1
|
Henningfield CM, Ngo M, Murray KM, Kwang NE, Tsourmas KI, Neumann J, Pashkutz ZA, Kawauchi S, Swarup V, Lane TE, MacGregor GR, Green KN. Generation of an Inducible Destabilized-Domain Cre Mouse Line to Target Disease Associated Microglia. Glia 2025; 73:1272-1287. [PMID: 39988890 DOI: 10.1002/glia.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/25/2025]
Abstract
The function of microglia during progression of Alzheimer's disease (AD) can be investigated using mouse models that enable genetic manipulation of microglial subpopulations in a temporal manner. We developed mouse lines that express either Cre recombinase (Cre) for constitutive targeting, or destabilized-domain Cre recombinase (DD-Cre) for inducible targeting from the Cst7 locus (Cst7 DD-Cre) to specifically manipulate disease associated microglia (DAM) and crossed with Ai14 tdTomato cre-reporter line mice. Cst7Cre was found to target all brain resident myeloid cells, due to transient developmental expression of Cst7, but no expression was found in the inducible Cst7 DD-Cre mice. Further crossing of this line with 5xFAD mice combined with dietary administration of trimethoprim to induce DD-Cre activity produces long-term labeling in DAM without evidence of leakiness, with tdTomato-expression restricted to cells surrounding plaques. Using this model, we found that DAMs are a subset of plaque-associated microglia (PAMs) and their transition to DAM increases with age and disease stage. Spatial transcriptomic analysis revealed that tdTomato+ cells show higher expression of disease and inflammatory genes compared to other microglial populations, including non-labeled PAMs. These models allow either complete cre-loxP targeting of all brain myeloid cells (Cst7Cre), or inducible targeting of DAMs, without leakiness (Cst7 DD-Cre).
Collapse
Affiliation(s)
- Caden M Henningfield
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Minh Ngo
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Kaitlin M Murray
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Nellie E Kwang
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Kate I Tsourmas
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Jonathan Neumann
- Transgenic Mouse Facility, University Laboratory Animal Services, Office of Research, University of California, Irvine, California, USA
| | - Zachary A Pashkutz
- Transgenic Mouse Facility, University Laboratory Animal Services, Office of Research, University of California, Irvine, California, USA
| | - Shimako Kawauchi
- Transgenic Mouse Facility, University Laboratory Animal Services, Office of Research, University of California, Irvine, California, USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Thomas E Lane
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
- Center for Virus Research, University of California, Irvine, USA
| | - Grant R MacGregor
- Transgenic Mouse Facility, University Laboratory Animal Services, Office of Research, University of California, Irvine, California, USA
- Department of Developmental and Cell Biology, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| | - Kim N Green
- Department of Neurobiology and Behavior, Charlie Dunlop School of Biological Sciences, University of California, Irvine, California, USA
| |
Collapse
|
2
|
Chen W, Liu X, Muñoz VR, Kahn CR. Loss of insulin signaling in microglia impairs cellular uptake of Aβ and neuroinflammatory response exacerbating AD-like neuropathology. Proc Natl Acad Sci U S A 2025; 122:e2501527122. [PMID: 40388612 DOI: 10.1073/pnas.2501527122] [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: 01/21/2025] [Accepted: 04/07/2025] [Indexed: 05/21/2025] Open
Abstract
Insulin receptors are present on cells throughout the body, including the brain. Dysregulation of insulin signaling in neurons and astrocytes has been implicated in altered mood, cognition, and the pathogenesis of Alzheimer's disease (AD). To define the role of insulin signaling in microglia, the primary phagocytes in the brain critical for maintenance and damage repair, we created mice with an inducible microglia-specific insulin receptor knockout (MG-IRKO). RiboTag profiling of microglial mRNAs revealed that loss of insulin signaling results in alterations of gene expression in pathways related to innate immunity and cellular metabolism. In vitro, loss of insulin signaling in microglia results in metabolic reprogramming with an increase in glycolysis and impaired uptake of Aβ. In vivo, MG-IRKO mice exhibit alterations in mood and social behavior, and when crossed with the 5xFAD mouse model of AD, the resultant mice exhibit increased levels of Aβ plaque and elevated neuroinflammation. Thus, insulin signaling in microglia plays a key role in microglial cellular metabolism and the ability of the cells to take up Aβ, such that reduced insulin signaling in microglia alters mood and social behavior and accelerates AD pathogenesis. Together, these data indicate key roles of insulin action in microglia and the potential of targeting insulin signaling in microglia in treatment of AD.
Collapse
Affiliation(s)
- Wenqiang Chen
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02215
- Clinical and Translational Research, Steno Diabetes Center Copenhagen, Herlev 2730, Denmark
| | - Xiangyu Liu
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02215
| | - Vitor Rosetto Muñoz
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02215
- Laboratory of Molecular Biology of Exercise, University of Campinas, Limeira, São Paulo 13484-350, Brazil
| | - C Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center and Department of Medicine, Harvard Medical School, Boston, MA 02215
| |
Collapse
|
3
|
Theis T, Kumar S, Shah P, Patel M, Tadmori I, Ayala C, Tschang M, Young W, Schachner M. Depletion of Cell Adhesion Molecule L1 from Microglia and Macrophages Reduces Recovery After Spinal Cord Injury. Int J Mol Sci 2025; 26:3285. [PMID: 40244153 PMCID: PMC11989546 DOI: 10.3390/ijms26073285] [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: 12/29/2024] [Revised: 03/27/2025] [Accepted: 03/29/2025] [Indexed: 04/18/2025] Open
Abstract
The young mammalian central nervous system regenerates after spinal cord injury and recovers locomotion, whereas adult mice only show limited recovery that depends on the injury severity, genetic background, and physical therapy. At the molecular level, key regulators that contribute to recovery are cell adhesion molecules, such as L1CAM (L1). At the cell surface, L1 functions as a homotypic receptor that signal-transduces crucial functions in neuronal migration and survival, neurite outgrowth, myelination, formation of synapses, and synaptic plasticity. In the adult central nervous system, L1 is expressed only by neurons. We now show that L1 is unexpectedly also expressed by 26% microglia, freshly isolated from a 7-day-old mouse brain. At postnatal day 21, only 3% of microglia are L1-positive. Using a mouse mutant in which L1 is deleted specifically in monocytes of 10- to 14-week-old mice, functional recovery was reduced up to 4 weeks after injury at lower thoracic spinal levels. Also, NF200-immunoreactive and 5-HT-immunoreactive fibers were found decreased below the injury site as compared to wild-type mice. In conclusion, microglial cells that express L1 stimulate neurite outgrowth in vitro, improve functional recovery after spinal cord injury in adult mice, and increase fiber densities caudal to the lesion site.
Collapse
Affiliation(s)
- Thomas Theis
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| | - Suneel Kumar
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08844, USA;
| | - Pratiksha Shah
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| | - Mukti Patel
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| | - Iman Tadmori
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| | - Carlos Ayala
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| | - Monica Tschang
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| | - Wise Young
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| |
Collapse
|
4
|
Jiang L, Zhang X, Wang S, Zhang J, Chen J, Lu J, Yao L, Jin W, Li N, Li Q. Functional Monomers Equipped Microgel System for Managing Parkinson's Disease by Intervening Chemokine Axis-mediated Nerve Cell Communications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410070. [PMID: 39721010 PMCID: PMC11831437 DOI: 10.1002/advs.202410070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/15/2024] [Indexed: 12/28/2024]
Abstract
The complex pathology of Parkinson's disease (PD) requires comprehensive understanding and multi-pronged interventions for communication between nerve cells. Despite new developments in nanotechnology in the treatment of PD, in-depth exploration of their biological effects, in particular, the specific mechanisms of inflammation inhibition are lacking. Herein, using the stable cascade catalysis channel formed by polydopamine (PDA), imidazole groups, and Cu ions, a microgel system comprising functional monomers [superoxide dismutase (SOD) with double bonds, PDA, 2-methacryloyloxy ethyl phosphorylcholine (MPC), and Cu ions] is proposed for managing PD. The microgel can be efficiently delivered to the brain aided by MPC, after which a multi-level regulatory strategy targeting neurons and microglia can be initiated. The catalytic activity cascade elicited by SOD and Cu ions can regulate the anti-inflammatory phenotypic transformation of microglia by relieving oxidative stress. Meanwhile, the dopamine (DA) released from PDA can facilitate DA storage and neurogenesis, inhibiting CX3CL1 release and the CX3CR1 receptor on microglia and further regulating the CX3CL1/CX3CR1-NF-κB-NLRP3 signaling pathway in microglia to inhibit neuroinflammation. Therefore, the proposed microgel delivery system with functional monomers represents a promising therapeutic strategy for managing neuroinflammation and promoting neurogenesis in PD by intervening chemokine axis-mediated communication between neurons and microglia.
Collapse
Affiliation(s)
- Lin Jiang
- College of Life SciencesChina Jiliang UniversityHangzhou310018China
| | - Xu Zhang
- Department of NeurologyThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhou450052China
| | - Shun Wang
- Department of NeurologyThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhou450052China
| | - Jiangkuan Zhang
- Department of NeurologyThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhou450052China
| | - Junyang Chen
- School of Life SciencesZhengzhou UniversityZhengzhou450001China
| | - Jiachuan Lu
- Department of NeurologyThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhou450052China
| | - Liting Yao
- College of Life SciencesChina Jiliang UniversityHangzhou310018China
| | - Weiwei Jin
- College of Life SciencesChina Jiliang UniversityHangzhou310018China
| | - Nan Li
- Department of NeurologyThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhou450052China
| | - Qing Li
- Department of NeurologyThe Second Affiliated Hospital of Zhengzhou UniversityZhengzhou UniversityZhengzhou450052China
| |
Collapse
|
5
|
Pang X, Gu L, Han QY, Xing JQ, Zhao M, Huang SY, Yi JX, Pan J, Hong H, Xue W, Zhou XQ, Su ZH, Zhang XR, Sun LM, Jiang SZ, Luo D, Chen L, Wang ZJ, Yu Y, Xia T, Zhang XM, Li AL, Zhou T, Cai H, Li T. RGS22 maintains the physiological function of ependymal cells to prevent hydrocephalus. SCIENCE CHINA. LIFE SCIENCES 2025; 68:441-453. [PMID: 39400871 DOI: 10.1007/s11427-024-2720-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 08/30/2024] [Indexed: 10/15/2024]
Abstract
Ependymal cells line the wall of cerebral ventricles and ensure the unidirectional cerebrospinal fluid (CSF) flow by beating their motile cilia coordinately. The ependymal denudation or ciliary dysfunction causes hydrocephalus. Here, we report that the deficiency of regulator of G-protein signaling 22 (RGS22) results in severe congenital hydrocephalus in both mice and rats. Interestingly, RGS22 is specifically expressed in ependymal cells within the brain. Using conditional knock-out mice, we further demonstrate that the deletion of Rgs22 exclusively in nervous system is sufficient to induce hydrocephalus. Mechanistically, we show that Rgs22 deficiency leads to the ependymal denudation and impaired ciliogenesis. This phenomenon can be attributed to the excessive activation of lysophosphatidic acid receptor (LPAR) signaling under Rgs22-/- condition, as the LPAR blockade effectively alleviates hydrocephalus in Rgs22-/- rats. Therefore, our findings unveil a previously unrecognized role of RGS22 in the central nervous system, and present RGS22 as a potential diagnostic and therapeutic target for hydrocephalus.
Collapse
Affiliation(s)
- Xue Pang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Lin Gu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Qiu-Ying Han
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Jia-Qing Xing
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Ming Zhao
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Shao-Yi Huang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Jun-Xi Yi
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Jie Pan
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Hao Hong
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Wen Xue
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Xue-Qing Zhou
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Zhi-Hui Su
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Xin-Ran Zhang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Li-Ming Sun
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Shao-Zhen Jiang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
- School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Dan Luo
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Ling Chen
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Zheng-Jie Wang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Yu Yu
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Tian Xia
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Xue-Min Zhang
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
- School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Ai-Ling Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
- School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Tao Zhou
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China
| | - Hong Cai
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China.
| | - Tao Li
- Nanhu Laboratory, National Center of Biomedical Analysis, Beijing, 100039, China.
- School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
6
|
Chen SW, Chu Y, Chu CH, Pham XDT, Ng HP, Guo CL, Cheng PL. ECM29/proteasome-mediated self-antigen generation by CNS-resident neuroglia promotes regulatory T cell activation. Cell Rep 2025; 44:115161. [PMID: 39786996 DOI: 10.1016/j.celrep.2024.115161] [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: 02/20/2024] [Revised: 11/08/2024] [Accepted: 12/16/2024] [Indexed: 01/12/2025] Open
Abstract
Proteasomes generate antigenic peptides presented on cell surfaces-a process that, in neuroglia, is highly responsive to external stimuli. However, the function of the self-antigens presented by CNS parenchymal cells remains unclear. Here, we report that the fidelity of neuroglial self-antigens is crucial to suppress encephalitogenic T cell responses by elevating regulatory T (Treg) cell populations. We demonstrate that loss of the proteasome adaptor protein Ecm29 alters the efficacy and accuracy of antigen generation. Inducible oligodendroglia- or microglia-conditional Ecm29 knockout mice exhibit higher susceptibility to experimental autoimmune encephalomyelitis (EAE) than control counterparts do, coincident with reduced Treg cell populations in the spinal cord. Immunopeptidome profiling identifies self-antigens that modulate myelin-reactive T cell responses. Intraspinal adeno-associated virus (AAV)/Olig001-mediated expression of the self-antigen NDUFA1p ameliorates EAE and expands NDUFA1p-recognizing CD103+CD8+CD122+ Treg cells. Thus, Ecm29/proteasome-controlled, neuroglia-derived self-antigens modulate CNS immune tolerance.
Collapse
Affiliation(s)
- Sheng-Wen Chen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Ying Chu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Chien-Hsin Chu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | | | - Hang Pong Ng
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Chin-Lin Guo
- Institute of Physics, Academia Sinica, Taipei, Taiwan
| | - Pei-Lin Cheng
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
7
|
Robb JL, Boisjoly F, Machuca-Parra AI, Coursan A, Manceau R, Majeur D, Rodaros D, Bouyakdan K, Greffard K, Bilodeau JF, Forest A, Daneault C, Ruiz M, Laurent C, Arbour N, Layé S, Fioramonti X, Madore C, Fulton S, Alquier T. Blockage of ATGL-mediated breakdown of lipid droplets in microglia alleviates neuroinflammatory and behavioural responses to lipopolysaccharides. Brain Behav Immun 2025; 123:315-333. [PMID: 39326768 DOI: 10.1016/j.bbi.2024.09.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 08/21/2024] [Accepted: 09/21/2024] [Indexed: 09/28/2024] Open
Abstract
Lipid droplets (LD) are triglyceride storing organelles that have emerged as an important component of cellular inflammatory responses. LD lipolysis via adipose triglyceride lipase (ATGL), the enzyme that catalyses the rate-limiting step of triglyceride lipolysis, regulates inflammation in peripheral immune and non-immune cells. ATGL elicits both pro- and anti-inflammatory responses in the periphery in a cell-type dependent manner. The present study determined the impact of ATGL inhibition and microglia-specific ATGL genetic loss-of-function on acute inflammatory and behavioural responses to pro-inflammatory insult. First, we evaluated the impact of lipolysis inhibition on lipopolysaccharide (LPS)-induced expression and secretion of cytokines and phagocytosis in mouse primary microglia cultures. Lipase inhibitors (ORlistat and ATGListatin) and LPS led to LD accumulation in microglia. Pan-lipase inhibition with ORlistat alleviated LPS-induced expression of IL-1β and IL-6. Specific inhibition of ATGL had a similar action on CCL2, IL-1β and IL-6 expression in both neonatal and adult microglia cultures. CCL2 and IL-6 secretion were also reduced by ATGListatin or knockdown of ATGL. ATGListatin increased phagocytosis in neonatal cultures independently from LPS treatment. Second, targeted and untargeted lipid profiling revealed that ATGListatin reduced LPS-induced generation of pro-inflammatory prostanoids and modulated ceramide species in neonatal microglia. Finally, the role of microglial ATGL in neuroinflammation was assessed using a novel microglia-specific and inducible ATGL knockout mouse model. Loss of microglial ATGL in adult male mice dampened LPS-induced expression of IL-6 and IL-1β and microglial density. LPS-induced sickness- and anxiety-like behaviours were also reduced in male mice with loss of ATGL in microglia. Together, our results demonstrate potent anti-inflammatory effects produced by pharmacological or genetic inhibition of ATGL-mediated triglyceride lipolysis and thereby propose that supressing microglial LD lipolysis has beneficial actions in acute neuroinflammatory conditions.
Collapse
Affiliation(s)
- Josephine Louise Robb
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada; Département de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Frédérick Boisjoly
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada; Département de Neurosciences, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Arturo Israel Machuca-Parra
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada; Département de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Adeline Coursan
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - Romane Manceau
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada; Département de Neurosciences, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Danie Majeur
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada; Département de Neurosciences, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Demetra Rodaros
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Khalil Bouyakdan
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Karine Greffard
- Axe Endocrinologie et Néphrologie, CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
| | - Jean-François Bilodeau
- Axe Endocrinologie et Néphrologie, CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada; Département de Médecine, Faculté de Médecine, Université Laval, Québec, QC, G1K 7P4, Canada
| | - Anik Forest
- Institut de Cardiologie de Montréal, Plateforme de métabolomique, Montréal, QC H1T1C8, Canada
| | - Caroline Daneault
- Institut de Cardiologie de Montréal, Plateforme de métabolomique, Montréal, QC H1T1C8, Canada
| | - Matthieu Ruiz
- Département de Nutrition, Université de Montréal, Montréal, QC H3T 1J4, Canada; Institut de Cardiologie de Montréal, Plateforme de métabolomique, Montréal, QC H1T1C8, Canada
| | - Cyril Laurent
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada; Département de Neurosciences, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Nathalie Arbour
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada; Département de Neurosciences, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Sophie Layé
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France; Food4BrainHealth France-Canada International Research Network, Bordeaux, France
| | - Xavier Fioramonti
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France; Food4BrainHealth France-Canada International Research Network, Bordeaux, France
| | - Charlotte Madore
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France; Food4BrainHealth France-Canada International Research Network, Bordeaux, France
| | - Stephanie Fulton
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada; Département de Nutrition, Université de Montréal, Montréal, QC H3T 1J4, Canada; Food4BrainHealth France-Canada International Research Network, Bordeaux, France
| | - Thierry Alquier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada; Département de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada; Food4BrainHealth France-Canada International Research Network, Bordeaux, France.
| |
Collapse
|
8
|
Jacquet RG, González Ibáñez F, Picard K, Funes L, Khakpour M, Gouras GK, Tremblay MÈ, Maxfield FR, Solé-Domènech S. Microglia degrade Alzheimer's amyloid-beta deposits extracellularly via digestive exophagy. Cell Rep 2024; 43:115052. [PMID: 39644493 PMCID: PMC11760508 DOI: 10.1016/j.celrep.2024.115052] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/17/2024] [Accepted: 11/20/2024] [Indexed: 12/09/2024] Open
Abstract
How microglia digest Alzheimer's fibrillar amyloid-beta (Aβ) plaques that are too large to be phagocytosed is not well understood. Here, we show that primary microglial cells create acidic extracellular compartments, lysosomal synapses, on model plaques and digest them with exocytosed lysosomal enzymes. This mechanism, called digestive exophagy, is confirmed by electron microscopy in 5xFAD mouse brains, which shows that a lysosomal enzyme, acid phosphatase, is secreted toward the plaques in structures resembling lysosomal synapses. Signaling studies demonstrate that the PI3K-AKT pathway modulates the formation of lysosomal synapses, as inhibition of PI3K1β or AKT1/2 reduces both lysosome exocytosis and actin polymerization, both required for the formation of the compartments. Finally, we show that small fibrils of Aβ previously internalized and trafficked to lysosomes are exocytosed toward large Aβ aggregates by microglia. Thus, the release of lysosomal contents during digestive exophagy may also contribute to the spread and growth of fibrillar Aβ.
Collapse
Affiliation(s)
- Rudy G Jacquet
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Fernando González Ibáñez
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC G1E 6W2, Canada; Département de Médecine Moléculaire, Université Laval, Québec City, QC G1V 0A6, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Katherine Picard
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC G1E 6W2, Canada; Département de Médecine Moléculaire, Université Laval, Québec City, QC G1V 0A6, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Lucy Funes
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Mohammadparsa Khakpour
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC G1E 6W2, Canada; Département de Médecine Moléculaire, Université Laval, Québec City, QC G1V 0A6, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada
| | - Gunnar K Gouras
- Experimental Dementia Unit, BMC, Lund University, 221 84 Lund, Sweden
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC G1E 6W2, Canada; Département de Médecine Moléculaire, Université Laval, Québec City, QC G1V 0A6, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC V8W 2Y2, Canada; Neurology and Neurosurgery Department, McGill University, Montréal, QC H3A 2B4, Canada; Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Centre for Advanced Materials and Related Technology (CAMTEC) and Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, BC V8W 2Y2, Canada
| | | | | |
Collapse
|
9
|
Zhang L, Huang L, Zhou Y, Meng J, Zhang L, Zhou Y, Zheng N, Guo T, Zhao S, Wang Z, Huo Y, Zhao Y, Chen XF, Zheng H, Holtzman DM, Zhang YW. Microglial CD2AP deficiency exerts protection in an Alzheimer's disease model of amyloidosis. Mol Neurodegener 2024; 19:95. [PMID: 39695808 DOI: 10.1186/s13024-024-00789-7] [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: 08/29/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The CD2-associated protein (CD2AP) was initially identified in peripheral immune cells and regulates cytoskeleton and protein trafficking. Single nucleotide polymorphisms (SNPs) in the CD2AP gene have been associated with Alzheimer's disease (AD). However, the functional role of CD2AP, especially its role in microglia during AD onset, remains elusive. METHODS CD2AP protein levels in cultured primary cells and in 5xFAD mice was studied. Microglial CD2AP-deficient mice were crossed with 5xFAD mice and the offspring were subjected to neuropathological assessment, behavioral tests, electrophysiology, RNA-seq, Golgi staining, and biochemistry analysis. Primary microglia were also isolated for assessing their uptake and morphology changes. RESULTS We find that CD2AP is abundantly expressed in microglia and its levels are elevated in the brain of AD patients and the 5xFAD model mice at pathological stages. We demonstrate that CD2AP haploinsufficiency in microglia significantly attenuates cognitive and synaptic deficits, weakens the response of microglia to Aβ and the formation of disease-associated microglia (DAM), and alleviates synapse loss in 5xFAD mice. We show that CD2AP-deficient microglia exhibit compromised uptake ability. In addition, we find that CD2AP expression is positively correlated with the expression of the complement C1q that is important for synapse phagocytosis and the formation of DAM in response to Aβ deposition. Moreover, we reveal that CD2AP interacts with colony stimulating factor 1 receptor (CSF1R) and regulates CSF1R cell surface levels, which may further affect C1q expression. CONCLUSIONS Our results demonstrate that CD2AP regulates microgliosis and identify a protective function of microglial CD2AP deficiency against Aβ deposition, suggesting the importance of detailed investigation of AD-associated genes in different brain cells for thoroughly understanding their exact contribution to AD.
Collapse
Affiliation(s)
- Lingliang Zhang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Lingling Huang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yuhang Zhou
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Jian Meng
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Liang Zhang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yunqiang Zhou
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Naizhen Zheng
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Tiantian Guo
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Shanshan Zhao
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Zijie Wang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yuanhui Huo
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yingjun Zhao
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Xiao-Fen Chen
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Honghua Zheng
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Yun-Wu Zhang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China.
| |
Collapse
|
10
|
Mroue-Ruiz FH, Desai B, Garvin M, Shehu J, Kamau F, Kar U, Bolton JL. Constitutive expression of CX3CR1-BAC-Cre introduces minimal off-target effects in microglia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.01.621625. [PMID: 39554070 PMCID: PMC11566009 DOI: 10.1101/2024.11.01.621625] [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: 11/19/2024]
Abstract
CX3CR1-Cre mouse lines have produced important advancements in our understanding of microglial biology. Recent studies have demonstrated the adverse effects of tamoxifen-induced CX3CR1-Cre expression during development, which include changes in microglial density, phenotype, and DNA damage, as well as anxiety-like behavior. However, the unintended effects of constitutive CX3CR1-BAC-Cre expression remain unexplored. Here, we characterized the effects of CX3CR1-BAC-Cre expression on microglia in CX3CR1-BAC-Cre+/- and CX3CR1-BAC-Cre-/- male and female littermates during early postnatal development and adulthood in multiple brain regions. Additionally, we performed anxiety-like behavior tests to assess changes caused by Cre expression. We found that CX3CR1-BAC-Cre expression causes subtle region- and sex-specific changes in microglial density, volume, and morphology during development, but these changes normalized by adulthood in all brain regions except the hippocampus. No behavioral effects were found. Our findings suggest that the constitutive-Cre model might be less detrimental than the inducible model, and highlight the need for proper controls.
Collapse
Affiliation(s)
| | - Bhoomi Desai
- Neuroscience Institute, Georgia State University
| | | | - Jonila Shehu
- Neuroscience Institute, Georgia State University
| | - Faith Kamau
- Neuroscience Institute, Georgia State University
| | - Urjoshi Kar
- Neuroscience Institute, Georgia State University
| | | |
Collapse
|
11
|
Bi Y, Xie Z, Cao X, Ni H, Xia S, Bao X, Huang Q, Xu Y, Zhang Q. Cedrol attenuates acute ischemic injury through inhibition of microglia-associated neuroinflammation via ERβ-NF-κB signaling pathways. Brain Res Bull 2024; 218:111102. [PMID: 39414157 DOI: 10.1016/j.brainresbull.2024.111102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 09/15/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024]
Abstract
Microglia-associated neuroinflammation plays essential roles in pathology of acute stroke. Cedrol, a natural compound extracted from ginger, has been shown to confer inhibitory effects on inflammation in various diseases. However, whether Cedrol suppresses neuroinflammation and protects brains from acute ischemic injury still remains unclear. In this study, we found that Cedrol inhibited microglia activation and the production of inflammatory factors in LPS-challenged microglia and the penumbra region of middle cerebral artery occlusion (MCAO) mice. We also found that Cedrol reduced the infarct size and mNSS scores and improved acute cerebral ischemia-induced behavioral outcomes, suggesting remarked neuroprotection of Cedrol. Molecular docking analysis showed that Cedrol bound to estrogen receptor β (ERβ) with moderate-strong affinity. Intriguingly, treatment with fulvestrant, an ER blocker, abolished the anti-inflammatory effects of Cedrol. Cedrol significantly reversed the LPS- and MCAO-induced increases in phosphorylation levels of IκB and NF-κB P65 in primary microglia and MCAO mice, respectively. Additionally, Cedrol was observed to rescue LPS-induced shuttling of NF-κB P65 from cytoplasm to nuclei in primary microglia, indicating inhibitory effects of Cedrol on NF-κB signaling. These results suggest microglia associated neuroinflammation may be mediated by ERβ-NF-κB signaling pathway. Together, our study reveals that Cedrol protected brain function from acute cerebral ischemia through inhibition of microglia-associated neuroinflammation via ERβ-NF-κB signaling pathways, and Cedrol may serve as an alternative option for treatment of acute stroke injury.
Collapse
Affiliation(s)
- Yu Bi
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, China; Department of Neurology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China; Department of Neurology,Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Ziyi Xie
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, China; Department of Neurology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China; Department of Neurology,Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Xiang Cao
- Department of Neurology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China; Department of Neurology,Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Huanyu Ni
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Shengnan Xia
- Department of Neurology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China; Department of Neurology,Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Xinyu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China; Department of Neurology,Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Qinyue Huang
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China; Department of Neurology,Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China
| | - Qingxiu Zhang
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, Nanjing, China; Department of Neurology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China; Department of Neurology,Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, China.
| |
Collapse
|
12
|
Ahmed S, Polis B, Jamwal S, Sanganahalli BG, MacDowell Kaswan Z, Islam R, Kim D, Bowers C, Giuliano L, Biederer T, Hyder F, Kaffman A. Transient impairment in microglial function causes sex-specific deficits in synaptic maturity and hippocampal function in mice exposed to early adversity. Brain Behav Immun 2024; 122:95-109. [PMID: 39134183 PMCID: PMC11402597 DOI: 10.1016/j.bbi.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 08/04/2024] [Accepted: 08/08/2024] [Indexed: 08/17/2024] Open
Abstract
Abnormal development and function of the hippocampus are two of the most consistent findings in humans and rodents exposed to early-life adversity (ELA), with males often being more affected than females. Using the limited bedding (LB) paradigm as a rodent model of ELA, we found that male adolescent mice that had been exposed to LB exhibit significant deficits in contextual fear conditioning and synaptic connectivity in the hippocampus, which are not observed in females. This is linked to altered developmental refinement of connectivity, with LB severely impairing microglial-mediated synaptic pruning in the hippocampus of male and female pups on postnatal day 17 (P17), but not in adolescent P33 mice when levels of synaptic engulfment by microglia are substantially lower. Since the rodent hippocampus undergoes intense synaptic pruning during the second and third weeks of life, we investigated whether microglia are required for the synaptic and behavioral aberrations observed in adolescent LB mice. Indeed, transient ablation of microglia from P13-21 in normally developing mice caused sex-specific behavioral and synaptic abnormalities similar to those observed in adolescent LB mice. Furthermore, chemogenetic activation of microglia during the same period reversed the microglial-mediated phagocytic deficits at P17 and restored normal contextual fear conditioning and synaptic connectivity in adolescent LB male mice. Our data support an additional contribution of astrocytes in the sex-specific effects of LB, with increased expression of the membrane receptor MEGF10 and enhanced synaptic engulfment in hippocampal astrocytes of 17-day-old LB females, but not in LB male littermates. These findings suggest a potential compensatory mechanism that may explain the relative resilience of LB females. Collectively, our study highlights a novel role for glial cells in mediating sex-specific hippocampal deficits in a mouse model of ELA.
Collapse
Affiliation(s)
- Sahabuddin Ahmed
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Baruh Polis
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Sumit Jamwal
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Basavaraju G Sanganahalli
- Department of Radiology & Biomedical Imaging and Magnetic Resonance Research Center, Yale University, New Haven, CT, 06520, USA
| | - Zoe MacDowell Kaswan
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Rafiad Islam
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Dana Kim
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Christian Bowers
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Lauryn Giuliano
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA
| | - Thomas Biederer
- Department of Neurology, Yale School of Medicine, 100 College Street, New Haven, CT 06510, USA
| | - Fahmeed Hyder
- Department of Radiology & Biomedical Imaging and Magnetic Resonance Research Center, Yale University, New Haven, CT, 06520, USA; Department of Biomedical Engineering, Yale University, New Haven, CT, 06519, USA
| | - Arie Kaffman
- Department of Psychiatry, Yale University School of Medicine, 300 George Street, Suite 901, New Haven CT, 06511, USA.
| |
Collapse
|
13
|
Huang HY, Chen YZ, Zhao C, Zheng XN, Yu K, Yue JX, Ju HQ, Shi YX, Tian L. Alternations in inflammatory macrophage niche drive phenotypic and functional plasticity of Kupffer cells. Nat Commun 2024; 15:9337. [PMID: 39472435 PMCID: PMC11522483 DOI: 10.1038/s41467-024-53659-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 10/18/2024] [Indexed: 11/02/2024] Open
Abstract
Inflammatory signals lead to recruitment of circulating monocytes and induce their differentiation into pro-inflammatory macrophages. Therefore, whether blocking inflammatory monocytes can mitigate disease progression is being actively evaluated. Here, we employ multiple lineage-tracing models and show that monocyte-derived macrophages (mo-mac) are the major population of immunosuppressive, liver metastasis-associated macrophages (LMAM), while the proportion of Kupffer cells (KC) as liver-resident macrophages is diminished in metastatic nodules. Paradoxically, genetic ablation of mo-macs results in only a marginal decrease in LMAMs. Using a proliferation-recording system and a KC-tracing model in a monocyte-deficient background, we find that LMAMs can be replenished either via increased local macrophage proliferation or by promoting KC infiltration. In the latter regard, KCs undergo transient proliferation and exhibit substantial phenotypic and functional alterations through epigenetic reprogramming following the vacating of macrophage niches by monocyte depletion. Our data thus suggest that a simultaneous blockade of monocyte recruitment and macrophage proliferation may effectively target immunosuppressive myelopoiesis and reprogram the microenvironment towards an immunostimulatory state.
Collapse
Affiliation(s)
- Han-Ying Huang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yan-Zhou Chen
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Chuang Zhao
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Xin-Nan Zheng
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Kai Yu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jia-Xing Yue
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Huai-Qiang Ju
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Yan-Xia Shi
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China
| | - Lin Tian
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P. R. China.
| |
Collapse
|
14
|
Ramos A, Ishizuka K, Hayashida A, Namkung H, Hayes LN, Srivastava R, Zhang M, Kariya T, Elkins N, Palen T, Carloni E, Tsujimura T, Calva C, Ikemoto S, Rais R, Slusher BS, Niwa M, Saito A, Saitoh T, Takimoto E, Sawa A. Nuclear GAPDH in cortical microglia mediates cellular stress-induced cognitive inflexibility. Mol Psychiatry 2024; 29:2967-2978. [PMID: 38615102 PMCID: PMC11449656 DOI: 10.1038/s41380-024-02553-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 03/12/2024] [Accepted: 04/05/2024] [Indexed: 04/15/2024]
Abstract
We report a mechanism that underlies stress-induced cognitive inflexibility at the molecular level. In a mouse model under subacute cellular stress in which deficits in rule shifting tasks were elicited, the nuclear glyceraldehyde dehydrogenase (N-GAPDH) cascade was activated specifically in microglia in the prelimbic cortex. The cognitive deficits were normalized with a pharmacological intervention with a compound (the RR compound) that selectively blocked the initiation of N-GAPDH cascade without affecting glycolytic activity. The normalization was also observed with a microglia-specific genetic intervention targeting the N-GAPDH cascade. At the mechanistic levels, the microglial secretion of High-Mobility Group Box (HMGB), which is known to bind with and regulate the NMDA-type glutamate receptors, was elevated. Consequently, the hyperactivation of the prelimbic layer 5 excitatory neurons, a neural substrate for cognitive inflexibility, was also observed. The upregulation of the microglial HMGB signaling and neuronal hyperactivation were normalized by the pharmacological and microglia-specific genetic interventions. Taken together, we show a pivotal role of cortical microglia and microglia-neuron interaction in stress-induced cognitive inflexibility. We underscore the N-GAPDH cascade in microglia, which causally mediates stress-induced cognitive alteration.
Collapse
Affiliation(s)
- Adriana Ramos
- Departments of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Koko Ishizuka
- Departments of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Arisa Hayashida
- Departments of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- International Collaborative Research Administration, Juntendo University, Tokyo, Japan
| | - Ho Namkung
- Departments of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lindsay N Hayes
- Departments of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rupali Srivastava
- Departments of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Manling Zhang
- Departments of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Taro Kariya
- Departments of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Noah Elkins
- Departments of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Trexy Palen
- Departments of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elisa Carloni
- Departments of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tsuyoshi Tsujimura
- Departments of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Coleman Calva
- Neurocircuitry of Motivation Section, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Satoshi Ikemoto
- Neurocircuitry of Motivation Section, National Institute on Drug Abuse, Baltimore, MD, USA
| | - Rana Rais
- Departments of Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Departments of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Barbara S Slusher
- Departments of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Departments of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Departments of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Departments of Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Departments of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Minae Niwa
- Departments of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Atsushi Saito
- Departments of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Eiki Takimoto
- Departments of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Akira Sawa
- Departments of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Departments of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Departments of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Departments of Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Departments of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA.
| |
Collapse
|
15
|
Robertson KV, Rodriguez AS, Cartailler JP, Shrestha S, Schleh MW, Schroeder KR, Valenti AM, Kramer AT, Harrison FE, Hasty AH. Knockdown of microglial iron import gene, Slc11a2, worsens cognitive function and alters microglial transcriptional landscape in a sex-specific manner in the APP/PS1 model of Alzheimer's disease. J Neuroinflammation 2024; 21:238. [PMID: 39334471 PMCID: PMC11438269 DOI: 10.1186/s12974-024-03238-w] [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: 06/10/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Microglial cell iron load and inflammatory activation are significant hallmarks of late-stage Alzheimer's disease (AD). In vitro, microglia preferentially upregulate the iron importer, divalent metal transporter 1 (DMT1, gene name Slc11a2) in response to inflammatory stimuli, and excess iron can augment cellular inflammation, suggesting a feed-forward loop between iron import mechanisms and inflammatory signaling. However, it is not understood whether microglial iron import mechanisms directly contribute to inflammatory signaling and chronic disease in vivo. These studies determined the effects of microglial-specific knockdown of Slc11a2 on AD-related cognitive decline and microglial transcriptional phenotype. METHODS In vitro experiments and RT-qPCR were used to assess a role for DMT1 in amyloid-β-associated inflammation. To determine the effects of microglial Slc11a2 knockdown on AD-related phenotypes in vivo, triple-transgenic Cx3cr1Cre-ERT2;Slc11a2flfl;APP/PS1+or - mice were generated and administered corn oil or tamoxifen to induce knockdown at 5-6 months of age. Both sexes underwent behavioral analyses to assess cognition and memory (12-15 months of age). Hippocampal CD11b+ microglia were magnetically isolated from female mice (15-17 months) and bulk RNA-sequencing analysis was conducted. RESULTS DMT1 inhibition in vitro robustly decreased Aβ-induced inflammatory gene expression and cellular iron levels in conditions of excess iron. In vivo, Slc11a2KD APP/PS1 female, but not male, mice displayed a significant worsening of memory function in Morris water maze and a fear conditioning assay, along with significant hyperactivity compared to control WT and APP/PS1 mice. Hippocampal microglia from Slc11a2KD APP/PS1 females displayed significant increases in Enpp2, Ttr, and the iron-export gene, Slc40a1, compared to control APP/PS1 cells. Slc11a2KD cells from APP/PS1 females also exhibited decreased expression of markers associated with subsets of disease-associated microglia (DAMs), such as Apoe, Ctsb, Ly9, Csf1, and Hif1α. CONCLUSIONS This work suggests a sex-specific role for microglial iron import gene Slc11a2 in propagating behavioral and cognitive phenotypes in the APP/PS1 model of AD. These data also highlight an association between loss of a DAM-like phenotype in microglia and cognitive deficits in Slc11a2KD APP/PS1 female mice. Overall, this work illuminates an iron-related pathway in microglia that may serve a protective role during disease and offers insight into mechanisms behind disease-related sex differences.
Collapse
Affiliation(s)
- Katrina Volk Robertson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall, Nashville, TN, USA
| | - Alec S Rodriguez
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall, Nashville, TN, USA
| | | | - Shristi Shrestha
- Creative Data Solutions, Vanderbilt Center for Stem Cell Biology, Nashville, TN, USA
| | - Michael W Schleh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall, Nashville, TN, USA
| | - Kyle R Schroeder
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall, Nashville, TN, USA
| | - Arianna M Valenti
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall, Nashville, TN, USA
| | - Alec T Kramer
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Fiona E Harrison
- Department of Medicine, Vanderbilt University Medical Center, 7465 Medical Research Building IV, 2213 Garland Avenue, Nashville, TN, 37232, USA.
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 702 Light Hall, Nashville, TN, USA.
- VA Tennessee Valley Healthcare System, Nashville, TN, USA.
| |
Collapse
|
16
|
Henningfield CM, Kwang N, Tsourmas KI, Neumann J, Kawauchi S, Swarup V, MacGregor GR, Green KN. Generation of an inducible destabilized-domain Cre mouse line to target disease associated microglia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.613773. [PMID: 39345513 PMCID: PMC11429805 DOI: 10.1101/2024.09.18.613773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
The function of microglia during progression of Alzheimer's disease (AD) can be investigated using mouse models that enable genetic manipulation of microglial subpopulations in a temporal manner. We developed a mouse strain that expresses destabilized-domain Cre recombinase (DD-Cre) from the Cst7 locus ( Cst7 DD-Cre ) and tested this in 5xFAD amyloidogenic, Ai14 tdTomato cre-reporter line mice. Dietary administration of trimethoprim to induce DD-Cre activity produces long-term labeling in disease associated microglia (DAM) without evidence of leakiness, with tdTomato-expression restricted to cells surrounding plaques. Using this model, we found that DAMs are a subset of plaque-associated microglia (PAMs) and their transition to DAM increases with age and disease stage. Spatial transcriptomic analysis revealed that tdTomato+ cells show higher expression of disease and inflammatory genes compared to other microglial populations, including non-labeled PAMs. This model should allow inducible cre-loxP targeting of DAMs, without leakiness. Highlights We developed a new mouse strain which specifically enables recombination of loxP sites in disease associated microglia (DAMs) and can be used to manipulate DAM-gene expression.DAMs represent a subset of plaque associated microglia (PAMs), and DAM expression increases with disease progression.Spatial transcriptomic analyses reveal that DAMs have higher expression of disease and inflammatory genes compared to other PAMs.
Collapse
|
17
|
Joshi R, Brezani V, Mey GM, Guixé-Muntet S, Ortega-Ribera M, Zhuang Y, Zivny A, Werneburg S, Gracia-Sancho J, Szabo G. IRF3 regulates neuroinflammatory responses and the expression of genes associated with Alzheimer's disease. J Neuroinflammation 2024; 21:212. [PMID: 39215356 PMCID: PMC11363437 DOI: 10.1186/s12974-024-03203-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
The pathological role of interferon signaling is emerging in neuroinflammatory disorders, yet, the specific role of Interferon Regulatory Factor 3 (IRF3) in neuroinflammation remains poorly understood. Here, we show that global IRF3 deficiency delays TLR4-mediated signaling in microglia and attenuates the hallmark features of LPS-induced inflammation such as cytokine release, microglial reactivity, astrocyte activation, myeloid cell infiltration, and inflammasome activation. Moreover, expression of a constitutively active IRF3 (S388D/S390D: IRF3-2D) in microglia induces a transcriptional program reminiscent of the Activated Response Microglia and the expression of genes associated with Alzheimer's disease, notably apolipoprotein-e. Using bulk-RNAseq of IRF3-2D brain myeloid cells, we identified Z-DNA binding protein-1 (ZBP1) as a target of IRF3 that is relevant across various neuroinflammatory disorders. Lastly, we show IRF3 phosphorylation and IRF3-dependent ZBP1 induction in response to Aβ in primary microglia cultures. Together, our results identify IRF3 as an important regulator of LPS and Aβ -mediated neuroinflammatory responses and highlight IRF3 as a central regulator of disease-specific gene activation in different neuroinflammatory diseases.
Collapse
Affiliation(s)
- Radhika Joshi
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA
| | - Veronika Brezani
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA
| | - Gabrielle M Mey
- Department of Opthalmology and Visual Sciences, Kellogg Eye Center Michigan Neuroscience Institute, University of Michigan, Ann Arbor, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Sergi Guixé-Muntet
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute-CIBEREHD, Barcelona, Spain
| | - Marti Ortega-Ribera
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA
| | - Yuan Zhuang
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA
| | - Adam Zivny
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA
| | - Sebastian Werneburg
- Department of Opthalmology and Visual Sciences, Kellogg Eye Center Michigan Neuroscience Institute, University of Michigan, Ann Arbor, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jordi Gracia-Sancho
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute-CIBEREHD, Barcelona, Spain
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Gyongyi Szabo
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, USA.
| |
Collapse
|
18
|
Alves M, Gil B, Villegas-Salmerón J, Salari V, Martins-Ferreira R, Arribas Blázquez M, Menéndez Méndez A, Da Rosa Gerbatin R, Smith J, de Diego-Garcia L, Conte G, Sierra-Marquez J, Merino Serrais P, Mitra M, Fernandez Martin A, Wang Y, Kesavan J, Melia C, Parras A, Beamer E, Zimmer B, Heiland M, Cavanagh B, Parcianello Cipolat R, Morgan J, Teng X, Prehn JHM, Fabene PF, Bertini G, Artalejo AR, Ballestar E, Nicke A, Olivos-Oré LA, Connolly NMC, Henshall DC, Engel T. Opposing effects of the purinergic P2X7 receptor on seizures in neurons and microglia in male mice. Brain Behav Immun 2024; 120:121-140. [PMID: 38777288 DOI: 10.1016/j.bbi.2024.05.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 04/28/2024] [Accepted: 05/19/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND The purinergic ATP-gated P2X7 receptor (P2X7R) is increasingly recognized to contribute to pathological neuroinflammation and brain hyperexcitability. P2X7R expression has been shown to be increased in the brain, including both microglia and neurons, in experimental models of epilepsy and patients. To date, the cell type-specific downstream effects of P2X7Rs during seizures remain, however, incompletely understood. METHODS Effects of P2X7R signaling on seizures and epilepsy were analyzed in induced seizure models using male mice including the kainic acid model of status epilepticus and pentylenetetrazole model and in male and female mice in a genetic model of Dravet syndrome. RNA sequencing was used to analyze P2X7R downstream signaling during seizures. To investigate the cell type-specific role of the P2X7R during seizures and epilepsy, we generated mice lacking exon 2 of the P2rx7 gene in either microglia (P2rx7:Cx3cr1-Cre) or neurons (P2rx7:Thy-1-Cre). To investigate the protective potential of overexpressing P2X7R in GABAergic interneurons, P2X7Rs were overexpressed using adeno-associated virus transduction under the mDlx promoter. RESULTS RNA sequencing of hippocampal tissue from wild-type and P2X7R knock-out mice identified both glial and neuronal genes, in particular genes involved in GABAergic signaling, under the control of the P2X7R following seizures. Mice with deleted P2rx7 in microglia displayed less severe acute seizures and developed a milder form of epilepsy, and microglia displayed an anti-inflammatory molecular profile. In contrast, mice lacking P2rx7 in neurons showed a more severe seizure phenotype when compared to epileptic wild-type mice. Analysis of single-cell expression data revealed that human P2RX7 expression is elevated in the hippocampus of patients with temporal lobe epilepsy in excitatory and inhibitory neurons. Functional studies determined that GABAergic interneurons display increased responses to P2X7R activation in experimental epilepsy. Finally, we show that viral transduction of P2X7R in GABAergic interneurons protects against evoked and spontaneous seizures in experimental temporal lobe epilepsy and in mice lacking Scn1a, a model of Dravet syndrome. CONCLUSIONS Our results suggest a dual and opposing action of P2X7R in epilepsy and suggest P2X7R overexpression in GABAergic interneurons as a novel therapeutic strategy for acquired and, possibly, genetic forms of epilepsy.
Collapse
Affiliation(s)
- Mariana Alves
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
| | - Beatriz Gil
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
| | - Javier Villegas-Salmerón
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland; The SFI Centre for Research Training in Genomics Data Science, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
| | - Valentina Salari
- Department of Neurosciences, Biomedicine and Movement Sciences, School of Medicine, University of Verona, 37134 Verona, Italy
| | - Ricardo Martins-Ferreira
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916 Badalona, Barcelona, Spain; Immunogenetics Laboratory, Molecular Pathology and Immunology, Instituto de Ciências Biomédicas Abel Salazar - Universidade do Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; Autoimmunity and Neuroscience Group, UMIB - Unit for Multidisciplinary Research in Biomedicine, ICBAS - School of Medicine and Biomedical Sciences, University of Porto, Porto, Portugal; ITR - Laboratory for Integrative and Translational Research in Population Health, Porto, Portugal
| | - Marina Arribas Blázquez
- Department of Pharmacology and Toxicology, Veterinary Faculty, Universidad Complutense de Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - Aida Menéndez Méndez
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; Department of Medicine, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28670, Villaviciosa de Odon, Spain
| | - Rogerio Da Rosa Gerbatin
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland
| | - Jonathon Smith
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland
| | - Laura de Diego-Garcia
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; Ocupharm Research Group, Faculty of Optics and Optometry, Complutense University of Madrid, Avda. Arcos de Jalon, 118 (28037), Madrid, Spain
| | - Giorgia Conte
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
| | - Juan Sierra-Marquez
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany; Laboratorio Cajal de Circuitos Corticales (CTB), Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Campus Montegancedo S/N, Pozuelo de Alarcon, 28223 Madrid, Spain; Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid 28002, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Instituto de Salud Carlos III, Madrid 28031, Spain
| | - Paula Merino Serrais
- Laboratorio Cajal de Circuitos Corticales (CTB), Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Campus Montegancedo S/N, Pozuelo de Alarcon, 28223 Madrid, Spain
| | - Meghma Mitra
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
| | - Ana Fernandez Martin
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
| | - Yitao Wang
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jaideep Kesavan
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland
| | - Ciara Melia
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; VivoArchitect, Route de la Corniche 5, 1066 Epalinges, Vaud, Switzerland
| | - Alberto Parras
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
| | - Edward Beamer
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Béla Zimmer
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Mona Heiland
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland
| | - Brenton Cavanagh
- Cellular and Molecular Imaging Core, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Rafael Parcianello Cipolat
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland
| | - James Morgan
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; Division of Developmental Biology and Medicine, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, UK
| | - Xinchen Teng
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jochen H M Prehn
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland
| | - Paolo F Fabene
- Department of Neurosciences, Biomedicine and Movement Sciences, School of Medicine, University of Verona, 37134 Verona, Italy; Section of Anatomy and Histology, Department of Neurosciences, Biomedicine, and Movement Science, Faculty of Medicine, University of Verona, Verona, Italy; Section of Innovation Biomedicine, Department of Engineering for Innovation Medicine, Faculty of Medicine, University of Verona, Verona, Italy
| | - Giuseppe Bertini
- Department of Neurosciences, Biomedicine and Movement Sciences, School of Medicine, University of Verona, 37134 Verona, Italy
| | - Antonio R Artalejo
- Department of Pharmacology and Toxicology, Veterinary Faculty, Universidad Complutense de Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - Esteban Ballestar
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916 Badalona, Barcelona, Spain; Epigenetics in Inflammatory and Metabolic Diseases Laboratory, Health Science Center (HSC), East China Normal University (ECNU), Shanghai 200241, China
| | - Annette Nicke
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Luis A Olivos-Oré
- Department of Pharmacology and Toxicology, Veterinary Faculty, Universidad Complutense de Madrid, Avda. Puerta de Hierro s/n, 28040 Madrid, Spain
| | - Niamh M C Connolly
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland
| | - David C Henshall
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland
| | - Tobias Engel
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland; FutureNeuro, SFI Research Centre for Chronic and Rare Neurological Diseases, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland.
| |
Collapse
|
19
|
Kaiser T, Dattero J, Li L, Chen M, Jiang M, Harrahill A, Butovsky O, Feng G. Transgenic Targeting of Fcrls Creates a Highly Efficient Constitutively Active Microglia Cre Line with Differentiated Specificity. eNeuro 2024; 11:ENEURO.0549-23.2024. [PMID: 38926085 PMCID: PMC11236590 DOI: 10.1523/eneuro.0549-23.2024] [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: 12/26/2023] [Revised: 04/27/2024] [Accepted: 05/13/2024] [Indexed: 06/28/2024] Open
Abstract
Microglia carry out important functions as the resident macrophages of the brain. To study their role in health and disease, the research community needs tools to genetically modify them with maximum completeness in a manner that distinguishes them from closely related cell types, such as monocytes. While currently available tamoxifen-inducible CreERT2 lines can achieve the differentiation from other cells, the field needs improved and publicly available constitutively active Cre lines, especially ones with favorable efficiency and specificity profiles for studies where high recombination efficiency is imperative and where tamoxifen administration is contraindicated. Here, we leverage the microglia-specific Fcrls gene to generate mice expressing Cre. Using genomic methods, we show correct positioning of the transgene and intact microglia homeostasis in Fcrls-2A-Cre mice. Crossing Fcrls-2A-Cre mice to four different reporters, we demonstrate highly efficient recombination in microglia across differentially sensitive loxP alleles in different genomic contexts, indicating robust applicability of the line. Further, we show that microglia recombine a loxP reporter during early embryonic development, supporting the use of the line for developmental studies. Finally, using immunofluorescence and flow cytometry, we reveal that most border-associated macrophages are also targeted whereas only few liver and spleen macrophages and virtually no white blood cell subsets exhibit Cre activity, distinguishing this line from another publicly available Cre line, Cx3cr1-CreM Fcrls-2A-Cre mice are immediately available (JAX #036591) and serve as a valuable addition to the community's microglia toolbox by providing highly efficient constitutive Cre activity with excellent specificity, particularly for studies where tamoxifen administration is undesirable.
Collapse
Affiliation(s)
- Tobias Kaiser
- McGovern Institute for Brain Research at MIT, Cambridge, Massachusetts 02139
- Departments of Brain and Cognitive Sciences, Cambridge, Massachusetts 02139
| | - Jordan Dattero
- McGovern Institute for Brain Research at MIT, Cambridge, Massachusetts 02139
- Bioengineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Liang Li
- McGovern Institute for Brain Research at MIT, Cambridge, Massachusetts 02139
- Departments of Brain and Cognitive Sciences, Cambridge, Massachusetts 02139
| | - Mandy Chen
- McGovern Institute for Brain Research at MIT, Cambridge, Massachusetts 02139
- Departments of Brain and Cognitive Sciences, Cambridge, Massachusetts 02139
| | - Minqing Jiang
- McGovern Institute for Brain Research at MIT, Cambridge, Massachusetts 02139
- Departments of Brain and Cognitive Sciences, Cambridge, Massachusetts 02139
| | - Andrew Harrahill
- McGovern Institute for Brain Research at MIT, Cambridge, Massachusetts 02139
- Departments of Brain and Cognitive Sciences, Cambridge, Massachusetts 02139
| | - Oleg Butovsky
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Guoping Feng
- McGovern Institute for Brain Research at MIT, Cambridge, Massachusetts 02139
- Departments of Brain and Cognitive Sciences, Cambridge, Massachusetts 02139
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142
| |
Collapse
|
20
|
Robertson KV, Rodriguez AS, Cartailler JP, Shrestha S, Schroeder KR, Valenti AM, Harrison FE, Hasty AH. Knockdown of microglial iron import gene, DMT1, worsens cognitive function and alters microglial transcriptional landscape in a sex-specific manner in the APP/PS1 model of Alzheimer's disease. RESEARCH SQUARE 2024:rs.3.rs-4559940. [PMID: 38978579 PMCID: PMC11230470 DOI: 10.21203/rs.3.rs-4559940/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Background Microglial cell iron load and inflammatory activation are significant hallmarks of late-stage Alzheimer's disease (AD). In vitro, microglia preferentially upregulate the iron importer, divalent metal transporter 1 (DMT1, gene name Slc11a2) in response to inflammatory stimuli, and excess iron can augment cellular inflammation, suggesting a feed-forward loop between iron import mechanisms and inflammatory signaling. However, it is not understood whether microglial iron import mechanisms directly contribute to inflammatory signaling and chronic disease in vivo. These studies determined the effects of microglial-specific knockdown of Slc11a2 on AD-related cognitive decline and microglial transcriptional phenotype. Methods In vitro experiments and RT-qPCR were used to assess a role for DMT1 in amyloid-β-associated inflammation. To determine the effects of microglial Slc11a2 knockdown on AD-related phenotypes in vivo, triple-transgenic Cx3cr1 Cre - ERT2 ;Slc11a2 flfl;APP/PS1 + or - mice were generated and administered corn oil or tamoxifen to induce knockdown at 5-6 months of age. Both sexes underwent behavioral analyses to assess cognition and memory (12-15 months of age). Hippocampal CD11b + microglia were magnetically isolated from female mice (15-17 months) and bulk RNA-sequencing analysis was conducted. Results DMT1 inhibition in vitro robustly decreased Aβ-induced inflammatory gene expression and cellular iron levels in conditions of excess iron. In vivo, Slc11a2 KD APP/PS1 female, but not male, mice displayed a significant worsening of memory function in Morris water maze and a fear conditioning assay, along with significant hyperactivity compared to control WT and APP/PS1 mice. Hippocampal microglia from Slc11a2 KD APP/PS1 females displayed significant increases in Enpp2, Ttr, and the iron-export gene, Slc40a1, compared to control APP/PS1 cells. Slc11a2 KD cells from APP/PS1 females also exhibited decreased expression of markers associated with disease-associated microglia (DAMs), such as Apoe, Ctsb, Csf1, and Hif1α. Conclusions This work suggests a sex-specific role for microglial iron import gene Slc11a2 in propagating behavioral and cognitive phenotypes in the APP/PS1 model of AD. These data also highlight an association between loss of a DAM-like phenotype in microglia and cognitive deficits in Slc11a2 KD APP/PS1 female mice. Overall, this work illuminates an iron-related pathway in microglia that may serve a protective role during disease and offers insight into mechanisms behind disease-related sex differences.
Collapse
|
21
|
Arvanitaki ES, Goulielmaki E, Gkirtzimanaki K, Niotis G, Tsakani E, Nenedaki E, Rouska I, Kefalogianni M, Xydias D, Kalafatakis I, Psilodimitrakopoulos S, Karagogeos D, Schumacher B, Stratakis E, Garinis GA. Microglia-derived extracellular vesicles trigger age-related neurodegeneration upon DNA damage. Proc Natl Acad Sci U S A 2024; 121:e2317402121. [PMID: 38635632 PMCID: PMC11047102 DOI: 10.1073/pnas.2317402121] [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: 10/09/2023] [Accepted: 03/22/2024] [Indexed: 04/20/2024] Open
Abstract
DNA damage and neurodegenerative disorders are intimately linked but the underlying mechanism remains elusive. Here, we show that persistent DNA lesions in tissue-resident macrophages carrying an XPF-ERCC1 DNA repair defect trigger neuroinflammation and neuronal cell death in mice. We find that microglia accumulate dsDNAs and chromatin fragments in the cytosol, which are sensed thereby stimulating a viral-like immune response in Er1Cx/- and naturally aged murine brain. Cytosolic DNAs are packaged into extracellular vesicles (EVs) that are released from microglia and discharge their dsDNA cargo into IFN-responsive neurons triggering cell death. To remove cytosolic dsDNAs and prevent inflammation, we developed targeting EVs to deliver recombinant DNase I to Er1Cx/- brain microglia in vivo. We show that EV-mediated elimination of cytosolic dsDNAs is sufficient to prevent neuroinflammation, reduce neuronal apoptosis, and delay the onset of neurodegenerative symptoms in Er1Cx/- mice. Together, our findings unveil a causal mechanism leading to neuroinflammation and provide a rationalized therapeutic strategy against age-related neurodegeneration.
Collapse
Affiliation(s)
- Ermioni S. Arvanitaki
- Department of Biology, University of Crete, HeraklionGR71409, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| | - Evi Goulielmaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| | - Katerina Gkirtzimanaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| | - George Niotis
- Department of Biology, University of Crete, HeraklionGR71409, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| | - Edisona Tsakani
- Department of Biology, University of Crete, HeraklionGR71409, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| | - Electra Nenedaki
- Department of Biology, University of Crete, HeraklionGR71409, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| | - Iliana Rouska
- Department of Biology, University of Crete, HeraklionGR71409, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| | - Mary Kefalogianni
- Department of Physics, University of Crete, HeraklionGR71003, Crete, Greece
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, HeraklionGR71110, Crete, Greece
| | - Dionysios Xydias
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, HeraklionGR71110, Crete, Greece
- Materials Science and Technology Department, University of Crete, HeraklionGR70013, Crete, Greece
| | - Ilias Kalafatakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
- Medical School, Division of Basic Sciences, University of Crete, HeraklionGR71003, Crete, Greece
| | - Sotiris Psilodimitrakopoulos
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, HeraklionGR71110, Crete, Greece
| | - Domna Karagogeos
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
- Medical School, Division of Basic Sciences, University of Crete, HeraklionGR71003, Crete, Greece
| | - Björn Schumacher
- Institute for Genome Stability in Ageing and Disease, Medical Faculty, University and University Hospital of Cologne, Cologne50931, Germany
- Cologne Excellence Cluster for Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne50931, Germany
| | - Emmanuel Stratakis
- Institute of Electronic Structure and Laser, Foundation for Research and Technology-Hellas, HeraklionGR71110, Crete, Greece
| | - George A. Garinis
- Department of Biology, University of Crete, HeraklionGR71409, Crete, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, HeraklionGR70013, Crete, Greece
| |
Collapse
|
22
|
Conedera FM, Kokona D, Zinkernagel MS, Stein JV, Lin CP, Alt C, Enzmann V. Macrophages coordinate immune response to laser-induced injury via extracellular traps. J Neuroinflammation 2024; 21:68. [PMID: 38500151 PMCID: PMC10949579 DOI: 10.1186/s12974-024-03064-0] [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: 01/10/2024] [Accepted: 03/13/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Retinal degeneration results from disruptions in retinal homeostasis due to injury, disease, or aging and triggers peripheral leukocyte infiltration. Effective immune responses rely on coordinated actions of resident microglia and recruited macrophages, critical for tissue remodeling and repair. However, these phagocytes also contribute to chronic inflammation in degenerated retinas, yet the precise coordination of immune response to retinal damage remains elusive. Recent investigations have demonstrated that phagocytic cells can produce extracellular traps (ETs), which are a source of self-antigens that alter the immune response, which can potentially lead to tissue injury. METHODS Innovations in experimental systems facilitate real-time exploration of immune cell interactions and dynamic responses. We integrated in vivo imaging with ultrastructural analysis, transcriptomics, pharmacological treatments, and knockout mice to elucidate the role of phagocytes and their modulation of the local inflammatory response through extracellular traps (ETs). Deciphering these mechanisms is essential for developing novel and enhanced immunotherapeutic approaches that can redirect a specific maladaptive immune response towards favorable wound healing in the retina. RESULTS Our findings underscore the pivotal role of innate immune cells, especially macrophages/monocytes, in regulating retinal repair and inflammation. The absence of neutrophil and macrophage infiltration aids parenchymal integrity restoration, while their depletion, particularly macrophages/monocytes, impedes vascular recovery. We demonstrate that macrophages/monocytes, when recruited in the retina, release chromatin and granular proteins, forming ETs. Furthermore, the pharmacological inhibition of ETosis support retinal and vascular repair, surpassing the effects of blocking innate immune cell recruitment. Simultaneously, the absence of ETosis reshapes the inflammatory response, causing neutrophils, helper, and cytotoxic T-cells to be restricted primarily in the superficial capillary plexus instead of reaching the damaged photoreceptor layer. CONCLUSIONS Our data offer novel insights into innate immunity's role in responding to retinal damage and potentially help developing innovative immunotherapeutic approaches that can shift the immune response from maladaptive to beneficial for retinal regeneration.
Collapse
Affiliation(s)
- Federica M Conedera
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland.
- Department of Ophthalmology, Bern University Hospital and Department of BioMedical Research, University of Bern, Bern, Switzerland.
| | - Despina Kokona
- Department of Ophthalmology, Bern University Hospital and Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Martin S Zinkernagel
- Department of Ophthalmology, Bern University Hospital and Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Charles P Lin
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Clemens Alt
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Volker Enzmann
- Department of Ophthalmology, Bern University Hospital and Department of BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
23
|
Joshi R, Brezani V, Mey GM, Guixé-Muntet S, Ortega-Ribera M, Zhuang Y, Zivny A, Werneburg S, Gracia-Sancho J, Szabo G. IRF3 regulates neuroinflammatory responses and the expression of genes associated with Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.582968. [PMID: 38654824 PMCID: PMC11037866 DOI: 10.1101/2024.03.08.582968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
The pathological role of interferon signaling is emerging in neuroinflammatory disorders, yet, the specific role of Interferon Regulatory Factor 3 (IRF3) in neuroinflammation remains poorly understood. Here, we show that global IRF3 deficiency delays TLR4-mediated signaling in microglia and attenuates the hallmark features of LPS-induced inflammation such as cytokine release, microglial reactivity, astrocyte activation, myeloid cell infiltration, and inflammasome activation. Moreover, expression of a constitutively active IRF3 (S388D/S390D:IRF3-2D) in microglia induces a transcriptional program reminiscent of the Activated Response Microglia and the expression of genes associated with Alzheimer's Disease, notably apolipoprotein-e. Lastly, using bulk-RNAseq of IRF3-2D brain myeloid cells, we identified Z-DNA binding protein-1 as a target of IRF3 that is relevant across various neuroinflammatory disorders. Together, our results identify IRF3 as an important regulator of LPS-mediated neuroinflammatory responses and highlight IRF3 as a central regulator of disease-specific gene activation in different neuroinflammatory diseases.
Collapse
Affiliation(s)
- Radhika Joshi
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, USA
| | - Veronika Brezani
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, USA
| | - Gabrielle M Mey
- Department of Opthalmology and Visual Sciences, Kellogg Eye Center Michigan Neuroscience Institute, University of Michigan, Ann Arbor, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Sergi Guixé-Muntet
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute- CIBEREHD, Barcelona, Spain
| | - Marti Ortega-Ribera
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, USA
| | - Yuan Zhuang
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, USA
| | - Adam Zivny
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, USA
| | - Sebastian Werneburg
- Department of Opthalmology and Visual Sciences, Kellogg Eye Center Michigan Neuroscience Institute, University of Michigan, Ann Arbor, USA
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jordi Gracia-Sancho
- Liver Vascular Biology, IDIBAPS Biomedical Research Institute- CIBEREHD, Barcelona, Spain
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Gyongyi Szabo
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, USA
| |
Collapse
|
24
|
Zhuang T, Chen MH, Wu RX, Wang J, Hu XD, Meng T, Wu AH, Li Y, Yang YF, Lei Y, Hu DH, Li YX, Zhang L, Sun AJ, Lu W, Zhang GN, Zuo JL, Ruan CC. ALKBH5-mediated m6A modification of IL-11 drives macrophage-to-myofibroblast transition and pathological cardiac fibrosis in mice. Nat Commun 2024; 15:1995. [PMID: 38443404 PMCID: PMC10914760 DOI: 10.1038/s41467-024-46357-x] [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: 08/10/2023] [Accepted: 02/24/2024] [Indexed: 03/07/2024] Open
Abstract
Cardiac macrophage contributes to the development of cardiac fibrosis, but factors that regulate cardiac macrophages transition and activation during this process remains elusive. Here we show, by single-cell transcriptomics, lineage tracing and parabiosis, that cardiac macrophages from circulating monocytes preferentially commit to macrophage-to-myofibroblast transition (MMT) under angiotensin II (Ang II)-induced hypertension, with accompanying increased expression of the RNA N6-methyladenosine demethylases, ALKBH5. Meanwhile, macrophage-specific knockout of ALKBH5 inhibits Ang II-induced MMT, and subsequently ameliorates cardiac fibrosis and dysfunction. Mechanistically, RNA immunoprecipitation sequencing identifies interlukin-11 (IL-11) mRNA as a target for ALKBH5-mediated m6A demethylation, leading to increased IL-11 mRNA stability and protein levels. By contrast, overexpression of IL11 in circulating macrophages reverses the phenotype in ALKBH5-deficient mice and macrophage. Lastly, targeted delivery of ALKBH5 or IL-11 receptor α (IL11RA1) siRNA to monocytes/macrophages attenuates MMT and cardiac fibrosis under hypertensive stress. Our results thus suggest that the ALKBH5/IL-11/IL11RA1/MMT axis alters cardiac macrophage and contributes to hypertensive cardiac fibrosis and dysfunction in mice, and thereby identify potential targets for cardiac fibrosis therapy in patients.
Collapse
Affiliation(s)
- Tao Zhuang
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, Shanghai, China
| | - Mei-Hua Chen
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, Shanghai, China
- Institute of Metabolism and Regenerative Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruo-Xi Wu
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, Shanghai, China
| | - Jing Wang
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, Shanghai, China
| | - Xi-De Hu
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, Shanghai, China
| | - Ting Meng
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, Shanghai, China
| | - Ai-Hua Wu
- Minhang Hospital and School of Pharmacy, Key Laboratory of Smart Drug Delivery Ministry of Education, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, China
| | - Yan Li
- Department of Cardiology, RuiJin Hospital/LuWan Branch, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yong-Feng Yang
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, Shanghai, China
| | - Yu Lei
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, Shanghai, China
| | - Dong-Hua Hu
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, Shanghai, China
| | - Yan-Xiu Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Li Zhang
- Department of Cardiology and Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ai-Jun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Wei Lu
- Minhang Hospital and School of Pharmacy, Key Laboratory of Smart Drug Delivery Ministry of Education, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, China.
| | - Guan-Nan Zhang
- Department of Immunology, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Jun-Li Zuo
- Department of Geriatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Cheng-Chao Ruan
- Department of Physiology and Pathophysiology, Shanghai Key Laboratory of Bioactive Small Molecules, State Key Laboratory of Medical Neurobiology, School of Basic Medical Sciences, and Jinshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
25
|
Huang R, Sun M, Wang W, Yu X, Liu F. YTHDF2 alleviates microglia activation via promoting circHIPK2 degradation. J Neuroimmunol 2024; 387:578265. [PMID: 38147784 DOI: 10.1016/j.jneuroim.2023.578265] [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: 11/22/2023] [Revised: 12/06/2023] [Accepted: 12/12/2023] [Indexed: 12/28/2023]
Abstract
Microglial activation is a common cellular dysfunction in central nervous system inflammation, accompanied by abnormal expression of circular RNAs (circRNAs). YTHDF2, a N6-methyladenosine (m6A) reader, is known as a key element in RNA degradation. Here, lipolysaccharide induced microglia activation in mouse cortex and BV2 cells, accompanied by the decreased YTHDF2 and elevated circHIPK2. YTHDF2 overexpression or circHIPK2 knockdown in BV2 microglia inhibited the expressions of iNOS protein, IL-1β mRNA and IL-6 mRNA. Subsequent experiments revealed that YTHDF2 facilitated circHIPK2 degradation, thereby alleviating microglia activation. These findings suggest that YTHDF2 overexpression could serve as a therapeutic approach for inhibiting microglia activation.
Collapse
Affiliation(s)
- Rongrong Huang
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Ming Sun
- Department of Ultrasound, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Weiwei Wang
- Department of Pathology, Qingdao Eighth People's Hospital, Qingdao 266100, China
| | - Xiaoyu Yu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214122, China.
| | - Fan Liu
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong 226001, China.
| |
Collapse
|
26
|
Bobotis BC, Halvorson T, Carrier M, Tremblay MÈ. Established and emerging techniques for the study of microglia: visualization, depletion, and fate mapping. Front Cell Neurosci 2024; 18:1317125. [PMID: 38425429 PMCID: PMC10902073 DOI: 10.3389/fncel.2024.1317125] [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: 10/10/2023] [Accepted: 01/15/2024] [Indexed: 03/02/2024] Open
Abstract
The central nervous system (CNS) is an essential hub for neuronal communication. As a major component of the CNS, glial cells are vital in the maintenance and regulation of neuronal network dynamics. Research on microglia, the resident innate immune cells of the CNS, has advanced considerably in recent years, and our understanding of their diverse functions continues to grow. Microglia play critical roles in the formation and regulation of neuronal synapses, myelination, responses to injury, neurogenesis, inflammation, and many other physiological processes. In parallel with advances in microglial biology, cutting-edge techniques for the characterization of microglial properties have emerged with increasing depth and precision. Labeling tools and reporter models are important for the study of microglial morphology, ultrastructure, and dynamics, but also for microglial isolation, which is required to glean key phenotypic information through single-cell transcriptomics and other emerging approaches. Strategies for selective microglial depletion and modulation can provide novel insights into microglia-targeted treatment strategies in models of neuropsychiatric and neurodegenerative conditions, cancer, and autoimmunity. Finally, fate mapping has emerged as an important tool to answer fundamental questions about microglial biology, including their origin, migration, and proliferation throughout the lifetime of an organism. This review aims to provide a comprehensive discussion of these established and emerging techniques, with applications to the study of microglia in development, homeostasis, and CNS pathologies.
Collapse
Affiliation(s)
- Bianca Caroline Bobotis
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology, Victoria, BC, Canada
| | - Torin Halvorson
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
- British Columbia Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Micaël Carrier
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Département de Psychiatrie et de Neurosciences, Faculté de Médecine, Université Laval, Québec City, QC, Canada
- Axe neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology, Victoria, BC, Canada
- Axe neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada
| |
Collapse
|
27
|
Keshvari S, Masson JJR, Ferrari-Cestari M, Bodea LG, Nooru-Mohamed F, Tse BWC, Sokolowski KA, Batoon L, Patkar OL, Sullivan MA, Ebersbach H, Stutz C, Parton RG, Summers KM, Pettit AR, Hume DA, Irvine KM. Reversible expansion of tissue macrophages in response to macrophage colony-stimulating factor (CSF1) transforms systemic lipid and carbohydrate metabolism. Am J Physiol Endocrinol Metab 2024; 326:E149-E165. [PMID: 38117267 DOI: 10.1152/ajpendo.00347.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/01/2023] [Accepted: 12/17/2023] [Indexed: 12/21/2023]
Abstract
Macrophages regulate metabolic homeostasis in health and disease. Macrophage colony-stimulating factor (CSF1)-dependent macrophages contribute to homeostatic control of the size of the liver. This study aimed to determine the systemic metabolic consequences of elevating circulating CSF1. Acute administration of a CSF1-Fc fusion protein to mice led to monocytosis, increased resident tissue macrophages in the liver and all major organs, and liver growth. These effects were associated with increased hepatic glucose uptake and extensive mobilization of body fat. The impacts of CSF1 on macrophage abundance, liver size, and body composition were rapidly reversed to restore homeostasis. The effects of CSF1 on metabolism were independent of several known endocrine regulators and did not impact the physiological fasting response. Analysis using implantable telemetry in metabolic cages revealed progressively reduced body temperature and physical activity with no change in diurnal food intake. These results demonstrate the existence of a dynamic equilibrium between CSF1, the mononuclear phagocyte system, and control of liver-to-body weight ratio, which in turn controls systemic metabolic homeostasis. This novel macrophage regulatory axis has the potential to promote fat mobilization, without changes in appetence, which may have novel implications for managing metabolic syndrome.NEW & NOTEWORTHY CSF1 administration expands tissue macrophages, which transforms systemic metabolism. CSF1 drives fat mobilization and glucose uptake to support liver growth. The effects of CSF1 are independent of normal hormonal metabolic regulation. The effects of CSF1 are rapidly reversible, restoring homeostatic body composition. CSF1-dependent macrophages and liver size are coupled in a dynamic equilibrium.
Collapse
Affiliation(s)
- Sahar Keshvari
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Jesse J R Masson
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Michelle Ferrari-Cestari
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Liviu-Gabriel Bodea
- Clem Jones Centre for Ageing and Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Fathima Nooru-Mohamed
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Brian W C Tse
- Preclinical Imaging Facility, Translational Research Institute, Brisbane, Queensland, Australia
| | - Kamil A Sokolowski
- Preclinical Imaging Facility, Translational Research Institute, Brisbane, Queensland, Australia
| | - Lena Batoon
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Omkar L Patkar
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Mitchell A Sullivan
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Hilmar Ebersbach
- Novartis Institutes for Biomedical Research (NIBR), Basel, Switzerland
| | - Cian Stutz
- Novartis Institutes for Biomedical Research (NIBR), Basel, Switzerland
| | - Robert G Parton
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
- Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, Queensland, Australia
| | - Kim M Summers
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Allison R Pettit
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - David A Hume
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| | - Katharine M Irvine
- Mater Research Institute-The University of Queensland, Translational Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
28
|
Volk Robertson K, Schleh MW, Harrison FE, Hasty AH. Microglial-specific knockdown of iron import gene, Slc11a2, blunts LPS-induced neuroinflammatory responses in a sex-specific manner. Brain Behav Immun 2024; 116:370-384. [PMID: 38141840 PMCID: PMC10874246 DOI: 10.1016/j.bbi.2023.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023] Open
Abstract
Neuroinflammation and microglial iron load are significant hallmarks found in several neurodegenerative diseases. In in vitro systems, microglia preferentially upregulate the iron importer, divalent metal transporter 1 (DMT1, gene name Slc11a2) in response to inflammatory stimuli, and it has been shown that iron can augment cellular inflammation, suggesting a feed-forward loop between mechanisms involved in iron import and inflammatory signaling. However, it is not understood how microglial iron import mechanisms contribute to inflammation in vivo, or whether altering a microglial iron-related gene affects the inflammatory response. These studies aimed to determine the effect of knocking down microglial iron import gene Slc11a2 on the inflammatory response in vivo. We generated a novel model of tamoxifen-inducible, microglial-specific Slc11a2 knockdown using Cx3cr1Cre-ERT2 mice. Transgenic male and female mice were administered intraperitoneal saline or lipopolysaccharide (LPS) and assessed for sickness behavior post-injection. Plasma cytokines and microglial bulk RNA sequencing (RNASeq) analyses were performed at 4 h post-LPS, and microglia were collected for gene expression analysis after 24 h. A subset of mice was assessed in a behavioral test battery following LPS-induced sickness recovery. Control male, but not female, mice significantly upregulated microglial Slc11a2 at 4 and 24 h following LPS. In Slc11a2 knockdown mice, we observed an improvement in the acute behavioral sickness response post-LPS in male, but not female, animals. Microglia from male, but not female, knockdown animals exhibited a significant decrease in LPS-provoked pro-inflammatory cytokine expression after 24 h. RNASeq data from male knockdown microglia 4 h post-LPS revealed a robust downregulation in inflammatory genes including Il6, Tnfα, and Il1β, and an increase in anti-inflammatory and homeostatic markers (e.g., Tgfbr1, Cx3cr1, and Trem2). This corresponded with a profound decrease in plasma pro-inflammatory cytokines 4 h post-LPS. At 4 h, male knockdown microglia also upregulated expression of markers of iron export, iron recycling, and iron homeostasis and decreased iron storage and import genes, along with pro-oxidant markers such as Cybb, Nos2, and Hif1α. Overall, this work elucidates how manipulating a specific gene involved in iron import in microglia alters acute inflammatory signaling and overall cell activation state in male mice. These data highlight a sex-specific link between a microglial iron import gene and the pro-inflammatory response to LPS in vivo, providing further insight into the mechanisms driving neuroinflammatory disease.
Collapse
Affiliation(s)
- Katrina Volk Robertson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Michael W Schleh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Fiona E Harrison
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; VA Tennessee Valley Healthcare System, Nashville, TN, USA.
| |
Collapse
|
29
|
Bedolla AM, McKinsey GL, Ware K, Santander N, Arnold TD, Luo Y. A comparative evaluation of the strengths and potential caveats of the microglial inducible CreER mouse models. Cell Rep 2024; 43:113660. [PMID: 38217856 PMCID: PMC10874587 DOI: 10.1016/j.celrep.2023.113660] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 10/02/2023] [Accepted: 12/20/2023] [Indexed: 01/15/2024] Open
Abstract
The recent proliferation of new Cre and CreER recombinase lines provides researchers with a diverse toolkit to study microglial gene function. To determine how best to apply these lines in studies of microglial gene function, a thorough and detailed comparison of their properties is needed. Here, we examined four different microglial CreER lines (Cx3cr1YFP-CreER(Litt), Cx3cr1CreER(Jung), P2ry12CreER, and Tmem119CreER), focusing on (1) recombination specificity, (2) leakiness (the degree of tamoxifen-independent recombination in microglia and other cells), (3) the efficiency of tamoxifen-induced recombination, (4) extraneural recombination (the degree of recombination in cells outside of the CNS, particularly myelo/monocyte lineages), and (5) off-target effects in the context of neonatal brain development. We identify important caveats and strengths for these lines, which will provide broad significance for researchers interested in performing conditional gene deletion in microglia. We also provide data emphasizing the potential of these lines for injury models that result in the recruitment of splenic immune cells.
Collapse
Affiliation(s)
- Alicia M Bedolla
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA; Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Gabriel L McKinsey
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kierra Ware
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Nicolas Santander
- Instituto de Ciencias de la Salud, Universidad de O'Higgins, Rancagua, Chile
| | - Thomas D Arnold
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yu Luo
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA; Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45229, USA; Immunology Graduate Program, Cincinnati Children's Hospital Medical Center.
| |
Collapse
|
30
|
Rusin D, Vahl Becirovic L, Lyszczarz G, Krueger M, Benmamar-Badel A, Vad Mathiesen C, Sigurðardóttir Schiöth E, Lykke Lambertsen K, Wlodarczyk A. Microglia-Derived Insulin-like Growth Factor 1 Is Critical for Neurodevelopment. Cells 2024; 13:184. [PMID: 38247874 PMCID: PMC10813844 DOI: 10.3390/cells13020184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/10/2024] [Accepted: 01/16/2024] [Indexed: 01/23/2024] Open
Abstract
Insulin-like growth factor 1 (IGF-1) is a peptide hormone essential for the proper development and growth of the organism, as a complete knockout of Igf1 in mice is lethal, causing microcephaly, growth retardation and the defective development of organs. In the central nervous system, neurons and glia have been reported to express Igf1, but their relative importance for postnatal development has not yet been fully defined. In order to address this, here, we obtained mice with a microglia-specific inducible conditional knockout of Igf1. We show that the deficiency in microglial Igf1, starting in the first postnatal week, leads to body and brain growth retardation, severely impaired myelination, changes in microglia numbers, and behavioral abnormalities. These results emphasize the importance of microglial-derived Igf1 for brain development and function and open new perspectives for the investigation of the role of microglial-Igf1 in neurological diseases.
Collapse
Affiliation(s)
- Dominika Rusin
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Lejla Vahl Becirovic
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Gabriela Lyszczarz
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Martin Krueger
- Institute for Anatomy, University of Leipzig, 04103 Leipzig, Germany
| | - Anouk Benmamar-Badel
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Cecilie Vad Mathiesen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
- Neuroscience Academy Denmark, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Eydís Sigurðardóttir Schiöth
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
- Department of Clinical Research, BRIDGE—Brain Research Interdisciplinary Guided Excellence, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
- Department of Neurology, Odense University Hospital, 5000 Odense C, Denmark
| | - Agnieszka Wlodarczyk
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
- Department of Clinical Research, BRIDGE—Brain Research Interdisciplinary Guided Excellence, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| |
Collapse
|
31
|
DePaula-Silva AB. The Contribution of Microglia and Brain-Infiltrating Macrophages to the Pathogenesis of Neuroinflammatory and Neurodegenerative Diseases during TMEV Infection of the Central Nervous System. Viruses 2024; 16:119. [PMID: 38257819 PMCID: PMC10819099 DOI: 10.3390/v16010119] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/06/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
The infection of the central nervous system (CNS) with neurotropic viruses induces neuroinflammation and is associated with the development of neuroinflammatory and neurodegenerative diseases, including multiple sclerosis and epilepsy. The activation of the innate and adaptive immune response, including microglial, macrophages, and T and B cells, while required for efficient viral control within the CNS, is also associated with neuropathology. Under healthy conditions, resident microglia play a pivotal role in maintaining CNS homeostasis. However, during pathological events, such as CNS viral infection, microglia become reactive, and immune cells from the periphery infiltrate into the brain, disrupting CNS homeostasis and contributing to disease development. Theiler's murine encephalomyelitis virus (TMEV), a neurotropic picornavirus, is used in two distinct mouse models: TMEV-induced demyelination disease (TMEV-IDD) and TMEV-induced seizures, representing mouse models of multiple sclerosis and epilepsy, respectively. These murine models have contributed substantially to our understanding of the pathophysiology of MS and seizures/epilepsy following viral infection, serving as critical tools for identifying pharmacological targetable pathways to modulate disease development. This review aims to discuss the host-pathogen interaction during a neurotropic picornavirus infection and to shed light on our current understanding of the multifaceted roles played by microglia and macrophages in the context of these two complexes viral-induced disease.
Collapse
Affiliation(s)
- Ana Beatriz DePaula-Silva
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
32
|
Ahlback A, Gentek R. Fate-Mapping Macrophages: From Ontogeny to Functions. Methods Mol Biol 2024; 2713:11-43. [PMID: 37639113 DOI: 10.1007/978-1-0716-3437-0_2] [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] [Indexed: 08/29/2023]
Abstract
Macrophages are vital to the physiological function of most tissues, but also contribute to disease through a multitude of pathological roles. They are thus highly plastic and heterogeneous. It is now well recognized that macrophages develop from several distinct progenitors from embryogenesis onwards and extending throughout life. Tissue-resident macrophages largely originate from embryonic sources and in many cases self-maintain independently without monocyte input. However, in certain tissues, monocyte-derived macrophages replace these over time or as a result of tissue injury and inflammation. This additional layer of heterogeneity has introduced many questions regarding the influence of origin on fate and function of macrophages in health and disease. To comprehensively address these questions, appropriate methods of tracing macrophage ontogeny are required. This chapter explores why ontogeny is of vital importance in macrophage biology and how to delineate macrophage populations by origin through genetic fate mapping. First, we summarize the current view of macrophage ontogeny and briefly discuss how origin may influence macrophage function in homeostasis and pathology. We go on to make the case for genetic fate mapping as the gold standard and briefly review different fate-mapping models. We then put forward our recommendations for fate-mapping strategies best suited to answer specific research questions and finally discuss the strengths and limitations of currently available models.
Collapse
Affiliation(s)
- Anna Ahlback
- The University of Edinburgh, Institute for Regeneration and Repair, Centre for Reproductive Health & Centre for Inflammation Research, Edinburgh, UK
| | - Rebecca Gentek
- The University of Edinburgh, Institute for Regeneration and Repair, Centre for Reproductive Health & Centre for Inflammation Research, Edinburgh, UK.
| |
Collapse
|
33
|
Yang EJ, Frolinger T, Iqbal U, Estill M, Shen L, Trageser KJ, Pasinetti GM. The role of the Toll like receptor 4 signaling in sex-specific persistency of depression-like behavior in response to chronic stress. Brain Behav Immun 2024; 115:169-178. [PMID: 37838079 PMCID: PMC11146676 DOI: 10.1016/j.bbi.2023.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 10/03/2023] [Accepted: 10/07/2023] [Indexed: 10/16/2023] Open
Abstract
Chronic stress is a major risk factor for Major Depressive Disorder (MDD), and it has been shown to impact the immune system and cause microglia activation in the medial prefrontal cortex (mPFC) involved in the pathogenesis of depression. The aim of this study is to further investigate cellular and molecular mechanisms underlying persistent depression behavior in sex specific manner, which is observed clinically. Here, we report that both male and female mice exhibited depression-like behavior following exposure to chronic stress. However, only female mice showed persistent depression-like behavior, which was associated with microglia activation in mPFC, characterized by distinctive alterations in the phenotype of microglia. Given these findings, to further investigate the underlying molecular mechanisms associated with persistent depression-like behavior and microglia activation in female mice, we used translating-ribosome affinity purification (TRAP). We find that Toll like receptor 4 (TLR4) signaling is casually related to persistent depression-like behavior in female mice. This is supported by the evidence that the fact that genetic ablation of TLR4 expression in microglia significantly reduced the persistent depression-like behavior to baseline levels in female mice. This study tentatively supports the hypothesis that the TLR4 signaling in microglia may be responsible for the sex differences in persistent depression-like behavior in female.
Collapse
Affiliation(s)
- Eun-Jeong Yang
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Tal Frolinger
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Umar Iqbal
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Molly Estill
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Li Shen
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Kyle J Trageser
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Giulio M Pasinetti
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States; Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, New York 10468, United States.
| |
Collapse
|
34
|
Socodato R, Almeida TO, Portugal CC, Santos ECS, Tedim-Moreira J, Galvão-Ferreira J, Canedo T, Baptista FI, Magalhães A, Ambrósio AF, Brakebusch C, Rubinstein B, Moreira IS, Summavielle T, Pinto IM, Relvas JB. Microglial Rac1 is essential for experience-dependent brain plasticity and cognitive performance. Cell Rep 2023; 42:113447. [PMID: 37980559 DOI: 10.1016/j.celrep.2023.113447] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 08/14/2023] [Accepted: 10/31/2023] [Indexed: 11/21/2023] Open
Abstract
Microglia, the largest population of brain immune cells, continuously interact with synapses to maintain brain homeostasis. In this study, we use conditional cell-specific gene targeting in mice with multi-omics approaches and demonstrate that the RhoGTPase Rac1 is an essential requirement for microglia to sense and interpret the brain microenvironment. This is crucial for microglia-synapse crosstalk that drives experience-dependent plasticity, a fundamental brain property impaired in several neuropsychiatric disorders. Phosphoproteomics profiling detects a large modulation of RhoGTPase signaling, predominantly of Rac1, in microglia of mice exposed to an environmental enrichment protocol known to induce experience-dependent brain plasticity and cognitive performance. Ablation of microglial Rac1 affects pathways involved in microglia-synapse communication, disrupts experience-dependent synaptic remodeling, and blocks the gains in learning, memory, and sociability induced by environmental enrichment. Our results reveal microglial Rac1 as a central regulator of pathways involved in the microglia-synapse crosstalk required for experience-dependent synaptic plasticity and cognitive performance.
Collapse
Affiliation(s)
- Renato Socodato
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal.
| | - Tiago O Almeida
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal; ICBAS - School of Medicine and Biomedical Sciences, Porto, Portugal
| | - Camila C Portugal
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
| | - Evelyn C S Santos
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal; Department of Biomedicine, Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - Joana Tedim-Moreira
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal; Department of Biomedicine, Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - João Galvão-Ferreira
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal; Department of Biomedicine, Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - Teresa Canedo
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
| | - Filipa I Baptista
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra Institute for Clinical and Biomedical Research (iCBR), and Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
| | - Ana Magalhães
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
| | - António F Ambrósio
- Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra Institute for Clinical and Biomedical Research (iCBR), and Clinical Academic Center of Coimbra (CACC), University of Coimbra, Coimbra, Portugal
| | - Cord Brakebusch
- Molecular Pathology Section, BRIC, Københavns Biocenter, Copenhagen, Denmark
| | | | - Irina S Moreira
- Department of Life Sciences, Center for Innovative Biomedicine and Biotechnology (CIBB) and CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Teresa Summavielle
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal; ESS.PP, Escola Superior de Saúde do Politécnico do Porto, Porto, Portugal
| | - Inês Mendes Pinto
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal; Department of Biomedicine, Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - João B Relvas
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal; Department of Biomedicine, Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal.
| |
Collapse
|
35
|
Kellogg CM, Pham K, Ko S, Cox JE, Machalinski AH, Stout MB, Sharpe AL, Beckstead MJ, Chucair-Elliott AJ, Ocañas SR, Freeman WM. Specificity and efficiency of tamoxifen-mediated Cre induction is equivalent regardless of age. iScience 2023; 26:108413. [PMID: 38058312 PMCID: PMC10696116 DOI: 10.1016/j.isci.2023.108413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/15/2023] [Accepted: 11/02/2023] [Indexed: 12/08/2023] Open
Abstract
Temporally controlling Cre recombination through tamoxifen (Tam) induction has many advantages for biomedical research. Most studies report early post-natal/juvenile (<2 m.o.) Tam induction, but age-related neurodegeneration and aging studies can require Cre induction in older mice (>12 m.o.). While anecdotally reported as problematic, there are no published comparisons of Tam-mediated Cre induction at early and late ages. Here, microglial-specific Cx3cr1creERT2 mice were crossed to a floxed NuTRAP reporter to compare Cre induction at early (3-6 m.o.) and late (20 m.o.) ages. Specificity and efficiency of microglial labeling at 21-22 m.o. were identical in mice induced with Tam at early and late ages. Age-related microglial translatomic changes were also similar regardless of Tam induction age. Each Cre and flox mouse line should be independently validated, however, these findings demonstrate that Tam-mediated Cre induction can be performed even into older mouse ages and should be generalizable to other inducible Cre models.
Collapse
Affiliation(s)
- Collyn M. Kellogg
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kevin Pham
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Sunghwan Ko
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Neuroscience Graduate Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jillian E.J. Cox
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Neuroscience Graduate Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Adeline H. Machalinski
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Michael B. Stout
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Amanda L. Sharpe
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Neuroscience Graduate Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael J. Beckstead
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Ana J. Chucair-Elliott
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Sarah R. Ocañas
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Neuroscience Graduate Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Willard M. Freeman
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
- Neuroscience Graduate Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
36
|
Morrison V, Houpert M, Trapani J, Brockman A, Kingsley P, Katdare K, Layden H, Nguena-Jones G, Trevisan A, Maguire-Zeiss K, Marnett L, Bix G, Ihrie R, Carter B. Jedi-1/MEGF12-mediated phagocytosis controls the pro-neurogenic properties of microglia in the ventricular-subventricular zone. Cell Rep 2023; 42:113423. [PMID: 37952151 PMCID: PMC10842823 DOI: 10.1016/j.celrep.2023.113423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 10/03/2023] [Accepted: 10/25/2023] [Indexed: 11/14/2023] Open
Abstract
Microglia are the primary phagocytes in the central nervous system and clear dead cells generated during development or disease. The phagocytic process shapes the microglia phenotype, which affects the local environment. A unique population of microglia resides in the ventricular-subventricular zone (V-SVZ) of neonatal mice, but how they influence the neurogenic niche is not well understood. Here, we demonstrate that phagocytosis contributes to a pro-neurogenic microglial phenotype in the V-SVZ and that these microglia phagocytose apoptotic cells via the engulfment receptor Jedi-1. Deletion of Jedi-1 decreases apoptotic cell clearance, triggering a neuroinflammatory microglia phenotype that resembles dysfunctional microglia in neurodegeneration and aging and that reduces neural precursor proliferation via elevated interleukin-1β signaling; interleukin-1 receptor inhibition rescues precursor proliferation in vivo. Together, these results reveal a critical role for Jedi-1 in connecting microglial phagocytic activity to the maintenance of a pro-neurogenic phenotype in the developing V-SVZ.
Collapse
Affiliation(s)
- Vivianne Morrison
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37235, USA; Department of Cell and Molecular Biology, Tulane University, New Orleans, LA 70118, USA
| | - Matthew Houpert
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37235, USA
| | - Jonathan Trapani
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37235, USA
| | - Asa Brockman
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37235, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37235, USA
| | - Philip Kingsley
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Ketaki Katdare
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37235, USA
| | - Hillary Layden
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA
| | - Gabriela Nguena-Jones
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37235, USA
| | - Alexandra Trevisan
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA; Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | - Lawrence Marnett
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA; Department of Chemistry, Vanderbilt University, Nashville, TN 37235, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37235, USA; A.B. Hancock Jr. Memorial Laboratory for Cancer Research, Vanderbilt-Ingram Cancer Center, Nashville, TN 37232, USA
| | - Gregory Bix
- Center for Clinical Neuroscience Research, Tulane University, New Orleans, LA 70118, USA
| | - Rebecca Ihrie
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37235, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37235, USA
| | - Bruce Carter
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, USA; Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37235, USA.
| |
Collapse
|
37
|
Kellogg CM, Pham K, Machalinski AH, Porter HL, Blankenship HE, Tooley KB, Stout MB, Rice HC, Sharpe AL, Beckstead MJ, Chucair-Elliott AJ, Ocañas SR, Freeman WM. Microglial MHC-I induction with aging and Alzheimer's is conserved in mouse models and humans. GeroScience 2023; 45:3019-3043. [PMID: 37393197 PMCID: PMC10643718 DOI: 10.1007/s11357-023-00859-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 06/21/2023] [Indexed: 07/03/2023] Open
Abstract
Major histocompatibility complex I (MHC-I) CNS cellular localization and function is still being determined after previously being thought to be absent from the brain. MHC-I expression has been reported to increase with brain aging in mouse, rat, and human whole tissue analyses, but the cellular localization was undetermined. Neuronal MHC-I is proposed to regulate developmental synapse elimination and tau pathology in Alzheimer's disease (AD). Here, we report that across newly generated and publicly available ribosomal profiling, cell sorting, and single-cell data, microglia are the primary source of classical and non-classical MHC-I in mice and humans. Translating ribosome affinity purification-qPCR analysis of 3-6- and 18-22-month-old (m.o.) mice revealed significant age-related microglial induction of MHC-I pathway genes B2m, H2-D1, H2-K1, H2-M3, H2-Q6, and Tap1 but not in astrocytes and neurons. Across a timecourse (12-23 m.o.), microglial MHC-I gradually increased until 21 m.o. and then accelerated. MHC-I protein was enriched in microglia and increased with aging. Microglial expression, and absence in astrocytes and neurons, of MHC-I-binding leukocyte immunoglobulin-like (Lilrs) and paired immunoglobin-like type 2 (Pilrs) receptor families could enable cell -autonomous MHC-I signaling and increased with aging in mice and humans. Increased microglial MHC-I, Lilrs, and Pilrs were observed in multiple AD mouse models and human AD data across methods and studies. MHC-I expression correlated with p16INK4A, suggesting an association with cellular senescence. Conserved induction of MHC-I, Lilrs, and Pilrs with aging and AD opens the possibility of cell-autonomous MHC-I signaling to regulate microglial reactivation with aging and neurodegeneration.
Collapse
Affiliation(s)
- Collyn M Kellogg
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kevin Pham
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
| | - Adeline H Machalinski
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
| | - Hunter L Porter
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
| | - Harris E Blankenship
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kyla B Tooley
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael B Stout
- Aging and Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Heather C Rice
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Amanda L Sharpe
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael J Beckstead
- Aging and Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Ana J Chucair-Elliott
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
| | - Sarah R Ocañas
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Willard M Freeman
- Genes and Human Disease Program, Oklahoma Medical Research Foundation, 825 NE 13Th Street, Oklahoma City, OK, USA.
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
38
|
Faust TE, Feinberg PA, O'Connor C, Kawaguchi R, Chan A, Strasburger H, Frosch M, Boyle MA, Masuda T, Amann L, Knobeloch KP, Prinz M, Schaefer A, Schafer DP. A comparative analysis of microglial inducible Cre lines. Cell Rep 2023; 42:113031. [PMID: 37635351 PMCID: PMC10591718 DOI: 10.1016/j.celrep.2023.113031] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 07/12/2023] [Accepted: 08/09/2023] [Indexed: 08/29/2023] Open
Abstract
Cre/loxP technology has revolutionized genetic studies and allowed for spatial and temporal control of gene expression in specific cell types. Microglial biology has particularly benefited because microglia historically have been difficult to transduce with virus or electroporation methods for gene delivery. Here, we investigate five of the most widely available microglial inducible Cre lines. We demonstrate varying degrees of recombination efficiency, cell-type specificity, and spontaneous recombination, depending on the Cre line and inter-loxP distance. We also establish best practice guidelines and protocols to measure recombination efficiency, particularly in microglia. There is increasing evidence that microglia are key regulators of neural circuits and major drivers of a broad range of neurological diseases. Reliable manipulation of their function in vivo is of utmost importance. Identifying caveats and benefits of all tools and implementing the most rigorous protocols are crucial to the growth of the field and the development of microglia-based therapeutics.
Collapse
Affiliation(s)
- Travis E Faust
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Philip A Feinberg
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Ciara O'Connor
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Riki Kawaguchi
- Department of Psychiatry and Neurology, UCLA, Los Angeles, CA 90095, USA
| | - Andrew Chan
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hayley Strasburger
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Maximilian Frosch
- Institute of Neuropathology, Medical Faculty, University of Freiburg, 79106 Freiburg, Germany
| | - Margaret A Boyle
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Takahiro Masuda
- Institute of Neuropathology, Medical Faculty, University of Freiburg, 79106 Freiburg, Germany; Division of Molecular Neuroimmunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Lukas Amann
- Institute of Neuropathology, Medical Faculty, University of Freiburg, 79106 Freiburg, Germany
| | - Klaus-Peter Knobeloch
- Institute of Neuropathology, Medical Faculty, University of Freiburg, 79106 Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, 79106 Freiburg, Germany; Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Anne Schaefer
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Max Planck Institute for Biology of Ageing, 50931 Cologne, Germany
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
39
|
Kellogg CM, Pham K, Ko S, Cox JEJ, Machalinski AH, Stout MB, Sharpe AL, Beckstead MJ, Chucair-Elliott AJ, Ocañas SR, Freeman WM. Consistent specificity and efficiency of tamoxifen-mediated cre induction across ages. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.19.558482. [PMID: 37781585 PMCID: PMC10541132 DOI: 10.1101/2023.09.19.558482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Temporally controlling cre recombination through tamoxifen (Tam) induction has many advantages for biomedical research. Most studies report Tam induction at early post-natal/juvenile (<2 m.o.) mouse ages, but age-related neurodegeneration and aging studies can require cre induction in older mice (>12 m.o.). While anecdotally reported as problematic, there are no published comparisons of Tam mediated cre induction at early and late ages. Here, microglial-specific Cx3cr1 creERT 2 mice were crossed to a floxed NuTRAP reporter to compare cre induction at early (3-6 m.o.) and late (20 m.o.) ages. Specificity and efficiency of microglial labeling at 21-22 m.o. were identical in mice induced with Tam at 3-6 m.o. or 20 m.o. of age. Age-related microglial translatomic changes were also similar regardless of Tam induction age. Each cre and flox mouse line should be validated independently, however, these findings demonstrate that Tam-mediated cre induction can be performed even into older mouse ages.
Collapse
|
40
|
Bedolla A, Wegman E, Weed M, Paranjpe A, Alkhimovitch A, Ifergan I, McClain L, Luo Y. Microglia-derived TGF-β1 ligand maintains microglia homeostasis via autocrine mechanism and is critical for normal cognitive function in adult mouse brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.05.547814. [PMID: 37461569 PMCID: PMC10349967 DOI: 10.1101/2023.07.05.547814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/23/2023]
Abstract
While TGF-β signaling is essential for microglial function, the cellular source of TGF-β ligand and its spatial regulation remains unclear in the adult CNS. Our data support that microglia, not astrocytes or neurons, are the primary producers of TGF-β1 ligands needed for microglial homeostasis. Microglia (MG)-Tgfb1 inducible knockout (iKO) leads to the activation of microglia featuring a dyshomeostatic transcriptomic profile that resembles disease-associated microglia (DAMs), injury-associated microglia, and aged microglia, suggesting that microglial self-produced TGF-β1 ligands are important in the adult CNS. Interestingly, astrocytes in MG-Tgfb1 iKO mice show a transcriptome profile that closely aligns with A1-like astrocytes. Additionally, using sparse mosaic single-cell microglia iKO of TGF-β1 ligand, we established an autocrine mechanism for TGF-β signaling. Importantly MG-Tgfb1 iKO mice show cognitive deficits, supporting that precise spatial regulation of TGF-β1 ligand derived from microglia is critical for the maintenance of brain homeostasis and normal cognitive function in the adult brain.
Collapse
Affiliation(s)
- Alicia Bedolla
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Elliot Wegman
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Max Weed
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Aditi Paranjpe
- Information Services, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Anastasia Alkhimovitch
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Igal Ifergan
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45229, USA
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Lucas McClain
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Yu Luo
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45229, USA
| |
Collapse
|
41
|
Vasek MJ, Mueller SM, Fass SB, Deajon-Jackson JD, Liu Y, Crosby HW, Koester SK, Yi J, Li Q, Dougherty JD. Local translation in microglial processes is required for efficient phagocytosis. Nat Neurosci 2023; 26:1185-1195. [PMID: 37277487 PMCID: PMC10580685 DOI: 10.1038/s41593-023-01353-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 05/03/2023] [Indexed: 06/07/2023]
Abstract
Neurons, astrocytes and oligodendrocytes locally regulate protein translation within distal processes. Here, we tested whether there is regulated local translation within peripheral microglial processes (PeMPs) from mouse brain. We show that PeMPs contain ribosomes that engage in de novo protein synthesis, and these are associated with transcripts involved in pathogen defense, motility and phagocytosis. Using a live slice preparation, we further show that acute translation blockade impairs the formation of PeMP phagocytic cups, the localization of lysosomal proteins within them, and phagocytosis of apoptotic cells and pathogen-like particles. Finally, PeMPs severed from their somata exhibit and require de novo local protein synthesis to effectively surround pathogen-like particles. Collectively, these data argue for regulated local translation in PeMPs and indicate a need for new translation to support dynamic microglial functions.
Collapse
Affiliation(s)
- Michael J Vasek
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Shayna M Mueller
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Stuart B Fass
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jelani D Deajon-Jackson
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Yating Liu
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Haley W Crosby
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
| | - Sarah K Koester
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jiwon Yi
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, Saint Louis, MO, USA
| | - Qingyun Li
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Neuroscience, Washington University School of Medicine, Saint Louis, MO, USA
| | - Joseph D Dougherty
- Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Psychiatry, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
42
|
Kellogg CM, Pham K, Machalinski AH, Porter HL, Blankenship HE, Tooley K, Stout MB, Rice HC, Sharpe AL, Beckstead MJ, Chucair-Elliott AJ, Ocañas SR, Freeman WM. Microglial MHC-I induction with aging and Alzheimer's is conserved in mouse models and humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531435. [PMID: 36945372 PMCID: PMC10028873 DOI: 10.1101/2023.03.07.531435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Major Histocompatibility Complex I (MHC-I) CNS cellular localization and function is still being determined after previously being thought to be absent from the brain. MHC-I expression has been reported to increase with brain aging in mouse, rat, and human whole tissue analyses but the cellular localization was undetermined. Neuronal MHC-I is proposed to regulate developmental synapse elimination and tau pathology in Alzheimer's disease (AD). Here we report that across newly generated and publicly available ribosomal profiling, cell sorting, and single-cell data, microglia are the primary source of classical and non-classical MHC-I in mice and humans. Translating Ribosome Affinity Purification-qPCR analysis of 3-6 and 18-22 month old (m.o.) mice revealed significant age-related microglial induction of MHC-I pathway genes B2m , H2-D1 , H2-K1 , H2-M3 , H2-Q6 , and Tap1 but not in astrocytes and neurons. Across a timecourse (12-23 m.o.), microglial MHC-I gradually increased until 21 m.o. and then accelerated. MHC-I protein was enriched in microglia and increased with aging. Microglial expression, and absence in astrocytes and neurons, of MHC-I binding Leukocyte Immunoglobulin-like (Lilrs) and Paired immunoglobin-like type 2 (Pilrs) receptor families could enable cell-autonomous MHC-I signaling and increased with aging in mice and humans. Increased microglial MHC-I, Lilrs, and Pilrs were observed in multiple AD mouse models and human AD data across methods and studies. MHC-I expression correlated with p16INK4A , suggesting an association with cellular senescence. Conserved induction of MHC-I, Lilrs, and Pilrs with aging and AD opens the possibility of cell-autonomous MHC-I signaling to regulate microglial reactivation with aging and neurodegeneration.
Collapse
Affiliation(s)
- Collyn M. Kellogg
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Kevin Pham
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Adeline H. Machalinski
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Hunter L. Porter
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Harris E. Blankenship
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kyla Tooley
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael B. Stout
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Heather C. Rice
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Amanda L. Sharpe
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael J. Beckstead
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK USA
| | - Ana J. Chucair-Elliott
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
| | - Sarah R. Ocañas
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Willard M. Freeman
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK USA
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK USA
| |
Collapse
|
43
|
Castranio EL, Hasel P, Haure-Mirande JV, Ramirez Jimenez AV, Hamilton BW, Kim RD, Glabe CG, Wang M, Zhang B, Gandy S, Liddelow SA, Ehrlich ME. Microglial INPP5D limits plaque formation and glial reactivity in the PSAPP mouse model of Alzheimer's disease. Alzheimers Dement 2023; 19:2239-2252. [PMID: 36448627 PMCID: PMC10481344 DOI: 10.1002/alz.12821] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/23/2022] [Accepted: 09/13/2022] [Indexed: 12/05/2022]
Abstract
INTRODUCTION The inositol polyphosphate-5-phosphatase D (INPP5D) gene encodes a dual-specificity phosphatase that can dephosphorylate both phospholipids and phosphoproteins. Single nucleotide polymorphisms in INPP5D impact risk for developing late onset sporadic Alzheimer's disease (LOAD). METHODS To assess the consequences of inducible Inpp5d knockdown in microglia of APPKM670/671NL /PSEN1Δexon9 (PSAPP) mice, we injected 3-month-old Inpp5dfl/fl /Cx3cr1CreER/+ and PSAPP/Inpp5dfl/fl /Cx3cr1CreER/+ mice with either tamoxifen (TAM) or corn oil (CO) to induce recombination. RESULTS At age 6 months, we found that the percent area of 6E10+ deposits and plaque-associated microglia in Inpp5d knockdown mice were increased compared to controls. Spatial transcriptomics identified a plaque-specific expression profile that was extensively altered by Inpp5d knockdown. DISCUSSION These results demonstrate that conditional Inpp5d downregulation in the PSAPP mouse increases plaque burden and recruitment of microglia to plaques. Spatial transcriptomics highlighted an extended gene expression signature associated with plaques and identified CST7 (cystatin F) as a novel marker of plaques. HIGHLIGHTS Inpp5d knockdown increases plaque burden and plaque-associated microglia number. Spatial transcriptomics identifies an expanded plaque-specific gene expression profile. Plaque-induced gene expression is altered by Inpp5d knockdown in microglia. Our plaque-associated gene signature overlaps with human Alzheimer's disease gene networks.
Collapse
Affiliation(s)
- Emilie L. Castranio
- Department of Neurology, Icahn School of Medicine at Mount
Sinai, New York, New York, USA
| | - Philip Hasel
- Neuroscience Institute, NYU Grossman School of Medicine,
New York, New York, USA
| | | | | | - B. Wade Hamilton
- Department of Neurology, Icahn School of Medicine at Mount
Sinai, New York, New York, USA
| | - Rachel D. Kim
- Neuroscience Institute, NYU Grossman School of Medicine,
New York, New York, USA
| | - Charles G. Glabe
- Department of Molecular Biology and Biochemistry,
University of California, Irvine, Irvine, California, USA
| | - Minghui Wang
- Department of Genetics and Genomic Sciences, Icahn School
of Medicine at Mount Sinai, New York, New York, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School
of Medicine at Mount Sinai, New York, New York, USA
| | - Sam Gandy
- Department of Neurology, Icahn School of Medicine at Mount
Sinai, New York, New York, USA
- Department of Psychiatry and Alzheimer’s Disease
Research Center, Icahn School of Medicine at Mount Sinai, New York, New York,
USA
- James J. Peters VA Medical Center, Bronx, New York,
USA
| | - Shane A. Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine,
New York, New York, USA
- Department of Neuroscience & Physiology, NYU Grossman
School of Medicine, New York, New York, USA
- Department of Ophthalmology, NYU Grossman School of
Medicine, New York, New York, USA
- Parekh Center for Interdisciplinary Neurology, NYU Grossman
School of Medicine, New York, New York, USA
| | - Michelle E. Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount
Sinai, New York, New York, USA
- Department of Genetics and Genomic Sciences, Icahn School
of Medicine at Mount Sinai, New York, New York, USA
- Department of Pediatrics, Icahn School of Medicine at
Mount Sinai, New York, New York, USA
| |
Collapse
|
44
|
Osse AML, Pandey RS, Wirt RA, Ortiz AA, Salazar A, Kimmich M, Toledano Strom EN, Oblak A, Lamb B, Hyman JM, Carter GW, Kinney J. Reduction in GABAB on glia induce Alzheimer's disease related changes. Brain Behav Immun 2023; 110:260-275. [PMID: 36906075 PMCID: PMC10115139 DOI: 10.1016/j.bbi.2023.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 02/26/2023] [Accepted: 03/01/2023] [Indexed: 03/13/2023] Open
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disorder characterized by beta-amyloid plaques (Aβ), neurofibrillary tangles (NFT), and neuroinflammation. Data have demonstrated that neuroinflammation contributes to Aβ and NFT onset and progression, indicating inflammation and glial signaling is vital to understanding AD. A previous investigation demonstrated a significant decrease of the GABAB receptor (GABABR) in APP/PS1 mice (Salazar et al., 2021). To determine if changes in GABABR restricted to glia serve a role in AD, we developed a mouse model with a reduction of GABABR restricted to macrophages, GAB/CX3ert. This model exhibits changes in gene expression and electrophysiological alterations similar to amyloid mouse models of AD. Crossing the GAB/CX3ert mouse with APP/PS1 resulted in significant increases in Aβ pathology. Our data demonstrates that decreased GABABR on macrophages leads to several changes observed in AD mouse models, as well as exacerbation of AD pathology when crossed with existing models. These data suggest a novel mechanism in AD pathogenesis.
Collapse
Affiliation(s)
- Amanda M Leisgang Osse
- University of Nevada, Las Vegas, Department of Brain Health, 4505 S. Maryland Parkway, Las Vegas, NV 89154, United States.
| | - Ravi S Pandey
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032, United States
| | - Ryan A Wirt
- University of Nevada, Las Vegas, Department of Psychology, 4505 S. Maryland Parkway, Las Vegas, NV 89154, United States
| | - Andrew A Ortiz
- University of Nevada, Las Vegas, Department of Brain Health, 4505 S. Maryland Parkway, Las Vegas, NV 89154, United States
| | - Arnold Salazar
- University of Nevada, Las Vegas, Department of Brain Health, 4505 S. Maryland Parkway, Las Vegas, NV 89154, United States
| | - Michael Kimmich
- University of Nevada, Las Vegas, Department of Brain Health, 4505 S. Maryland Parkway, Las Vegas, NV 89154, United States
| | - Erin N Toledano Strom
- University of Nevada, Las Vegas, Department of Brain Health, 4505 S. Maryland Parkway, Las Vegas, NV 89154, United States
| | - Adrian Oblak
- Indiana University, School of Medicine, 340 W 10(th) Street, Indianapolis, IN 46202, United States
| | - Bruce Lamb
- Indiana University, School of Medicine, 340 W 10(th) Street, Indianapolis, IN 46202, United States
| | - James M Hyman
- University of Nevada, Las Vegas, Department of Psychology, 4505 S. Maryland Parkway, Las Vegas, NV 89154, United States
| | - Gregory W Carter
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032, United States
| | - Jefferson Kinney
- University of Nevada, Las Vegas, Department of Brain Health, 4505 S. Maryland Parkway, Las Vegas, NV 89154, United States
| |
Collapse
|
45
|
Sun J, Zheng Y, Hu J. Targeting Microglia with Adeno-associated Viruses. Neurosci Bull 2023; 39:863-865. [PMID: 36333483 PMCID: PMC10169966 DOI: 10.1007/s12264-022-00975-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/13/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Jing Sun
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yufei Zheng
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Ji Hu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| |
Collapse
|
46
|
Bedolla A, Mckinsey G, Ware K, Santander N, Arnold T, Luo Y. Finding the right tool: a comprehensive evaluation of microglial inducible cre mouse models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.17.536878. [PMID: 37131606 PMCID: PMC10153116 DOI: 10.1101/2023.04.17.536878] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The recent proliferation of new Cre and CreER recombinase lines provides researchers with a diverse toolkit to study microglial gene function. To determine how best to apply these lines in studies of microglial gene function, a thorough and detailed comparison of their properties is needed. Here, we examined four different microglial CreER lines (Cx3cr1CreER(Litt), Cx3cr1CreER(Jung), P2ry12CreER, Tmem119CreER), focusing on (1) recombination specificity; (2) leakiness - degree of non-tamoxifen recombination in microglia and other cells; (3) efficiency of tamoxifen-induced recombination; (4) extra-neural recombination -the degree of recombination in cells outside the CNS, particularly myelo/monocyte lineages (5) off-target effects in the context of neonatal brain development. We identify important caveats and strengths for these lines which will provide broad significance for researchers interested in performing conditional gene deletion in microglia. We also provide data emphasizing the potential of these lines for injury models that result in the recruitment of splenic immune cells.
Collapse
Affiliation(s)
- Alicia Bedolla
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Gabriel Mckinsey
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - Kierra Ware
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Nicolas Santander
- Instituto de Ciencias de la Salud, Universidad de O´Higgins, Rancagua, Chile
| | - Thomas Arnold
- Department of Pediatrics, University of California San Francisco, San Francisco, CA 94143, USA
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - Yu Luo
- Department of Molecular and Cellular Biosciences, University of Cincinnati, Cincinnati, OH 45229, USA
- Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45229, USA
| |
Collapse
|
47
|
Ocañas SR, Ansere VA, Kellogg CM, Isola JVV, Chucair-Elliott AJ, Freeman WM. Chromosomal and gonadal factors regulate microglial sex effects in the aging brain. Brain Res Bull 2023; 195:157-171. [PMID: 36804773 PMCID: PMC10810555 DOI: 10.1016/j.brainresbull.2023.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/17/2023]
Abstract
Biological sex contributes to phenotypic sex effects through genetic (sex chromosomal) and hormonal (gonadal) mechanisms. There are profound sex differences in the prevalence and progression of age-related brain diseases, including neurodegenerative diseases. Inflammation of neural tissue is one of the most consistent age-related phenotypes seen with healthy aging and disease. The pro-inflammatory environment of the aging brain has primarily been attributed to microglial reactivity and adoption of heterogeneous reactive states dependent upon intrinsic (i.e., sex) and extrinsic (i.e., age, disease state) factors. Here, we review sex effects in microglia across the lifespan, explore potential genetic and hormonal molecular mechanisms of microglial sex effects, and discuss currently available models and methods to study sex effects in the aging brain. Despite recent attention to this area, significant further research is needed to mechanistically understand the regulation of microglial sex effects across the lifespan, which may open new avenues for sex informed prevention and treatment strategies.
Collapse
Affiliation(s)
- Sarah R Ocañas
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Victor A Ansere
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Collyn M Kellogg
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jose V V Isola
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Ana J Chucair-Elliott
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Willard M Freeman
- Genes & Human Disease Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Oklahoma City Veterans Affairs Medical Center, Oklahoma City, OK, USA; Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
48
|
Bridlance C, Thion MS. Multifaceted microglia during brain development: Models and tools. Front Neurosci 2023; 17:1125729. [PMID: 37034157 PMCID: PMC10076615 DOI: 10.3389/fnins.2023.1125729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/24/2023] [Indexed: 04/11/2023] Open
Abstract
Microglia, the brain resident macrophages, are multifaceted glial cells that belong to the central nervous and immune systems. As part of the immune system, they mediate innate immune responses, regulate brain homeostasis and protect the brain in response to inflammation or injury. At the same time, they can perform a wide array of cellular functions that relate to the normal functioning of the brain. Importantly, microglia are key actors of brain development. Indeed, these early brain invaders originate outside of the central nervous system from yolk sac myeloid progenitors, and migrate into the neural folds during early embryogenesis. Before the generation of oligodendrocytes and astrocytes, microglia thus occupy a unique position, constituting the main glial population during early development and participating in a wide array of embryonic and postnatal processes. During this developmental time window, microglia display remarkable features, being highly heterogeneous in time, space, morphology and transcriptional states. Although tremendous progress has been made in our understanding of their ontogeny and roles, there are several limitations for the investigation of specific microglial functions as well as their heterogeneity during development. This review summarizes the current murine tools and models used in the field to study the development of these peculiar cells. In particular, we focus on the methodologies used to label and deplete microglia, monitor their behavior through live-imaging and also discuss the progress currently being made by the community to unravel microglial functions in brain development and disorders.
Collapse
Affiliation(s)
- Cécile Bridlance
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Paris, France
- Collège Doctoral, Sorbonne Université, Paris, France
| | - Morgane Sonia Thion
- Institut de Biologie de l’École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Université PSL, Paris, France
| |
Collapse
|
49
|
Knockout of Rlim Results in a Sex Ratio Shift toward Males but Superovulation Cannot Compensate for the Reduced Litter Size. Animals (Basel) 2023; 13:ani13061079. [PMID: 36978620 PMCID: PMC10044649 DOI: 10.3390/ani13061079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/01/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
Technologies that can preselect offspring gender hold great promise for improving farm animal productivity and preventing human sex-related hereditary diseases. The maternal Rlim allele is required for imprinted X-chromosome inactivation, which is essential for the normal development of female mouse embryos. In this study, we inactivated the maternal Rlim allele in embryos by crossing a male transgenic mouse line carrying an X-linked CMV-Cre transgene with a female line carrying a loxP-flanked Rlim gene. Knockout of the maternal Rlim gene in embryos resulted in a male-biased sex ratio skew in the offspring. However, it also reduced litter size, and this effect was not compensated for by superovulation in the mother mice. In addition, we showed that siRNA-mediated knockdown of Rlim in mouse embryos leads to the birth of male-only progenies. This study provides a new promising method for male-biased sex selection, which may help to improve the productivity in livestock and prevent sex-associated hereditary diseases in humans.
Collapse
|
50
|
Microglia drive transient insult-induced brain injury by chemotactic recruitment of CD8 + T lymphocytes. Neuron 2023; 111:696-710.e9. [PMID: 36603584 DOI: 10.1016/j.neuron.2022.12.009] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 09/03/2022] [Accepted: 12/05/2022] [Indexed: 01/06/2023]
Abstract
The crosstalk between the nervous and immune systems has gained increasing attention for its emerging role in neurological diseases. Radiation-induced brain injury (RIBI) remains the most common medical complication of cranial radiotherapy, and its pathological mechanisms have yet to be elucidated. Here, using single-cell RNA and T cell receptor sequencing, we found infiltration and clonal expansion of CD8+ T lymphocytes in the lesioned brain tissues of RIBI patients. Furthermore, by strategies of genetic or pharmacologic interruption, we identified a chemotactic action of microglia-derived CCL2/CCL8 chemokines in mediating the infiltration of CCR2+/CCR5+ CD8+ T cells and tissue damage in RIBI mice. Such a chemotactic axis also participated in the progression of cerebral infarction in the mouse model of ischemic injury. Our findings therefore highlight the critical role of microglia in mediating the dysregulation of adaptive immune responses and reveal a potential therapeutic strategy for non-infectious brain diseases.
Collapse
|