1
|
Alassaf M, Madan A, Ranganathan S, Marschall S, Wong JJ, Goldberg ZH, Brent AE, Rajan A. Adipocyte metabolic state regulates glial phagocytic function. Cell Rep 2025; 44:115704. [PMID: 40372917 DOI: 10.1016/j.celrep.2025.115704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 03/17/2025] [Accepted: 04/25/2025] [Indexed: 05/17/2025] Open
Abstract
Excess dietary sugar profoundly impacts organismal metabolism and health, yet it remains unclear how metabolic adaptations in adipose tissue influence other organs, including the brain. Here, we show that a high-sugar diet (HSD) in Drosophila reduces adipocyte glycolysis and mitochondrial pyruvate uptake, shifting metabolism toward fatty acid oxidation and ketogenesis. These metabolic changes trigger mitochondrial oxidation and elevate antioxidant responses. Adipocyte-specific manipulations of glycolysis, lipid metabolism, or mitochondrial dynamics non-autonomously modulate Draper expression in brain ensheathing glia, key cells responsible for neuronal debris clearance. Adipocyte-derived ApoB-containing lipoproteins maintain basal Draper levels in glia via LpR1, critical for effective glial phagocytic activity. Accordingly, reducing ApoB or LpR1 impairs glial clearance of degenerating neuronal debris after injury. Collectively, our findings demonstrate that dietary sugar-induced shifts in adipocyte metabolism substantially influence brain health by modulating glial phagocytosis, identifying adipocyte-derived ApoB lipoproteins as essential systemic mediators linking metabolic state with neuroprotective functions.
Collapse
Affiliation(s)
- Mroj Alassaf
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Aditi Madan
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Sunidhi Ranganathan
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Shannon Marschall
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Jordan J Wong
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Zachary H Goldberg
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Ava E Brent
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Akhila Rajan
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA.
| |
Collapse
|
2
|
Theisen EK, Rivas-Serna IM, Lee RJ, Jay TR, Kunduri G, Nguyen TT, Mazurak V, Clandinin MT, Clandinin TR, Vaughen JP. Glia phagocytose neuronal sphingolipids to infiltrate developing synapses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.14.648777. [PMID: 40313927 PMCID: PMC12045345 DOI: 10.1101/2025.04.14.648777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
The complex morphologies of mature neurons and glia emerge through profound rearrangements of cell membranes during development. Despite being integral components of these membranes, it is unclear whether lipids might actively sculpt these morphogenic processes. By analyzing lipid levels in the developing fruit fly brain, we discover dramatic increases in specific sphingolipids coinciding with neural circuit establishment. Disrupting this sphingolipid bolus via genetic perturbations of sphingolipid biosynthesis and catabolism leads to impaired glial autophagy. Remarkably, glia can obtain sphingolipid precursors needed for autophagy by phagocytosing neurons. These precursors are then converted into specific long-chain ceramide phosphoethanolamines (CPEs), invertebrate analogs of sphingomyelin. These lipids are essential for glia to arborize and infiltrate the brain, a critical step in circuit maturation that when disrupted leads to reduced synapse numbers. Taken together, our results demonstrate how spatiotemporal tuning of sphingolipid metabolism during development plays an instructive role in programming brain architecture. Highlights Brain sphingolipids (SLs) remodel to very long-chain species during circuit maturation Glial autophagy requires de novo SL biosynthesis coordinated across neurons and glia Glia evade a biosynthetic blockade by phagolysosomal salvage of neuronal SLsCeramide Phosphoethanolamine is critical for glial infiltration and synapse density.
Collapse
|
3
|
Peng L, Wang Z, Sun Q, Cao C, Li L, Zhang F, Chen G, Bu J, Wang Z, Li H. ATP11C as a key regulator of neuronal loss following intracerebral hemorrhage in mice. Biochem Biophys Res Commun 2025; 756:151531. [PMID: 40086357 DOI: 10.1016/j.bbrc.2025.151531] [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: 12/17/2024] [Revised: 02/14/2025] [Accepted: 02/22/2025] [Indexed: 03/16/2025]
Abstract
Intracerebral hemorrhage (ICH) is a severe form of stroke with high rates of mortality and morbidity. Neuronal loss following ICH is a critical factor influencing patient outcomes. Emerging evidence suggests that microglial phagocytic activity is enhanced after ICH, yet its role in neuronal loss remains unclear. In this study, we observed microglia engulfing viable neurons, characterized by high NeuN signals or intact nuclear morphology, in the perihematomal region of a murine autologous blood injection ICH model. This phenomenon was also observed in an in vitro ICH model, where microglia engulfed neurons in a neuron-microglia co-culture system treated with oxyhemoglobin. Furthermore, we found that oxyhemoglobin exposure induced phosphatidylserine (PS) externalization in non-apoptotic (PI-) neurons and led to a downregulation of the PS flippase ATP11C. Notably, lentivirus-mediated overexpression of ATP11C in neurons specifically prevented the ICH-induced decline in ATP11C levels and inhibited microglial engulfment of neurons. Furthermore, ATP11C overexpression significantly improved neurological outcomes in the mouse ICH model. These findings offer new insights into the mechanisms of neuronal loss after ICH, positioning ATP11C as a promising therapeutic target for attenuating brain injury by regulating PS externalization in neurons.
Collapse
Affiliation(s)
- Lu Peng
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Zilan Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Qing Sun
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Chang Cao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Lianxin Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Feiyang Zhang
- Suzhou Medical College, Soochow University, Suzhou, 215006, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou, 215006, China
| | - Jiyuan Bu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou, 215006, China.
| | - Zhong Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou, 215006, China.
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China; Institute of Stroke Research, Soochow University, 188 Shizi Street, Suzhou, 215006, China.
| |
Collapse
|
4
|
Pampuscenko K, Jankeviciute S, Morkuniene R, Sulskis D, Smirnovas V, Brown GC, Borutaite V. S100A9 protein activates microglia and stimulates phagocytosis, resulting in synaptic and neuronal loss. Neurobiol Dis 2025; 206:106817. [PMID: 39884585 DOI: 10.1016/j.nbd.2025.106817] [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: 08/22/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025] Open
Abstract
S100 calcium-binding protein A9 (S100A9, also known as calgranulin B) is expressed and secreted by myeloid cells under inflammatory conditions, and S100A9 can amplify inflammation. There is a large increase in S100A9 expression in the brains of patients with neurodegenerative diseases, such as Alzheimer's disease, and S100A9 has been suggested to contribute to neurodegeneration, but the mechanisms are unclear. Here we investigated the effects of extracellular recombinant S100A9 protein on microglia, neurons and synapses in primary rat brain neuronal-glial cell cultures. Incubation of cell cultures with 250-500 nM S100A9 caused neuronal loss without signs of apoptosis or necrosis, but accompanied by exposure of the "eat-me" signal - phosphatidylserine on neurons. S100A9 caused activation of microglial inflammation as evidenced by an increase in the microglial number, morphological changes, release of pro-inflammatory cytokines, and increased phagocytic activity. At lower concentrations, 10-100 nM S100A9 induced synaptic loss in the cultures. Depletion of microglia from the cultures prevented S100A9-induced neuronal and synaptic loss, indicating that neuronal and synaptic loss was mediated by microglia. These results suggest that extracellular S100A9 may contribute to neurodegeneration by activating microglial inflammation and phagocytosis, resulting in loss of synapses and neurons. This further suggests the possibility that neurodegeneration may be reduced by targeting S100A9 or microglia.
Collapse
Affiliation(s)
- Katryna Pampuscenko
- Neuroscience Institute, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania.
| | - Silvija Jankeviciute
- Neuroscience Institute, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania.
| | - Ramune Morkuniene
- Neuroscience Institute, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania.
| | - Darius Sulskis
- Life Sciences Center, Institute of Biotechnology, Vilnius University, LT-10257 Vilnius, Lithuania.
| | - Vytautas Smirnovas
- Life Sciences Center, Institute of Biotechnology, Vilnius University, LT-10257 Vilnius, Lithuania.
| | - Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1QW, United Kingdom.
| | - Vilmante Borutaite
- Neuroscience Institute, Lithuanian University of Health Sciences, LT-50161 Kaunas, Lithuania.
| |
Collapse
|
5
|
Nelson N, Miller V, Broadie K. Neuron-to-glia and glia-to-glia signaling directs critical period experience-dependent synapse pruning. Front Cell Dev Biol 2025; 13:1540052. [PMID: 40040788 PMCID: PMC11876149 DOI: 10.3389/fcell.2025.1540052] [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: 12/05/2024] [Accepted: 01/31/2025] [Indexed: 03/06/2025] Open
Abstract
Experience-dependent glial synapse pruning plays a pivotal role in sculpting brain circuit connectivity during early-life critical periods of development. Recent advances suggest a layered cascade of intercellular communication between neurons and glial phagocytes orchestrates this precise, targeted synapse elimination. We focus here on studies from the powerful Drosophila forward genetic model, with reference to complementary findings from mouse work. We present both neuron-to-glia and glia-to-glia intercellular signaling pathways directing experience-dependent glial synapse pruning. We discuss a putative hierarchy of secreted long-distance cues and cell surface short-distance cues that act to sequentially orchestrate glia activation, infiltration, target recognition, engulfment, and then phagocytosis for synapse pruning. Ligand-receptor partners mediating these stages in different contexts are discussed from recent Drosophila and mouse studies. Signaling cues include phospholipids, small neurotransmitters, insulin-like peptides, and proteins. Conserved receptors for these ligands are discussed, together with mechanisms where the receptor identity remains unknown. Potential mechanisms are proposed for the tight temporal-restriction of heightened experience-dependent glial synapse elimination during early-life critical periods, as well as potential means to re-open such plasticity at maturity.
Collapse
Affiliation(s)
- Nichalas Nelson
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN, United States
| | - Vanessa Miller
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN, United States
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN, United States
- Department of Cell and Developmental Biology, Vanderbilt University and Medical Center, Nashville, TN, United States
- Department of Pharmacology, Vanderbilt University and Medical Center, Nashville, TN, United States
- Kennedy Center for Research on Human Development, Vanderbilt University and Medical Center, Nashville, TN, United States
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, TN, United States
| |
Collapse
|
6
|
Silvera MC, Cantera R, Ferreiro MJ. Absence of pretaporter restrains features of the parkin phenotype in Drosophila. Exp Neurol 2024; 383:114997. [PMID: 39393670 DOI: 10.1016/j.expneurol.2024.114997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 09/13/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND Scientific research based on model organisms can help to understand the biology of Parkinson's Disease, the second most prevalent neurodegenerative disease. Drosophila melanogaster mutant for the gene parkin, homologous to human's PARK2, exhibit well-characterized phenotypes including loss of dopaminergic neurons, lower survival and motor defects. Through the transcriptomic analysis of an exceptional case of reversible neurodegeneration in Drosophila, our group identified that the gene pretaporter, homologous to TXNDC5 of humans, was downregulated in the reversal phase. Here, we explore the hypothesis that the lack of expression of pretaporter will restrain phenotypes observed in Drosophila parkin mutants. METHODS After establishing by immunochemistry that Pretaporter is expressed in PPL1 dopaminergic neurons, we constructed pretaporter-parkin double mutants flies to investigate the hypothesis through immunohistochemistry, survival and climbing assays. CONCLUSIONS It was found that the loss-of-function mutation in pretaporter significatively restrains the phenotype caused by the loss-of-function mutation in parkin in several key aspects: it abolished the loss of PPL1 neurons normally seen in parkin mutant flies, promoted their survival in both sexes and reduced the decay in motor ability in parkin female flies. We propose that the absence of Pretaporter in parkin mutant flies prevents the death of dopaminergic neurons by rendering them resistant to Draper-mediated-phagocytosis.
Collapse
Affiliation(s)
- María Constanza Silvera
- Departamento de Biología del Neurodesarrollo, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Avenida Italia 3318, 11600 Montevideo, Uruguay; Departamento de Neurofisiología Celular y Molecular, IIBCE, Avenida Italia 3318, 11600 Montevideo, Uruguay
| | - Rafael Cantera
- Departamento de Biología del Neurodesarrollo, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Avenida Italia 3318, 11600 Montevideo, Uruguay
| | - María José Ferreiro
- Departamento de Neurofarmacología Experimental, IIBCE, Avenida Italia 3318, 11600 Montevideo, Uruguay; Departamento de Biología del Neurodesarrollo, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Avenida Italia 3318, 11600 Montevideo, Uruguay.
| |
Collapse
|
7
|
Alassaf M, Rajan A. Adipocyte metabolic state regulates glial phagocytic function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.614765. [PMID: 39386724 PMCID: PMC11463506 DOI: 10.1101/2024.09.24.614765] [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: 10/12/2024]
Abstract
Obesity and type 2 diabetes are well-established risk factors for neurodegenerative disorders1-4, yet the underlying mechanisms remain poorly understood. The adipocyte-brain axis is crucial for brain function, as adipocytes secrete signaling molecules, including lipids and adipokines, that impinge on neural circuits to regulate feeding and energy expenditure5. Disruptions in the adipocyte-brain axis are associated with neurodegenerative conditions6, but the causal links are not fully understood. Neural debris accumulates with age and injury, and glial phagocytic function is crucial for clearing this debris and maintaining a healthy brain microenvironment7-9. Using adult Drosophila, we investigate how adipocyte metabolism influences glial phagocytic activity in the brain. We demonstrate that a prolonged obesogenic diet increases adipocyte fatty acid oxidation and ketogenesis. Genetic manipulations that mimic obesogenic diet-induced changes in adipocyte lipid and mitochondrial metabolism unexpectedly reduce the expression of the phagocytic receptor Draper in Drosophila microglia-like cells in the brain. We identify Apolpp-the Drosophila equivalent of human apolipoprotein B (ApoB)-as a critical adipocyte-derived signal that regulates glial phagocytosis. Additionally, we show that Lipoprotein Receptor 1 (LpR1), the LDL receptor on phagocytic glia, is required for glial capacity to clear injury-induced neuronal debris. Our findings establish that adipocyte-brain lipoprotein signaling regulates glial phagocytic function, revealing a novel pathway that links adipocyte metabolic disorders with neurodegeneration.
Collapse
Affiliation(s)
- Mroj Alassaf
- Basic Sciences Division, Fred Hutch, Seattle, WA-98109. The USA
| | - Akhila Rajan
- Basic Sciences Division, Fred Hutch, Seattle, WA-98109. The USA
| |
Collapse
|
8
|
Beachum AN, Salazar G, Nachbar A, Krause K, Klose H, Meyer K, Maserejian A, Ross G, Boyd H, Weigel T, Ambaye L, Miller H, Coutinho-Budd J. Glia multitask to compensate for neighboring glial cell dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.06.611719. [PMID: 39314422 PMCID: PMC11418964 DOI: 10.1101/2024.09.06.611719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
As glia mature, they undergo glial tiling to abut one another without invading each other's boundaries. Upon the loss of the secreted neurotrophin Spätzle3 (Spz3), Drosophila cortex glia transform morphologically and lose their intricate interactions with neurons and surrounding glial subtypes. Here, we reveal that all neighboring glial cell types (astrocytes, ensheathing glia, and subperineurial glia) react by extending processes into the previous cortex glial territory to compensate for lost cortex glial function and reduce the buildup of neuronal debris. However, the loss of Spz3 alone is not sufficient for glia to cross their natural borders, as blocking CNS growth via nutrient-restriction blocks the aberrant infiltration induced by the loss of Spz3. Surprisingly, even when these neighboring glia divert their cellular resources beyond their typical borders to take on new compensatory roles, they are able to multitask to continue to preserve their own normal functions to maintain CNS homeostasis.
Collapse
Affiliation(s)
- Allison N. Beachum
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | - Gabriela Salazar
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | - Amelia Nachbar
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | - Kevin Krause
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | - Hannah Klose
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | - Kate Meyer
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | | | - Grace Ross
- Department of Biology, University of Vermont, Burlington, VT 05405
| | - Hannah Boyd
- Department of Biology, University of Vermont, Burlington, VT 05405
| | - Thaddeus Weigel
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | - Lydia Ambaye
- Department of Biology, University of Vermont, Burlington, VT 05405
| | - Hayes Miller
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| | - Jaeda Coutinho-Budd
- Department of Neuroscience, University of Virginia, Charlottesville, VA 22903
| |
Collapse
|
9
|
Abstract
Cells can die as a consequence of being phagocytosed by other cells - a form of cell death that has been called phagotrophy, cell cannibalism, programmed cell removal and primary phagocytosis. However, these are all different manifestations of cell death by phagocytosis (termed 'phagoptosis' for short). The engulfed cells die as a result of cytotoxic oxidants, peptides and degradative enzymes within acidic phagolysosomes. Cell death by phagocytosis was discovered by Metchnikov in the 1880s, but was neglected until recently. It is now known to contribute to developmental cell death in nematodes, Drosophila and mammals, and is central to innate and adaptive immunity against pathogens. Cell death by phagocytosis mediates physiological turnover of erythrocytes and other leucocytes, making it the most abundant form of cell death in the mammalian body. Immunity against cancer is also partly mediated by macrophage phagocytosis of cancer cells, but cancer cells can also phagocytose host cells and other cancer cells in order to survive. Recent evidence indicates neurodegeneration and other neuropathologies can be mediated by microglial phagocytosis of stressed neurons. Thus, despite cell death by phagocytosis being poorly recognized, it is one of the oldest, commonest and most important forms of cell death.
Collapse
Affiliation(s)
- Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge, UK.
| |
Collapse
|
10
|
Alassaf M, Rajan A. Diet-induced glial insulin resistance impairs the clearance of neuronal debris in Drosophila brain. PLoS Biol 2023; 21:e3002359. [PMID: 37934726 PMCID: PMC10629620 DOI: 10.1371/journal.pbio.3002359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 10/03/2023] [Indexed: 11/09/2023] Open
Abstract
Obesity significantly increases the risk of developing neurodegenerative disorders, yet the precise mechanisms underlying this connection remain unclear. Defects in glial phagocytic function are a key feature of neurodegenerative disorders, as delayed clearance of neuronal debris can result in inflammation, neuronal death, and poor nervous system recovery. Mounting evidence indicates that glial function can affect feeding behavior, weight, and systemic metabolism, suggesting that diet may play a role in regulating glial function. While it is appreciated that glial cells are insulin sensitive, whether obesogenic diets can induce glial insulin resistance and thereby impair glial phagocytic function remains unknown. Here, using a Drosophila model, we show that a chronic obesogenic diet induces glial insulin resistance and impairs the clearance of neuronal debris. Specifically, obesogenic diet exposure down-regulates the basal and injury-induced expression of the glia-associated phagocytic receptor, Draper. Constitutive activation of systemic insulin release from Drosophila insulin-producing cells (IPCs) mimics the effect of diet-induced obesity on glial Draper expression. In contrast, genetically attenuating systemic insulin release from the IPCs rescues diet-induced glial insulin resistance and Draper expression. Significantly, we show that genetically stimulating phosphoinositide 3-kinase (Pi3k), a downstream effector of insulin receptor (IR) signaling, rescues high-sugar diet (HSD)-induced glial defects. Hence, we establish that obesogenic diets impair glial phagocytic function and delays the clearance of neuronal debris.
Collapse
Affiliation(s)
- Mroj Alassaf
- Basic Sciences Division, Fred Hutch, Seattle, Washington, United States of America
| | - Akhila Rajan
- Basic Sciences Division, Fred Hutch, Seattle, Washington, United States of America
| |
Collapse
|
11
|
Pampuscenko K, Morkuniene R, Krasauskas L, Smirnovas V, Brown GC, Borutaite V. Extracellular tau stimulates phagocytosis of living neurons by activated microglia via Toll-like 4 receptor-NLRP3 inflammasome-caspase-1 signalling axis. Sci Rep 2023; 13:10813. [PMID: 37402829 DOI: 10.1038/s41598-023-37887-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 06/29/2023] [Indexed: 07/06/2023] Open
Abstract
In tauopathies, abnormal deposition of intracellular tau protein followed by gradual elevation of tau in cerebrospinal fluids and neuronal loss has been documented, however, the mechanism how actually neurons die under tau pathology is largely unknown. We have previously shown that extracellular tau protein (2N4R isoform) can stimulate microglia to phagocytose live neurons, i.e. cause neuronal death by primary phagocytosis, also known as phagoptosis. Here we show that tau protein induced caspase-1 activation in microglial cells via 'Toll-like' 4 (TLR4) receptors and neutral sphingomyelinase. Tau-induced neuronal loss was blocked by caspase-1 inhibitors (Ac-YVAD-CHO and VX-765) as well as by TLR4 antibodies. Inhibition of caspase-1 by Ac-YVAD-CHO prevented tau-induced exposure of phosphatidylserine on the outer leaflet of neuronal membranes and reduced microglial phagocytic activity. We also show that suppression of NLRP3 inflammasome, which is down-stream of TLR4 receptors and mediates caspase-1 activation, by a specific inhibitor (MCC550) also prevented tau-induced neuronal loss. Moreover, NADPH oxidase is also involved in tau-induced neurotoxicity since neuronal loss was abolished by its pharmacological inhibitor. Overall, our data indicate that extracellular tau protein stimulates microglia to phagocytose live neurons via Toll-like 4 receptor-NLRP3 inflammasome-caspase-1 axis and NADPH oxidase, each of which may serve as a potential molecular target for pharmacological treatment of tauopathies.
Collapse
Affiliation(s)
- Katryna Pampuscenko
- Neuroscience Institute, Lithuanian University of Health Sciences, 50161, Kaunas, Lithuania.
| | - Ramune Morkuniene
- Neuroscience Institute, Lithuanian University of Health Sciences, 50161, Kaunas, Lithuania
| | - Lukas Krasauskas
- Life Sciences Center, Institute of Biotechnology, Vilnius University, 10257, Vilnius, Lithuania
| | - Vytautas Smirnovas
- Life Sciences Center, Institute of Biotechnology, Vilnius University, 10257, Vilnius, Lithuania
| | - Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QW, UK
| | - Vilmante Borutaite
- Neuroscience Institute, Lithuanian University of Health Sciences, 50161, Kaunas, Lithuania
| |
Collapse
|
12
|
Wu T, Deger JM, Ye H, Guo C, Dhindsa J, Pekarek BT, Al-Ouran R, Liu Z, Al-Ramahi I, Botas J, Shulman JM. Tau polarizes an aging transcriptional signature to excitatory neurons and glia. eLife 2023; 12:e85251. [PMID: 37219079 PMCID: PMC10259480 DOI: 10.7554/elife.85251] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 05/22/2023] [Indexed: 05/24/2023] Open
Abstract
Aging is a major risk factor for Alzheimer's disease (AD), and cell-type vulnerability underlies its characteristic clinical manifestations. We have performed longitudinal, single-cell RNA-sequencing in Drosophila with pan-neuronal expression of human tau, which forms AD neurofibrillary tangle pathology. Whereas tau- and aging-induced gene expression strongly overlap (93%), they differ in the affected cell types. In contrast to the broad impact of aging, tau-triggered changes are strongly polarized to excitatory neurons and glia. Further, tau can either activate or suppress innate immune gene expression signatures in a cell-type-specific manner. Integration of cellular abundance and gene expression pinpoints nuclear factor kappa B signaling in neurons as a marker for cellular vulnerability. We also highlight the conservation of cell-type-specific transcriptional patterns between Drosophila and human postmortem brain tissue. Overall, our results create a resource for dissection of dynamic, age-dependent gene expression changes at cellular resolution in a genetically tractable model of tauopathy.
Collapse
Affiliation(s)
- Timothy Wu
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
- Medical Scientist Training Program, Baylor College of MedicineHoustonUnited States
| | - Jennifer M Deger
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
- Medical Scientist Training Program, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Hui Ye
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
- Department of Neurology, Baylor College of MedicineHoustonUnited States
| | - Caiwei Guo
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
| | - Justin Dhindsa
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
- Medical Scientist Training Program, Baylor College of MedicineHoustonUnited States
| | - Brandon T Pekarek
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
| | - Rami Al-Ouran
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
| | - Zhandong Liu
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
- Department of Pediatrics, Baylor College of MedicineHoustonUnited States
- Center for Alzheimer’s and Neurodegenerative Diseases, Baylor College of MedicineHoustonUnited States
| | - Ismael Al-Ramahi
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
- Center for Alzheimer’s and Neurodegenerative Diseases, Baylor College of MedicineHoustonUnited States
| | - Juan Botas
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
- Center for Alzheimer’s and Neurodegenerative Diseases, Baylor College of MedicineHoustonUnited States
| | - Joshua M Shulman
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Department of Neuroscience, Baylor College of MedicineHoustonUnited States
- Department of Neurology, Baylor College of MedicineHoustonUnited States
- Center for Alzheimer’s and Neurodegenerative Diseases, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
13
|
Chung HL, Ye Q, Park YJ, Zuo Z, Mok JW, Kanca O, Tattikota SG, Lu S, Perrimon N, Lee HK, Bellen HJ. Very-long-chain fatty acids induce glial-derived sphingosine-1-phosphate synthesis, secretion, and neuroinflammation. Cell Metab 2023; 35:855-874.e5. [PMID: 37084732 PMCID: PMC10160010 DOI: 10.1016/j.cmet.2023.03.022] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 01/10/2023] [Accepted: 03/29/2023] [Indexed: 04/23/2023]
Abstract
VLCFAs (very-long-chain fatty acids) are the most abundant fatty acids in myelin. Hence, during demyelination or aging, glia are exposed to higher levels of VLCFA than normal. We report that glia convert these VLCFA into sphingosine-1-phosphate (S1P) via a glial-specific S1P pathway. Excess S1P causes neuroinflammation, NF-κB activation, and macrophage infiltration into the CNS. Suppressing the function of S1P in fly glia or neurons, or administration of Fingolimod, an S1P receptor antagonist, strongly attenuates the phenotypes caused by excess VLCFAs. In contrast, elevating the VLCFA levels in glia and immune cells exacerbates these phenotypes. Elevated VLCFA and S1P are also toxic in vertebrates based on a mouse model of multiple sclerosis (MS), experimental autoimmune encephalomyelitis (EAE). Indeed, reducing VLCFA with bezafibrate ameliorates the phenotypes. Moreover, simultaneous use of bezafibrate and fingolimod synergizes to improve EAE, suggesting that lowering VLCFA and S1P is a treatment avenue for MS.
Collapse
Affiliation(s)
- Hyung-Lok Chung
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Qi Ye
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ye-Jin Park
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Zhongyuan Zuo
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jung-Wan Mok
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Oguz Kanca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | | | - Shenzhao Lu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Nobert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute and Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Hyun Kyoung Lee
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Department of Pediatrics, Section of Neurology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA.
| |
Collapse
|
14
|
Wang R, Ma B, Shi K, Wu F, Zhou C. Effects of lithium on aggression in Drosophila. Neuropsychopharmacology 2023; 48:754-763. [PMID: 36253547 PMCID: PMC10066353 DOI: 10.1038/s41386-022-01475-2] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/15/2022] [Accepted: 10/05/2022] [Indexed: 11/08/2022]
Abstract
Lithium is a common medication used to treat mania and bipolar disorder, but the mechanisms by which lithium stabilizes mood and modifies aggression are still not fully understood. Here we found that acute but not chronic lithium significantly suppresses aggression without affecting locomotion in Drosophila melanogaster. Male flies treated with acute lithium are also less competitive than control males in establishing dominance. We also provided evidence that glycogen synthase kinase-3 (GSK-3), a well-known target of lithium, plays an important role in the anti-aggressive effect of lithium in Drosophila. Our genetic data showed that acute knockdown of GSK-3 in neurons can mimic the inhibitory effect of acute lithium on aggression, while specific overexpression of GSK-3 in a subset of P1 neurons profoundly promotes aggression which can be partially rescued by acute lithium application. Thus, these findings revealed the inhibitory effect of lithium on aggression in Drosophila and laid a groundwork for using Drosophila as a powerful model to investigate the mechanisms by which lithium reduces aggression.
Collapse
Affiliation(s)
- Rencong Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Baoxu Ma
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Kai Shi
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China
- University of Chinese Academy of Sciences, 100101, Beijing, China
| | - Fengming Wu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
- University of Chinese Academy of Sciences, 100101, Beijing, China.
| | - Chuan Zhou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China.
- University of Chinese Academy of Sciences, 100101, Beijing, China.
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, 518132, China.
| |
Collapse
|
15
|
Corty MM, Coutinho-Budd J. Drosophila glia take shape to sculpt the nervous system. Curr Opin Neurobiol 2023; 79:102689. [PMID: 36822142 PMCID: PMC10023329 DOI: 10.1016/j.conb.2023.102689] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/19/2022] [Accepted: 01/10/2023] [Indexed: 02/23/2023]
Abstract
The importance of glial cells has become increasingly apparent over the past 20 years, yet compared to neurons we still know relatively little about these essential cells. Most critical glial cell functions are conserved in Drosophila glia, often using the same key molecular players as their vertebrate counterparts. The relative simplicity of the Drosophila nervous system, combined with a vast array of powerful genetic tools, allows us to further dissect the molecular composition and functional roles of glia in ways that would be much more cumbersome or not possible in higher vertebrate systems. Importantly, Drosophila genetics allow for in vivo manipulation, and their transparent body wall enables in vivo imaging of glia in intact animals throughout early development. Here we discuss recent advances in Drosophila glial development detailing how these cells take on their mature morphologies and interact with neurons to perform their important functional roles in the nervous system.
Collapse
Affiliation(s)
- Megan M Corty
- Vollum Institute, Oregon Health & Science University, Portland, OR, USA. https://twitter.com/@megancphd
| | - Jaeda Coutinho-Budd
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
16
|
Hanson MA, Lemaitre B. Antimicrobial peptides do not directly contribute to aging in Drosophila, but improve lifespan by preventing dysbiosis. Dis Model Mech 2023; 16:dmm049965. [PMID: 36847474 PMCID: PMC10163324 DOI: 10.1242/dmm.049965] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/20/2023] [Indexed: 03/01/2023] Open
Abstract
Antimicrobial peptides (AMPs) are innate immune effectors first studied for their role in host defence. Recent studies have implicated these peptides in the clearance of aberrant cells and in neurodegenerative syndromes. In Drosophila, many AMPs are produced downstream of Toll and Imd NF-κB pathways upon infection. Upon aging, AMPs are upregulated, drawing attention to these molecules as possible causes of age-associated inflammatory diseases. However, functional studies overexpressing or silencing these genes have been inconclusive. Using an isogenic set of AMP gene deletions, we investigated the net impact of AMPs on aging. Overall, we found no major effect of individual AMPs on lifespan, with the possible exception of Defensin. However, ΔAMP14 flies lacking seven AMP gene families displayed reduced lifespan. Increased bacterial load in the food of aged ΔAMP14 flies suggested that their lifespan reduction was due to microbiome dysbiosis, consistent with a previous study. Moreover, germ-free conditions extended the lifespan of ΔAMP14 flies. Overall, our results did not point to an overt role of individual AMPs in lifespan. Instead, we found that AMPs collectively impact lifespan by preventing dysbiosis during aging.
Collapse
Affiliation(s)
- Mark A. Hanson
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Bruno Lemaitre
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
17
|
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
|
18
|
Zhang C, Raveney B, Takahashi F, Yeh TW, Hohjoh H, Yamamura T, Oki S. Pathogenic Microglia Orchestrate Neurotoxic Properties of Eomes-Expressing Helper T Cells. Cells 2023; 12:cells12060868. [PMID: 36980209 PMCID: PMC10047905 DOI: 10.3390/cells12060868] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/08/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
In addition to disease-associated microglia (DAM), microglia with MHC-II and/or IFN-I signatures may form additional pathogenic subsets that are relevant to neurodegeneration. However, the significance of such MHC-II and IFN-I signatures remains elusive. We demonstrate here that these microglial subsets play intrinsic roles in orchestrating neurotoxic properties of neurotoxic Eomes+ Th cells under the neurodegeneration-associated phase of experimental autoimmune encephalomyelitis (EAE) that corresponds to progressive multiple sclerosis (MS). Microglia acquire IFN-signature after sensing ectopically expressed long interspersed nuclear element-1 (L1) gene. Furthermore, ORF1, an L1-encoded protein aberrantly expressed in the diseased central nervous system (CNS), stimulated Eomes+ Th cells after Trem2-dependent ingestion and presentation in MHC-II context by microglia. Interestingly, administration of an L1 inhibitor significantly ameliorated neurodegenerative symptoms of EAE concomitant with reduced accumulation of Eomes+ Th cells in the CNS. Collectively, our data highlight a critical contribution of new microglia subsets as a neuroinflammatory hub in immune-mediated neurodegeneration.
Collapse
Affiliation(s)
- Chenyang Zhang
- Department of Immunology, National Institute of Neuroscience, NCNP, Tokyo 187-8502, Japan
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8549, Japan
| | - Ben Raveney
- Department of Immunology, National Institute of Neuroscience, NCNP, Tokyo 187-8502, Japan
| | - Fumio Takahashi
- Department of Immunology, National Institute of Neuroscience, NCNP, Tokyo 187-8502, Japan
| | - Tzu-wen Yeh
- Department of Immunology, National Institute of Neuroscience, NCNP, Tokyo 187-8502, Japan
| | - Hirohiko Hohjoh
- Department of Molecular Pharmacology, National Institute of Neuroscience, NCNP, Tokyo 187-8502, Japan
| | - Takashi Yamamura
- Department of Immunology, National Institute of Neuroscience, NCNP, Tokyo 187-8502, Japan
- Correspondence: (T.Y.); (S.O.); Tel.: +81-42-341-2711 (T.Y. & S.O.)
| | - Shinji Oki
- Department of Immunology, National Institute of Neuroscience, NCNP, Tokyo 187-8502, Japan
- Correspondence: (T.Y.); (S.O.); Tel.: +81-42-341-2711 (T.Y. & S.O.)
| |
Collapse
|
19
|
Davis J, Kolaski E, Babcock DT. Vexed mutations promote degeneration of dopaminergic neurons through excessive activation of the innate immune response. NPJ Parkinsons Dis 2022; 8:147. [PMID: 36323700 PMCID: PMC9630459 DOI: 10.1038/s41531-022-00417-5] [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] [Received: 05/10/2022] [Accepted: 10/14/2022] [Indexed: 11/11/2022] Open
Abstract
The hallmark of Parkinson's disease (PD) is the loss of dopaminergic (DA) neurons in the brain. However, little is known about why DA neurons are selectively vulnerable to PD. We previously completed a screen identifying genes associated with the progressive degeneration of DA neurons. Here we describe the role of a previously uncharacterized gene, CG42339, in the loss of DA neurons using Drosophila Melanogaster. CG42339 mutants display a progressive loss of DA neurons and locomotor dysfunction, along with an accumulation of advanced glycation end products (AGEs) in the brain. Based on this phenotype, we refer to CG42339 as vexed. We demonstrate that vexed is specifically required within cortex glia to maintain neuronal viability. Loss of vexed function results in excessive activation of the innate immune response in the brain, leading to loss of DA neurons. We show that activation of the innate immune response leads to increased nitric oxide signaling and accumulation of AGEs, which ultimately result in neurodegeneration. These results provide further insight into the relationship between the role of the immune response in the central nervous system and how this impacts neuronal viability.
Collapse
Affiliation(s)
- Jacinta Davis
- grid.259029.50000 0004 1936 746XDepartment of Biological Sciences, Lehigh University, Bethlehem, PA USA
| | - Elizabeth Kolaski
- grid.259029.50000 0004 1936 746XDepartment of Biological Sciences, Lehigh University, Bethlehem, PA USA
| | - Daniel T. Babcock
- grid.259029.50000 0004 1936 746XDepartment of Biological Sciences, Lehigh University, Bethlehem, PA USA
| |
Collapse
|
20
|
Serizier SB, Peterson JS, McCall K. Non-autonomous cell death induced by the Draper phagocytosis receptor requires signaling through the JNK and SRC pathways. J Cell Sci 2022; 135:jcs250134. [PMID: 36177600 PMCID: PMC10658789 DOI: 10.1242/jcs.250134] [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: 06/12/2020] [Accepted: 09/21/2022] [Indexed: 11/20/2022] Open
Abstract
The last step of cell death is cell clearance, a process critical for tissue homeostasis. For efficient cell clearance to occur, phagocytes and dead cells need to reciprocally signal to each other. One important phenomenon that is under-investigated, however, is that phagocytes not only engulf corpses but contribute to cell death progression. The aims of this study were to determine how the phagocytic receptor Draper non-autonomously induces cell death, using the Drosophila ovary as a model system. We found that Draper, expressed in epithelial follicle cells, requires its intracellular signaling domain to kill the adjacent nurse cell population. Kinases Src42A, Shark and JNK (Bsk) were required for Draper-induced nurse cell death. Signs of nurse cell death occurred prior to apparent engulfment and required the caspase Dcp-1, indicating that it uses a similar apoptotic pathway to starvation-induced cell death. These findings indicate that active signaling by Draper is required to kill nurse cells via the caspase Dcp-1, providing novel insights into mechanisms of phagoptosis driven by non-professional phagocytes.
Collapse
Affiliation(s)
- Sandy B. Serizier
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
- Program in Molecular Biology, Cell Biology, and Biochemistry, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Jeanne S. Peterson
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| | - Kimberly McCall
- Department of Biology, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
- Program in Molecular Biology, Cell Biology, and Biochemistry, Boston University, 5 Cummington Mall, Boston, MA 02215, USA
| |
Collapse
|
21
|
Barkaway A, Attwell D, Korte N. Immune-vascular mural cell interactions: consequences for immune cell trafficking, cerebral blood flow, and the blood-brain barrier. NEUROPHOTONICS 2022; 9:031914. [PMID: 35581998 PMCID: PMC9107322 DOI: 10.1117/1.nph.9.3.031914] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/13/2022] [Indexed: 06/15/2023]
Abstract
Brain barriers are crucial sites for cerebral energy supply, waste removal, immune cell migration, and solute exchange, all of which maintain an appropriate environment for neuronal activity. At the capillary level, where the largest area of brain-vascular interface occurs, pericytes adjust cerebral blood flow (CBF) by regulating capillary diameter and maintain the blood-brain barrier (BBB) by suppressing endothelial cell (EC) transcytosis and inducing tight junction expression between ECs. Pericytes also limit the infiltration of circulating leukocytes into the brain where resident microglia confine brain injury and provide the first line of defence against invading pathogens. Brain "waste" is cleared across the BBB into the blood, phagocytosed by microglia and astrocytes, or removed by the flow of cerebrospinal fluid (CSF) through perivascular routes-a process driven by respiratory motion and the pulsation of the heart, arteriolar smooth muscle, and possibly pericytes. "Dirty" CSF exits the brain and is probably drained around olfactory nerve rootlets and via the dural meningeal lymphatic vessels and possibly the skull bone marrow. The brain is widely regarded as an immune-privileged organ because it is accessible to few antigen-primed leukocytes. Leukocytes enter the brain via the meninges, the BBB, and the blood-CSF barrier. Advances in genetic and imaging tools have revealed that neurological diseases significantly alter immune-brain barrier interactions in at least three ways: (1) the brain's immune-privileged status is compromised when pericytes are lost or lymphatic vessels are dysregulated; (2) immune cells release vasoactive molecules to regulate CBF, modulate arteriole stiffness, and can plug and eliminate capillaries which impairs CBF and possibly waste clearance; and (3) immune-vascular interactions can make the BBB leaky via multiple mechanisms, thus aggravating the influx of undesirable substances and cells. Here, we review developments in these three areas and briefly discuss potential therapeutic avenues for restoring brain barrier functions.
Collapse
Affiliation(s)
- Anna Barkaway
- University College London, Department of Neuroscience, Physiology and Pharmacology, London, United Kingdom
| | - David Attwell
- University College London, Department of Neuroscience, Physiology and Pharmacology, London, United Kingdom
| | - Nils Korte
- University College London, Department of Neuroscience, Physiology and Pharmacology, London, United Kingdom
| |
Collapse
|
22
|
Zohar-Fux M, Ben-Hamo-Arad A, Arad T, Volin M, Shklyar B, Hakim-Mishnaevski K, Porat-Kuperstein L, Kurant E, Toledano H. The phagocytic cyst cells in Drosophila testis eliminate germ cell progenitors via phagoptosis. SCIENCE ADVANCES 2022; 8:eabm4937. [PMID: 35714186 PMCID: PMC9205596 DOI: 10.1126/sciadv.abm4937] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 05/04/2022] [Indexed: 06/15/2023]
Abstract
Phagoptosis is a frequently occurring nonautonomous cell death pathway in which phagocytes eliminate viable cells. While it is thought that phosphatidylserine (PS) "eat-me" signals on target cells initiate the process, the precise sequence of events is largely unknown. Here, we show that in Drosophila testes, progenitor germ cells are spontaneously removed by neighboring cyst cells through phagoptosis. Using live imaging with multiple markers, we demonstrate that cyst cell-derived early/late endosomes and lysosomes fused around live progenitors to acidify them, before DNA fragmentation and substantial PS exposure on the germ cell surface. Furthermore, the phagocytic receptor Draper is expressed on cyst cell membranes and is necessary for phagoptosis. Significantly, germ cell death is blocked by knockdown of either the endosomal component Rab5 or the lysosomal associated protein Lamp1, within the cyst cells. These data ascribe an active role for phagocytic cyst cells in removal of live germ cell progenitors.
Collapse
Affiliation(s)
- Maayan Zohar-Fux
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Aba Hushi Avenue, Mount Carmel, Haifa 3498838, Israel
| | - Aya Ben-Hamo-Arad
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Aba Hushi Avenue, Mount Carmel, Haifa 3498838, Israel
| | - Tal Arad
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Aba Hushi Avenue, Mount Carmel, Haifa 3498838, Israel
| | - Marina Volin
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Aba Hushi Avenue, Mount Carmel, Haifa 3498838, Israel
| | - Boris Shklyar
- Bioimaging Unit, Faculty of Natural Sciences, University of Haifa, 199 Aba Hushi Avenue, Mount Carmel, Haifa 3498838, Israel
| | - Ketty Hakim-Mishnaevski
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Aba Hushi Avenue, Mount Carmel, Haifa 3498838, Israel
| | - Lilach Porat-Kuperstein
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Aba Hushi Avenue, Mount Carmel, Haifa 3498838, Israel
| | - Estee Kurant
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Aba Hushi Avenue, Mount Carmel, Haifa 3498838, Israel
| | - Hila Toledano
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Aba Hushi Avenue, Mount Carmel, Haifa 3498838, Israel
| |
Collapse
|
23
|
Boulanger A, Dura JM. Neuron-glia crosstalk in neuronal remodeling and degeneration: Neuronal signals inducing glial cell phagocytic transformation in Drosophila. Bioessays 2022; 44:e2100254. [PMID: 35315125 DOI: 10.1002/bies.202100254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 03/04/2022] [Accepted: 03/07/2022] [Indexed: 11/09/2022]
Abstract
Neuronal remodeling is a conserved mechanism that eliminates unwanted neurites and can include the loss of cell bodies. In these processes, a key role for glial cells in events from synaptic pruning to neuron elimination has been clearly identified in the last decades. Signals sent from dying neurons or neurites to be removed are received by appropriate glial cells. After receiving these signals, glial cells infiltrate degenerating sites and then, engulf and clear neuronal debris through phagocytic mechanisms. There are few identified or proposed signals and receptors involved in neuron-glia crosstalk, which induces the transformation of glial cells to phagocytes during neuronal remodeling in Drosophila. Many of these signaling pathways are conserved in mammals. Here, we particularly emphasize the role of Orion, a recently identified neuronal CX3 C chemokine-like secreted protein, which induces astrocyte infiltration and engulfment during mushroom body neuronal remodeling. Although, chemokine signaling was not described previously in insects we propose that chemokine-like involvement in neuron/glial cell interaction is an evolutionarily ancient mechanism.
Collapse
Affiliation(s)
- Ana Boulanger
- IGH, Université de Montpellier, CNRS, Montpellier, France
| | | |
Collapse
|
24
|
Salazar JL, Yang SA, Lin YQ, Li-Kroeger D, Marcogliese PC, Deal SL, Neely GG, Yamamoto S. TM2D genes regulate Notch signaling and neuronal function in Drosophila. PLoS Genet 2021; 17:e1009962. [PMID: 34905536 PMCID: PMC8714088 DOI: 10.1371/journal.pgen.1009962] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 12/28/2021] [Accepted: 11/23/2021] [Indexed: 12/26/2022] Open
Abstract
TM2 domain containing (TM2D) proteins are conserved in metazoans and encoded by three separate genes in each model organism species that has been sequenced. Rare variants in TM2D3 are associated with Alzheimer's disease (AD) and its fly ortholog almondex is required for embryonic Notch signaling. However, the functions of this gene family remain elusive. We knocked-out all three TM2D genes (almondex, CG11103/amaretto, CG10795/biscotti) in Drosophila and found that they share the same maternal-effect neurogenic defect. Triple null animals are not phenotypically worse than single nulls, suggesting these genes function together. Overexpression of the most conserved region of the TM2D proteins acts as a potent inhibitor of Notch signaling at the γ-secretase cleavage step. Lastly, Almondex is detected in the brain and its loss causes shortened lifespan accompanied by progressive motor and electrophysiological defects. The functional links between all three TM2D genes are likely to be evolutionarily conserved, suggesting that this entire gene family may be involved in AD.
Collapse
Affiliation(s)
- Jose L. Salazar
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas, United States of America
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas, United States of America
| | - Sheng-An Yang
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas, United States of America
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas, United States of America
| | - Yong Qi Lin
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia
| | - David Li-Kroeger
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas, United States of America
- Department of Neurology, BCM, Houston, Texas, United States of America
- Center for Alzheimer’s and Neurodegenerative Diseases, BCM, Houston, Texas, United States of America
| | - Paul C. Marcogliese
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas, United States of America
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas, United States of America
| | - Samantha L. Deal
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas, United States of America
- Program in Developmental Biology, BCM, Houston, Texas, United States of America
| | - G. Gregory Neely
- The Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre and School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas, United States of America
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, Texas, United States of America
- Center for Alzheimer’s and Neurodegenerative Diseases, BCM, Houston, Texas, United States of America
- Program in Developmental Biology, BCM, Houston, Texas, United States of America
- Development, Disease Models & Therapeutics Graduate Program, BCM, Houston, Texas, United States of America
- Department of Neuroscience, BCM, Houston, Texas, United States of America
| |
Collapse
|
25
|
Phosphatidylserine synthase plays an essential role in glia and affects development, as well as the maintenance of neuronal function. iScience 2021; 24:102899. [PMID: 34401677 PMCID: PMC8358705 DOI: 10.1016/j.isci.2021.102899] [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: 04/21/2021] [Revised: 06/14/2021] [Accepted: 07/21/2021] [Indexed: 01/05/2023] Open
Abstract
Phosphatidylserine (PS) is an integral component of eukaryotic cell membranes and organelles. The Drosophila genome contains a single PS synthase (PSS)-encoding gene (Pss) homologous to mammalian PSSs. Flies with Pss loss-of-function alleles show a reduced life span, increased bang sensitivity, locomotor defects, and vacuolated brain, which are the signs associated with neurodegeneration. We observed defective mitochondria in mutant adult brain, as well as elevated production of reactive oxygen species, and an increase in autophagy and apoptotic cell death. Intriguingly, glial-specific knockdown or overexpression of Pss alters synaptogenesis and axonal growth in the larval stage, causes developmental arrest in pupal stages, and neurodegeneration in adults. This is not observed with pan-neuronal up- or down-regulation. These findings suggest that precisely regulated expression of Pss in glia is essential for the development and maintenance of brain function. We propose a mechanism that underlies these neurodegenerative phenotypes triggered by defective PS metabolism. Loss of Pss leads to developmental defects and neurodegeneration Loss of Pss causes a mitochondrial defect, elevated ROS, and secondary necrosis Pss functions in glia are essential for synaptogenesis and neuronal maintenance Glial Pss expression level must be tightly regulated to maintain a healthy nervous system
Collapse
|
26
|
Petrignani B, Rommelaere S, Hakim-Mishnaevski K, Masson F, Ramond E, Hilu-Dadia R, Poidevin M, Kondo S, Kurant E, Lemaitre B. A secreted factor NimrodB4 promotes the elimination of apoptotic corpses by phagocytes in Drosophila. EMBO Rep 2021; 22:e52262. [PMID: 34370384 PMCID: PMC8419693 DOI: 10.15252/embr.202052262] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 06/22/2021] [Accepted: 06/30/2021] [Indexed: 01/07/2023] Open
Abstract
Programmed cell death plays a fundamental role in development and tissue homeostasis. Professional and non‐professional phagocytes achieve the proper recognition, uptake, and degradation of apoptotic cells, a process called efferocytosis. Failure in efferocytosis leads to autoimmune and neurodegenerative diseases. In Drosophila, two transmembrane proteins of the Nimrod family, Draper and SIMU, mediate the recognition and internalization of apoptotic corpses. Beyond this early step, little is known about how apoptotic cell degradation is regulated. Here, we study the function of a secreted member of the Nimrod family, NimB4, and reveal its crucial role in the clearance of apoptotic cells. We show that NimB4 is expressed by macrophages and glial cells, the two main types of phagocytes in Drosophila. Similar to draper mutants, NimB4 mutants accumulate apoptotic corpses during embryogenesis and in the larval brain. Our study points to the role of NimB4 in phagosome maturation, more specifically in the fusion between the phagosome and lysosomes. We propose that similar to bridging molecules, NimB4 binds to apoptotic corpses to engage a phagosome maturation program dedicated to efferocytosis.
Collapse
Affiliation(s)
- Bianca Petrignani
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Samuel Rommelaere
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Ketty Hakim-Mishnaevski
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Florent Masson
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Elodie Ramond
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Reut Hilu-Dadia
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | | | - Shu Kondo
- Invertebrate Genetics Laboratory, Genetic Strains Research Center, National Institute of Genetics, Mishima, Japan
| | - Estee Kurant
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Bruno Lemaitre
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
27
|
Cai XT, Li H, Jensen MB, Maksoud E, Borneo J, Liang Y, Quake SR, Luo L, Haghighi P, Jasper H. Gut cytokines modulate olfaction through metabolic reprogramming of glia. Nature 2021; 596:97-102. [PMID: 34290404 PMCID: PMC8911385 DOI: 10.1038/s41586-021-03756-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 06/22/2021] [Indexed: 02/06/2023]
Abstract
Infection-induced aversion against enteropathogens is a conserved sickness behaviour that can promote host survival1,2. The aetiology of this behaviour remains poorly understood, but studies in Drosophila have linked olfactory and gustatory perception to avoidance behaviours against toxic microorganisms3-5. Whether and how enteric infections directly influence sensory perception to induce or modulate such behaviours remains unknown. Here we show that enteropathogen infection in Drosophila can modulate olfaction through metabolic reprogramming of ensheathing glia of the antennal lobe. Infection-induced unpaired cytokine expression in the intestine activates JAK-STAT signalling in ensheathing glia, inducing the expression of glial monocarboxylate transporters and the apolipoprotein glial lazarillo (GLaz), and affecting metabolic coupling of glia and neurons at the antennal lobe. This modulates olfactory discrimination, promotes the avoidance of bacteria-laced food and increases fly survival. Although transient in young flies, gut-induced metabolic reprogramming of ensheathing glia becomes constitutive in old flies owing to age-related intestinal inflammation, which contributes to an age-related decline in olfactory discrimination. Our findings identify adaptive glial metabolic reprogramming by gut-derived cytokines as a mechanism that causes lasting changes in a sensory system in ageing flies.
Collapse
Affiliation(s)
- Xiaoyu Tracy Cai
- Immunology Discovery, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.,Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945-1400, USA.,University of Southern California, Los Angeles, CA 90007, USA
| | - Hongjie Li
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA,Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Martin Borch Jensen
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945-1400, USA.,Gordian Biotechnology, 953 Indiana St., San Francisco, CA 94107, USA
| | - Elie Maksoud
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945-1400, USA
| | - Jovencio Borneo
- FACS lab, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Yuxin Liang
- NGS lab, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Stephen R. Quake
- Department of Bioengineering and Department of Applied Physics, Stanford University, Stanford, CA 94305, USA,Chan Zuckerberg Biohub, Stanford, CA 94305, USA
| | - Liqun Luo
- Department of Biology and Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Pejmun Haghighi
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945-1400, USA
| | - Heinrich Jasper
- Immunology Discovery, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.,Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945-1400, USA.,Corresponding author: Heinrich Jasper, Genentech, 1 DNA Way, South San Francisco, California 94080, USA.
| |
Collapse
|
28
|
Lebo DPV, McCall K. Murder on the Ovarian Express: A Tale of Non-Autonomous Cell Death in the Drosophila Ovary. Cells 2021; 10:cells10061454. [PMID: 34200604 PMCID: PMC8228772 DOI: 10.3390/cells10061454] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/05/2021] [Accepted: 06/07/2021] [Indexed: 12/30/2022] Open
Abstract
Throughout oogenesis, Drosophila egg chambers traverse the fine line between survival and death. After surviving the ten early and middle stages of oogenesis, egg chambers drastically change their size and structure to produce fully developed oocytes. The development of an oocyte comes at a cost, the price is the lives of the oocyte’s 15 siblings, the nurse cells. These nurse cells do not die of their own accord. Their death is dependent upon their neighbors—the stretch follicle cells. Stretch follicle cells are nonprofessional phagocytes that spend the final stages of oogenesis surrounding the nurse cells and subsequently forcing the nurse cells to give up everything for the sake of the oocyte. In this review, we provide an overview of cell death in the ovary, with a focus on recent findings concerning this phagocyte-dependent non-autonomous cell death.
Collapse
|
29
|
Raiders S, Black EC, Bae A, MacFarlane S, Klein M, Shaham S, Singhvi A. Glia actively sculpt sensory neurons by controlled phagocytosis to tune animal behavior. eLife 2021; 10:63532. [PMID: 33759761 PMCID: PMC8079151 DOI: 10.7554/elife.63532] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 03/23/2021] [Indexed: 02/07/2023] Open
Abstract
Glia in the central nervous system engulf neuron fragments to remodel synapses and recycle photoreceptor outer segments. Whether glia passively clear shed neuronal debris or actively prune neuron fragments is unknown. How pruning of single-neuron endings impacts animal behavior is also unclear. Here, we report our discovery of glia-directed neuron pruning in Caenorhabditis elegans. Adult C. elegans AMsh glia engulf sensory endings of the AFD thermosensory neuron by repurposing components of the conserved apoptotic corpse phagocytosis machinery. The phosphatidylserine (PS) flippase TAT-1/ATP8A functions with glial PS-receptor PSR-1/PSR and PAT-2/α-integrin to initiate engulfment. This activates glial CED-10/Rac1 GTPase through the ternary GEF complex of CED-2/CrkII, CED-5/DOCK180, CED-12/ELMO. Execution of phagocytosis uses the actin-remodeler WSP-1/nWASp. This process dynamically tracks AFD activity and is regulated by temperature, the AFD sensory input. Importantly, glial CED-10 levels regulate engulfment rates downstream of neuron activity, and engulfment-defective mutants exhibit altered AFD-ending shape and thermosensory behavior. Our findings reveal a molecular pathway underlying glia-dependent engulfment in a peripheral sense-organ and demonstrate that glia actively engulf neuron fragments, with profound consequences on neuron shape and animal sensory behavior. Neurons are tree-shaped cells that receive information through endings connected to neighbouring cells or the environment. Controlling the size, number and location of these endings is necessary to ensure that circuits of neurons get precisely the right amount of input from their surroundings. Glial cells form a large portion of the nervous system, and they are tasked with supporting, cleaning and protecting neurons. In humans, part of their duties is to ‘eat’ (or prune) unnecessary neuron endings. In fact, this role is so important that defects in glial pruning are associated with conditions such as Alzheimer’s disease. Yet it is still unknown how pruning takes place, and in particular whether it is the neuron or the glial cell that initiates the process. To investigate this question, Raiders et al. enlisted the common laboratory animal Caenorhabditis elegans, a tiny worm with a simple nervous system where each neuron has been meticulously mapped out. First, the experiments showed that glial cells in C. elegans actually prune the endings of sensory neurons. Focusing on a single glia-neuron pair then revealed that the glial cell could trim the endings of a living neuron by redeploying the same molecular machinery it uses to clear dead cell debris. Compared to this debris-clearing activity, however, the glial cell takes a more nuanced approach to pruning: specifically, it can adjust the amount of trimming based on the activity load of the neuron. When Raiders et al. disrupted the glial pruning for a single temperature-sensing neuron, the worm lost its normal temperature preferences; this demonstrated how the pruning activity of a single glial cell can be linked to behavior. Taken together the experiments showcase how C. elegans can be used to study glial pruning. Further work using this model could help to understand how disease emerges when glial cells cannot perform their role, and to spot the genetic factors that put certain individuals at increased risk for neurological and sensory disorders.
Collapse
Affiliation(s)
- Stephan Raiders
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States.,Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, United States
| | - Erik Calvin Black
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Andrea Bae
- Laboratory of Developmental Genetics, The Rockefeller University, New York, United States.,Cellular Imaging Shared Resources, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Stephen MacFarlane
- Department of Physics and Department of Biology, University of Miami, Coral Gables, United States
| | - Mason Klein
- Department of Physics and Department of Biology, University of Miami, Coral Gables, United States
| | - Shai Shaham
- Laboratory of Developmental Genetics, The Rockefeller University, New York, United States
| | - Aakanksha Singhvi
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States.,Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, United States.,Department of Biological Structure, University of Washington School of Medicine, Seattle, United States.,Brotman Baty Institute for Precision Medicine, Seattle, United States
| |
Collapse
|
30
|
Butler CA, Popescu AS, Kitchener EJA, Allendorf DH, Puigdellívol M, Brown GC. Microglial phagocytosis of neurons in neurodegeneration, and its regulation. J Neurochem 2021; 158:621-639. [PMID: 33608912 DOI: 10.1111/jnc.15327] [Citation(s) in RCA: 158] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/13/2021] [Accepted: 02/10/2021] [Indexed: 02/06/2023]
Abstract
There is growing evidence that excessive microglial phagocytosis of neurons and synapses contributes to multiple brain pathologies. RNA-seq and genome-wide association (GWAS) studies have linked multiple phagocytic genes to neurodegenerative diseases, and knock-out of phagocytic genes has been found to protect against neurodegeneration in animal models, suggesting that excessive microglial phagocytosis contributes to neurodegeneration. Here, we review recent evidence that microglial phagocytosis of live neurons and synapses causes neurodegeneration in animal models of Alzheimer's disease and other tauopathies, Parkinson's disease, frontotemporal dementias, multiple sclerosis, retinal degeneration and neurodegeneration induced by ischaemia, infection or ageing. We also review factors regulating microglial phagocytosis of neurons, including: nucleotides, frackalkine, phosphatidylserine, calreticulin, UDP, CD47, sialylation, complement, galectin-3, Apolipoprotein E, phagocytic receptors, Siglec receptors, cytokines, microglial epigenetics and expression profile. Some of these factors may be potential treatment targets to prevent neurodegeneration mediated by excessive microglial phagocytosis of live neurons and synapses.
Collapse
Affiliation(s)
- Claire A Butler
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Alma S Popescu
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | | | - Mar Puigdellívol
- Department of Biochemistry, University of Cambridge, Cambridge, UK.,Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Guy C Brown
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
31
|
Rigon L, De Filippis C, Napoli B, Tomanin R, Orso G. Exploiting the Potential of Drosophila Models in Lysosomal Storage Disorders: Pathological Mechanisms and Drug Discovery. Biomedicines 2021; 9:biomedicines9030268. [PMID: 33800050 PMCID: PMC8000850 DOI: 10.3390/biomedicines9030268] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/18/2021] [Accepted: 03/03/2021] [Indexed: 12/12/2022] Open
Abstract
Lysosomal storage disorders (LSDs) represent a complex and heterogeneous group of rare genetic diseases due to mutations in genes coding for lysosomal enzymes, membrane proteins or transporters. This leads to the accumulation of undegraded materials within lysosomes and a broad range of severe clinical features, often including the impairment of central nervous system (CNS). When available, enzyme replacement therapy slows the disease progression although it is not curative; also, most recombinant enzymes cannot cross the blood-brain barrier, leaving the CNS untreated. The inefficient degradative capability of the lysosomes has a negative impact on the flux through the endolysosomal and autophagic pathways; therefore, dysregulation of these pathways is increasingly emerging as a relevant disease mechanism in LSDs. In the last twenty years, different LSD Drosophila models have been generated, mainly for diseases presenting with neurological involvement. The fruit fly provides a large selection of tools to investigate lysosomes, autophagy and endocytic pathways in vivo, as well as to analyse neuronal and glial cells. The possibility to use Drosophila in drug repurposing and discovery makes it an attractive model for LSDs lacking effective therapies. Here, ee describe the major cellular pathways implicated in LSDs pathogenesis, the approaches available for their study and the Drosophila models developed for these diseases. Finally, we highlight a possible use of LSDs Drosophila models for drug screening studies.
Collapse
Affiliation(s)
- Laura Rigon
- Fondazione Istituto di Ricerca Pediatrica “Città della Speranza”, Corso Stati Uniti 4, 35127 Padova, Italy; (C.D.F.); (R.T.)
- Correspondence:
| | - Concetta De Filippis
- Fondazione Istituto di Ricerca Pediatrica “Città della Speranza”, Corso Stati Uniti 4, 35127 Padova, Italy; (C.D.F.); (R.T.)
- Laboratory of Diagnosis and Therapy of Lysosomal Disorders, Department of Women’s and Children’s Health, University of Padova, Via Giustiniani 3, 35128 Padova, Italy
| | - Barbara Napoli
- Laboratory of Molecular Biology, Scientific Institute, IRCCS Eugenio Medea, Via Don Luigi Monza 20, Bosisio Parini, 23842 Lecco, Italy;
| | - Rosella Tomanin
- Fondazione Istituto di Ricerca Pediatrica “Città della Speranza”, Corso Stati Uniti 4, 35127 Padova, Italy; (C.D.F.); (R.T.)
- Laboratory of Diagnosis and Therapy of Lysosomal Disorders, Department of Women’s and Children’s Health, University of Padova, Via Giustiniani 3, 35128 Padova, Italy
| | - Genny Orso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via Marzolo 5, 35131 Padova, Italy;
| |
Collapse
|
32
|
Raiders S, Han T, Scott-Hewitt N, Kucenas S, Lew D, Logan MA, Singhvi A. Engulfed by Glia: Glial Pruning in Development, Function, and Injury across Species. J Neurosci 2021; 41:823-833. [PMID: 33468571 PMCID: PMC7880271 DOI: 10.1523/jneurosci.1660-20.2020] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/20/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023] Open
Abstract
Phagocytic activity of glial cells is essential for proper nervous system sculpting, maintenance of circuitry, and long-term brain health. Glial engulfment of apoptotic cells and superfluous connections ensures that neuronal connections are appropriately refined, while clearance of damaged projections and neurotoxic proteins in the mature brain protects against inflammatory insults. Comparative work across species and cell types in recent years highlights the striking conservation of pathways that govern glial engulfment. Many signaling cascades used during developmental pruning are re-employed in the mature brain to "fine tune" synaptic architecture and even clear neuronal debris following traumatic events. Moreover, the neuron-glia signaling events required to trigger and perform phagocytic responses are impressively conserved between invertebrates and vertebrates. This review offers a compare-and-contrast portrayal of recent findings that underscore the value of investigating glial engulfment mechanisms in a wide range of species and contexts.
Collapse
Affiliation(s)
- Stephan Raiders
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, Washington 98195
| | - Taeho Han
- UCSF Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California 94158
| | - Nicole Scott-Hewitt
- F.M. Kirby Center for Neurobiology, Boston Children's Hospital, Boston, Massachusetts 02115
- Stanley Center for Psychiatric Research, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02142
| | - Sarah Kucenas
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
| | - Deborah Lew
- Department of Biological Sciences, Fordham University, Bronx, New York 10458
| | - Mary A Logan
- Jungers Center, Department of Neurology, Oregon Health and Science University, Portland, Oregon 97239
| | - Aakanksha Singhvi
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, Washington 98195
| |
Collapse
|
33
|
Tsao CK, Huang YF, Sun YH. Early lineage segregation of the retinal basal glia in the Drosophila eye disc. Sci Rep 2020; 10:18522. [PMID: 33116242 PMCID: PMC7595039 DOI: 10.1038/s41598-020-75581-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 10/16/2020] [Indexed: 11/09/2022] Open
Abstract
The retinal basal glia (RBG) is a group of glia that migrates from the optic stalk into the third instar larval eye disc while the photoreceptor cells (PR) are differentiating. The RBGs are grouped into three major classes based on molecular and morphological characteristics: surface glia (SG), wrapping glia (WG) and carpet glia (CG). The SGs migrate and divide. The WGs are postmitotic and wraps PR axons. The CGs have giant nucleus and extensive membrane extension that each covers half of the eye disc. In this study, we used lineage tracing methods to determine the lineage relationships among these glia subtypes and the temporal profile of the lineage decisions for RBG development. We found that the CG lineage segregated from the other RBG very early in the embryonic stage. It has been proposed that the SGs migrate under the CG membrane, which prevented SGs from contacting with the PR axons lying above the CG membrane. Upon passing the front of the CG membrane, which is slightly behind the morphogenetic furrow that marks the front of PR differentiation, the migrating SG contact the nascent PR axon, which in turn release FGF to induce SGs' differentiation into WG. Interestingly, we found that SGs are equally distributed apical and basal to the CG membrane, so that the apical SGs are not prevented from contacting PR axons by CG membrane. Clonal analysis reveals that the apical and basal RBG are derived from distinct lineages determined before they enter the eye disc. Moreover, the basal SG lack the competence to respond to FGFR signaling, preventing its differentiation into WG. Our findings suggest that this novel glia-to-glia differentiation is both dependent on early lineage decision and on a yet unidentified regulatory mechanism, which can provide spatiotemporal coordination of WG differentiation with the progressive differentiation of photoreceptor neurons.
Collapse
Affiliation(s)
- Chia-Kang Tsao
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan, ROC.,Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, ROC
| | - Yu Fen Huang
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan, ROC.,Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, ROC.,, 64 Marvin Lane, Piscataway, NJ, 08854, USA
| | - Y Henry Sun
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan, ROC. .,Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan, ROC.
| |
Collapse
|
34
|
Dabool L, Hakim-Mishnaevski K, Juravlev L, Flint-Brodsly N, Mandel S, Kurant E. Drosophila Skp1 Homologue SkpA Plays a Neuroprotective Role in Adult Brain. iScience 2020; 23:101375. [PMID: 32739834 PMCID: PMC7399183 DOI: 10.1016/j.isci.2020.101375] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/14/2020] [Accepted: 07/14/2020] [Indexed: 11/17/2022] Open
Abstract
Skp1, a component of the ubiquitin E3 ligases, was found to be decreased in the brains of sporadic Parkinson's disease (PD) patients, and its overexpression prevented death of murine neurons in culture. Here we expose the neuroprotective role of the Drosophila skp1 homolog, skpA, in the adult brain. Neuronal knockdown of skpA leads to accumulation of ubiquitinated protein aggregates and loss of dopaminergic neurons accompanied by motor dysfunction and reduced lifespan. Conversely, neuronal overexpression of skpA reduces aggregate load, improves age-related motor decline, and prolongs lifespan. Moreover, SkpA rescues neurodegeneration in a Drosophila model of PD. We also show that a Drosophila homolog of FBXO7, the F Box protein, Nutcracker (Ntc), works in the same pathway with SkpA. However, skpA overexpression rescues ntc knockdown phenotype, suggesting that SkpA interacts with additional F box proteins in the adult brain neurons. Collectively, our study discloses Skp1/SkpA as a potential therapeutic target in neurodegenerative diseases. SkpA-mediated protein degradation is required for normal function of the adult brain SkpA overexpression rescues neurodegeneration in α-synuclein-induced fly PD model SkpA and Ntc work in the same pathway of protein degradation in adult brain neurons
Collapse
Affiliation(s)
- Lital Dabool
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Aba Khoushy Avenue, Mount Carmel, Haifa 34988-38, Israel; The Rappaport Family Institute for Research in the Medical Sciences, Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Ketty Hakim-Mishnaevski
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Aba Khoushy Avenue, Mount Carmel, Haifa 34988-38, Israel
| | - Liza Juravlev
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Aba Khoushy Avenue, Mount Carmel, Haifa 34988-38, Israel
| | - Naama Flint-Brodsly
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Aba Khoushy Avenue, Mount Carmel, Haifa 34988-38, Israel
| | - Silvia Mandel
- The Rappaport Family Institute for Research in the Medical Sciences, Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel
| | - Estee Kurant
- Department of Human Biology, Faculty of Natural Sciences, University of Haifa, 199 Aba Khoushy Avenue, Mount Carmel, Haifa 34988-38, Israel; The Rappaport Family Institute for Research in the Medical Sciences, Faculty of Medicine, Technion - Israel Institute of Technology, Haifa 31096, Israel.
| |
Collapse
|
35
|
Kim T, Song B, Lee IS. Drosophila Glia: Models for Human Neurodevelopmental and Neurodegenerative Disorders. Int J Mol Sci 2020; 21:E4859. [PMID: 32660023 PMCID: PMC7402321 DOI: 10.3390/ijms21144859] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/27/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
Glial cells are key players in the proper formation and maintenance of the nervous system, thus contributing to neuronal health and disease in humans. However, little is known about the molecular pathways that govern glia-neuron communications in the diseased brain. Drosophila provides a useful in vivo model to explore the conserved molecular details of glial cell biology and their contributions to brain function and disease susceptibility. Herein, we review recent studies that explore glial functions in normal neuronal development, along with Drosophila models that seek to identify the pathological implications of glial defects in the context of various central nervous system disorders.
Collapse
Affiliation(s)
| | | | - Im-Soon Lee
- Department of Biological Sciences, Center for CHANS, Konkuk University, Seoul 05029, Korea; (T.K.); (B.S.)
| |
Collapse
|
36
|
Márquez-Ropero M, Benito E, Plaza-Zabala A, Sierra A. Microglial Corpse Clearance: Lessons From Macrophages. Front Immunol 2020; 11:506. [PMID: 32292406 PMCID: PMC7135884 DOI: 10.3389/fimmu.2020.00506] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 03/05/2020] [Indexed: 12/11/2022] Open
Abstract
From development to aging and disease, the brain parenchyma is under the constant threat of debris accumulation, in the form of dead cells and protein aggregates. To prevent garbage buildup, the brain is equipped with efficient phagocytes: the microglia. Microglia are similar, but not identical to other tissue macrophages, and in this review, we will first summarize the differences in the origin, lineage and population maintenance of microglia and macrophages. Then, we will discuss several principles that govern macrophage phagocytosis of apoptotic cells (efferocytosis), including the existence of redundant recognition mechanisms ("find-me" and "eat-me") that lead to a tight coupling between apoptosis and phagocytosis. We will then describe that resulting from engulfment and degradation of apoptotic cargo, phagocytes undergo an epigenetic, transcriptional and metabolic rewiring that leads to trained immunity, and discuss its relevance for microglia and brain function. In summary, we will show that neuroimmunologists can learn many lessons from the well-developed field of macrophage phagocytosis biology.
Collapse
Affiliation(s)
- Mar Márquez-Ropero
- Achucarro Basque Center for Neuroscience, Parque Científico, University of the Basque Country (UPV/EHU), Leioa, Spain
- Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Eva Benito
- Achucarro Basque Center for Neuroscience, Parque Científico, University of the Basque Country (UPV/EHU), Leioa, Spain
- Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
- Ikerbasque Foundation, Bilbao, Spain
| | - Ainhoa Plaza-Zabala
- Achucarro Basque Center for Neuroscience, Parque Científico, University of the Basque Country (UPV/EHU), Leioa, Spain
- Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Amanda Sierra
- Achucarro Basque Center for Neuroscience, Parque Científico, University of the Basque Country (UPV/EHU), Leioa, Spain
- Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain
- Ikerbasque Foundation, Bilbao, Spain
| |
Collapse
|