1
|
Hu Y, Cao K, Wang F, Wu W, Mai W, Qiu L, Luo Y, Ge WP, Sun B, Shi L, Zhu J, Zhang J, Wu Z, Xie Y, Duan S, Gao Z. Dual roles of hexokinase 2 in shaping microglial function by gating glycolytic flux and mitochondrial activity. Nat Metab 2022; 4:1756-1774. [PMID: 36536134 DOI: 10.1038/s42255-022-00707-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 11/04/2022] [Indexed: 12/24/2022]
Abstract
Microglia continuously survey the brain parenchyma and actively shift status following stimulation. These processes demand a unique bioenergetic programme; however, little is known about the metabolic determinants in microglia. By mining large datasets and generating transgenic tools, here we show that hexokinase 2 (HK2), the most active isozyme associated with mitochondrial membrane, is selectively expressed in microglia in the brain. Genetic ablation of HK2 reduced microglial glycolytic flux and energy production, suppressed microglial repopulation, and attenuated microglial surveillance and damage-triggered migration in male mice. HK2 elevation is prominent in immune-challenged or disease-associated microglia. In ischaemic stroke models, however, HK2 deletion promoted neuroinflammation and potentiated cerebral damages. The enhanced inflammatory responses after HK2 ablation in microglia are associated with aberrant mitochondrial function and reactive oxygen species accumulation. Our study demonstrates that HK2 gates both glycolytic flux and mitochondrial activity to shape microglial functions, changes of which contribute to metabolic abnormalities and maladaptive inflammation in brain diseases.
Collapse
Affiliation(s)
- Yaling Hu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Kelei Cao
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Fang Wang
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Weiying Wu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Weihao Mai
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Liyao Qiu
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Yuxiang Luo
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
| | - Woo-Ping Ge
- Chinese Institute for Brain Research, Beijing, Beijing, China
| | - Binggui Sun
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
- Department of Neurobiology and Department of Anesthesiology, the Children's Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China
| | - Ligen Shi
- Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Junming Zhu
- Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianmin Zhang
- Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiying Wu
- Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yicheng Xie
- The Children's Hospital, Zhejiang, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Shumin Duan
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China.
| | - Zhihua Gao
- Department of Neurobiology and Department of Neurology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China.
| |
Collapse
|
2
|
Khakpour M, Ibáñez FG, Bordeleau M, Picard K, Mckee-Reid L, Ben-Azu B, Maggi L, Tremblay MÈ. Manual versus automatic analysis of microglial density and distribution: a comparison in the hippocampus of healthy and lipopolysaccharide-challenged mature male mice. Micron 2022; 161:103334. [DOI: 10.1016/j.micron.2022.103334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/19/2022] [Accepted: 07/29/2022] [Indexed: 10/16/2022]
|
3
|
Bosch LFP, Kierdorf K. The Shape of μ—How Morphological Analyses Shape the Study of Microglia. Front Cell Neurosci 2022; 16:942462. [PMID: 35846562 PMCID: PMC9276927 DOI: 10.3389/fncel.2022.942462] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/13/2022] [Indexed: 11/14/2022] Open
Abstract
Microglia, the innate immune cells of the CNS parenchyma, serve as the first line of defense in a myriad of neurodevelopmental, neurodegenerative, and neuroinflammatory conditions. In response to the peripheral inflammation, circulating mediators, and other external signals that are produced by these conditions, microglia dynamically employ different transcriptional programs as well as morphological adaptations to maintain homeostasis. To understand these cells’ function, the field has established a number of essential analysis approaches, such as gene expression, cell quantification, and morphological reconstruction. Although high-throughput approaches are becoming commonplace in regard to other types of analyses (e.g., single-cell scRNA-seq), a similar standard for morphological reconstruction has yet to be established. In this review, we offer an overview of microglial morphological analysis methods, exploring the advantages and disadvantages of each, highlighting a number of key studies, and emphasizing how morphological analysis has significantly contributed to our understanding of microglial function in the CNS parenchyma. In doing so, we advocate for the use of unbiased, automated morphological reconstruction approaches in future studies, in order to capitalize on the valuable information embedded in the cellular structures microglia inhabit.
Collapse
Affiliation(s)
- Lance Fredrick Pahutan Bosch
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Katrin Kierdorf
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS–Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- *Correspondence: Katrin Kierdorf,
| |
Collapse
|
4
|
Bolton JL, Short AK, Othy S, Kooiker CL, Shao M, Gunn BG, Beck J, Bai X, Law SM, Savage JC, Lambert JJ, Belelli D, Tremblay MÈ, Cahalan MD, Baram TZ. Early stress-induced impaired microglial pruning of excitatory synapses on immature CRH-expressing neurons provokes aberrant adult stress responses. Cell Rep 2022; 38:110600. [PMID: 35354026 PMCID: PMC9014810 DOI: 10.1016/j.celrep.2022.110600] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 02/10/2022] [Accepted: 03/08/2022] [Indexed: 12/12/2022] Open
Abstract
Several mental illnesses, characterized by aberrant stress reactivity, often arise after early-life adversity (ELA). However, it is unclear how ELA affects stress-related brain circuit maturation, provoking these enduring vulnerabilities. We find that ELA increases functional excitatory synapses onto stress-sensitive hypothalamic corticotropin-releasing hormone (CRH)-expressing neurons, resulting from disrupted developmental synapse pruning by adjacent microglia. Microglial process dynamics and synaptic element engulfment were attenuated in ELA mice, associated with deficient signaling of the microglial phagocytic receptor MerTK. Accordingly, selective chronic chemogenetic activation of ELA microglia increased microglial process dynamics and reduced excitatory synapse density to control levels. Notably, selective early-life activation of ELA microglia normalized adult acute and chronic stress responses, including stress-induced hormone secretion and behavioral threat responses, as well as chronic adrenal hypertrophy of ELA mice. Thus, microglial actions during development are powerful contributors to mechanisms by which ELA sculpts the connectivity of stress-regulating neurons, promoting vulnerability to stress and stress-related mental illnesses.
Collapse
Affiliation(s)
- Jessica L Bolton
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA.
| | - Annabel K Short
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA
| | - Shivashankar Othy
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Cassandra L Kooiker
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA
| | - Manlin Shao
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA
| | - Benjamin G Gunn
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA; Division of Neuroscience, Medical Research Institute, Dundee University, Ninewells Hospital and Medical School, Dundee, UK
| | - Jaclyn Beck
- Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA
| | - Xinglong Bai
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA
| | - Stephanie M Law
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA
| | - Julie C Savage
- Département de Médecine Moléculaire, Université Laval, Québec City, QC, Canada; Axe Neurosciences, Centre de recherche du CHU de Québec, Québec City, QC, Canada
| | - Jeremy J Lambert
- Division of Neuroscience, Medical Research Institute, Dundee University, Ninewells Hospital and Medical School, Dundee, UK
| | - Delia Belelli
- Division of Neuroscience, Medical Research Institute, Dundee University, Ninewells Hospital and Medical School, Dundee, UK
| | - Marie-Ève Tremblay
- Département de Médecine Moléculaire, Université Laval, Québec City, QC, Canada; Axe Neurosciences, Centre de recherche du CHU de Québec, Québec City, QC, Canada
| | - Michael D Cahalan
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Tallie Z Baram
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA; Department of Anatomy/Neurobiology, University of California, Irvine, Irvine, CA, USA; Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
5
|
Susceptibility of Female Mice to the Dietary Omega-3/Omega-6 Fatty-Acid Ratio: Effects on Adult Hippocampal Neurogenesis and Glia. Int J Mol Sci 2022; 23:ijms23063399. [PMID: 35328825 PMCID: PMC8950413 DOI: 10.3390/ijms23063399] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/15/2022] [Accepted: 03/18/2022] [Indexed: 02/04/2023] Open
Abstract
Maternal intake of omega-3 (n-3 PUFAs) and omega-6 (n-6 PUFAs) polyunsaturated fatty acids impacts hippocampal neurogenesis during development, an effect that may extend to adulthood by altering adult hippocampal neurogenesis (AHN). The n-3 PUFAs and n-6 PUFAs are precursors of inflammatory regulators that potentially affect AHN and glia. Additionally, n-3 PUFA dietary supplementation may present a sexually dimorphic action in the brain. Therefore, we postulated that dietary n-6/n-3 PUFA balance shapes the adult DG in a sex-dependent manner influencing AHN and glia. We test our hypothesis by feeding adult female and male mice with n-3 PUFA balanced or deficient diets. To analyze the immunomodulatory potential of the diets, we injected mice with the bacterial endotoxin lipopolysaccharide (LPS). LPS reduced neuroblast number, and its effect was exacerbated by the n-3 PUFA-deficient diet. The n-3 PUFA-deficient diet reduced the DG volume, AHN, microglia number, and surveilled volume. The diet effect on most mature neuroblasts was exclusively significant in female mice. Colocalization and multivariate analysis revealed an association between microglia and AHN, as well as the sexual dimorphic effect of diet. Our study reveals that female mice are more susceptible than males to the effect of dietary n-6/n-3 PUFA ratio on AHN and microglia.
Collapse
|
6
|
Losenkova K, Takeda A, Ragauskas S, Cerrada-Gimenez M, Vähätupa M, Kaja S, Paul ML, Schmies CC, Rolshoven G, Müller CE, Sandholm J, Jalkanen S, Kalesnykas G, Yegutkin GG. CD73 controls ocular adenosine levels and protects retina from light-induced phototoxicity. Cell Mol Life Sci 2022; 79:152. [PMID: 35212809 PMCID: PMC8881442 DOI: 10.1007/s00018-022-04187-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 01/28/2022] [Accepted: 02/04/2022] [Indexed: 01/03/2023]
Abstract
ATP and adenosine have emerged as important signaling molecules involved in vascular remodeling, retinal functioning and neurovascular coupling in the mammalian eye. However, little is known about the regulatory mechanisms of purinergic signaling in the eye. Here, we used three-dimensional multiplexed imaging, in situ enzyme histochemistry, flow cytometric analysis, and single cell transcriptomics to characterize the whole pattern of purine metabolism in mouse and human eyes. This study identified ecto-nucleoside triphosphate diphosphohydrolase-1 (NTPDase1/CD39), NTPDase2, and ecto-5′-nucleotidase/CD73 as major ocular ecto-nucleotidases, which are selectively expressed in the photoreceptor layer (CD73), optic nerve head, retinal vasculature and microglia (CD39), as well as in neuronal processes and cornea (CD39, NTPDase2). Specifically, microglial cells can create a spatially arranged network in the retinal parenchyma by extending and retracting their branched CD39high/CD73low processes and forming local “purinergic junctions” with CD39low/CD73− neuronal cell bodies and CD39high/CD73− retinal blood vessels. The relevance of the CD73–adenosine pathway was confirmed by flash electroretinography showing that pharmacological inhibition of adenosine production by injection of highly selective CD73 inhibitor PSB-12489 in the vitreous cavity of dark-adapted mouse eyes rendered the animals hypersensitive to prolonged bright light, manifested as decreased a-wave and b-wave amplitudes. The impaired electrical responses of retinal cells in PSB-12489-treated mice were not accompanied by decrease in total thickness of the retina or death of photoreceptors and retinal ganglion cells. Our study thus defines ocular adenosine metabolism as a complex and spatially integrated network and further characterizes the critical role of CD73 in maintaining the functional activity of retinal cells.
Collapse
Affiliation(s)
- Karolina Losenkova
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Tykistökatu 6A, 20520, Turku, Finland
| | - Akira Takeda
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Tykistökatu 6A, 20520, Turku, Finland
| | | | | | | | - Simon Kaja
- Experimentica Ltd., Kuopio, Finland.,Department of Ophthalmology, Loyola University Chicago, Stritch School of Medicine, Maywood, IL, USA
| | - Marius L Paul
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Tykistökatu 6A, 20520, Turku, Finland.,Pharma Center Bonn, Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Constanze C Schmies
- Pharma Center Bonn, Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Georg Rolshoven
- Pharma Center Bonn, Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Christa E Müller
- Pharma Center Bonn, Pharmaceutical Institute, Pharmaceutical and Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Jouko Sandholm
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Sirpa Jalkanen
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Tykistökatu 6A, 20520, Turku, Finland
| | | | - Gennady G Yegutkin
- MediCity Research Laboratory and InFLAMES Flagship, University of Turku, Tykistökatu 6A, 20520, Turku, Finland.
| |
Collapse
|
7
|
Rotterman TM, Alvarez FJ. Microglia Dynamics and Interactions with Motoneurons Axotomized After Nerve Injuries Revealed By Two-Photon Imaging. Sci Rep 2020; 10:8648. [PMID: 32457369 PMCID: PMC7250868 DOI: 10.1038/s41598-020-65363-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 05/01/2020] [Indexed: 01/08/2023] Open
Abstract
The significance of activated microglia around motoneurons axotomized after nerve injuries has been intensely debated. In particular, whether microglia become phagocytic is controversial. To resolve these issues we directly observed microglia behaviors with two-photon microscopy in ex vivo spinal cord slices from CX3CR1-GFP mice complemented with confocal analyses of CD68 protein. Axotomized motoneurons were retrogradely-labeled from muscle before nerve injuries. Microglia behaviors close to axotomized motoneurons greatly differ from those within uninjured motor pools. They develop a phagocytic phenotype as early as 3 days after injury, characterized by frequent phagocytic cups, high phagosome content and CD68 upregulation. Interactions between microglia and motoneurons changed with time after axotomy. Microglia first extend processes that end in phagocytic cups at the motoneuron surface, then they closely attach to the motoneuron while extending filopodia over the cell body. Confocal 3D analyses revealed increased microglia coverage of the motoneuron cell body surface with time after injury and the presence of CD68 granules in microglia surfaces opposed to motoneurons. Some microglia formed macroclusters associated with dying motoneurons. Microglia in these clusters display the highest CD68 expression and associate with cytotoxic T-cells. These observations are discussed in relation to current theories on microglia function around axotomized motoneurons.
Collapse
Affiliation(s)
- Travis M Rotterman
- Department of Physiology, Emory University, Atlanta, GA, 30322, United States of America.,School of Biological Sciences, Georgia Tech, Atlanta, GA, 30318, United States of America
| | - Francisco J Alvarez
- Department of Physiology, Emory University, Atlanta, GA, 30322, United States of America.
| |
Collapse
|
8
|
Xu ZX, Kim GH, Tan JW, Riso AE, Sun Y, Xu EY, Liao GY, Xu H, Lee SH, Do NY, Lee CH, Clipperton-Allen AE, Kwon S, Page DT, Lee KJ, Xu B. Elevated protein synthesis in microglia causes autism-like synaptic and behavioral aberrations. Nat Commun 2020; 11:1797. [PMID: 32286273 PMCID: PMC7156673 DOI: 10.1038/s41467-020-15530-3] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 03/11/2020] [Indexed: 12/21/2022] Open
Abstract
Mutations that inactivate negative translation regulators cause autism spectrum disorders (ASD), which predominantly affect males and exhibit social interaction and communication deficits and repetitive behaviors. However, the cells that cause ASD through elevated protein synthesis resulting from these mutations remain unknown. Here we employ conditional overexpression of translation initiation factor eIF4E to increase protein synthesis in specific brain cells. We show that exaggerated translation in microglia, but not neurons or astrocytes, leads to autism-like behaviors in male mice. Although microglial eIF4E overexpression elevates translation in both sexes, it only increases microglial density and size in males, accompanied by microglial shift from homeostatic to a functional state with enhanced phagocytic capacity but reduced motility and synapse engulfment. Consequently, cortical neurons in the mice have higher synapse density, neuroligins, and excitation-to-inhibition ratio compared to control mice. We propose that functional perturbation of male microglia is an important cause for sex-biased ASD.
Collapse
Affiliation(s)
- Zhi-Xiang Xu
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA
| | - Gyu Hyun Kim
- Synaptic Circuit Plasticity Lab, Korea Brain Research Institute, Daegu, 41062, Korea
| | - Ji-Wei Tan
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA
| | - Anna E Riso
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA
- The Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Ye Sun
- Integrative Program in Biology and Neuroscience, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Ethan Y Xu
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA
- The Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Guey-Ying Liao
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA
| | - Haifei Xu
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA
| | - Sang-Hoon Lee
- Advanced Neural Imaging Center, Department of Structure & Function of Neural Network, Korea Brain Research Institute, Daegu, 41062, Korea
| | - Na-Young Do
- Synaptic Circuit Plasticity Lab, Korea Brain Research Institute, Daegu, 41062, Korea
| | - Chan Hee Lee
- Synaptic Circuit Plasticity Lab, Korea Brain Research Institute, Daegu, 41062, Korea
| | - Amy E Clipperton-Allen
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA
| | - Soonwook Kwon
- Department of Anatomy, Catholic University of Daegu, Daegu, 42472, Korea
| | - Damon T Page
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA
| | - Kea Joo Lee
- Synaptic Circuit Plasticity Lab, Korea Brain Research Institute, Daegu, 41062, Korea
| | - Baoji Xu
- Department of Neuroscience, The Scripps Research Institute Florida, Jupiter, FL, 33458, USA.
| |
Collapse
|
9
|
Communication, Cross Talk, and Signal Integration in the Adult Hippocampal Neurogenic Niche. Neuron 2020; 105:220-235. [PMID: 31972145 DOI: 10.1016/j.neuron.2019.11.029] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/21/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022]
Abstract
Radial glia-like neural stem cells (RGLs) in the dentate gyrus subregion of the hippocampus give rise to dentate granule cells (DGCs) and astrocytes throughout life, a process referred to as adult hippocampal neurogenesis. Adult hippocampal neurogenesis is sensitive to experiences, suggesting that it may represent an adaptive mechanism by which hippocampal circuitry is modified in response to environmental demands. Experiential information is conveyed to RGLs, progenitors, and adult-born DGCs via the neurogenic niche that is composed of diverse cell types, extracellular matrix, and afferents. Understanding how the niche performs its functions may guide strategies to maintain its health span and provide a permissive milieu for neurogenesis. Here, we first discuss representative contributions of niche cell types to regulation of neural stem cell (NSC) homeostasis and maturation of adult-born DGCs. We then consider mechanisms by which the activity of multiple niche cell types may be coordinated to communicate signals to NSCs. Finally, we speculate how NSCs integrate niche-derived signals to govern their regulation.
Collapse
|
10
|
Neurons, Glia, Extracellular Matrix and Neurovascular Unit: A Systems Biology Approach to the Complexity of Synaptic Plasticity in Health and Disease. Int J Mol Sci 2020; 21:ijms21041539. [PMID: 32102370 PMCID: PMC7073232 DOI: 10.3390/ijms21041539] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 02/06/2023] Open
Abstract
The synaptic cleft has been vastly investigated in the last decades, leading to a novel and fascinating model of the functional and structural modifications linked to synaptic transmission and brain processing. The classic neurocentric model encompassing the neuronal pre- and post-synaptic terminals partly explains the fine-tuned plastic modifications under both pathological and physiological circumstances. Recent experimental evidence has incontrovertibly added oligodendrocytes, astrocytes, and microglia as pivotal elements for synapse formation and remodeling (tripartite synapse) in both the developing and adult brain. Moreover, synaptic plasticity and its pathological counterpart (maladaptive plasticity) have shown a deep connection with other molecular elements of the extracellular matrix (ECM), once considered as a mere extracellular structural scaffold altogether with the cellular glue (i.e., glia). The ECM adds another level of complexity to the modern model of the synapse, particularly, for the long-term plasticity and circuit maintenance. This model, called tetrapartite synapse, can be further implemented by including the neurovascular unit (NVU) and the immune system. Although they were considered so far as tightly separated from the central nervous system (CNS) plasticity, at least in physiological conditions, recent evidence endorsed these elements as structural and paramount actors in synaptic plasticity. This scenario is, as far as speculations and evidence have shown, a consistent model for both adaptive and maladaptive plasticity. However, a comprehensive understanding of brain processes and circuitry complexity is still lacking. Here we propose that a better interpretation of the CNS complexity can be granted by a systems biology approach through the construction of predictive molecular models that enable to enlighten the regulatory logic of the complex molecular networks underlying brain function in health and disease, thus opening the way to more effective treatments.
Collapse
|
11
|
Savage JC, St-Pierre MK, Hui CW, Tremblay ME. Microglial Ultrastructure in the Hippocampus of a Lipopolysaccharide-Induced Sickness Mouse Model. Front Neurosci 2019; 13:1340. [PMID: 31920505 PMCID: PMC6932978 DOI: 10.3389/fnins.2019.01340] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 11/27/2019] [Indexed: 12/13/2022] Open
Abstract
Sickness behavior is a set of behavioral changes induced by infections and mediated by pro-inflammatory cytokines. It is characterized by fatigue, decreased appetite and weight loss, changes in sleep patterns, cognitive functions, and lost interest in social activity. It can expedite recovery by conserving energy to mount an immune response involving innate immunity. To provide insights into microglial implication in sickness behavior with special focus on cognitive and social impairment, we investigated changes in their ultrastructure and interactions with synapses using a toxemia mouse model. Adult mice were injected with 1 mg/kg lipopolysaccharide (LPS) or saline, and assayed for signs of sickness behavior. LPS treated mice displayed reduced activity in open-field tests 24 h post-injection, while social avoidance and weight gain/loss were not significantly different between treatment groups. Microglia were investigated using electron microscopy to describe changes in their structure and function at nanoscale resolution. Microglial cell bodies and processes were investigated in the hippocampus CA1, a region responsible for learning and memory that is often impacted after peripheral LPS administration. Microglia in LPS treated animals displayed larger cell bodies as well as less complex processes at the time point examined. Strikingly, microglial processes in LPS injected animals were also more likely to contact excitatory synapses and contained more phagocytic material compared with saline injected controls. We have identified at the ultrastructural level significant changes in microglia-synapse interactions shortly after LPS administration, which draws attention to studying the roles of microglia in synaptic rewiring after inflammatory stimuli.
Collapse
Affiliation(s)
- Julie C Savage
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Quebec, QC, Canada.,Départment de Médecine Moléculaire, Faculté de Médecine, Université Laval, Quebec, QC, Canada
| | - Marie-Kim St-Pierre
- Départment de Médecine Moléculaire, Faculté de Médecine, Université Laval, Quebec, QC, Canada
| | - Chin Wai Hui
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Quebec, QC, Canada.,Départment de Médecine Moléculaire, Faculté de Médecine, Université Laval, Quebec, QC, Canada.,Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong
| | - Marie-Eve Tremblay
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Quebec, QC, Canada.,Départment de Médecine Moléculaire, Faculté de Médecine, Université Laval, Quebec, QC, Canada
| |
Collapse
|
12
|
Miller-Rhodes P, Kong C, Baht GS, Saminathan P, Rodriguiz RM, Wetsel WC, Gelbard HA, Terrando N. The broad spectrum mixed-lineage kinase 3 inhibitor URMC-099 prevents acute microgliosis and cognitive decline in a mouse model of perioperative neurocognitive disorders. J Neuroinflammation 2019; 16:193. [PMID: 31660984 PMCID: PMC6816182 DOI: 10.1186/s12974-019-1582-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 09/10/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Patients with pre-existing neurodegenerative disease commonly experience fractures that require orthopedic surgery. Perioperative neurocognitive disorders (PND), including delirium and postoperative cognitive dysfunction, are serious complications that can result in increased 1-year mortality when superimposed on dementia. Importantly, there are no disease-modifying therapeutic options for PND. Our lab developed the "broad spectrum" mixed-lineage kinase 3 inhibitor URMC-099 to inhibit pathological innate immune responses that underlie neuroinflammation-associated cognitive dysfunction. Here, we test the hypothesis that URMC-099 can prevent surgery-induced neuroinflammation and cognitive impairment. METHODS Orthopedic surgery was performed by fracturing the tibia of the left hindlimb with intramedullary fixation under general anesthesia and analgesia. In a pilot experiment, 9-month-old mice were treated five times with URMC-099 (10 mg/kg, i.p.), spaced 12 h apart, with three doses prior to surgery and two doses following surgery. In this experiment, microgliosis was evaluated using unbiased stereology and blood-brain barrier (BBB) permeability was assessed using immunoglobulin G (IgG) immunostaining. In follow-up experiments, 3-month-old mice were treated only three times with URMC-099 (10 mg/kg, i.p.), spaced 12 h apart, prior to orthopedic surgery. Two-photon scanning laser microscopy and CLARITY with light-sheet microscopy were used to define surgery-induced changes in microglial dynamics and morphology, respectively. Surgery-induced memory impairment was assessed using the "What-Where-When" and Memory Load Object Discrimination tasks. The acute peripheral immune response to surgery was assessed by cytokine/chemokine profiling and flow cytometry. Finally, long-term fracture healing was assessed in fracture callouses using micro-computerized tomography (microCT) and histomorphometry analyses. RESULTS Orthopedic surgery induced BBB disruption and microglial activation, but had no effect on microglial process motility. Surgically treated mice exhibited impaired object place and identity discrimination in the "What-Where-When" and Memory Load Object Discrimination tasks. Both URMC-099 dosing paradigms prevented the neuroinflammatory sequelae that accompanied orthopedic surgery. URMC-099 prophylaxis had no effect on the mobilization of the peripheral innate immune response and fracture healing. CONCLUSIONS These findings show that prophylactic URMC-099 treatment is sufficient to prevent surgery-induced microgliosis and cognitive impairment without affecting fracture healing. Together, these findings provide compelling evidence for the advancement of URMC-099 as a therapeutic option for PND.
Collapse
Affiliation(s)
- Patrick Miller-Rhodes
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, NY 14642 USA
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY 14642 USA
| | - Cuicui Kong
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710 USA
| | - Gurpreet S. Baht
- Department of Orthopedic Surgery and Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC 27710 USA
| | - Priyanka Saminathan
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642 USA
| | - Ramona M. Rodriguiz
- Department of Psychiatry and Behavioral Sciences, Mouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University Medical Center, Durham, NC 27710 USA
| | - William C. Wetsel
- Department of Psychiatry and Behavioral Sciences, Mouse Behavioral and Neuroendocrine Analysis Core Facility, Duke University Medical Center, Durham, NC 27710 USA
- Departments of Neurobiology and Cell Biology, Duke University Medical Center, Durham, NC 27710 USA
| | - Harris A. Gelbard
- Center for Neurotherapeutics Discovery, University of Rochester Medical Center, Rochester, NY 14642 USA
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY 14642 USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642 USA
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642 USA
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642 USA
| | - Niccolò Terrando
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710 USA
| |
Collapse
|
13
|
Stratoulias V, Venero JL, Tremblay M, Joseph B. Microglial subtypes: diversity within the microglial community. EMBO J 2019; 38:e101997. [PMID: 31373067 PMCID: PMC6717890 DOI: 10.15252/embj.2019101997] [Citation(s) in RCA: 340] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/29/2019] [Accepted: 05/03/2019] [Indexed: 12/03/2022] Open
Abstract
Microglia are brain-resident macrophages forming the first active immune barrier in the central nervous system. They fulfill multiple functions across development and adulthood and under disease conditions. Current understanding revolves around microglia acquiring distinct phenotypes upon exposure to extrinsic cues in their environment. However, emerging evidence suggests that microglia display differences in their functions that are not exclusively driven by their milieu, rather by the unique properties these cells possess. This microglial intrinsic heterogeneity has been largely overlooked, favoring the prevailing view that microglia are a single-cell type endowed with spectacular plasticity, allowing them to acquire multiple phenotypes and thereby fulfill their numerous functions in health and disease. Here, we review the evidence that microglia might form a community of cells in which each member (or "subtype") displays intrinsic properties and performs unique functions. Distinctive features and functional implications of several microglial subtypes are considered, across contexts of health and disease. Finally, we suggest that microglial subtype categorization shall be based on function and we propose ways for studying them. Hence, we advocate that plasticity (reaction states) and diversity (subtypes) should both be considered when studying the multitasking microglia.
Collapse
Affiliation(s)
- Vassilis Stratoulias
- Toxicology UnitInstitute of Environmental MedicineKarolinska InstitutetStockholmSweden
| | - Jose Luis Venero
- Departamento de Bioquímica y Biología MolecularFacultad de FarmaciaUniversidad de SevillaSevillaSpain
- Instituto de Biomedicina de Sevilla‐Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevillaSpain
| | - Marie‐Ève Tremblay
- Department of Molecular MedicineUniversité LavalQuebecQCCanada
- Axe NeurosciencesCentre de Recherche du CHU de Québec‐Université LavalQuebecQCCanada
| | - Bertrand Joseph
- Toxicology UnitInstitute of Environmental MedicineKarolinska InstitutetStockholmSweden
| |
Collapse
|
14
|
Rodríguez-Iglesias N, Sierra A, Valero J. Rewiring of Memory Circuits: Connecting Adult Newborn Neurons With the Help of Microglia. Front Cell Dev Biol 2019; 7:24. [PMID: 30891446 PMCID: PMC6411767 DOI: 10.3389/fcell.2019.00024] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 02/08/2019] [Indexed: 12/16/2022] Open
Abstract
New neurons are continuously generated from stem cells and integrated into the adult hippocampal circuitry, contributing to memory function. Several environmental, cellular, and molecular factors regulate the formation of new neurons, but the mechanisms that govern their incorporation into memory circuits are less explored. Herein we will focus on microglia, the resident immune cells of the CNS, which modulate the production of new neurons in the adult hippocampus and are also well suited to participate in their circuit integration. Microglia may contribute to the refinement of brain circuits during development and exert a role in physiological and pathological conditions by regulating axonal and dendritic growth; promoting the formation, elimination, and relocation of synapses; modulating excitatory synaptic maturation; and participating in functional synaptic plasticity. Importantly, microglia are able to sense subtle changes in their environment and may use this information to differently modulate hippocampal wiring, ultimately impacting on memory function. Deciphering the role of microglia in hippocampal circuitry constant rewiring will help to better understand the influence of microglia on memory function.
Collapse
Affiliation(s)
- Noelia Rodríguez-Iglesias
- Laboratory of Glial Cell Biology, Achucarro Basque Center for Neuroscience, Leioa, Spain.,Department of Neuroscience, University of the Basque Country UPV/EHU, Leioa, Spain
| | - Amanda Sierra
- Laboratory of Glial Cell Biology, Achucarro Basque Center for Neuroscience, Leioa, Spain.,Department of Neuroscience, University of the Basque Country UPV/EHU, Leioa, Spain.,Ikerbasque Foundation, Bilbao, Spain
| | - Jorge Valero
- Laboratory of Glial Cell Biology, Achucarro Basque Center for Neuroscience, Leioa, Spain.,Department of Neuroscience, University of the Basque Country UPV/EHU, Leioa, Spain.,Ikerbasque Foundation, Bilbao, Spain
| |
Collapse
|
15
|
Kyrargyri V, Attwell D, Jolivet RB, Madry C. Analysis of Signaling Mechanisms Regulating Microglial Process Movement. Methods Mol Biol 2019; 2034:191-205. [PMID: 31392686 DOI: 10.1007/978-1-4939-9658-2_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Microglia, the brain's innate immune cells, are extremely motile cells, continuously surveying the central nervous system (CNS) to serve homeostatic functions and to respond to pathological events. In the healthy brain, microglia exhibit a small cell body with long, branched, and highly motile processes, which constantly extend and retract, effectively "patrolling" the brain parenchyma. Over the last decade, methodological advances in microscopy and the availability of genetically encoded reporter mice have allowed us to probe microglial physiology in situ. Beyond their classical immunological roles, unexpected functions of microglia have been revealed, both in the developing and the adult brain: microglia regulate the generation of newborn neurons, control the formation and elimination of synapses, and modulate neuronal activity. Many of these newly ascribed functions depend directly on microglial process movement. Thus, elucidating the mechanisms underlying microglial motility is of great importance to understand their role in brain physiology and pathophysiology. Two-photon imaging of fluorescently labeled microglia, either in vivo or ex vivo in acute brain slices, has emerged as an indispensable tool for investigating microglial movements and their functional consequences. This chapter aims to provide a detailed description of the experimental data acquisition and analysis needed to address these questions, with a special focus on key dynamic and morphological metrics such as surveillance, directed motility, and ramification.
Collapse
Affiliation(s)
- Vasiliki Kyrargyri
- Department of Immunology, Laboratory of Molecular Genetics, Hellenic Pasteur Institute, Athens, Greece
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Renaud Blaise Jolivet
- Département de Physique Nucléaire et Corpusculaire, University of Geneva, Geneva, Switzerland
- CERN, Geneva, Switzerland
| | - Christian Madry
- Institute of Neurophysiology, Charité-Universitätsmedizin, Berlin, Germany.
| |
Collapse
|
16
|
3DMorph Automatic Analysis of Microglial Morphology in Three Dimensions from Ex Vivo and In Vivo Imaging. eNeuro 2018; 5:eN-MNT-0266-18. [PMID: 30627639 PMCID: PMC6325541 DOI: 10.1523/eneuro.0266-18.2018] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 10/18/2018] [Accepted: 10/28/2018] [Indexed: 11/21/2022] Open
Abstract
Microglia are dynamic immune cells of the central nervous system, and their morphology is commonly used as a readout of cellular function. However, current morphological analysis techniques rely on either tracing of cells or two-dimensional projection analysis, which are time-consuming, subject to bias, and may ignore important three-dimensional (3D) information. Therefore, we have created 3DMorph, a MATLAB-based script that analyzes microglial morphology from 3D data. The program initially requires input of threshold levels, cell size expectations, and preferred methods of skeletonization. This makes 3DMorph easily scalable and adaptable to different imaging parameters or cell types. After these settings are defined, the program is completely automatic and can batch process files without user input. Output data includes cell volume, territorial volume, branch length, number of endpoints and branch points, and average distance between cells. We show that 3DMorph is accurate compared to manual tracing, with significantly decreased user input time. Importantly, 3DMorph is capable of processing in vivo microglial morphology, as well as other 3D branching cell types, from mouse cranial windows or acute hippocampal slices. Therefore, we present a novel, user-friendly, scalable, and semiautomatic method of analyzing cell morphology in 3 dimensions. This method should improve the accuracy of cell measurements, remove user bias between conditions, increase reproducibility between experimenters and labs, and reduce user input time. We provide this open source code on GitHub so that it is free and accessible to all investigators.
Collapse
|