2025.102862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 05/25/2025]
Abstract
Harmful algal blooms (HABs), driven by warming oceans and increased eutrophication, are negatively affecting aquatic ecosystems as well as human health. Blooms of diatom Pseudo-nitzschia spp. produce a neurotoxin, domoic acid (DA), which can be concentrated by filter feeders, including shellfish. Consumption of DA-contaminated seafood causes amnesic shellfish poisoning. DA causes well-established effects on neurons, inducing neurotoxicity by binding to ionotropic glutamate receptors. However, its effects on non-neuronal cells including microglia, the resident immune cells in the brain, are less well understood. Microglia play critical roles in brain health, and disruptions in microglial activity during development can have long-term impacts on brain function and disease risk. The objective of this study was to examine the effects of developmental DA exposure on microglia using zebrafish (Danio rerio). We characterized effects of DA exposure on microglial abundance and morphology at two developmental stages (2 and 4 days post-fertilization (dpf)). We additionally assessed impacts on cell death, cytokine expression, and startle response behavior. In older larvae (4 dpf), microglial effects occurred only at the highest dose (0.3 ng DA per larvae) and coincided with severe morphological defects. In contrast, 2 dpf exposure to lower doses (0.1 ng DA per embryo) caused transient tremors without gross abnormalities but exposure to either 0.1 or 0.3 ng DA per larvae increased microglial abundance and altered microglial morphology. In contrast, 4 dpf exposure to 0.3 ng DA per larvae reduced microglial numbers. Increases in mRNA levels of il1b, il4, and tgfb were observed after 4 dpf exposure, but no significant cytokine changes were detected at 2 dpf. Overall, the effects of DA are highly developmental stage-specific, with microglial reactivity occurring at doses that do not cause gross morphological changes. These findings suggest that neuroinflammation may arise at lower DA doses, indicating a potential mechanism for subtle toxic effects and emphasizing the need for further research to better understand the consequences of low-dose DA exposure on microglial function.
Collapse
Affiliation(s)
- Alia S Hidayat
- Biology Department and Woods Hole Center for Oceans and Human Health, Woods Hole Oceanographic Institution, Woods Hole, MA 02543, USA; Massachusetts Institute of Technology (MIT) - Woods Hole Oceanographic Institution (WHOI) Joint Graduate Program in Oceanography and Oceanographic Engineering, USA; Center for American Progress, WA, DC 20002, USA.
| | - Jordan A Pitt
- Biology Department and Woods Hole Center for Oceans and Human Health, Woods Hole Oceanographic Institution, Woods Hole, MA 02543, USA; Massachusetts Institute of Technology (MIT) - Woods Hole Oceanographic Institution (WHOI) Joint Graduate Program in Oceanography and Oceanographic Engineering, USA; Exponent Inc, 1075 Worcester Street, Natick, MA, 01760, USA
| | - Helen Fredricks
- Marine Chemistry and Geochemistry department, Woods Hole Oceanographic Institution, Woods Hole, MA 02543, USA
| | - Mark E Hahn
- Biology Department and Woods Hole Center for Oceans and Human Health, Woods Hole Oceanographic Institution, Woods Hole, MA 02543, USA
| | - Neelakanteswar Aluru
- Biology Department and Woods Hole Center for Oceans and Human Health, Woods Hole Oceanographic Institution, Woods Hole, MA 02543, USA
| |
Collapse
|
2
|
Barber HM, Robbins CG, Cutler Z, Brown RI, Werkman I, Kucenas S. Radial astroglia cooperate with microglia to clear neuronal cell bodies during zebrafish optic tectum development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643334. [PMID: 40161638 PMCID: PMC11952540 DOI: 10.1101/2025.03.14.643334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The clearance of dead cells by phagocytes is an essential component of neural development in many organisms. Microglia are the main phagocytes in the central nervous system (CNS), but the extent of participation by other glial cells remains unclear, especially under homeostatic conditions. During zebrafish optic tectum (OT) development, we observed radial astroglia forming dynamic, spherical projections from their basal processes. These projections, which we call scyllate heads, coincide with a wave of neuronal cell death in the OT. We show that scyllate heads surround the majority of dying neurons soon after phosphatidylserine exposure. However, unlike traditional phagosomes, scyllate heads persist for many hours and are rarely acidified or internalized. Instead, microglia invade scyllate heads and remove their contents for terminal degradation. Our study reveals an active role for radial astroglia in homeostatic cell clearance and cooperation between microglia and radial astroglia during zebrafish OT development. Highlights Optic tectum astroglia form large, dynamic projections called scyllate headsScyllate heads surround the majority of dying neurons during a wave of apoptosisScyllate heads are intermediate containers of dying cells rather than phagosomesMicroglia invade scyllate heads to remove their contents for terminal degradation.
Collapse
|
3
|
Abhilash PL, Bharti U, Rashmi SK, Philip M, Raju TR, Kutty BM, Sagar BKC, Alladi PA. Aging and MPTP Sensitivity Depend on Molecular and Ultrastructural Signatures of Astroglia and Microglia in Mice Substantia Nigra. Cell Mol Neurobiol 2025; 45:13. [PMID: 39833644 PMCID: PMC11753320 DOI: 10.1007/s10571-024-01528-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 12/24/2024] [Indexed: 01/22/2025]
Abstract
Both astroglia and microglia show region-specific distribution in CNS and often maladapt to age-associated alterations within their niche. Studies on autopsied substantia nigra (SN) of Parkinson's disease (PD) patients and experimental models propose gliosis as a trigger for neuronal loss. Epidemiological studies propose an ethnic bias in PD prevalence, since Caucasians are more susceptible than non-whites. Similarly, different mice strains are variably sensitive to MPTP. We had earlier likened divergent MPTP sensitivity of C57BL/6 J and CD-1 mice with differential susceptibility to PD, based on the numbers of SN neurons. We examined whether the variability was incumbent to inter-strain differences in glial features of male C57BL/6 J and CD-1 mice. Stereological counts showed relatively more microglia and fewer astrocytes in the SN of normal C57BL/6 J mice, suggesting persistence of an immune-vigilant state. MPTP-induced microgliosis and astrogliosis in both strains suggest their involvement in pathogenesis. ELISA of pro-inflammatory cytokines in the ventral-midbrain revealed augmentation of TNF-α and IL-6 at middle age in both strains that reduced at old age, suggesting middle age as a critical, inflamm-aging-associated time point. TNF-α levels were high in C57BL/6 J, through aging and post-MPTP, while IL-6 and IL-1β were upregulated at old age. CD-1 had higher levels of anti-inflammatory cytokine TGF-β. MPTP challenge caused upregulation of enzymes MAO-A, MAO-B, and iNOS in both strains. Post-MPTP enhancement in fractalkine and hemeoxygenase-1 may be neuron-associated compensatory signals. Ultrastructural observations of elongated astroglial/microglial mitochondria vis-à-vis the shrunken ones in neurons suggest a scale-up of their functions with neurotoxic consequences. Thus, astroglia and microglia may modulate aging and PD susceptibility.
Collapse
Affiliation(s)
- P L Abhilash
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bengaluru, 560029, India
| | - Upasna Bharti
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neurosciences, Bengaluru, 560029, India
| | - Santhosh Kumar Rashmi
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bengaluru, 560029, India
| | - Mariamma Philip
- Department of Biostatistics, National Institute of Mental Health and Neurosciences, Bengaluru, 560029, India
| | - T R Raju
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bengaluru, 560029, India
| | - Bindu M Kutty
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bengaluru, 560029, India
| | - B K Chandrasekhar Sagar
- Department of Biostatistics, National Institute of Mental Health and Neurosciences, Bengaluru, 560029, India
| | - Phalguni Anand Alladi
- Department of Neurophysiology, National Institute of Mental Health and Neurosciences, Hosur Road, Bengaluru, 560029, India.
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neurosciences, Bengaluru, 560029, India.
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neuro Sciences, Hosur Road, Bangalore, India.
| |
Collapse
|
4
|
Gobbo D, Kirchhoff F. Animal-based approaches to understanding neuroglia physiology in vitro and in vivo. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:229-263. [PMID: 40122627 DOI: 10.1016/b978-0-443-19104-6.00012-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
This chapter describes the pivotal role of animal models for unraveling the physiology of neuroglial cells in the central nervous system (CNS). The two rodent species Mus musculus (mice) and Rattus norvegicus (rats) have been indispensable in scientific research due to their remarkable resemblance to humans anatomically, physiologically, and genetically. Their ease of maintenance, short gestation times, and rapid development make them ideal candidates for studying the physiology of astrocytes, oligodendrocyte-lineage cells, and microglia. Moreover, their genetic similarity to humans facilitates the investigation of molecular mechanisms governing neural physiology. Mice are largely the predominant model of neuroglial research, owing to advanced genetic manipulation techniques, whereas rats remain invaluable for applications requiring larger CNS structures for surgical manipulations. Next to rodents, other animal models, namely, Danio rerio (zebrafish) and Drosophila melanogaster (fruit fly), will be discussed to emphasize their critical role in advancing our understanding of glial physiology. Each animal model provides distinct advantages and disadvantages. By combining the strengths of each of them, researchers can gain comprehensive insights into glial function across species, ultimately promoting the understanding of glial physiology in the human CNS and driving the development of novel therapeutic interventions for CNS disorders.
Collapse
Affiliation(s)
- Davide Gobbo
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany.
| | - Frank Kirchhoff
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany; Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, Homburg, Germany.
| |
Collapse
|
5
|
Silva NJ, Anderson S, Mula SA, Escoubas CC, Nakajo H, Molofsky AV. Microglial cathepsin B promotes neuronal efferocytosis during brain development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.03.626596. [PMID: 39677624 PMCID: PMC11642881 DOI: 10.1101/2024.12.03.626596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Half of all newborn neurons in the developing brain are removed via efferocytosis - the phagocytic clearance of apoptotic cells. Microglia are brain-resident professional phagocytes that play important roles in neural circuit development including as primary effectors of efferocytosis. While the mechanisms through which microglia recognize potential phagocytic cargo are widely studied, the lysosomal mechanisms that are necessary for efficient digestion are less well defined. Here we show that the lysosomal protease cathepsin B promotes microglial efferocytosis of neurons and restricts the accumulation of apoptotic cells during brain development. We show that cathepsin B is microglia-specific and enriched in brain regions where neuronal turnover is high in both zebrafish and mouse. Myeloid-specific cathepsin B knockdown in zebrafish led to dysmorphic microglia containing undigested dead cells, as well as an accumulation of dead cells in surrounding tissue. These effects where phenocopied in mice globally deficient for Ctsb using markers for apoptosis. We also observed behavioral impairments in both models. Live imaging studies in zebrafish revealed deficits in phagolysosomal fusion and acidification, and live imaging of cultured mouse microglia reveal delayed phagocytosis consistent with impairments in digestion and resolution of phagocytosis rather than initial uptake. These data reveal a novel role for microglial cathepsin B in mediating neuronal efferocytosis during typical brain development.
Collapse
|
6
|
Czopka T, Monk K, Peri F. Glial Cell Development and Function in the Zebrafish Central Nervous System. Cold Spring Harb Perspect Biol 2024; 16:a041350. [PMID: 38692835 PMCID: PMC11529855 DOI: 10.1101/cshperspect.a041350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Over the past decades the zebrafish has emerged as an excellent model organism with which to study the biology of all glial cell types in nervous system development, plasticity, and regeneration. In this review, which builds on the earlier work by Lyons and Talbot in 2015, we will summarize how the relative ease to manipulate the zebrafish genome and its suitability for intravital imaging have helped understand principles of glial cell biology with a focus on oligodendrocytes, microglia, and astrocytes. We will highlight recent findings on the diverse properties and functions of these glial cell types in the central nervous system and discuss open questions and future directions of the field.
Collapse
Affiliation(s)
- Tim Czopka
- Centre for Clinical Brain Sciences, The University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| | - Kelly Monk
- Vollum Institute, Oregon Health and Science University, Portland, Oregon 97239, USA
| | - Francesca Peri
- Department of Molecular Life Sciences, University of Zürich, 8057 Zürich, Switzerland
| |
Collapse
|
7
|
Zareba J, Cattaneo EF, Villani A, Othman A, Streb S, Peri F. NPC1 links cholesterol trafficking to microglial morphology via the gastrosome. Nat Commun 2024; 15:8638. [PMID: 39366931 PMCID: PMC11452621 DOI: 10.1038/s41467-024-52874-6] [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: 03/11/2024] [Accepted: 09/19/2024] [Indexed: 10/06/2024] Open
Abstract
Microglia play important roles in brain development and homeostasis by removing dying neurons through efferocytosis. Morphological changes in microglia are hallmarks of many neurodegenerative conditions, such as Niemann-Pick disease type C. Here, NPC1 loss causes microglia to shift from a branched to an ameboid form, though the cellular basis and functional impact of this change remain unclear. Using zebrafish, we show that NPC1 deficiency causes an efferocytosis-dependent expansion of the microglial gastrosome, a collection point for engulfed material. In vivo and in vitro experiments on microglia and mammalian macrophages demonstrate that NPC1 localizes to the gastrosome, and its absence leads to cholesterol accumulation in this compartment. NPC1 loss and neuronal cell death synergistically affect gastrosome size and cell shape, increasing the sensitivity of NPC1-deficient cells to neuronal cell death. Finally, we demonstrate conservation of cholesterol accumulation and gastrosome expansion in NPC patient-derived fibroblasts, offering an interesting target for further disease investigation.
Collapse
Affiliation(s)
- Joanna Zareba
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Elena F Cattaneo
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Ambra Villani
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Alaa Othman
- Functional Genomic Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Sebastian Streb
- Functional Genomic Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Francesca Peri
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
8
|
Gordon H, Schafer ZT, Smith CJ. Microglia cannibalism and efferocytosis leads to shorter lifespans of developmental microglia. PLoS Biol 2024; 22:e3002819. [PMID: 39475879 PMCID: PMC11524473 DOI: 10.1371/journal.pbio.3002819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 08/29/2024] [Indexed: 11/02/2024] Open
Abstract
The overproduction of cells and subsequent production of debris is a universal principle of neurodevelopment. Here, we show an additional feature of the developing nervous system that causes neural debris-promoted by the sacrificial nature of embryonic microglia that irreversibly become phagocytic after clearing other neural debris. Described as long-lived, microglia colonize the embryonic brain and persist into adulthood. Using transgenic zebrafish to investigate the microglia debris during brain construction, we identified that unlike other neural cell types that die in developmental stages after they have expanded, necroptosis-dependent microglial debris is prevalent when microglia are expanding in the zebrafish brain. Time-lapse imaging of microglia demonstrates that this debris is cannibalized by other microglia. To investigate features that promote microglia death and cannibalism, we used time-lapse imaging and fate-mapping strategies to track the lifespan of individual developmental microglia. These approaches revealed that instead of embryonic microglia being long-lived cells that completely digest their phagocytic debris, once most developmental microglia in zebrafish become phagocytic they eventually die, including ones that are cannibalistic. These results establish a paradox-which we tested by increasing neural debris and manipulating phagocytosis-that once most microglia in the embryo become phagocytic, they die, create debris, and then are cannibalized by other microglia, resulting in more phagocytic microglia that are destined to die.
Collapse
Affiliation(s)
- Hannah Gordon
- Department of Biological Sciences at the University of Notre Dame, Notre Dame, Indiana, United States of America
- The Center for Stem Cells and Regenerative Medicine at the University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Zachary T. Schafer
- Department of Biological Sciences at the University of Notre Dame, Notre Dame, Indiana, United States of America
| | - Cody J. Smith
- Department of Biological Sciences at the University of Notre Dame, Notre Dame, Indiana, United States of America
- The Center for Stem Cells and Regenerative Medicine at the University of Notre Dame, Notre Dame, Indiana, United States of America
| |
Collapse
|
9
|
Gordon H, Schafer Z, Smith CJ. Microglia cannibalism and efferocytosis leads to shorter lifespans of developmental microglia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.15.532426. [PMID: 36993267 PMCID: PMC10055159 DOI: 10.1101/2023.03.15.532426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
The overproduction of cells and subsequent production of debris is a universal principle of neurodevelopment. Here we show an additional feature of the developing nervous system that causes neural debris - promoted by the sacrificial nature of embryonic microglia that irreversibly become phagocytic after clearing other neural debris. Described as long-lived, microglia colonize the embryonic brain and persist into adulthood. Using transgenic zebrafish to investigate the microglia debris during brain construction, we identified that unlike other neural cell-types that die in developmental stages after they have expanded, necroptosis-dependent microglial debris is prevalent when microglia are expanding in the zebrafish brain. Time-lapse imaging of microglia demonstrates that this debris is cannibalized by other microglia. To investigate features that promote microglia death and cannibalism, we used time-lapse imaging and fate-mapping strategies to track the lifespan of individual developmental microglia. These approaches revealed that instead of embryonic microglia being long-lived cells that completely digest their phagocytic debris, once most developmental microglia in zebrafish become phagocytic they eventually die, including ones that are cannibalistic. These results establish a paradox -- which we tested by increasing neural debris and manipulating phagocytosis -- that once most microglia in the embryo become phagocytic, they die, create debris and then are cannibalized by other microglia, resulting in more phagocytic microglia that are destined to die.
Collapse
Affiliation(s)
- Hannah Gordon
- Department of Biological Sciences, at the University of Notre Dame, Notre Dame, IN
- The Center for Stem Cells and Regenerative Medicine at the University of Notre Dame, Notre Dame, IN
| | - Zachary Schafer
- Department of Biological Sciences, at the University of Notre Dame, Notre Dame, IN
| | - Cody J. Smith
- Department of Biological Sciences, at the University of Notre Dame, Notre Dame, IN
- The Center for Stem Cells and Regenerative Medicine at the University of Notre Dame, Notre Dame, IN
| |
Collapse
|
10
|
Barry-Carroll L, Gomez-Nicola D. The molecular determinants of microglial developmental dynamics. Nat Rev Neurosci 2024; 25:414-427. [PMID: 38658739 DOI: 10.1038/s41583-024-00813-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2024] [Indexed: 04/26/2024]
Abstract
Microglia constitute the largest population of parenchymal macrophages in the brain and are considered a unique subset of central nervous system glial cells owing to their extra-embryonic origins in the yolk sac. During development, microglial progenitors readily proliferate and eventually colonize the entire brain. In this Review, we highlight the origins of microglial progenitors and their entry routes into the brain and discuss the various molecular and non-molecular determinants of their fate, which may inform their specific functions. Specifically, we explore recently identified mechanisms that regulate microglial colonization of the brain, including the availability of space, and describe how the expansion of highly proliferative microglial progenitors facilitates the occupation of the microglial niche. Finally, we shed light on the factors involved in establishing microglial identity in the brain.
Collapse
Affiliation(s)
- Liam Barry-Carroll
- Nutrineuro, UMR 1286 INRAE, Bordeaux University, Bordeaux INP, Bordeaux, France
| | - Diego Gomez-Nicola
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK.
| |
Collapse
|
11
|
Rutherford HA, Candeias D, Duncan CJA, Renshaw SA, Hamilton N. Macrophage transplantation rescues RNASET2-deficient leukodystrophy by replacing deficient microglia in a zebrafish model. Proc Natl Acad Sci U S A 2024; 121:e2321496121. [PMID: 38753517 PMCID: PMC11126979 DOI: 10.1073/pnas.2321496121] [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: 12/20/2023] [Accepted: 04/05/2024] [Indexed: 05/18/2024] Open
Abstract
RNASET2-deficient leukodystrophy is a rare infantile white matter disorder mimicking a viral infection and resulting in severe psychomotor impairments. Despite its severity, there is little understanding of cellular mechanisms of pathogenesis and no treatments. Recent research using the rnaset2 mutant zebrafish model has suggested that microglia may be the drivers of the neuropathology, due to their failure to digest apoptotic debris during neurodevelopment. Therefore, we developed a strategy for microglial replacement through transplantation of adult whole kidney marrow-derived macrophages into embryonic hosts. Using live imaging, we revealed that transplant-derived macrophages can engraft within host brains and express microglia-specific markers, suggesting the adoption of a microglial phenotype. Tissue-clearing strategies revealed the persistence of transplanted cells in host brains beyond embryonic stages. We demonstrated that transplanted cells clear apoptotic cells within the brain, as well as rescue overactivation of the antiviral response otherwise seen in mutant larvae. RNA sequencing at the point of peak transplant-derived cell engraftment confirms that transplantation can reduce the brain-wide immune response and particularly, the antiviral response, in rnaset2-deficient brains. Crucially, this reduction in neuroinflammation resulted in behavioral rescue-restoring rnaset2 mutant motor activity to wild-type (WT) levels in embryonic and juvenile stages. Together, these findings demonstrate the role of microglia as the cellular drivers of neuropathology in rnaset2 mutants and that macrophage transplantation is a viable strategy for microglial replacement in the zebrafish. Therefore, microglia-targeted interventions may have therapeutic benefits in RNASET2-deficient leukodystrophy.
Collapse
Affiliation(s)
- Holly A. Rutherford
- Department of Infection and Immunity, School of Medicine and Population Health, University of Sheffield, SheffieldS10 2RX, United Kingdom
- Bateson Centre, University of Sheffield, SheffieldS10 2TN, United Kingdom
| | - Diogo Candeias
- Department of Biology, University of York, YorkYO10 5DD, United Kingdom
- York Biomedical research Institute, University of York, YorkYO10 5DD, United Kingdom
| | - Christopher J. A. Duncan
- Immunology and Inflammation Theme, Translational and Clinical Research Institute, Newcastle University, NewcastleNE2 4HH, United Kingdom
- Department of Infection and Tropical Medicine, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals National Health Services Foundation Trust, NewcastleNE2 4HH, United Kingdom
| | - Stephen A. Renshaw
- Department of Infection and Immunity, School of Medicine and Population Health, University of Sheffield, SheffieldS10 2RX, United Kingdom
- Bateson Centre, University of Sheffield, SheffieldS10 2TN, United Kingdom
| | - Noémie Hamilton
- Department of Biology, University of York, YorkYO10 5DD, United Kingdom
- York Biomedical research Institute, University of York, YorkYO10 5DD, United Kingdom
| |
Collapse
|
12
|
Tufan T, Comertpay G, Villani A, Nelson GM, Terekhova M, Kelley S, Zakharov P, Ellison RM, Shpynov O, Raymond M, Sun J, Chen Y, Bockelmann E, Stremska M, Peterson LW, Boeckaerts L, Goldman SR, Etchegaray JI, Artyomov MN, Peri F, Ravichandran KS. Rapid unleashing of macrophage efferocytic capacity via transcriptional pause release. Nature 2024; 628:408-415. [PMID: 38480883 DOI: 10.1038/s41586-024-07172-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 02/07/2024] [Indexed: 03/18/2024]
Abstract
During development, inflammation or tissue injury, macrophages may successively engulf and process multiple apoptotic corpses via efferocytosis to achieve tissue homeostasis1. How macrophages may rapidly adapt their transcription to achieve continuous corpse uptake is incompletely understood. Transcriptional pause/release is an evolutionarily conserved mechanism, in which RNA polymerase (Pol) II initiates transcription for 20-60 nucleotides, is paused for minutes to hours and is then released to make full-length mRNA2. Here we show that macrophages, within minutes of corpse encounter, use transcriptional pause/release to unleash a rapid transcriptional response. For human and mouse macrophages, the Pol II pause/release was required for continuous efferocytosis in vitro and in vivo. Interestingly, blocking Pol II pause/release did not impede Fc receptor-mediated phagocytosis, yeast uptake or bacterial phagocytosis. Integration of data from three genomic approaches-precision nuclear run-on sequencing, RNA sequencing, and assay for transposase-accessible chromatin using sequencing (ATAC-seq)-on efferocytic macrophages at different time points revealed that Pol II pause/release controls expression of select transcription factors and downstream target genes. Mechanistic studies on transcription factor EGR3, prominently regulated by pause/release, uncovered EGR3-related reprogramming of other macrophage genes involved in cytoskeleton and corpse processing. Using lysosomal probes and a new genetic fluorescent reporter, we identify a role for pause/release in phagosome acidification during efferocytosis. Furthermore, microglia from egr3-deficient zebrafish embryos displayed reduced phagocytosis of apoptotic neurons and fewer maturing phagosomes, supporting defective corpse processing. Collectively, these data indicate that macrophages use Pol II pause/release as a mechanism to rapidly alter their transcriptional programs for efficient processing of the ingested apoptotic corpses and for successive efferocytosis.
Collapse
Affiliation(s)
- Turan Tufan
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gamze Comertpay
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ambra Villani
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Geoffrey M Nelson
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Marina Terekhova
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Shannon Kelley
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Pavel Zakharov
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rochelle M Ellison
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Oleg Shpynov
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- JetBrains Research, Munich, Germany
| | - Michael Raymond
- Department of Neuroscience and MIC, University of Virginia, Charlottesville, VA, USA
| | - Jerry Sun
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yitan Chen
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Enno Bockelmann
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Marta Stremska
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lance W Peterson
- Department of Pediatrics, Division of Rheumatology and Immunology, Washington University in St. Louis, St. Louis, MO, USA
| | - Laura Boeckaerts
- Unit for Cell Clearance in Health and Disease, VIB-UGent Center for Inflammation Research and the Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Seth R Goldman
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - J Iker Etchegaray
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Maxim N Artyomov
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Francesca Peri
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Kodi S Ravichandran
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Neuroscience and MIC, University of Virginia, Charlottesville, VA, USA.
- Unit for Cell Clearance in Health and Disease, VIB-UGent Center for Inflammation Research and the Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| |
Collapse
|
13
|
Chen F, Tang H, Cai X, Lin J, Xiang L, Kang R, Liu J, Tang D. Targeting paraptosis in cancer: opportunities and challenges. Cancer Gene Ther 2024; 31:349-363. [PMID: 38177306 DOI: 10.1038/s41417-023-00722-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/07/2023] [Accepted: 12/12/2023] [Indexed: 01/06/2024]
Abstract
Cell death can be classified into two primary categories: accidental cell death and regulated cell death (RCD). Within RCD, there are distinct apoptotic and non-apoptotic cell death pathways. Among the various forms of non-apoptotic RCD, paraptosis stands out as a unique mechanism characterized by distinct morphological changes within cells. These alterations encompass cytoplasmic vacuolization, organelle swelling, notably in the endoplasmic reticulum and mitochondria, and the absence of typical apoptotic features, such as cell shrinkage and DNA fragmentation. Biochemically, paraptosis distinguishes itself by its independence from caspases, which are conventionally associated with apoptotic death. This intriguing cell death pathway can be initiated by various cellular stressors, including oxidative stress, protein misfolding, and specific chemical compounds. Dysregulated paraptosis plays a pivotal role in several critical cancer-related processes, such as autophagic degradation, drug resistance, and angiogenesis. This review provides a comprehensive overview of recent advancements in our understanding of the mechanisms and regulation of paraptosis. Additionally, it delves into the potential of paraptosis-related compounds for targeted cancer treatment, with the aim of enhancing treatment efficacy while minimizing harm to healthy cells.
Collapse
Affiliation(s)
- Fangquan Chen
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510150, China
| | - Hu Tang
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510150, China
| | - Xiutao Cai
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510150, China
| | - Junhao Lin
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510150, China
| | - Limin Xiang
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510150, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jiao Liu
- DAMP Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510150, China.
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
14
|
Tewari M, Michalski S, Egan TM. Modulation of Microglial Function by ATP-Gated P2X7 Receptors: Studies in Rat, Mice and Human. Cells 2024; 13:161. [PMID: 38247852 PMCID: PMC10814008 DOI: 10.3390/cells13020161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
P2X receptors are a family of seven ATP-gated ion channels that trigger physiological and pathophysiological responses in a variety of cells. Five of the family members are sensitive to low concentrations of extracellular ATP, while the P2X6 receptor has an unknown affinity. The last subtype, the P2X7 receptor, is unique in requiring millimolar concentrations to fully activate in humans. This low sensitivity imparts the agonist with the ability to act as a damage-associated molecular pattern that triggers the innate immune response in response to the elevated levels of extracellular ATP that accompany inflammation and tissue damage. In this review, we focus on microglia because they are the primary immune cells of the central nervous system, and they activate in response to ATP or its synthetic analog, BzATP. We start by introducing purinergic receptors and then briefly consider the roles that microglia play in neurodevelopment and disease by referencing both original works and relevant reviews. Next, we move to the role of extracellular ATP and P2X receptors in initiating and/or modulating innate immunity in the central nervous system. While most of the data that we review involve work on mice and rats, we highlight human studies of P2X7R whenever possible.
Collapse
|
15
|
Marín-Teva JL, Sepúlveda MR, Neubrand VE, Cuadros MA. Microglial Phagocytosis During Embryonic and Postnatal Development. ADVANCES IN NEUROBIOLOGY 2024; 37:151-161. [PMID: 39207691 DOI: 10.1007/978-3-031-55529-9_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia play decisive roles during the development of the central nervous system (CNS). Phagocytosis is one of the classical functions attributed to microglia, being involved in nearly all phases of the embryonic and postnatal development of the brain, such as rapid clearance of cell debris to avoid an inflammatory response, controlling the number of neuronal and glial cells or their precursors, contribution to axon guidance and to refinement of synaptic connections. To carry out all these tasks, microglial cells are equipped with a panoply of receptors, that convert microglia to the "professional phagocytes" of the nervous parenchyma. These receptors are modulated by spatiotemporal cues that adapt the properties of microglia to the needs of the developing CNS. Thus, in this chapter, we will discuss the role of microglial phagocytosis in all the aforementioned processes. First, we will explain the general phagocytic process, to describe afterward the performance of microglial cells in detail.
Collapse
Affiliation(s)
- José L Marín-Teva
- Department of Cell Biology, Faculty of Sciences, University of Granada, Granada, Spain.
| | - M Rosario Sepúlveda
- Department of Cell Biology, Faculty of Sciences, University of Granada, Granada, Spain
| | - Veronika E Neubrand
- Department of Cell Biology, Faculty of Sciences, University of Granada, Granada, Spain
| | - Miguel A Cuadros
- Department of Cell Biology, Faculty of Sciences, University of Granada, Granada, Spain
| |
Collapse
|
16
|
Morales M, Findley AP, Mitchell DM. Intercellular contact and cargo transfer between Müller glia and to microglia precede apoptotic cell clearance in the developing retina. Development 2024; 151:dev202407. [PMID: 38174987 PMCID: PMC10820749 DOI: 10.1242/dev.202407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024]
Abstract
To clarify our understanding of glial phagocytosis in retinal development, we used real-time imaging of larval zebrafish to provide cell-type specific resolution of this process. We show that radial Müller glia frequently participate in microglial phagocytosis while also completing a subset of phagocytic events. Müller glia actively engage with dying cells through initial target cell contact and phagocytic cup formation, after which an exchange of the dying cell from Müller glia to microglia often takes place. In addition, we find evidence that Müller glia cellular material, possibly from the initial Müller cell phagocytic cup, is internalized into microglial compartments. Previously undescribed Müller cell behaviors were seen, including cargo splitting, wrestling for targets and lateral passing of cargo to neighbors. Collectively, our work provides new insight into glial functions and intercellular interactions, which will allow future work to understand these behaviors on a molecular level.
Collapse
Affiliation(s)
- Michael Morales
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844, USA
| | - Anna P. Findley
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844, USA
| | - Diana M. Mitchell
- Department of Biological Sciences, University of Idaho, Moscow, ID 83844, USA
| |
Collapse
|
17
|
Valamparamban GF, Spéder P. Homemade: building the structure of the neurogenic niche. Front Cell Dev Biol 2023; 11:1275963. [PMID: 38107074 PMCID: PMC10722289 DOI: 10.3389/fcell.2023.1275963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/16/2023] [Indexed: 12/19/2023] Open
Abstract
Neural stem/progenitor cells live in an intricate cellular environment, the neurogenic niche, which supports their function and enables neurogenesis. The niche is made of a diversity of cell types, including neurons, glia and the vasculature, which are able to signal to and are structurally organised around neural stem/progenitor cells. While the focus has been on how individual cell types signal to and influence the behaviour of neural stem/progenitor cells, very little is actually known on how the niche is assembled during development from multiple cellular origins, and on the role of the resulting topology on these cells. This review proposes to draw a state-of-the art picture of this emerging field of research, with the aim to expose our knowledge on niche architecture and formation from different animal models (mouse, zebrafish and fruit fly). We will span its multiple aspects, from the existence and importance of local, adhesive interactions to the potential emergence of larger-scale topological properties through the careful assembly of diverse cellular and acellular components.
Collapse
Affiliation(s)
| | - Pauline Spéder
- Institut Pasteur, Université Paris Cité, CNRS UMR3738, Structure and Signals in the Neurogenic Niche, Paris, France
| |
Collapse
|
18
|
Morales M, Findley AP, Mitchell DM. Intercellular contact and cargo transfer between Müller glia and to microglia precede apoptotic cell clearance in the developing retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.06.561302. [PMID: 37873206 PMCID: PMC10592698 DOI: 10.1101/2023.10.06.561302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
To clarify our understanding of glial phagocytosis in retinal development, we used real time imaging of larval zebrafish to provide cell-type specific resolution of this process. We show that radial Müller glia frequently participate in microglial phagocytosis while also completing a subset of phagocytic events. Müller glia (MG) actively engage with dying cells through initial target cell contact and phagocytic cup formation after which an exchange of the dying cell from MG to microglia often takes place. Additionally, we find evidence that Müller glia cellular material, possibly from the initial Müller cell's phagocytic cup, is internalized into microglial compartments. Previously undescribed Müller cell behaviors were seen, including cargo splitting, wrestling for targets, lateral passing of cargo to neighbors, and engulfment of what is possibly synaptic puncta. Collectively, our work provides new insight into glial functions and intercellular interactions, which will allow future work to understand these behaviors on a molecular level.
Collapse
Affiliation(s)
| | - Anna P Findley
- Biological Sciences, University of Idaho, Moscow, ID 83844
| | | |
Collapse
|
19
|
Martins TG, Soliman R, Cordero-Maldonado ML, Donato C, Ameli C, Mombaerts L, Skupin A, Peri F, Crawford AD. Seizure-induced increase in microglial cell population in the developing zebrafish brain. Epilepsy Res 2023; 195:107203. [PMID: 37572541 DOI: 10.1016/j.eplepsyres.2023.107203] [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: 03/03/2023] [Revised: 06/21/2023] [Accepted: 07/31/2023] [Indexed: 08/14/2023]
Abstract
Epilepsy is a chronic brain disorder characterized by unprovoked and recurrent seizures, of which 60% are of unknown etiology. Recent studies implicate microglia in the pathophysiology of epilepsy. However, their role in this process, in particular following early-life seizures, remains poorly understood due in part to the lack of suitable experimental models allowing the in vivo imaging of microglial activity. Given the advantage of zebrafish larvae for minimally-invasive imaging approaches, we sought for the first time to describe the microglial responses after acute seizures in two different zebrafish larval models: a chemically-induced epileptic model by the systemic injection of kainate at 3 days post-fertilization, and the didys552 genetic epilepsy model, which carries a mutation in scn1lab that leads to spontaneous epileptiform discharges. Kainate-treated larvae exhibited transient brain damage as shown by increased numbers of apoptotic nuclei as early as one day post-injection, which was followed by an increase in the number of microglia in the brain. A similar microglial phenotype was also observed in didys552-/- mutants, suggesting that microglia numbers change in response to seizure-like activity in the brain. Interestingly, kainate-treated larvae also displayed a decreased seizure threshold towards subsequent pentylenetetrazole-induced seizures, as shown by higher locomotor and encephalographic activity in comparison with vehicle-injected larvae. These results are comparable to kainate-induced rodent seizure models and suggest the suitability of these zebrafish seizure models for future studies, in particular to elucidate the links between epileptogenesis and microglial dynamic changes after seizure induction in the developing brain, and to understand how these modulate seizure susceptibility.
Collapse
Affiliation(s)
- Teresa G Martins
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg.
| | - Remon Soliman
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | | | - Cristina Donato
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Corrado Ameli
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Laurent Mombaerts
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Alexander Skupin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg; University of California, San Diego (UCSD), La Jolla, CA, United States
| | - Francesca Peri
- Developmental Biology Group, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Alexander D Crawford
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg; Institute for Orphan Drug Discovery, Bremerhaven, Germany.
| |
Collapse
|
20
|
Boghozian R, Sharma S, Narayana K, Cheema M, Brown CE. Sex and interferon gamma signaling regulate microglia migration in the adult mouse cortex in vivo. Proc Natl Acad Sci U S A 2023; 120:e2302892120. [PMID: 37428916 PMCID: PMC10629543 DOI: 10.1073/pnas.2302892120] [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: 02/19/2023] [Accepted: 06/02/2023] [Indexed: 07/12/2023] Open
Abstract
Although microglia possess the unique ability to migrate, whether mobility is evident in all microglia, is sex dependent, and what molecular mechanisms drive this, is not well understood in the adult brain. Using longitudinal in vivo two-photon imaging of sparsely labeled microglia, we find a relatively small population of microglia (~5%) are mobile under normal conditions. Following injury (microbleed), the fraction of mobile microglia increased in a sex-dependent manner, with male microglia migrating significantly greater distances toward the microbleed relative to their female counterparts. To understand the signaling pathways involved, we interrogated the role of interferon gamma (IFNγ). Our data show that in male mice, stimulating microglia with IFNγ promotes migration whereas inhibiting IFNγ receptor 1 signaling inhibits them. By contrast, female microglia were generally unaffected by these manipulations. These findings highlight the diversity of microglia migratory responses to injury, its dependence on sex and the signaling mechanisms that modulate this behavior.
Collapse
Affiliation(s)
- Roobina Boghozian
- Division of Medical Sciences, University of Victoria, Victoria, BCV8P 5C2, Canada
| | - Sorabh Sharma
- Division of Medical Sciences, University of Victoria, Victoria, BCV8P 5C2, Canada
| | - Kamal Narayana
- Division of Medical Sciences, University of Victoria, Victoria, BCV8P 5C2, Canada
| | - Manjinder Cheema
- Division of Medical Sciences, University of Victoria, Victoria, BCV8P 5C2, Canada
| | - Craig E. Brown
- Division of Medical Sciences, University of Victoria, Victoria, BCV8P 5C2, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, BCV6T 2A1, Canada
| |
Collapse
|
21
|
Barry-Carroll L, Greulich P, Marshall AR, Riecken K, Fehse B, Askew KE, Li K, Garaschuk O, Menassa DA, Gomez-Nicola D. Microglia colonize the developing brain by clonal expansion of highly proliferative progenitors, following allometric scaling. Cell Rep 2023; 42:112425. [PMID: 37099424 DOI: 10.1016/j.celrep.2023.112425] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 01/25/2023] [Accepted: 04/06/2023] [Indexed: 04/27/2023] Open
Abstract
Microglia arise from the yolk sac and enter the brain during early embryogenesis. Upon entry, microglia undergo in situ proliferation and eventually colonize the entire brain by the third postnatal week in mice. However, the intricacies of their developmental expansion remain unclear. Here, we characterize the proliferative dynamics of microglia during embryonic and postnatal development using complementary fate-mapping techniques. We demonstrate that the developmental colonization of the brain is facilitated by clonal expansion of highly proliferative microglial progenitors that occupy spatial niches throughout the brain. Moreover, the spatial distribution of microglia switches from a clustered to a random pattern between embryonic and late postnatal development. Interestingly, the developmental increase in microglial numbers follows the proportional growth of the brain in an allometric manner until a mosaic distribution has been established. Overall, our findings offer insight into how the competition for space may drive microglial colonization by clonal expansion during development.
Collapse
Affiliation(s)
- Liam Barry-Carroll
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Philip Greulich
- School of Mathematical Sciences, University of Southampton, Southampton, UK; Institute for Life Sciences (IfLS), University of Southampton, Southampton, UK
| | - Abigail R Marshall
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharine E Askew
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Kaizhen Li
- Department of Neurophysiology, University of Tübingen, Tübingen, Germany
| | - Olga Garaschuk
- Department of Neurophysiology, University of Tübingen, Tübingen, Germany
| | - David A Menassa
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK; The Queen's College, University of Oxford, Oxford, UK
| | - Diego Gomez-Nicola
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK; Institute for Life Sciences (IfLS), University of Southampton, Southampton, UK.
| |
Collapse
|
22
|
Peterman E, Quitevis EJA, Black EC, Horton EC, Aelmore RL, White E, Sagasti A, Rasmussen JP. Zebrafish cutaneous injury models reveal that Langerhans cells engulf axonal debris in adult epidermis. Dis Model Mech 2023; 16:dmm049911. [PMID: 36876992 PMCID: PMC10110399 DOI: 10.1242/dmm.049911] [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: 09/29/2022] [Accepted: 02/28/2023] [Indexed: 03/07/2023] Open
Abstract
Somatosensory neurons extend enormous peripheral axons to the skin, where they detect diverse environmental stimuli. Somatosensory peripheral axons are easily damaged due to their small caliber and superficial location. Axonal damage results in Wallerian degeneration, creating vast quantities of cellular debris that phagocytes must remove to maintain organ homeostasis. The cellular mechanisms that ensure efficient clearance of axon debris from stratified adult skin are unknown. Here, we established zebrafish scales as a tractable model to study axon degeneration in the adult epidermis. Using this system, we demonstrated that skin-resident immune cells known as Langerhans cells engulf the majority of axon debris. In contrast to immature skin, adult keratinocytes did not significantly contribute to debris removal, even in animals lacking Langerhans cells. Our study establishes a powerful new model for studying Wallerian degeneration and identifies a new function for Langerhans cells in maintenance of adult skin homeostasis following injury. These findings have important implications for pathologies that trigger somatosensory axon degeneration.
Collapse
Affiliation(s)
- Eric Peterman
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | | | - Erik C. Black
- Department of Biology, University of Washington, Seattle, WA 98195, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Emma C. Horton
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Rune L. Aelmore
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Ethan White
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Alvaro Sagasti
- Molecular, Cell and Developmental Biology Department, University of California, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| | - Jeffrey P. Rasmussen
- Department of Biology, University of Washington, Seattle, WA 98195, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
23
|
Sierra-Martín A, Navascués J, Neubrand VE, Sepúlveda MR, Martín-Oliva D, Cuadros MA, Marín-Teva JL. LPS-stimulated microglial cells promote ganglion cell death in organotypic cultures of quail embryo retina. Front Cell Neurosci 2023; 17:1120400. [PMID: 37006469 PMCID: PMC10050569 DOI: 10.3389/fncel.2023.1120400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/27/2023] [Indexed: 03/17/2023] Open
Abstract
During development microglia colonize the central nervous system (CNS) and play an important role in programmed cell death, not only because of their ability to remove dead cells by phagocytosis, but also because they can promote the death of neuronal and glial cells. To study this process, we used as experimental systems the developing in situ quail embryo retina and organotypic cultures of quail embryo retina explants (QEREs). In both systems, immature microglia show an upregulation of certain inflammatory markers, e.g., inducible NO synthase (iNOS), and nitric oxide (NO) under basal conditions, which can be further enhanced with LPS-treatment. Hence, we investigated in the present study the role of microglia in promoting ganglion cell death during retinal development in QEREs. Results showed that LPS-stimulation of microglia in QEREs increases (i) the percentage of retinal cells with externalized phosphatidylserine, (ii) the frequency of phagocytic contacts between microglial and caspase-3-positive ganglion cells, (iii) cell death in the ganglion cell layer, and (iv) microglial production of reactive oxygen/nitrogen species, such as NO. Furthermore, iNOS inhibition by L-NMMA decreases cell death of ganglion cells and increases the number of ganglion cells in LPS-treated QEREs. These data demonstrate that LPS-stimulated microglia induce ganglion cell death in cultured QEREs by a NO-dependent mechanism. The fact that phagocytic contacts between microglial and caspase-3-positive ganglion cells increase suggests that this cell death might be mediated by microglial engulfment, although a phagocytosis-independent mechanism cannot be excluded.
Collapse
|
24
|
Sun LWH, Asana Marican HT, Shen H. In Vivo Imaging of Radiation-Induced Apoptosis at Single-Cell Resolution in Transgenic Zebrafish Embryos. Radiat Res 2023; 199:229-239. [PMID: 36745564 DOI: 10.1667/rade-22-00174.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 01/17/2023] [Indexed: 02/07/2023]
Abstract
Among the various types of cell death induced by ionizing radiation, apoptosis is a highly regulated and well-characterized form. Investigating radiation-induced apoptosis in an intact organism offers advantages in capturing the dynamics of apoptosis under preserved physiology, although high resolution imaging remains challenging. Owing to their optical transparency and genetic amenability, zebrafish is an ideal animal model for research into this aspect. In this study, we present a secA5 transgenic zebrafish expressing genetically encoded secreted ANNEXIN V fused with mVenus, a yellow fluorescent protein that enables reporting of radiation-induced apoptosis. Using in vivo imaging approach, we show that after 2 Gy total-body irradiation, apoptosis could be visualized at single-cell resolution in different cell types throughout the embryo. Elevated apoptosis could be imaged and quantified in the neuroepithelium of the embryonic brain, as well as the proliferative zone and parenchyma of the larval brain. In addition, clearance of apoptotic cells by microglia, the professional phagocytes residing in the brain, could be imaged at single-cell resolution in irradiated larvae. These results establish transgenic secA5 zebrafish as a useful and versatile in vivo system for investigating the dynamic process of radiation-induced apoptosis.
Collapse
Affiliation(s)
| | | | - Hongyuan Shen
- Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore
| |
Collapse
|
25
|
Pitts KM, Margeta MA. Myeloid masquerade: Microglial transcriptional signatures in retinal development and disease. Front Cell Neurosci 2023; 17:1106547. [PMID: 36779012 PMCID: PMC9909491 DOI: 10.3389/fncel.2023.1106547] [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: 11/23/2022] [Accepted: 01/12/2023] [Indexed: 01/27/2023] Open
Abstract
Microglia are dynamic guardians of neural tissue and the resident immune cells of the central nervous system (CNS). The disease-associated microglial signature (DAM), also known as the microglial neurodegenerative phenotype (MGnD), has gained significant attention in recent years as a fundamental microglial response common to various neurodegenerative disease pathologies. Interestingly, this signature shares many features in common with developmental microglia, suggesting the existence of recycled gene programs which play a role both in early neural circuit formation as well as in response to aging and disease. In addition, recent advances in single cell RNA sequencing have revealed significant heterogeneity within the original DAM signature, with contributions from both yolk sac-derived microglia as well as bone marrow-derived macrophages. In this review, we examine the role of the DAM signature in retinal development and disease, highlighting crosstalk between resident microglia and infiltrating monocytes which may critically contribute to the underlying mechanisms of age-related neurodegeneration.
Collapse
Affiliation(s)
- Kristen M. Pitts
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
- Schepens Eye Research Institute of Mass, Eye and Ear, Boston, MA, United States
| | - Milica A. Margeta
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
- Schepens Eye Research Institute of Mass, Eye and Ear, Boston, MA, United States
| |
Collapse
|
26
|
Ahmad I, Subramani M. Microglia: Friends or Foes in Glaucoma? A Developmental Perspective. Stem Cells Transl Med 2022; 11:1210-1218. [PMID: 36426733 PMCID: PMC9801300 DOI: 10.1093/stcltm/szac077] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/29/2022] [Indexed: 11/27/2022] Open
Abstract
Glaucoma is the most prevalent form of optic neuropathy where a progressive degeneration of retinal ganglion cells (RGCs) leads to irreversible loss of vision. The mechanism underlying glaucomatous degeneration remains poorly understood. However, evidence suggests that microglia, which regulate RGC numbers and synaptic integrity during development and provide homeostatic support in adults, may contribute to the disease process. Hence, microglia represent a valid cellular target for therapeutic approaches in glaucoma. Here, we provide an overview of the role of microglia in RGC development and degeneration in the backdrop of neurogenesis and neurodegeneration in the central nervous system and discuss how pathological recapitulation of microglia-mediated developmental mechanisms may help initiate or exacerbate glaucomatous degeneration.
Collapse
Affiliation(s)
- Iqbal Ahmad
- Corresponding author: Iqbal Ahmad, Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Murali Subramani
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
27
|
Narra SS, Rondeau P, Fernezelian D, Gence L, Ghaddar B, Bourdon E, Lefebvre d'Hellencourt C, Rastegar S, Diotel N. Distribution of microglia/immune cells in the brain of adult zebrafish in homeostatic and regenerative conditions: Focus on oxidative stress during brain repair. J Comp Neurol 2022; 531:238-255. [PMID: 36282721 DOI: 10.1002/cne.25421] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 09/14/2022] [Accepted: 09/16/2022] [Indexed: 11/08/2022]
Abstract
Microglia are macrophage-like cells exerting determinant roles in neuroinflammatory and oxidative stress processes during brain regeneration. We used zebrafish as a model of brain plasticity and repair. First, by performing L-plastin (Lcp1) immunohistochemistry and using transgenic Tg(mpeg1.1:GFP) or Tg(mpeg1.1:mCherry) fish, we analyzed the distribution of microglia/immune cells in the whole brain. Specific regional differences were evidenced in terms of microglia/immune cell density and morphology (elongated, branched, highly branched, and amoeboid). Taking advantage of Tg(fli:GFP) and Tg(GFAP::GFP) enabling the detection of endothelial cells and neural stem cells (NSCs), we highlighted the association of elongated microglia/immune cells with blood vessels and rounded/amoeboid microglia with NSCs. Second, after telencephalic injury, we showed that L-plastin cells were still abundantly present at 5 days post-lesion (dpl) and were associated with regenerative neurogenesis. Finally, RNA-sequencing analysis from injured telencephalon (5 dpl) confirmed the upregulation of microglia/immune cell markers and highlighted a significant increase of genes involved in oxidative stress (nox2, nrf2a, and gsr). The analysis of antioxidant activities at 5 dpl also revealed an upregulation of superoxide dismutase and persistent H2 O2 generation in the injured telencephalon. Also, microglia/immune cells were shown to be a source of oxidative stress at 5 dpl. Overall, our data provide a better characterization of microglia/immune cell distribution in the healthy zebrafish brain, highlighting some evolutionarily conserved features with mammals. They also emphasize that 5 days after injury, microglia/immune cells are still activated and are associated to a persistent redox imbalance. Together, these data raise the question of the role of oxidative stress in regenerative neurogenesis in zebrafish.
Collapse
Affiliation(s)
- Sai Sandhya Narra
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI) Saint‐Denis de La Réunion France
| | - Philippe Rondeau
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI) Saint‐Denis de La Réunion France
| | - Danielle Fernezelian
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI) Saint‐Denis de La Réunion France
| | - Laura Gence
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI) Saint‐Denis de La Réunion France
| | - Batoul Ghaddar
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI) Saint‐Denis de La Réunion France
| | - Emmanuel Bourdon
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI) Saint‐Denis de La Réunion France
| | - Christian Lefebvre d'Hellencourt
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI) Saint‐Denis de La Réunion France
| | - Sepand Rastegar
- Institute of Biological and Chemical Systems‐Biological Information Processing (IBCS‐BIP), Karlsruhe Institute of Technology (KIT) Karlsruhe Germany
| | - Nicolas Diotel
- Université de La Réunion, INSERM, UMR 1188, Diabète athérothrombose Thérapies Réunion Océan Indien (DéTROI) Saint‐Denis de La Réunion France
| |
Collapse
|
28
|
Vidal-Itriago A, Radford RAW, Aramideh JA, Maurel C, Scherer NM, Don EK, Lee A, Chung RS, Graeber MB, Morsch M. Microglia morphophysiological diversity and its implications for the CNS. Front Immunol 2022; 13:997786. [PMID: 36341385 PMCID: PMC9627549 DOI: 10.3389/fimmu.2022.997786] [Citation(s) in RCA: 155] [Impact Index Per Article: 51.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/20/2022] [Indexed: 07/30/2023] Open
Abstract
Microglia are mononuclear phagocytes of mesodermal origin that migrate to the central nervous system (CNS) during the early stages of embryonic development. After colonizing the CNS, they proliferate and remain able to self-renew throughout life, maintaining the number of microglia around 5-12% of the cells in the CNS parenchyma. They are considered to play key roles in development, homeostasis and innate immunity of the CNS. Microglia are exceptionally diverse in their morphological characteristics, actively modifying the shape of their processes and soma in response to different stimuli. This broad morphological spectrum of microglia responses is considered to be closely correlated to their diverse range of functions in health and disease. However, the morphophysiological attributes of microglia, and the structural and functional features of microglia-neuron interactions, remain largely unknown. Here, we assess the current knowledge of the diverse microglial morphologies, with a focus on the correlation between microglial shape and function. We also outline some of the current challenges, opportunities, and future directions that will help us to tackle unanswered questions about microglia, and to continue unravelling the mysteries of microglia, in all its shapes.
Collapse
Affiliation(s)
- Andrés Vidal-Itriago
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Rowan A. W. Radford
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Jason A. Aramideh
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Cindy Maurel
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Natalie M. Scherer
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Emily K. Don
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Albert Lee
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Roger S. Chung
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Manuel B. Graeber
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Marco Morsch
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
29
|
Zhao F, He J, Tang J, Cui N, Shi Y, Li Z, Liu S, Wang Y, Ma M, Zhao C, Luo L, Li L. Brain milieu induces early microglial maturation through the BAX-Notch axis. Nat Commun 2022; 13:6117. [PMID: 36253375 PMCID: PMC9576735 DOI: 10.1038/s41467-022-33836-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 09/30/2022] [Indexed: 12/24/2022] Open
Abstract
Microglia are derived from primitive myeloid cells and gain their early identity in the embryonic brains. However, the mechanism by which the brain milieu confers microglial maturation signature remains elusive. Here, we demonstrate that the baxcq55 zebrafish and Baxtm1Sjk mouse embryos exhibit similarly defective early microglial maturation. BAX, a typical pro-apoptotic factor, is highly enriched in neuronal cells and regulates microglial maturation through both pro-apoptotic and non-apoptotic mechanisms. BAX regulates dlb via the CaMKII-CREB axis calcium-dependently in living neurons while ensuring the efficient Notch activation in the immigrated pre-microglia by apoptotic neurons. Notch signaling is conserved in supporting embryonic microglia maturation. Compromised microglial development occurred in the Cx3cr1Cre/+Rbpjfl/fl embryonic mice; however, microglia acquire their appropriate signature when incubated with DLL3 in vitro. Thus, our findings elucidate a BAX-CaMKII-CREB-Notch network triggered by the neuronal milieu in microglial development, which may provide innovative insights for targeting microglia in neuronal disorder treatment.
Collapse
Affiliation(s)
- Fangying Zhao
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, 400715, Chongqing, P.R. China
| | - Jiangyong He
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, 400715, Chongqing, P.R. China
- Research Center of Stem Cells and Ageing, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, 400714, Chongqing, P.R. China
| | - Jun Tang
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, 400715, Chongqing, P.R. China
| | - Nianfei Cui
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, 400715, Chongqing, P.R. China
| | - Yanyan Shi
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, 400715, Chongqing, P.R. China
| | - Zhifan Li
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, 400715, Chongqing, P.R. China
| | - Shengnan Liu
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, 400715, Chongqing, P.R. China
| | - Yazhou Wang
- Department of Neurobiology and Institute of Neurosciences, School of Basic Medicine, Fourth Military Medical University, 169 Chang Le Xi Road, 710032, Xi'an, Shaanxi, P.R. China
| | - Ming Ma
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, 400715, Chongqing, P.R. China
| | - Congjian Zhao
- Chongqing Engineering Research Center of Medical Electronics and Information Technology, School of Bioinformatics, Chongqing University of Posts and Telecommunications, 400065, Chongqing, P.R. China
| | - Lingfei Luo
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, 400715, Chongqing, P.R. China.
| | - Li Li
- Institute of Developmental Biology and Regenerative Medicine, Key Laboratory of Freshwater Fish Reproduction and Development, Ministry of Education, Southwest University, 400715, Chongqing, P.R. China.
- Research Center of Stem Cells and Ageing, Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, 400714, Chongqing, P.R. China.
| |
Collapse
|
30
|
Guedes JR, Ferreira PA, Costa JM, Cardoso AL, Peça J. Microglia-dependent remodeling of neuronal circuits. J Neurochem 2022; 163:74-93. [PMID: 35950924 PMCID: PMC9826178 DOI: 10.1111/jnc.15689] [Citation(s) in RCA: 28] [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: 03/23/2022] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 01/11/2023]
Abstract
Microglia are tissue-resident macrophages responsible for the surveillance, neuronal support, and immune defense of the brain parenchyma. Recently, the role played by microglia in the formation and function of neuronal circuits has garnered substantial attention. During development, microglia have been shown to engulf neuronal precursors and participate in pruning mechanisms while, in the mature brain, they influence synaptic signaling, provide trophic support and shape synaptic plasticity. Recently, studies have unveiled different microglial characteristics associated with specific brain regions. This emerging view suggests that the maturation and function of distinct neuronal circuits may be potentially associated with the molecular identity microglia adopts across the brain. Here, we review and summarize the known role of these cells in the thalamus, hippocampus, cortex, and cerebellum. We focus on in vivo studies to highlight the characteristics of microglia that may be important in the remodeling of these neuronal circuits and in relation to neurodevelopmental and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Joana R. Guedes
- CNC—Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- Institute of Interdisciplinary Research (IIIUC), University of CoimbraCoimbraPortugal
| | - Pedro A. Ferreira
- CNC—Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- Department of Life SciencesUniversity of CoimbraCoimbraPortugal
| | - Jéssica M. Costa
- CNC—Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- Institute of Interdisciplinary Research (IIIUC), University of CoimbraCoimbraPortugal
| | - Ana L. Cardoso
- CNC—Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- Institute of Interdisciplinary Research (IIIUC), University of CoimbraCoimbraPortugal
| | - João Peça
- CNC—Center for Neuroscience and Cell BiologyUniversity of CoimbraCoimbraPortugal
- Department of Life SciencesUniversity of CoimbraCoimbraPortugal
| |
Collapse
|
31
|
Mafba and Mafbb regulate microglial colonization of zebrafish brain via controlling chemotaxis receptor expression. Proc Natl Acad Sci U S A 2022; 119:e2203273119. [PMID: 36122226 PMCID: PMC9522419 DOI: 10.1073/pnas.2203273119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Microglia are a subpopulation of macrophages residing in the central nervous system (CNS). Because microglial precursors/peripheral macrophages are born in peripheral hematopoietic tissues, the establishment of a microglia pool in the CNS involves two processes: colonization, the homing of macrophages from peripheral tissues to the CNS, and maturation, the differentiation of brain-colonizing macrophages into microglia. This study aims to investigate the molecular mechanisms underlying microglial colonization during early development. Utilizing a zebrafish model system, we show that Mafba and Mafbb, two zebrafish orthologs of mammalian MAFB essential for macrophage differentiation and phagocytosis, regulate microglial colonization of the brain via modulating the lysoPS-Gpr34a signaling pathway during early embryogenesis. Our findings reveal a previously unappreciated genetic mechanism involved in microglial colonization of the brain. Microglia are the central nervous system (CNS)–resident macrophages involved in neural inflammation, neurogenesis, and neural activity regulation. Previous studies have shown that naturally occurring neuronal apoptosis plays a critical role in regulating microglial colonization of the brain in zebrafish. However, the molecular signaling cascades underlying neuronal apoptosis-mediated microglial colonization and the regulation of these cascades remain undefined. Here, we show that basic leucine zipper (b-Zip) transcription factors, Mafba and Mafbb, two zebrafish orthologs of mammalian MAFB, are key regulators in neuronal apoptosis-mediated microglial colonization of the brain in zebrafish. We document that the loss of Mafba and Mafbb function perturbs microglial colonization of the brain. We further demonstrate that Mafba and Mafbb act cell-autonomously and cooperatively to orchestrate microglial colonization, at least in part, by regulating the expression of G protein–coupled receptor 34a (Gpr34a), which directs peripheral macrophage recruitment into the brain through sensing the lysophosphatidylserine (lysoPS) released by the apoptotic neurons. Our study reveals that Mafba and Mafbb regulate neuronal apoptosis-mediated microglial colonization of the brain in zebrafish via the lysoPS-Gpr34a pathway.
Collapse
|
32
|
Wieghofer P, Engelbert M, Chui TYP, Rosen RB, Sakamoto T, Sebag J. Hyalocyte origin, structure, and imaging. EXPERT REVIEW OF OPHTHALMOLOGY 2022; 17:233-248. [PMID: 36632192 PMCID: PMC9831111 DOI: 10.1080/17469899.2022.2100762] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 07/08/2022] [Indexed: 01/14/2023]
Abstract
Introduction Hyalocytes have been recognized as resident tissue macrophages of the vitreous body since the mid-19th century. Despite this, knowledge about their origin, turnover, and dynamics is limited. Areas covered Historically, initial studies on the origin of hyalocytes used light and electron microscopy. Modern investigations across species including rodents and humans will be described. Novel imaging is now available to study human hyalocytes in vivo. The shared ontogeny with retinal microglia and their eventual interdependence as well as differences will be discussed. Expert opinion Owing to a common origin as myeloid cells, hyalocytes and retinal microglia have similarities, but hyalocytes appear to be distinct as resident macrophages of the vitreous body.
Collapse
Affiliation(s)
- Peter Wieghofer
- Cellular Neuroanatomy, Institute of Theoretical Medicine, Medical Faculty, University of Augsburg, Universitätsstraße 2, 86159 Augsburg, Germany
| | - Michael Engelbert
- Vitreous Retina Macula Consultants of New York, New York, NY 10022, USA
- LuEsther T. Mertz Retinal Research Center, Manhattan Eye, Ear and Throat Hospital, New York, NY 10065, USA
- Department of Ophthalmology, New York University School of Medicine, New York, NY 10016, USA
| | - Toco YP Chui
- Department of Ophthalmology, New York Eye and Ear Infirmary of Mount Sinai, New York, New York; Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Richard B Rosen
- Department of Ophthalmology, New York Eye and Ear Infirmary of Mount Sinai, New York, New York; Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Taiji Sakamoto
- Department of Ophthalmology, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, Japan
| | - J Sebag
- Doheny Eye Institute, UCLA, Los Angeles, CA, USA
- Clinical Ophthalmology, Stein Eye Institute, Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- VMR Institute for Vitreous Macula Retina, Huntington Beach, CA, USA
| |
Collapse
|
33
|
Iyer H, Shen K, Meireles AM, Talbot WS. A lysosomal regulatory circuit essential for the development and function of microglia. SCIENCE ADVANCES 2022; 8:eabp8321. [PMID: 36044568 PMCID: PMC9432849 DOI: 10.1126/sciadv.abp8321] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 07/18/2022] [Indexed: 05/17/2023]
Abstract
As the primary phagocytic cells of the central nervous system, microglia exquisitely regulate their lysosomal activity to facilitate brain development and homeostasis. However, mechanisms that coordinate lysosomal activity with microglia development, chemotaxis, and function remain unclear. Here, we show that embryonic macrophages require the lysosomal guanosine triphosphatase (GTPase) RagA and the GTPase-activating protein Folliculin to colonize the brain in zebrafish. We demonstrate that embryonic macrophages in rraga mutants show increased expression of lysosomal genes but display significant down-regulation of immune- and chemotaxis-related genes. Furthermore, we find that RagA and Folliculin repress the key lysosomal transcription factor Tfeb and its homologs Tfe3a and Tfe3b in the macrophage lineage. Using RNA sequencing, we establish that Tfeb and Tfe3 are required for activation of lysosomal target genes under conditions of stress but not for basal expression of lysosomal pathways. Collectively, our data define a lysosomal regulatory circuit essential for macrophage development and function in vivo.
Collapse
|
34
|
Wu L, Xue R, Chen J, Xu J. dock8 deficiency attenuates microglia colonization in early zebrafish larvae. Cell Death Dis 2022; 8:366. [PMID: 35977943 PMCID: PMC9386030 DOI: 10.1038/s41420-022-01155-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 07/30/2022] [Accepted: 08/02/2022] [Indexed: 11/09/2022]
Abstract
Microglia are tissue-resident macrophages that carry out immune functions in the brain. The deficiency or dysfunction of microglia has been implicated in many neurodegenerative disorders. DOCK8, a member of the DOCK family, functions as a guanine nucleotide exchange factor and plays key roles in immune regulation and neurological diseases. The functions of DOCK8 in microglia development are not fully understood. Here, we generated zebrafish dock8 mutants by CRISPR/Cas9 genome editing and showed that dock8 mutations attenuate microglia colonization in the zebrafish midbrain at early larvae stages. In vivo time-lapse imaging revealed that the motility of macrophages was reduced in the dock8 mutant. We further found that cdc42/cdc42l, which encode the small GTPase activated by Dock8, also regulate microglia colonization in zebrafish. Collectively, our study suggests that the Dock8-Cdc42 pathway is required for microglia colonization in zebrafish larvae.
Collapse
Affiliation(s)
- Linxiu Wu
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Rongtao Xue
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jiahao Chen
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China.
| | - Jin Xu
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China.
| |
Collapse
|
35
|
Chen XK, Kwan JSK, Wong GTC, Yi ZN, Ma ACH, Chang RCC. Leukocyte invasion of the brain after peripheral trauma in zebrafish (Danio rerio). EXPERIMENTAL & MOLECULAR MEDICINE 2022; 54:973-987. [PMID: 35831435 PMCID: PMC9356012 DOI: 10.1038/s12276-022-00801-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 11/29/2022]
Abstract
Despite well-known systemic immune reactions in peripheral trauma, little is known about their roles in posttraumatic neurological disorders, such as anxiety, sickness, and cognitive impairment. Leukocyte invasion of the brain, a common denominator of systemic inflammation, is involved in neurological disorders that occur in peripheral inflammatory diseases, whereas the influences of peripheral leukocytes on the brain after peripheral trauma remain largely unclear. In this study, we found that leukocytes, largely macrophages, transiently invaded the brain of zebrafish larvae after peripheral trauma through vasculature-independent migration, which was a part of the systemic inflammation and was mediated by interleukin-1b (il1b). Notably, myeloid cells in the brain that consist of microglia and invading macrophages were implicated in posttraumatic anxiety-like behaviors, such as hyperactivity (restlessness) and thigmotaxis (avoidance), while a reduction in systemic inflammation or myeloid cells can rescue these behaviors. In addition, invading leukocytes together with microglia were found to be responsible for the clearance of apoptotic cells in the brain; however, they also removed the nonapoptotic cells, which suggested that phagocytes have dual roles in the brain after peripheral trauma. More importantly, a category of conserved proteins between zebrafish and humans or rodents that has been featured in systemic inflammation and neurological disorders was determined in the zebrafish brain after peripheral trauma, which supported that zebrafish is a translational model of posttraumatic neurological disorders. These findings depicted leukocyte invasion of the brain during systemic inflammation after peripheral trauma and its influences on the brain through il1b-dependent mechanisms. Invasion of the brain by white blood cells followed tail amputation in zebrafish, the resulting systemic inflammation producing anxiety-like behaviors. Scientists have long recognised an association between systemic inflammation following peripheral traumatic injury such as limb loss and post-traumatic neurological disorders such as anxiety and depression. Raymond Chuen-Chung Chang at the University of Hong Kong, Alvin Chun-Hang Ma at Hong Kong Polytechnic University, China, and co-workers found that following trauma, white cells, mainly macrophages, flowed from neighboring tissues into the hindbrain, before spreading throughout the brain. This influx of white cells, mediated by the small signaling protein interleukin-1b, triggered anxiety-like behaviors such as hyperactivity and avoidance in the zebrafish. The researchers emphasize that the links between systemic inflammation following peripheral trauma and neurological responses require extensive further research.
Collapse
Affiliation(s)
- Xiang-Ke Chen
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | | | - Gordon Tin-Chun Wong
- Department of Anaesthesiology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Zhen-Ni Yi
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Alvin Chun-Hang Ma
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China.
| | - Raymond Chuen-Chung Chang
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China. .,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.
| |
Collapse
|
36
|
Green LA, O'Dea MR, Hoover CA, DeSantis DF, Smith CJ. The embryonic zebrafish brain is seeded by a lymphatic-dependent population of mrc1 + microglia precursors. Nat Neurosci 2022; 25:849-864. [PMID: 35710983 PMCID: PMC10680068 DOI: 10.1038/s41593-022-01091-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 05/06/2022] [Indexed: 02/02/2023]
Abstract
Microglia are the resident macrophages of the CNS that serve critical roles in brain construction. Although human brains contain microglia by 4 weeks gestation, an understanding of the earliest microglia that seed the brain during its development remains unresolved. Using time-lapse imaging in zebrafish, we discovered a mrc1a+ microglia precursor population that seeds the brain before traditionally described microglia. These early microglia precursors are dependent on lymphatic vasculature that surrounds the brain and are independent of pu1+ yolk sac-derived microglia. Single-cell RNA-sequencing datasets reveal Mrc1+ microglia in the embryonic brains of mice and humans. We then show in zebrafish that these early mrc1a+ microglia precursors preferentially expand during pathophysiological states in development. Taken together, our results identify a critical role of lymphatics in the microglia precursors that seed the early embryonic brain.
Collapse
Affiliation(s)
- Lauren A Green
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
- The Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, USA
| | - Michael R O'Dea
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Camden A Hoover
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
- The Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, USA
| | - Dana F DeSantis
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
- The Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, USA
| | - Cody J Smith
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA.
- The Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, USA.
| |
Collapse
|
37
|
Levraud JP, Rawls JF, Clatworthy AE. Using zebrafish to understand reciprocal interactions between the nervous and immune systems and the microbial world. J Neuroinflammation 2022; 19:170. [PMID: 35765004 PMCID: PMC9238045 DOI: 10.1186/s12974-022-02506-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 06/01/2022] [Indexed: 11/10/2022] Open
Abstract
Animals rely heavily on their nervous and immune systems to perceive and survive within their environment. Despite the traditional view of the brain as an immunologically privileged organ, these two systems interact with major consequences. Furthermore, microorganisms within their environment are major sources of stimuli and can establish relationships with animal hosts that range from pathogenic to mutualistic. Research from a variety of human and experimental animal systems are revealing that reciprocal interactions between microbiota and the nervous and immune systems contribute significantly to normal development, homeostasis, and disease. The zebrafish has emerged as an outstanding model within which to interrogate these interactions due to facile genetic and microbial manipulation and optical transparency facilitating in vivo imaging. This review summarizes recent studies that have used the zebrafish for analysis of bidirectional control between the immune and nervous systems, the nervous system and the microbiota, and the microbiota and immune system in zebrafish during development that promotes homeostasis between these systems. We also describe how the zebrafish have contributed to our understanding of the interconnections between these systems during infection in fish and how perturbations may result in pathology.
Collapse
Affiliation(s)
- Jean-Pierre Levraud
- Université Paris-Saclay, CNRS, Institut Pasteur, Université Paris-Cité, Institut des Neurosciences Paris-Saclay, 91400, Saclay, France.
| | - John F. Rawls
- grid.26009.3d0000 0004 1936 7961Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, 213 Research Drive, Durham, NC 27710 USA
| | - Anne E. Clatworthy
- grid.66859.340000 0004 0546 1623Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142 USA ,grid.32224.350000 0004 0386 9924Department of Molecular Biology and Center for Computational and Integrative Biology, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114 USA
| |
Collapse
|
38
|
Lucas CD, Medina CB, Bruton FA, Dorward DA, Raymond MH, Tufan T, Etchegaray JI, Barron B, Oremek ME, Arandjelovic S, Farber E, Onngut-Gumuscu S, Ke E, Whyte MKB, Rossi AG, Ravichandran KS. Pannexin 1 drives efficient epithelial repair after tissue injury. Sci Immunol 2022; 7:eabm4032. [PMID: 35559667 PMCID: PMC7612772 DOI: 10.1126/sciimmunol.abm4032] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Epithelial tissues such as lung and skin are exposed to the environment and therefore particularly vulnerable to damage during injury or infection. Rapid repair is therefore essential to restore function and organ homeostasis. Dysregulated epithelial tissue repair occurs in several human disease states, yet how individual cell types communicate and interact to coordinate tissue regeneration is incompletely understood. Here, we show that pannexin 1 (Panx1), a cell membrane channel activated by caspases in dying cells, drives efficient epithelial regeneration after tissue injury by regulating injury-induced epithelial proliferation. Lung airway epithelial injury promotes the Panx1-dependent release of factors including ATP, from dying epithelial cells, which regulates macrophage phenotype after injury. This process, in turn, induces a reparative response in tissue macrophages that includes the induction of the soluble mitogen amphiregulin, which promotes injury-induced epithelial proliferation. Analysis of regenerating lung epithelium identified Panx1-dependent induction of Nras and Bcas2, both of which positively promoted epithelial proliferation and tissue regeneration in vivo. We also established that this role of Panx1 in boosting epithelial repair after injury is conserved between mouse lung and zebrafish tailfin. These data identify a Panx1-mediated communication circuit between epithelial cells and macrophages as a key step in promoting epithelial regeneration after injury.
Collapse
Affiliation(s)
- Christopher D. Lucas
- Center for Cell Clearance, Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh BioQuarter, UK
- Institute for Regeneration and Repair, Edinburgh BioQuarter, UK
| | - Christopher B. Medina
- Center for Cell Clearance, Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Finnius A. Bruton
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh BioQuarter, UK
| | - David A. Dorward
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh BioQuarter, UK
| | - Michael H. Raymond
- Center for Cell Clearance, Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Turan Tufan
- Center for Cell Clearance, Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - J. Iker Etchegaray
- Center for Cell Clearance, Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Brady Barron
- Center for Cell Clearance, Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Magdalena E.M. Oremek
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh BioQuarter, UK
| | - Sanja Arandjelovic
- Center for Cell Clearance, Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Emily Farber
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Suna Onngut-Gumuscu
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Eugene Ke
- Center for Cell Clearance, Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Moira KB Whyte
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh BioQuarter, UK
| | - Adriano G. Rossi
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, Edinburgh BioQuarter, UK
| | - Kodi S. Ravichandran
- Center for Cell Clearance, Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
- Inflammation Research Centre, VIB, and the Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
39
|
Romero-Morales AI, Gama V. Revealing the Impact of Mitochondrial Fitness During Early Neural Development Using Human Brain Organoids. Front Mol Neurosci 2022; 15:840265. [PMID: 35571368 PMCID: PMC9102998 DOI: 10.3389/fnmol.2022.840265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Mitochondrial homeostasis -including function, morphology, and inter-organelle communication- provides guidance to the intrinsic developmental programs of corticogenesis, while also being responsive to environmental and intercellular signals. Two- and three-dimensional platforms have become useful tools to interrogate the capacity of cells to generate neuronal and glia progeny in a background of metabolic dysregulation, but the mechanistic underpinnings underlying the role of mitochondria during human neurogenesis remain unexplored. Here we provide a concise overview of cortical development and the use of pluripotent stem cell models that have contributed to our understanding of mitochondrial and metabolic regulation of early human brain development. We finally discuss the effects of mitochondrial fitness dysregulation seen under stress conditions such as metabolic dysregulation, absence of developmental apoptosis, and hypoxia; and the avenues of research that can be explored with the use of brain organoids.
Collapse
Affiliation(s)
| | - Vivian Gama
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, United States
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
40
|
Anderson SR, Roberts JM, Ghena N, Irvin EA, Schwakopf J, Cooperstein IB, Bosco A, Vetter ML. Neuronal apoptosis drives remodeling states of microglia and shifts in survival pathway dependence. eLife 2022; 11:e76564. [PMID: 35481836 PMCID: PMC9071266 DOI: 10.7554/elife.76564] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 04/27/2022] [Indexed: 01/13/2023] Open
Abstract
Microglia serve critical remodeling roles that shape the developing nervous system, responding to the changing neural environment with phagocytosis or soluble factor secretion. Recent single-cell sequencing (scRNAseq) studies have revealed the context-dependent diversity in microglial properties and gene expression, but the cues promoting this diversity are not well defined. Here, we ask how interactions with apoptotic neurons shape microglial state, including lysosomal and lipid metabolism gene expression and dependence on Colony-stimulating factor 1 receptor (CSF1R) for survival. Using early postnatal mouse retina, a CNS region undergoing significant developmental remodeling, we performed scRNAseq on microglia from mice that are wild-type, lack neuronal apoptosis (Bax KO), or are treated with CSF1R inhibitor (PLX3397). We find that interactions with apoptotic neurons drive multiple microglial remodeling states, subsets of which are resistant to CSF1R inhibition. We find that TAM receptor Mer and complement receptor 3 are required for clearance of apoptotic neurons, but that Mer does not drive expression of remodeling genes. We show TAM receptor Axl is negligible for phagocytosis or remodeling gene expression but is consequential for microglial survival in the absence of CSF1R signaling. Thus, interactions with apoptotic neurons shift microglia toward distinct remodeling states and through Axl, alter microglial dependence on survival pathway, CSF1R.
Collapse
Affiliation(s)
| | | | - Nathaniel Ghena
- Department of Neurobiology, University of UtahSalt Lake CityUnited States
- Interdepartmental Program in Neuroscience, University of UtahSalt Lake CityUnited States
| | - Emmalyn A Irvin
- Department of Neurobiology, University of UtahSalt Lake CityUnited States
| | - Joon Schwakopf
- Department of Neurobiology, University of UtahSalt Lake CityUnited States
| | | | - Alejandra Bosco
- Department of Neurobiology, University of UtahSalt Lake CityUnited States
| | - Monica L Vetter
- Department of Neurobiology, University of UtahSalt Lake CityUnited States
| |
Collapse
|
41
|
Akhmanova M, Emtenani S, Krueger D, Gyoergy A, Guarda M, Vlasov M, Vlasov F, Akopian A, Ratheesh A, De Renzis S, Siekhaus DE. Cell division in tissues enables macrophage infiltration. Science 2022; 376:394-396. [PMID: 35446632 DOI: 10.1101/2021.04.19.438995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Cells migrate through crowded microenvironments within tissues during normal development, immune response, and cancer metastasis. Although migration through pores and tracks in the extracellular matrix (ECM) has been well studied, little is known about cellular traversal into confining cell-dense tissues. We find that embryonic tissue invasion by Drosophila macrophages requires division of an epithelial ectodermal cell at the site of entry. Dividing ectodermal cells disassemble ECM attachment formed by integrin-mediated focal adhesions next to mesodermal cells, allowing macrophages to move their nuclei ahead and invade between two immediately adjacent tissues. Invasion efficiency depends on division frequency, but reduction of adhesion strength allows macrophage entry independently of division. This work demonstrates that tissue dynamics can regulate cellular infiltration.
Collapse
Affiliation(s)
- Maria Akhmanova
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Shamsi Emtenani
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Daniel Krueger
- Developmental Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Attila Gyoergy
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Mariana Guarda
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | | | - Fedor Vlasov
- Bundesgymnasium Klosterneuburg, Klosterneuburg, Austria
| | | | - Aparna Ratheesh
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | - Stefano De Renzis
- Developmental Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Daria E Siekhaus
- Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| |
Collapse
|
42
|
Thiel WA, Blume ZI, Mitchell DM. Compensatory engulfment and Müller glia reactivity in the absence of microglia. Glia 2022; 70:1402-1425. [PMID: 35451181 DOI: 10.1002/glia.24182] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/05/2022] [Accepted: 04/08/2022] [Indexed: 01/25/2023]
Abstract
Microglia are known for important phagocytic functions in the vertebrate retina. Reports also suggest that Müller glia have phagocytic capacity, though the relative levels and contexts in which this occurs remain to be thoroughly examined. Here, we investigate Müller glial engulfment of dying cells in the developing zebrafish retina in the presence and absence of microglia, using a genetic mutant in which microglia do not develop. We show that in normal conditions clearance of dying cells is dominated by microglia; however, Müller glia do have a limited clearance role. In retinas lacking intact microglial populations, we found a striking increase in the engulfment load assumed by the Müller glia, which displayed prominent cellular compartments containing apoptotic cells, several of which localized with the early phagosome/endosome marker Rab5. Consistent with increased engulfment, lysosomal staining was also increased in Müller glia in the absence of microglia. Increased engulfment load led to evidence of Müller glia reactivity including upregulation of gfap but did not trigger cell cycle re-entry by differentiated Müller glia. Our work provides important insight into the phagocytic capacity of Müller glia and the ability for compensatory functions and downstream effects. Therefore, effects of microglial deficiency or depletion on other glial cell types should be well-considered in experimental manipulations, in neurodegenerative disease, and in therapeutic approaches that target microglia. Our findings further justify future work to understand differential mechanisms and contexts of phagocytosis by glial cells in the central nervous system, and the significance of these mechanisms in health and disease.
Collapse
Affiliation(s)
- Whitney A Thiel
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, USA
| | - Zachary I Blume
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, USA
| | - Diana M Mitchell
- Department of Biological Sciences, University of Idaho, Moscow, Idaho, USA
| |
Collapse
|
43
|
Pharmacological assessment of zebrafish-based cardiotoxicity models. Biomed Pharmacother 2022; 148:112695. [DOI: 10.1016/j.biopha.2022.112695] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/14/2022] [Accepted: 02/02/2022] [Indexed: 01/03/2023] Open
|
44
|
Ahmed M, Kojima Y, Masai I. Strip1 regulates retinal ganglion cell survival by suppressing Jun-mediated apoptosis to promote retinal neural circuit formation. eLife 2022; 11:74650. [PMID: 35314028 PMCID: PMC8940179 DOI: 10.7554/elife.74650] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 02/04/2022] [Indexed: 12/13/2022] Open
Abstract
In the vertebrate retina, an interplay between retinal ganglion cells (RGCs), amacrine (AC), and bipolar (BP) cells establishes a synaptic layer called the inner plexiform layer (IPL). This circuit conveys signals from photoreceptors to visual centers in the brain. However, the molecular mechanisms involved in its development remain poorly understood. Striatin-interacting protein 1 (Strip1) is a core component of the striatin-interacting phosphatases and kinases (STRIPAK) complex, and it has shown emerging roles in embryonic morphogenesis. Here, we uncover the importance of Strip1 in inner retina development. Using zebrafish, we show that loss of Strip1 causes defects in IPL formation. In strip1 mutants, RGCs undergo dramatic cell death shortly after birth. AC and BP cells subsequently invade the degenerating RGC layer, leading to a disorganized IPL. Mechanistically, zebrafish Strip1 interacts with its STRIPAK partner, Striatin 3 (Strn3), and both show overlapping functions in RGC survival. Furthermore, loss of Strip1 or Strn3 leads to activation of the proapoptotic marker, Jun, within RGCs, and Jun knockdown rescues RGC survival in strip1 mutants. In addition to its function in RGC maintenance, Strip1 is required for RGC dendritic patterning, which likely contributes to proper IPL formation. Taken together, we propose that a series of Strip1-mediated regulatory events coordinates inner retinal circuit formation by maintaining RGCs during development, which ensures proper positioning and neurite patterning of inner retinal neurons. The back of the eye is lined with an intricate tissue known as the retina, which consists of carefully stacked neurons connecting to each other in well-defined ‘synaptic’ layers. Near the surface, photoreceptors cells detect changes in light levels, before passing this information through the inner plexiform layer to retinal ganglion cells (or RGCs) below. These neurons will then relay the visual signals to the brain. Despite the importance of this inner retinal circuit, little is known about how it is created as an organism develops. As a response, Ahmed et al. sought to identify which genes are essential to establish the inner retinal circuit, and how their absence affects retinal structure. To do this, they introduced random errors in the genetic code of zebrafish and visualised the resulting retinal circuits in these fast-growing, translucent fish. Initial screening studies found fish with mutations in a gene encoding a protein called Strip1 had irregular layering of the inner retina. Further imaging experiments to pinpoint the individual neurons affected showed that in zebrafish without Strip1, RGCs died in the first few days of development. Consequently, other neurons moved into the RGC layer to replace the lost cells, leading to layering defects. Ahmed et al. concluded that Strip1 promotes RGC survival and thereby coordinates proper positioning of neurons in the inner retina. In summary, these findings help to understand how the inner retina is wired; they could also shed light on the way other layered structures are established in the nervous system. Moreover, this study paves the way for future research investigating Strip1 as a potential therapeutic target to slow down the death of RGCs in conditions such as glaucoma.
Collapse
Affiliation(s)
- Mai Ahmed
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University
| | - Yutaka Kojima
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University
| | - Ichiro Masai
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University
| |
Collapse
|
45
|
Liu KE, Raymond MH, Ravichandran KS, Kucenas S. Clearing Your Mind: Mechanisms of Debris Clearance After Cell Death During Neural Development. Annu Rev Neurosci 2022; 45:177-198. [PMID: 35226828 PMCID: PMC10157384 DOI: 10.1146/annurev-neuro-110920-022431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neurodevelopment and efferocytosis have fascinated scientists for decades. How an organism builds a nervous system that is precisely tuned for efficient behaviors and survival and how it simultaneously manages constant somatic cell turnover are complex questions that have resulted in distinct fields of study. Although neurodevelopment requires the overproduction of cells that are subsequently pruned back, very few studies marry these fields to elucidate the cellular and molecular mechanisms that drive nervous system development through the lens of cell clearance. In this review, we discuss these fields to highlight exciting areas of future synergy. We first review neurodevelopment from the perspective of overproduction and subsequent refinement and then discuss who clears this developmental debris and the mechanisms that control these events. We then end with how a more deliberate merger of neurodevelopment and efferocytosis could reframe our understanding of homeostasis and disease and discuss areas of future study. Expected final online publication date for the Annual Review of Neuroscience, Volume 45 is July 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Kendra E Liu
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA; .,Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia, USA
| | - Michael H Raymond
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA; .,Center for Clearance, University of Virginia, Charlottesville, Virginia, USA
| | - Kodi S Ravichandran
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA; .,Center for Clearance, University of Virginia, Charlottesville, Virginia, USA.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia, USA.,VIB-UGent Center for Inflammation Research and the Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Sarah Kucenas
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia, USA; .,Program in Fundamental Neuroscience, University of Virginia, Charlottesville, Virginia, USA.,Department of Biology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
46
|
Cuadros MA, Sepulveda MR, Martin-Oliva D, Marín-Teva JL, Neubrand VE. Microglia and Microglia-Like Cells: Similar but Different. Front Cell Neurosci 2022; 16:816439. [PMID: 35197828 PMCID: PMC8859783 DOI: 10.3389/fncel.2022.816439] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
Microglia are the tissue-resident macrophages of the central nervous parenchyma. In mammals, microglia are thought to originate from yolk sac precursors and posteriorly maintained through the entire life of the organism. However, the contribution of microglial cells from other sources should also be considered. In addition to “true” or “bona-fide” microglia, which are of embryonic origin, the so-called “microglia-like cells” are hematopoietic cells of bone marrow origin that can engraft the mature brain mainly under pathological conditions. These cells implement great parts of the microglial immune phenotype, but they do not completely adopt the “true microglia” features. Because of their pronounced similarity, true microglia and microglia-like cells are usually considered together as one population. In this review, we discuss the origin and development of these two distinct cell types and their differences. We will also review the factors determining the appearance and presence of microglia-like cells, which can vary among species. This knowledge might contribute to the development of therapeutic strategies aiming at microglial cells for the treatment of diseases in which they are involved, for example neurodegenerative disorders like Alzheimer’s and Parkinson’s diseases.
Collapse
Affiliation(s)
- Miguel A Cuadros
- Department of Cell Biology, Faculty of Science, University of Granada, Granada, Spain
| | - M Rosario Sepulveda
- Department of Cell Biology, Faculty of Science, University of Granada, Granada, Spain
| | - David Martin-Oliva
- Department of Cell Biology, Faculty of Science, University of Granada, Granada, Spain
| | - José L Marín-Teva
- Department of Cell Biology, Faculty of Science, University of Granada, Granada, Spain
| | - Veronika E Neubrand
- Department of Cell Biology, Faculty of Science, University of Granada, Granada, Spain
| |
Collapse
|
47
|
Liu J, Guo Y, Zhang C, Zeng Y, Luo Y, Wang G. Clearance Systems in the Brain, From Structure to Function. Front Cell Neurosci 2022; 15:729706. [PMID: 35173581 PMCID: PMC8841422 DOI: 10.3389/fncel.2021.729706] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 12/17/2021] [Indexed: 12/13/2022] Open
Abstract
As the most metabolically active organ in the body, there is a recognized need for pathways that remove waste proteins and neurotoxins from the brain. Previous research has indicated potential associations between the clearance system in the brain and the pathological conditions of the central nervous system (CNS), due to its importance, which has attracted considerable attention recently. In the last decade, studies of the clearance system have been restricted to the glymphatic system. However, removal of toxic and catabolic waste by-products cannot be completed independently by the glymphatic system, while no known research or article has focused on a comprehensive overview of the structure and function of the clearance system. This thesis addresses a neglected aspect of linkage between the structural composition and main components as well as the role of neural cells throughout the clearance system, which found evidence that the components of CNS including the glymphatic system and the meningeal lymphatic system interact with a neural cell, such as astrocytes and microglia, to carry out vital clearance functions. As a result of this evidence that can contribute to a better understanding of the clearance system, suggestions were identified for further clinical intervention development of severe conditions caused by the accumulation of metabolic waste products and neurotoxins in the brain, such as Alzheimer’s disease (AD) and Parkinson’s disease (PD).
Collapse
Affiliation(s)
- Jiachen Liu
- Xiangya Medical College of Central South University, Changsha, China
| | - Yunzhi Guo
- Xiangya Medical College of Central South University, Changsha, China
| | - Chengyue Zhang
- Xiangya Medical College of Central South University, Changsha, China
| | - Yang Zeng
- Xiangya Medical College of Central South University, Changsha, China
| | - Yongqi Luo
- Xiangya Medical College of Central South University, Changsha, China
| | - Gaiqing Wang
- Shanxi Medical University, Taiyuan, China
- Department of Neurology, Affiliated Sanya Central Hospital of Hainan Medical University, Sanya, China
- *Correspondence: Gaiqing Wang, ,
| |
Collapse
|
48
|
Möller K, Brambach M, Villani A, Gallo E, Gilmour D, Peri F. A role for the centrosome in regulating the rate of neuronal efferocytosis by microglia in vivo. eLife 2022; 11:82094. [PMID: 36398880 PMCID: PMC9674339 DOI: 10.7554/elife.82094] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/25/2022] [Indexed: 11/19/2022] Open
Abstract
During brain development, many newborn neurons undergo apoptosis and are engulfed by microglia, the tissue-resident phagocytes of the brain, in a process known as efferocytosis. A hallmark of microglia is their highly branched morphology characterized by the presence of numerous dynamic extensions that these cells use for scanning the brain parenchyma and engulfing unwanted material. The mechanisms driving branch formation and apoptotic cell engulfment in microglia are unclear. By taking a live-imaging approach in zebrafish, we show that while microglia generate multiple microtubule-based branches, they only successfully engulf one apoptotic neuron at a time. Further investigation into the mechanism underlying this sequential engulfment revealed that targeted migration of the centrosome into one branch is predictive of phagosome formation and polarized vesicular trafficking. Moreover, experimentally doubling centrosomal numbers in microglia increases the rate of engulfment and even allows microglia to remove two neurons simultaneously, providing direct supporting evidence for a model where centrosomal migration is a rate-limiting step in branch-mediated efferocytosis. Conversely, light-mediated depolymerization of microtubules causes microglia to lose their typical branched morphology and switch to an alternative mode of engulfment, characterized by directed migration towards target neurons, revealing unexpected plasticity in their phagocytic ability. Finally, building on work focusing on the establishment of the immunological synapse, we identified a conserved signalling pathway underlying centrosomal movement in engulfing microglia.
Collapse
Affiliation(s)
- Katrin Möller
- Department of Molecular Life Sciences, University of ZürichZürichSwitzerland
| | - Max Brambach
- Department of Molecular Life Sciences, University of ZürichZürichSwitzerland
| | - Ambra Villani
- Department of Molecular Life Sciences, University of ZürichZürichSwitzerland
| | - Elisa Gallo
- Department of Molecular Life Sciences, University of ZürichZürichSwitzerland
| | - Darren Gilmour
- Department of Molecular Life Sciences, University of ZürichZürichSwitzerland
| | - Francesca Peri
- Department of Molecular Life Sciences, University of ZürichZürichSwitzerland
| |
Collapse
|
49
|
Thiel W, Esposito EJ, Findley AP, Blume ZI, Mitchell DM. Modulation of retinoid-X-receptors differentially regulates expression of apolipoprotein genes apoc1 and apoeb by zebrafish microglia. Biol Open 2021; 11:273656. [PMID: 34878094 PMCID: PMC8822359 DOI: 10.1242/bio.058990] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 11/30/2021] [Indexed: 11/20/2022] Open
Abstract
Transcriptome analyses performed in both human and zebrafish indicate strong expression of Apoe and Apoc1 by microglia. Apoe expression by microglia is well appreciated, but Apoc1 expression has not been well-examined. PPAR/RXR and LXR/RXR receptors appear to regulate expression of the apolipoprotein gene cluster in macrophages, but a similar role in microglia in vivo has not been studied. Here, we characterized microglial expression of apoc1 in the zebrafish central nervous system (CNS) in situ and demonstrate that in the CNS, apoc1 expression is unique to microglia. We then examined the effects of PPAR/RXR and LXR/RXR modulation on microglial expression of apoc1 and apoeb during early CNS development using a pharmacological approach. Changes in apoc1 and apoeb transcripts in response to pharmacological modulation were quantified by RT-qPCR in whole heads, and in individual microglia using hybridization chain reaction (HCR) in situ hybridization. We found that expression of apoc1 and apoeb by microglia were differentially regulated by LXR/RXR and PPAR/RXR modulating compounds, respectively, during development. Our results also suggest RXR receptors could be involved in endogenous induction of apoc1 expression by microglia. Collectively, our work supports the use of zebrafish to better understand regulation and function of these apolipoproteins in the CNS. Summary: Here we investigate expression of two apolipoprotein genes by microglia in the zebrafish model during normal development, and in contexts of pharmacological manipulations that target candidate regulatory receptors.
Collapse
Affiliation(s)
- Whitney Thiel
- Biological Sciences, University of Idaho, Moscow, ID 83844, Russia
| | - Emma J Esposito
- Biological Sciences, University of Idaho, Moscow, ID 83844, Russia
| | - Anna P Findley
- Biological Sciences, University of Idaho, Moscow, ID 83844, Russia
| | - Zachary I Blume
- Biological Sciences, University of Idaho, Moscow, ID 83844, Russia
| | - Diana M Mitchell
- Biological Sciences, University of Idaho, Moscow, ID 83844, Russia
| |
Collapse
|
50
|
Ranawat N, Masai I. Mechanisms underlying microglial colonization of developing neural retina in zebrafish. eLife 2021; 10:70550. [PMID: 34872632 PMCID: PMC8651297 DOI: 10.7554/elife.70550] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 11/02/2021] [Indexed: 12/13/2022] Open
Abstract
Microglia are brain-resident macrophages that function as the first line of defense in brain. Embryonic microglial precursors originate in peripheral mesoderm and migrate into the brain during development. However, the mechanism by which they colonize the brain is incompletely understood. The retina is one of the first brain regions to accommodate microglia. In zebrafish, embryonic microglial precursors use intraocular hyaloid blood vessels as a pathway to migrate into the optic cup via the choroid fissure. Once retinal progenitor cells exit the cell cycle, microglial precursors associated with hyaloid blood vessels start to infiltrate the retina preferentially through neurogenic regions, suggesting that colonization of retinal tissue depends upon the neurogenic state. Along with blood vessels and retinal neurogenesis, IL34 also participates in microglial precursor colonization of the retina. Altogether, CSF receptor signaling, blood vessels, and neuronal differentiation function as cues to create an essential path for microglial migration into developing retina. The immune system is comprised of many different cells which protect our bodies from infection and other illnesses. The brain contains its own population of immune cells called microglia. Unlike neurons, these cells form outside the brain during development. They then travel to the brain and colonize specific regions like the retina, the light-sensing part of the eye in vertebrates. It is poorly understood how newly formed microglia migrate to the retina and whether their entry depends on the developmental state of nerve cells (also known as neurons) in this region. To help answer these questions, Ranawat and Masai attached fluorescent labels that can be seen under a microscope to microglia in the embryos of zebrafish. Developing zebrafish are transparent, making it easy to trace the fluorescent microglia as they travel to the retina and insert themselves among its neurons. Ranawat and Masai found that blood vessels around the retina act as a pathway that microglia move along. Once they reach the retina, the microglia remain attached and only enter the retina at sites where brain cells are starting to mature in to adult neurons. Further experiments showed that microglia fail to infiltrate and colonize the retina when blood vessels are damaged or neuron maturation is blocked. These findings reveal some of the key elements that guide microglia to the retina during development. However, further work is needed to establish the molecular and biochemical processes that allow microglia to attach to blood vessels and detect when cells in the retina are starting to mature.
Collapse
Affiliation(s)
- Nishtha Ranawat
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Japan
| | - Ichiro Masai
- Developmental Neurobiology Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Japan
| |
Collapse
|