201
|
Umpierre AD, Bystrom LL, Ying Y, Liu YU, Worrell G, Wu LJ. Microglial calcium signaling is attuned to neuronal activity in awake mice. eLife 2020; 9:56502. [PMID: 32716294 PMCID: PMC7402678 DOI: 10.7554/elife.56502] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 07/14/2020] [Indexed: 12/14/2022] Open
Abstract
Microglial calcium signaling underlies a number of key physiological and pathological processes in situ, but has not been studied in vivo in awake mice. Using multiple GCaMP6 variants targeted to microglia, we assessed how microglial calcium signaling responds to alterations in neuronal activity across a wide range. We find that only a small subset of microglial somata and processes exhibited spontaneous calcium transients in a chronic window preparation. However, hyperactive shifts in neuronal activity (kainate status epilepticus and CaMKIIa Gq DREADD activation) triggered increased microglial process calcium signaling, often concomitant with process extension. Additionally, hypoactive shifts in neuronal activity (isoflurane anesthesia and CaMKIIa Gi DREADD activation) also increased microglial process calcium signaling. Under hypoactive neuronal conditions, microglia also exhibited process extension and outgrowth with greater calcium signaling. Our work reveals that microglia have highly distinct microdomain signaling, and that processes specifically respond to bi-directional shifts in neuronal activity through increased calcium signaling.
Collapse
Affiliation(s)
| | | | - Yanlu Ying
- Department of Neurology, Mayo Clinic, Rochester, United States
| | - Yong U Liu
- Department of Neurology, Mayo Clinic, Rochester, United States
| | - Gregory Worrell
- Department of Neurology, Mayo Clinic, Rochester, United States
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, United States.,Department of Neuroscience, Mayo Clinic, Jacksonville, United States.,Department of Immunology, Mayo Clinic, Rochester, United States
| |
Collapse
|
202
|
Abstract
Microglia are the major immune cells in the central nervous system and play a key role in the normal function of the brain. Microglia exhibit functional diversity, and they control the inflammation in central nervous system through releasing inflammatory cytokine, clearing apoptotic cells via phagocytosis, regulating synaptic plasticity and the formation of neural network by synapse pruning. Recent studies have strongly indicated that the microglial dysfunction is associated with a variety of neuropsychiatric diseases such as depression, which have been termed as "microgliopathy". The emergency of advanced technologies and tools has enabled us to comprehensively understand the role of microglia in physiology and pathology, and growing studies have targetted microglia to explore the treatment of neuropsychiatric diseases. Here, we describe the key progress of microglia research, and review the recent developments in the understanding of the role of microglia in physiology and etiology of depression.
Collapse
|
203
|
Yu L, Su X, Li S, Zhao F, Mu D, Qu Y. Microglia and Their Promising Role in Ischemic Brain Injuries: An Update. Front Cell Neurosci 2020; 14:211. [PMID: 32754016 PMCID: PMC7365911 DOI: 10.3389/fncel.2020.00211] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/16/2020] [Indexed: 12/14/2022] Open
Abstract
Ischemic brain injuries are common diseases with high morbidity, disability, and mortality rates, which have significant impacts on human health and life. Microglia are resident cells of the central nervous system (CNS). The inflammatory responses mediated by microglia play an important role in the occurrence and development of ischemic brain injuries. This article summarizes the activation, polarization, depletion, and repopulation of microglia after ischemic brain injuries, proposing new treatment strategies for such injuries through the modulation of microglial function.
Collapse
Affiliation(s)
- Luting Yu
- Department of Paediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Xiaojuan Su
- Department of Paediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Shiping Li
- Department of Paediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Fengyan Zhao
- Department of Paediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Dezhi Mu
- Department of Paediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Yi Qu
- Department of Paediatrics, West China Second University Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| |
Collapse
|
204
|
Park D, Kim S, Kim H, Shin J, Jung H, Um JW. Seizure progression triggered by
IQSEC3
loss is mitigated by reducing activated microglia in mice. Glia 2020; 68:2661-2673. [DOI: 10.1002/glia.23876] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/29/2020] [Accepted: 06/04/2020] [Indexed: 01/02/2023]
Affiliation(s)
- Dongseok Park
- Department of Brain and Cognitive Sciences Daegu Gyeongbuk Institute of Science and Technology (DGIST) Daegu South Korea
| | - Seungjoon Kim
- Department of Brain and Cognitive Sciences Daegu Gyeongbuk Institute of Science and Technology (DGIST) Daegu South Korea
| | - Hyeonho Kim
- Department of Brain and Cognitive Sciences Daegu Gyeongbuk Institute of Science and Technology (DGIST) Daegu South Korea
| | - Jungsu Shin
- Department of Brain and Cognitive Sciences Daegu Gyeongbuk Institute of Science and Technology (DGIST) Daegu South Korea
| | - Hyeji Jung
- Department of Brain and Cognitive Sciences Daegu Gyeongbuk Institute of Science and Technology (DGIST) Daegu South Korea
| | - Ji Won Um
- Department of Brain and Cognitive Sciences Daegu Gyeongbuk Institute of Science and Technology (DGIST) Daegu South Korea
- Core Protein Resources Center, DGIST Daegu South Korea
| |
Collapse
|
205
|
Nguyen PT, Dorman LC, Pan S, Vainchtein ID, Han RT, Nakao-Inoue H, Taloma SE, Barron JJ, Molofsky AB, Kheirbek MA, Molofsky AV. Microglial Remodeling of the Extracellular Matrix Promotes Synapse Plasticity. Cell 2020; 182:388-403.e15. [PMID: 32615087 DOI: 10.1016/j.cell.2020.05.050] [Citation(s) in RCA: 328] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 04/20/2020] [Accepted: 05/27/2020] [Indexed: 12/21/2022]
Abstract
Synapse remodeling is essential to encode experiences into neuronal circuits. Here, we define a molecular interaction between neurons and microglia that drives experience-dependent synapse remodeling in the hippocampus. We find that the cytokine interleukin-33 (IL-33) is expressed by adult hippocampal neurons in an experience-dependent manner and defines a neuronal subset primed for synaptic plasticity. Loss of neuronal IL-33 or the microglial IL-33 receptor leads to impaired spine plasticity, reduced newborn neuron integration, and diminished precision of remote fear memories. Memory precision and neuronal IL-33 are decreased in aged mice, and IL-33 gain of function mitigates age-related decreases in spine plasticity. We find that neuronal IL-33 instructs microglial engulfment of the extracellular matrix (ECM) and that its loss leads to impaired ECM engulfment and a concomitant accumulation of ECM proteins in contact with synapses. These data define a cellular mechanism through which microglia regulate experience-dependent synapse remodeling and promote memory consolidation.
Collapse
Affiliation(s)
- Phi T Nguyen
- Department of Psychiatry and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Leah C Dorman
- Department of Psychiatry and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Simon Pan
- Department of Psychiatry and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Ilia D Vainchtein
- Department of Psychiatry and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Rafael T Han
- Department of Psychiatry and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Hiromi Nakao-Inoue
- Department of Psychiatry and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Sunrae E Taloma
- Department of Psychiatry and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jerika J Barron
- Department of Psychiatry and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Ari B Molofsky
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Mazen A Kheirbek
- Department of Psychiatry and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| | - Anna V Molofsky
- Department of Psychiatry and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA; Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
206
|
Gómez-Budia M, Konttinen H, Saveleva L, Korhonen P, Jalava PI, Kanninen KM, Malm T. Glial smog: Interplay between air pollution and astrocyte-microglia interactions. Neurochem Int 2020; 136:104715. [DOI: 10.1016/j.neuint.2020.104715] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 12/15/2022]
|
207
|
Regulation of Microglial Functions by Purinergic Mechanisms in the Healthy and Diseased CNS. Cells 2020; 9:cells9051108. [PMID: 32365642 PMCID: PMC7290360 DOI: 10.3390/cells9051108] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/25/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
Microglial cells, the resident macrophages of the central nervous system (CNS), exist in a process-bearing, ramified/surveying phenotype under resting conditions. Upon activation by cell-damaging factors, they get transformed into an amoeboid phenotype releasing various cell products including pro-inflammatory cytokines, chemokines, proteases, reactive oxygen/nitrogen species, and the excytotoxic ATP and glutamate. In addition, they engulf pathogenic bacteria or cell debris and phagocytose them. However, already resting/surveying microglia have a number of important physiological functions in the CNS; for example, they shield small disruptions of the blood–brain barrier by their processes, dynamically interact with synaptic structures, and clear surplus synapses during development. In neurodegenerative illnesses, they aggravate the original disease by a microglia-based compulsory neuroinflammatory reaction. Therefore, the blockade of this reaction improves the outcome of Alzheimer’s Disease, Parkinson’s Disease, multiple sclerosis, amyotrophic lateral sclerosis, etc. The function of microglia is regulated by a whole array of purinergic receptors classified as P2Y12, P2Y6, P2Y4, P2X4, P2X7, A2A, and A3, as targets of endogenous ATP, ADP, or adenosine. ATP is sequentially degraded by the ecto-nucleotidases and 5′-nucleotidase enzymes to the almost inactive inosine as an end product. The appropriate selective agonists/antagonists for purinergic receptors as well as the respective enzyme inhibitors may profoundly interfere with microglial functions and reconstitute the homeostasis of the CNS disturbed by neuroinflammation.
Collapse
|
208
|
Elliot A, Myllymäki H, Feng Y. Inflammatory Responses during Tumour Initiation: From Zebrafish Transgenic Models of Cancer to Evidence from Mouse and Man. Cells 2020; 9:cells9041018. [PMID: 32325966 PMCID: PMC7226149 DOI: 10.3390/cells9041018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/08/2020] [Accepted: 04/14/2020] [Indexed: 12/12/2022] Open
Abstract
The zebrafish is now an important model organism for cancer biology studies and provides unique and complementary opportunities in comparison to the mammalian equivalent. The translucency of zebrafish has allowed in vivo live imaging studies of tumour initiation and progression at the cellular level, providing novel insights into our understanding of cancer. Here we summarise the available transgenic zebrafish tumour models and discuss what we have gleaned from them with respect to cancer inflammation. In particular, we focus on the host inflammatory response towards transformed cells during the pre-neoplastic stage of tumour development. We discuss features of tumour-associated macrophages and neutrophils in mammalian models and present evidence that supports the idea that these inflammatory cells promote early stage tumour development and progression. Direct live imaging of tumour initiation in zebrafish models has shown that the intrinsic inflammation induced by pre-neoplastic cells is tumour promoting. Signals mediating leukocyte recruitment to pre-neoplastic cells in zebrafish correspond to the signals that mediate leukocyte recruitment in mammalian tumours. The activation state of macrophages and neutrophils recruited to pre-neoplastic cells in zebrafish appears to be heterogenous, as seen in mammalian models, which provides an opportunity to study the plasticity of innate immune cells during tumour initiation. Although several potential mechanisms are described that might mediate the trophic function of innate immune cells during tumour initiation in zebrafish, there are several unknowns that are yet to be resolved. Rapid advancement of genetic tools and imaging technologies for zebrafish will facilitate research into the mechanisms that modulate leukocyte function during tumour initiation and identify targets for cancer prevention.
Collapse
Affiliation(s)
| | | | - Yi Feng
- Correspondence: ; Tel.: +44-(0)131-242-6685
| |
Collapse
|
209
|
Microglial metabolic flexibility supports immune surveillance of the brain parenchyma. Nat Commun 2020; 11:1559. [PMID: 32214088 PMCID: PMC7096448 DOI: 10.1038/s41467-020-15267-z] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 02/27/2020] [Indexed: 02/07/2023] Open
Abstract
Microglia are highly motile cells that continuously monitor the brain environment and respond to damage-associated cues. While glucose is the main energy substrate used by neurons in the brain, the nutrients metabolized by microglia to support surveillance of the parenchyma remain unexplored. Here, we use fluorescence lifetime imaging of intracellular NAD(P)H and time-lapse two-photon imaging of microglial dynamics in vivo and in situ, to show unique aspects of the microglial metabolic signature in the brain. Microglia are metabolically flexible and can rapidly adapt to consume glutamine as an alternative metabolic fuel in the absence of glucose. During insulin-induced hypoglycemia in vivo or in aglycemia in acute brain slices, glutaminolysis supports the maintenance of microglial process motility and damage-sensing functions. This metabolic shift sustains mitochondrial metabolism and requires mTOR-dependent signaling. This remarkable plasticity allows microglia to maintain their critical surveillance and phagocytic roles, even after brain neuroenergetic homeostasis is compromised. Glucose is the main source of fuel in the brain. Here, the authors show that in the absence of glucose, glutamine is required for microglia to maintain their immune surveillance function.
Collapse
|
210
|
Abstract
Microglia dynamically interact with neurons influencing the development, structure, and function of neuronal networks. Recent studies suggest microglia may also influence neuronal activity by physically interacting with axonal domains responsible for action potential initiation and propagation. However, the nature of these microglial process interactions is not well understood. Microglial-axonal contacts are present early in development and persist through adulthood, implicating microglial interactions in the regulation of axonal integrity in both the developing and mature central nervous system. Moreover, changes in microglial-axonal contact have been described in disease states such as multiple sclerosis (MS) and traumatic brain injury (TBI). Depending on the disease state, there are increased associations with specific axonal segments. In MS, there is enhanced contact with the axon initial segment and node of Ranvier, while, in TBI, microglia alter interactions with axons at the site of injury, as well as at the axon initial segment. In this article, we review the interactions of microglial processes with axonal segments, analyzing their associations with various axonal domains and how these interactions may differ between MS and TBI. Furthermore, we discuss potential functional consequences and molecular mechanisms of these interactions and how these may differ among various types of microglial-axonal interactions.
Collapse
Affiliation(s)
- Savannah D Benusa
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Audrey D Lafrenaye
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
211
|
Sato K, Mochida S, Tomimoto D, Konuma T, Kiyota N, Tsuda S, Shiga Y, Omodaka K, Nakazawa T. A pyruvate dehydrogenase kinase inhibitor prevents retinal cell death and improves energy metabolism in rat retinas after ischemia/reperfusion injury. Exp Eye Res 2020; 193:107997. [PMID: 32165157 DOI: 10.1016/j.exer.2020.107997] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 03/06/2020] [Accepted: 03/06/2020] [Indexed: 12/19/2022]
Abstract
We aimed to assess the neuroprotective effect of a pyruvate dehydrogenase kinase (PDK) inhibitor, Nov3r after ischemia/reperfusion (IR) injury in rats. IR injury was induced by applying 150 mmHg of intraocular pressure for 50 min. Nov3r was orally administered (100 mg/kg) 3 h before and 24 h after IR injury. TUNEL-positive cells increased and immunoreactive RBPMS-positive cells decreased in the rat retinas after IR injury. Administration of Nov3r significantly ameliorated the increase in TUNEL-positive cells and prevented the RBPMS-positive cell decrease. Similarly, the number of IR-induced Iba1-positive microglial cells was significantly reduced with Nov3r treatment. Among metabolic parameters, IR damage induced the elevation of lactate and pyruvate, and the reduction of ATP. Oral administration of Nov3r ameliorated these changes. Our data suggest that the Nov3r had a retinal neuroprotective effect in IR injury in rats. This finding suggests that the regulation of pyruvate dehydrogenase (PDH) activity has potential therapeutic value by enabling metabolic reprograming in diseases associated with ischemic retinal damage, such as diabetic retinopathy, retinopathy of prematurity, retinal vein occlusion, ischemic optic neuropathy and glaucoma.
Collapse
Affiliation(s)
- Kota Sato
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan; Collaborative Program for Ophthalmic Drug Discovery, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Seiya Mochida
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka, Japan
| | - Daisuke Tomimoto
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka, Japan
| | - Takahiro Konuma
- Biological/Pharmacological Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1 Murasaki-cho, Takatsuki, Osaka, Japan
| | - Naoki Kiyota
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Satoru Tsuda
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Yukihiro Shiga
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Kazuko Omodaka
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Toru Nakazawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Miyagi, Japan; Collaborative Program for Ophthalmic Drug Discovery, Tohoku University Graduate School of Medicine, Miyagi, Japan; Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Miyagi, Japan; Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Miyagi, Japan; Department of Ophthalmic Imaging and Information Analytics, Tohoku University Graduate School of Medicine, Miyagi, Japan.
| |
Collapse
|
212
|
Inhibition of NMDA Receptors Downregulates Astrocytic AQP4 to Suppress Seizures. Cell Mol Neurobiol 2020; 40:1283-1295. [DOI: 10.1007/s10571-020-00813-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 02/14/2020] [Indexed: 01/28/2023]
|
213
|
Umpierre AD, Wu LJ. Microglia Research in the 100th Year Since Its Discovery. Neurosci Bull 2020; 36:303-306. [PMID: 32103452 DOI: 10.1007/s12264-020-00477-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 01/09/2020] [Indexed: 12/30/2022] Open
Affiliation(s)
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, 55905, USA. .,Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA. .,Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
214
|
Affiliation(s)
- Dilek Mercan
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Bonn, Germany
| | - Michael T Heneka
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University of Bonn Medical Center, Bonn, Germany. .,German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany. .,Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
215
|
Abstract
In vivo two-photon imaging of microglia in the intact brain has revealed that microglia constantly survey neuronal soma. Research over the past decade and a recent paper by Cserép et al. published in Science are now uncovering the nature, mechanisms, and consequences of these interactions in health and injury.
Collapse
|
216
|
Raghunatha P, Vosoughi A, Kauppinen TM, Jackson MF. Microglial NMDA receptors drive pro-inflammatory responses via PARP-1/TRMP2 signaling. Glia 2020; 68:1421-1434. [PMID: 32036619 DOI: 10.1002/glia.23790] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/16/2020] [Accepted: 01/23/2020] [Indexed: 12/16/2022]
Abstract
Chronic neuroinflammation driven by microglia is a characteristic feature associated with numerous neurodegenerative diseases. While acute inflammation can assist with recovery and repair, prolonged microglial pro-inflammatory responses are known to exacerbate neurodegenerative processes. Yet, detrimental outcomes of extended microglial activation are counterbalanced by beneficial outcomes including phagocytosis and release of trophic factors promoting neuronal viability. Our past work has shown that the nuclear enzyme poly(ADP-ribose) polymerase-1 (PARP-1) is a key signaling hub driving pro-inflammatory microglia responses, but the signaling pathway maintaining PARP-1 activation remains elusive. While best understood for its role in promoting DNA repair, our group has shown that PARP-1 activity can be stimulated via Ca2+ influx-dependent ERK1/2-mediated phosphorylation. However, to date, the route of Ca2+ entry responsible for stimulating PARP-1 has not been identified. A likely candidate is via Ca2+ -permeable transient receptor potential melastatin 2 (TRPM2) channels activated downstream of PARP-1 in a cascade that involves ADP-ribose (ADPR) production by poly(ADP-ribose) glycohydrolase (PARG). Here we demonstrate that NMDA receptor (NMDAR) stimulation in primary cultured microglia induces their proliferation, morphological activation and release of pro-inflammatory mediators. These responses were contingent on the recruitment of PARP-1, PARG and Ca2+ permeable TRPM2 channels. Furthermore, we show that Ca2+ influx is necessary to activate PARP-1/TRPM2 signaling, in an ERK1/2-dependent, but DNA damage independent, manner. Our findings, showing that PARP-1/TRPM2 mediate the pro-inflammatory effects of NMDAR stimulation, provides a unifying mechanism linking elevated glutamate levels to chronic neuroinflammation.
Collapse
Affiliation(s)
- Prajwal Raghunatha
- Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada.,Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, Manitoba, Canada
| | - Amir Vosoughi
- Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, Manitoba, Canada
| | - Tiina M Kauppinen
- Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada.,Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, Manitoba, Canada.,The Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
| | - Michael F Jackson
- Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada.,Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, Manitoba, Canada
| |
Collapse
|
217
|
Grovola MR, Paleologos N, Wofford KL, Harris JP, Browne KD, Johnson V, Duda JE, Wolf JA, Cullen DK. Mossy cell hypertrophy and synaptic changes in the hilus following mild diffuse traumatic brain injury in pigs. J Neuroinflammation 2020; 17:44. [PMID: 32005260 PMCID: PMC6993507 DOI: 10.1186/s12974-020-1720-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 01/19/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Each year in the USA, over 2.4 million people experience mild traumatic brain injury (TBI), which can induce long-term neurological deficits. The dentate gyrus of the hippocampus is notably susceptible to damage following TBI, as hilar mossy cell changes in particular may contribute to post-TBI dysfunction. Moreover, microglial activation after TBI may play a role in hippocampal circuit and/or synaptic remodeling; however, the potential effects of chronic microglial changes are currently unknown. The objective of the current study was to assess neuropathological and neuroinflammatory changes in subregions of the dentate gyrus at acute to chronic time points following mild TBI using an established model of closed-head rotational acceleration induced TBI in pigs. METHODS This study utilized archival tissue of pigs which were subjected to sham conditions or rapid head rotation in the coronal plane to generate mild TBI. A quantitative assessment of neuropathological changes in the hippocampus was performed via immunohistochemical labeling of whole coronal tissue sections at 3 days post-injury (DPI), 7 DPI, 30 DPI, and 1 year post-injury (YPI), with a focus on mossy cell atrophy and synaptic reorganization, in context with microglial alterations (e.g., density, proximity to mossy cells) in the dentate gyrus. RESULTS There were no changes in mossy cell density between sham and injured animals, indicating no frank loss of mossy cells at the mild injury level evaluated. However, we found significant mossy cell hypertrophy at 7 DPI and 30 DPI in anterior (> 16% increase in mean cell area at each time; p = < 0.001 each) and 30 DPI in posterior (8.3% increase; p = < 0.0001) hippocampus. We also found dramatic increases in synapsin staining around mossy cells at 7 DPI in both anterior (74.7% increase in synapsin labeling; p = < 0.0001) and posterior (82.7% increase; p = < 0.0001) hippocampus. Interestingly, these morphological and synaptic alterations correlated with a significant change in microglia in proximity to mossy cells at 7 DPI in anterior and at 30 DPI in the posterior hippocampus. For broader context, while we found that there were significant increases in microglia density in the granule cell layer at 30 DPI (anterior and posterior) and 1 YPI (posterior only) and in the molecular layer at 1 YPI (anterior only), we found no significant changes in overall microglial density in the hilus at any of the time points evaluated post-injury. CONCLUSIONS The alterations of mossy cell size and synaptic inputs paired with changes in microglia density around the cells demonstrate the susceptibility of hilar mossy cells after even mild TBI. This subtle hilar mossy cell pathology may play a role in aberrant hippocampal function post-TBI, although additional studies are needed to characterize potential physiological and cognitive alterations.
Collapse
Affiliation(s)
- Michael R Grovola
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, 105E Hayden Hall/3320 Smith Walk, Philadelphia, PA, 19104, USA
| | - Nicholas Paleologos
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, 105E Hayden Hall/3320 Smith Walk, Philadelphia, PA, 19104, USA
| | - Kathryn L Wofford
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, 105E Hayden Hall/3320 Smith Walk, Philadelphia, PA, 19104, USA
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - James P Harris
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, 105E Hayden Hall/3320 Smith Walk, Philadelphia, PA, 19104, USA
| | - Kevin D Browne
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, 105E Hayden Hall/3320 Smith Walk, Philadelphia, PA, 19104, USA
| | - Victoria Johnson
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, 105E Hayden Hall/3320 Smith Walk, Philadelphia, PA, 19104, USA
| | - John E Duda
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Parkinson's Disease Research, Education and Clinical Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - John A Wolf
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, 105E Hayden Hall/3320 Smith Walk, Philadelphia, PA, 19104, USA
| | - D Kacy Cullen
- Center for Neurotrauma, Neurodegeneration & Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA.
- Center for Brain Injury & Repair, Department of Neurosurgery, University of Pennsylvania, 105E Hayden Hall/3320 Smith Walk, Philadelphia, PA, 19104, USA.
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, 105E Hayden Hall/3320 Smith Walk, Philadelphia, PA, 19104, USA.
| |
Collapse
|
218
|
Alves M, Smith J, Engel T. Differential Expression of the Metabotropic P2Y Receptor Family in the Cortex Following Status Epilepticus and Neuroprotection via P2Y 1 Antagonism in Mice. Front Pharmacol 2020; 10:1558. [PMID: 32009961 PMCID: PMC6976538 DOI: 10.3389/fphar.2019.01558] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 12/03/2019] [Indexed: 12/18/2022] Open
Abstract
Purinergic signaling via P2 receptors is now widely accepted to play a critical role during increased states of hyperexcitability and seizure-induced pathology. In the setting of seizures and epilepsy, most attention has been paid to investigating the fast-acting ATP-gated P2X receptor family. More recent evidence has now also provided compelling evidence of an involvement of the slower-acting P2Y receptor family during seizures. This includes data demonstrating expression changes of P2Y receptors in the hippocampus following acute seizures and during epilepsy and anticonvulsive properties of P2Y-targeting drugs; in particular drugs targeting the P2Y1 subtype. Seizures, however, also involve damage to extra-hippocampal brain regions such as the cortex, which is thought to contribute to the epileptic phenotype. To analyze expressional changes of the P2Y receptor family in the cortex following status epilepticus and to determine the impact of drugs interfering with P2Y1 signaling on cortical damage, we used a unilateral mouse model of intraamygdala kainic acid-induced status epilepticus. Analysis of cortical tissue showed that status epilepticus leads to a global up-regulation of the P2Y receptor family in the cortex including P2Y1, P2Y2, P2Y4, and P2Y6, with the P2Y1 and P2Y4 receptor subtypes showing the strongest increase. Supporting a detrimental role of P2Y1 activation during status epilepticus, treatment with the P2Y1 agonist MRS2365 exacerbated high frequency high amplitude spiking, synonymous with injury-causing electrographic activity, and treatment with the P2Y1 antagonists MRS2500 protected against seizure-induced cortical damage. Suggesting P2Y1-mediated effects are predominantly due to increased microglia activation, treatment with the broad-spectrum anti-inflammatory drug minocycline abolished the observed neuroprotective effects of P2Y1 antagonism. In conclusion, our results further support a role for P2Y1-mediated signaling during seizure generation and seizure-induced neurodegeneration, suggesting P2Y1-targeting therapies as novel treatment for drug-refractory status epilepticus.
Collapse
Affiliation(s)
- Mariana Alves
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jonathon Smith
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,FutureNeuro SFI Research Centre, Dublin, Ireland
| | - Tobias Engel
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.,FutureNeuro SFI Research Centre, Dublin, Ireland
| |
Collapse
|
219
|
Distinct P2Y Receptors Mediate Extension and Retraction of Microglial Processes in Epileptic and Peritumoral Human Tissue. J Neurosci 2020; 40:1373-1388. [PMID: 31896671 DOI: 10.1523/jneurosci.0218-19.2019] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022] Open
Abstract
Microglia exhibit multiple, phenotype-dependent motility patterns often triggered by purinergic stimuli. However, little data exist on motility of human microglia in pathological situations. Here we examine motility of microglia stained with a fluorescent lectin in tissue slices from female and male epileptic patients diagnosed with mesial temporal lobe epilepsy or cortical glioma (peritumoral cortex). Microglial shape varied from ramified to amoeboid cells predominantly in regions of high neuronal loss or closer to a tumor. Live imaging revealed unstimulated or purine-induced microglial motilities, including surveillance movements, membrane ruffling, and process extension or retraction. At different concentrations, ADP triggered opposing motilities. Low doses triggered process extension. It was suppressed by P2Y12 receptor antagonists, which also reduced process length and surveillance movements. Higher purine doses caused process retraction and membrane ruffling, which were blocked by joint application of P2Y1 and P2Y13 receptor antagonists. Purinergic effects on motility were similar for all microglia tested. Both amoeboid and ramified cells from mesial temporal lobe epilepsy or peritumoral cortex tissue expressed P2Y12 receptors. A minority of microglia expressed the adenosine A2A receptor, which has been linked with process withdrawal of rodent cells. Laser-mediated tissue damage let us test the functional significance of these effects. Moderate damage induced microglial process extension, which was blocked by P2Y12 receptor antagonists. Overall, the purine-induced motility of human microglia in epileptic tissue is similar to that of rodent microglia in that the P2Y12 receptor initiates process extension. It differs in that retraction is triggered by joint activation of P2Y1/P2Y13 receptors.SIGNIFICANCE STATEMENT Microglial cells are brain-resident immune cells with multiple functions in healthy or diseased brains. These diverse functions are associated with distinct phenotypes, including different microglial shapes. In the rodent, purinergic signaling is associated with changes in cell shape, such as process extension toward tissue damage. However, there are little data on living human microglia, especially in diseased states. We developed a reliable technique to stain microglia from epileptic and glioma patients to examine responses to purines. Low-intensity purinergic stimuli induced process extension, as in rodents. In contrast, high-intensity stimuli triggered a process withdrawal mediated by both P2Y1 and P2Y13 receptors. P2Y1/P2Y13 receptor activation has not previously been linked to microglial morphological changes.
Collapse
|
220
|
Abstract
Microglia are the resident immune cells and professional phagocytes of the central nervous system. However, little is known about the contribution of their phagocytic signaling to the neuropathology and pathophysiology of epilepsy. Here, we summarize and discuss the implications of recent evidence supporting that aberrant microglia phagocytic activity and alterations in phagocytosis signaling molecules occur in association with microglia-neuronal contacts, neuronal/synaptic loss, and spontaneous recurrent seizures in human and preclinical models of epilepsy. This body of evidence provides strong support that the microglial contribution to epileptogenic networks goes beyond inflammation, and suggests that phagocytic signaling molecules may be novel therapeutic targets for epilepsy.
Collapse
Affiliation(s)
| | - Amy L. Brewster
- Department of Psychological Sciences, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
221
|
Stearoylethanolamide interferes with retrograde endocannabinoid signalling and supports the blood-brain barrier integrity under acute systemic inflammation. Biochem Pharmacol 2019; 174:113783. [PMID: 31881191 DOI: 10.1016/j.bcp.2019.113783] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/23/2019] [Indexed: 02/06/2023]
Abstract
Neuroinflammation plays a prominent role in the onset of demyelinating diseases, major depressive disorder and delayed neurodegeneration. An open question remains whether pharmacological suppression of inflammation can effectively reduce the progression of these states. Bioactive lipid mediators such as N-acylethanolamines (NAEs) have an anti-inflammatory activity and are of pharmacological interest due to their endogenous on-demand production and the existence of distinct biological targets in humans and animals. Here we demonstrate for the first time, that treatment with stearoylethanolamide (SEA), a prevailing endogenously formed NAE, is neuroprotective against LPS-induced neuroinflammation in C57BL/6 male mice. SEA restricted the spreading of peripheral inflammation to the brain, and averted the activation of resident microglia and leukocyte trafficking to the brain parenchyma. Treatment with SEA per se increased the neuronal expression of cannabinoid receptors CB1/2 and brain levels of the most potent endogenous CB1/2 agonist 2-arachidonoylglycerol in vivo. SEA enhanced the amplitude of synaptic vesicle release, supported the balanced signal-to-noise ratio in glutamate- and GABAergic neurotransmission and decreased the excitotoxic risk associated with higher extracellular glutamate levels under neuroinflammation. The interference of SEA with the endocannabinoid system and presynaptic neurotransmitter release may represent an intrinsic neuroprotective mechanism that is triggered by inflammation and glutamate excitotoxicity. Thus, our data allows to consider SEA for the preventive therapy of acute and late-onset neuroinflammation-associated synaptic dysfunction and neurodegeneration.
Collapse
|
222
|
Synaptic Pruning by Microglia in Epilepsy. J Clin Med 2019; 8:jcm8122170. [PMID: 31818018 PMCID: PMC6947403 DOI: 10.3390/jcm8122170] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 11/27/2019] [Accepted: 12/05/2019] [Indexed: 12/16/2022] Open
Abstract
Structural and functional collapse of the balance between excitatory (E) and inhibitory (I) synapses, i.e., synaptic E/I balance, underlies the pathogeneses of various central nervous system (CNS) disorders. In epilepsy, the synaptic E/I balance tips toward excitation; thus, most of the existing epileptic remedies have focused on how to directly suppress the activity of neurons. However, because as many as 30% of patients with epilepsy are drug resistant, the discovery of new therapeutic targets is strongly desired. Recently, the roles of glial cells in epilepsy have gained attention because glial cells manipulate synaptic structures and functions in addition to supporting neuronal survival and growth. Among glial cells, microglia, which are brain-resident immune cells, have been shown to mediate inflammation, neuronal death and aberrant neurogenesis after epileptic seizures. However, few studies have investigated the involvement of synaptic pruning—one of the most important roles of microglia—in the epileptic brain. In this review, we propose and discuss the hypothesis that synaptic pruning by microglia is enhanced in the epileptic brain, drawing upon the findings of previous studies. We further discuss the possibility that aberrant synaptic pruning by microglia induces synaptic E/I imbalance, promoting the development and aggravation of epilepsy.
Collapse
|
223
|
Araki T, Ikegaya Y, Koyama R. Microglia attenuate the kainic acid-induced death of hippocampal neurons in slice cultures. Neuropsychopharmacol Rep 2019; 40:85-91. [PMID: 31794154 PMCID: PMC7292224 DOI: 10.1002/npr2.12086] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/03/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022] Open
Abstract
Background Status epilepticus‐induced hippocampal neuronal death, astrogliosis, and the activation of microglia are common pathological changes in mesial temporal lobe epilepsy (mTLE) with resistance to antiepileptic drugs. Neuronal death in mTLE gradually progresses and is involved in the aggravation of epilepsy and the impairment of hippocampus‐dependent memory. Thus, clarifying the cellular mechanisms by which neurons are protected in mTLE will significantly contribute to the treatment of epilepsy. Here, mainly using hippocampal slice cultures with or without the pharmacological depletion of microglia, we directly examined whether microglia, the resident immune cells of the brain that can act either neurotoxically or in a neuroprotective manner, accelerate or attenuate kainic acid (KA)‐induced neuronal death in vitro. Methods Hippocampal slice cultures were treated with KA to induce neuronal death in vitro. Clodronate‐containing liposomes or PLX3397 was used to deplete microglia in hippocampal slice cultures, and the effect on KA‐induced neuronal death was immunohistochemically assessed. Results The loss of microglia significantly promoted a decrease in neuronal density in KA‐treated hippocampal slice cultures. Conclusion Our results suggest that microglia are neuroprotective against KA‐induced neuronal death in slice cultures. We investigated the role of microglia in kainic acid‐induced neuronal death using hippocampal slice cultures.We found that pharmacological removal of microglia from cultured hippocampal slices enhanced kainic acid‐induced neuronal death. These results suggest that microglia are neuroprotective against kainic acid‐induced neuronal death.![]()
Collapse
Affiliation(s)
- Tasuku Araki
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan.,Center for Information and Neural Networks, Suita City, Japan
| | - Ryuta Koyama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
224
|
Kluge MG, Abdolhoseini M, Zalewska K, Ong LK, Johnson SJ, Nilsson M, Walker FR. Spatiotemporal analysis of impaired microglia process movement at sites of secondary neurodegeneration post-stroke. J Cereb Blood Flow Metab 2019; 39:2456-2470. [PMID: 30204044 PMCID: PMC6893987 DOI: 10.1177/0271678x18797346] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
It has recently been identified that after motor cortex stroke, the ability of microglia processes to respond to local damage cues is lost from the thalamus, a major site of secondary neurodegeneration (SND). In this study, we combine a photothrombotic stroke model in mice, acute slice and fluorescent imaging to analyse the loss of microglia process responsiveness. The peri-infarct territories and thalamic areas of SND were investigated at time-points 3, 7, 14, 28 and 56 days after stroke. We confirmed the highly specific nature of non-responsive microglia processes to sites of SND. Non-responsiveness was at no time observed at the peri-infarct but started in the thalamus seven days post-stroke and persisted for 56 days. Loss of directed process extension is not a reflection of general functional paralysis as phagocytic function continued to increase over time. Additionally, we identified that somal P2Y12 was present on non-responsive microglia in the first two weeks after stroke but not at later time points. Finally, both classical microglia activation and loss of process extension are highly correlated with neuronal damage. Our findings highlight the importance of microglia, specifically microglia dynamic functions, to the progression of SND post-stroke, and their potential relevance as modulators or therapeutic targets during stroke recovery.
Collapse
Affiliation(s)
- Murielle G Kluge
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Mahmoud Abdolhoseini
- School of Electrical Engineering and Computer Science, University of Newcastle, Callaghan, NSW, Australia
| | - Katarzyna Zalewska
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia
| | - Lin Kooi Ong
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia.,NHMRC Centre of Research Excellence Stroke Rehabilitation and Brain Recovery, Heidelberg, VIC, Australia
| | - Sarah J Johnson
- School of Electrical Engineering and Computer Science, University of Newcastle, Callaghan, NSW, Australia
| | - Michael Nilsson
- Hunter Medical Research Institute, Newcastle, NSW, Australia.,NHMRC Centre of Research Excellence Stroke Rehabilitation and Brain Recovery, Heidelberg, VIC, Australia
| | - Frederick R Walker
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, University of Newcastle, Callaghan, NSW, Australia.,Hunter Medical Research Institute, Newcastle, NSW, Australia.,NHMRC Centre of Research Excellence Stroke Rehabilitation and Brain Recovery, Heidelberg, VIC, Australia
| |
Collapse
|
225
|
What Do Microglia Really Do in Healthy Adult Brain? Cells 2019; 8:cells8101293. [PMID: 31652490 PMCID: PMC6829860 DOI: 10.3390/cells8101293] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/17/2019] [Accepted: 10/18/2019] [Indexed: 12/27/2022] Open
Abstract
Microglia originate from yolk sac-primitive macrophages and auto-proliferate into adulthood without replacement by bone marrow-derived circulating cells. In inflammation, stroke, aging, or infection, microglia have been shown to contribute to brain pathology in both deleterious and beneficial ways, which have been studied extensively. However, less is known about their role in the healthy adult brain. Astrocytes and oligodendrocytes are widely accepted to strongly contribute to the maintenance of brain homeostasis and to modulate neuronal function. On the other hand, contribution of microglia to cognition and behavior is only beginning to be understood. The ability to probe their function has become possible using microglial depletion assays and conditional mutants. Studies have shown that the absence of microglia results in cognitive and learning deficits in rodents during development, but this effect is less pronounced in adults. However, evidence suggests that microglia play a role in cognition and learning in adulthood and, at a cellular level, may modulate adult neurogenesis. This review presents the case for repositioning microglia as key contributors to the maintenance of homeostasis and cognitive processes in the healthy adult brain, in addition to their classical role as sentinels coordinating the neuroinflammatory response to tissue damage and disease.
Collapse
|
226
|
Neuronal network activity controls microglial process surveillance in awake mice via norepinephrine signaling. Nat Neurosci 2019; 22:1771-1781. [PMID: 31636449 PMCID: PMC6858573 DOI: 10.1038/s41593-019-0511-3] [Citation(s) in RCA: 226] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 09/11/2019] [Indexed: 01/23/2023]
Abstract
Microglia dynamically survey the brain parenchyma. Microglial processes interact with neuronal elements; however, what role neuronal network activity plays in regulating microglial dynamics is not entirely clear. Most studies of microglial dynamics use either slice preparations or in vivo imaging in anesthetized mice. Here we demonstrate that microglia in awake mice have a relatively reduced process area and surveillance territory and that reduced neuronal activity under general anesthesia increases microglial process velocity, extension and territory surveillance. Similarly, reductions in local neuronal activity through sensory deprivation or optogenetic inhibition increase microglial process surveillance. Using pharmacological and chemogenetic approaches, we demonstrate that reduced norepinephrine signaling is necessary for these increases in microglial process surveillance. These findings indicate that under basal physiological conditions, noradrenergic tone in awake mice suppresses microglial process surveillance. Our results emphasize the importance of awake imaging for studying microglia-neuron interactions and demonstrate how neuronal activity influences microglial process dynamics.
Collapse
|
227
|
Microglial P2Y12 Receptor Regulates Seizure-Induced Neurogenesis and Immature Neuronal Projections. J Neurosci 2019; 39:9453-9464. [PMID: 31597724 DOI: 10.1523/jneurosci.0487-19.2019] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 10/01/2019] [Accepted: 10/02/2019] [Indexed: 11/21/2022] Open
Abstract
Seizures are common in humans with various etiologies ranging from congenital aberrations to acute injuries that alter the normal balance of brain excitation and inhibition. A notable consequence of seizures is the induction of aberrant neurogenesis and increased immature neuronal projections. However, regulatory mechanisms governing these features during epilepsy development are not fully understood. Recent studies show that microglia, the brain's resident immune cell, contribute to normal neurogenesis and regulate seizure phenotypes. However, the role of microglia in aberrant neurogenic seizure contexts has not been adequately investigated. To address this question, we coupled the intracerebroventricular kainic acid model with current pharmacogenetic approaches to eliminate microglia in male mice. We show that microglia promote seizure-induced neurogenesis and subsequent seizure-induced immature neuronal projections above and below the pyramidal neurons between the DG and the CA3 regions. Furthermore, we identify microglial P2Y12 receptors (P2Y12R) as a participant in this neurogenic process. Together, our results implicate microglial P2Y12R signaling in epileptogenesis and provide further evidence for targeting microglia in general and microglial P2Y12R in specific to ameliorate proepileptogenic processes.SIGNIFICANCE STATEMENT Epileptogenesis is a process by which the brain develops epilepsy. Several processes have been identified that confer the brain with such epileptic characteristics, including aberrant neurogenesis and increased immature neuronal projections. Understanding the mechanisms that promote such changes is critical in developing therapies to adequately restrain epileptogenesis. We investigated the role of purinergic P2Y12 receptors selectively expressed by microglia, the resident brain immune cells. We report, for the first time, that microglia in general and microglial P2Y12 receptors in specific promote both aberrant neurogenesis and increased immature neuronal projections. These results indicate that microglia enhance epileptogenesis by promoting these processes and suggest that targeting this immune axis could be a novel therapeutic strategy in the clinic.
Collapse
|
228
|
Suzuki T, Kohyama K, Moriyama K, Ozaki M, Hasegawa S, Ueno T, Saitoe M, Morio T, Hayashi M, Sakuma H. Extracellular ADP augments microglial inflammasome and NF-κB activation via the P2Y12 receptor. Eur J Immunol 2019; 50:205-219. [PMID: 31549730 DOI: 10.1002/eji.201848013] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 07/31/2019] [Accepted: 09/23/2019] [Indexed: 12/11/2022]
Abstract
The NLRP3 inflammasome is a molecular complex that translates signals from pathogens and tissue damage into inflammatory responses, and plays crucial roles in numerous neurological diseases. Activation of the NLRP3 inflammasome leads to caspase-1 dependent cleavage of pro-IL-1β to form mature IL-1β. By acting on the P2X7 purinergic receptor, extracellular ATP is one of the major stimuli that activates the NLRP3 inflammasome. Although microglia express multiple purinergic receptors, their roles in inflammasome-mediated inflammation are largely unknown. We studied the role of the P2Y12 receptor, a metabotropic P2Y receptor enriched in microglia, on inflammation in vitro. Inhibition of the microglial P2Y12 receptor by PSB0739 or siRNA knockdown suppressed IL-1β release. P2Y12 receptor-deficient microglia displayed markedly attenuated IL-1β mRNA expression and release. P2Y12 receptor blockade also suppressed IL-6 production. Both IL-1β and IL-6 responses were augmented by extracellular ADP or ADP-βS and were abrogated by PSB0739. Mechanistically, ADP-βS potentiated NF-κB activation. In addition, ADP altered mitochondrial membrane potential in combination with ATP and increased the number of caspase-1 positive cells through the P2Y12 receptor. These results elucidate a novel inflammatory mechanism by which extracellular ADP acts on the P2Y12 receptor to activate NF-κB and the NLRP3 inflammasome to enhance microglial inflammation.
Collapse
Affiliation(s)
- Tomonori Suzuki
- Developmental Neuroimmunology Project, Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Pediatrics and Developmental Biology, Bio-Environmental Response Division, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kuniko Kohyama
- Developmental Neuroimmunology Project, Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kengo Moriyama
- Developmental Neuroimmunology Project, Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Mariko Ozaki
- Developmental Neuroimmunology Project, Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Setsuko Hasegawa
- Developmental Neuroimmunology Project, Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Taro Ueno
- Learning and Memory Project, Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Minoru Saitoe
- Learning and Memory Project, Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Tomohiro Morio
- Department of Pediatrics and Developmental Biology, Bio-Environmental Response Division, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masaharu Hayashi
- Developmental Neuroimmunology Project, Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Hiroshi Sakuma
- Developmental Neuroimmunology Project, Department of Brain Development and Neural Regeneration, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
229
|
Kalitzin S, Petkov G, Suffczynski P, Grigorovsky V, Bardakjian BL, Lopes da Silva F, Carlen PL. Epilepsy as a manifestation of a multistate network of oscillatory systems. Neurobiol Dis 2019; 130:104488. [DOI: 10.1016/j.nbd.2019.104488] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 12/18/2022] Open
|
230
|
Eyo UB, Mo M, Yi MH, Murugan M, Liu J, Yarlagadda R, Margolis DJ, Xu P, Wu LJ. P2Y12R-Dependent Translocation Mechanisms Gate the Changing Microglial Landscape. Cell Rep 2019; 23:959-966. [PMID: 29694903 PMCID: PMC5965271 DOI: 10.1016/j.celrep.2018.04.001] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 02/21/2018] [Accepted: 03/30/2018] [Indexed: 01/01/2023] Open
Abstract
Microglia are an exquisitely tiled and self-contained population in the CNS that do not receive contributions from circulating monocytes in the periphery. While microglia are long-lived cells, the extent to which their cell bodies are fixed and the molecular mechanisms by which the microglial landscape is regulated have not been determined. Using chronic in vivo two-photon imaging to follow the microglial population in young adult mice, we document a daily rearrangement of the microglial landscape. Furthermore, we show that the microglial landscape can be modulated by severe seizures, acute injury, and sensory deprivation. Finally, we demonstrate a critical role for microglial P2Y12Rs in regulating the microglial landscape through cellular translocation independent of proliferation. These findings suggest that microglial patrol the CNS through both process motility and soma translocation.
Collapse
Affiliation(s)
- Ukpong B Eyo
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Mingshu Mo
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA; Department of Neurology, First Affiliated Hospital of Guangzhou Medical University, Guangdong 510120, China
| | - Min-Hee Yi
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Madhuvika Murugan
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Junting Liu
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Rohan Yarlagadda
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - David J Margolis
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA
| | - Pingyi Xu
- Department of Neurology, First Affiliated Hospital of Guangzhou Medical University, Guangdong 510120, China.
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08854, USA; Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA.
| |
Collapse
|
231
|
Peng J, Liu Y, Umpierre AD, Xie M, Tian DS, Richardson JR, Wu LJ. Microglial P2Y12 receptor regulates ventral hippocampal CA1 neuronal excitability and innate fear in mice. Mol Brain 2019; 12:71. [PMID: 31426845 PMCID: PMC6700820 DOI: 10.1186/s13041-019-0492-x] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 08/13/2019] [Indexed: 12/31/2022] Open
Abstract
The P2Y12 receptor (P2Y12R) is a purinoceptor that is selectively expressed in microglia in the central nervous system. As a signature receptor, microglial P2Y12R mediates process chemotaxis towards ADP/ATP gradients and is engaged in several neurological diseases including chronic pain, stroke and seizures. However, the role of microglial P2Y12R in regulating neuronal excitability and innate behaviors is not fully understood. Here, we generated P2Y12-floxed mice to delete microglial P2Y12R beginning in development (CX3CR1Cre/+:P2Y12f/f; "constitutive knockout"), or after normal development in adult mice (CX3CR1CreER/+:P2Y12f/f; "induced knockout"). Using a battery of behavioral tests, we found that both constitutive and induced P2Y12R knockout mice exhibited innate fear but not learned fear behaviors. After mice were exposed to the elevated plus maze, the c-fos expression in ventral hippocampus CA1 neurons was robustly increased in P2Y12R knockout mice compared with wild-type mice. Consistently, using whole cell patch clamp recording, we found the excitability of ventral hippocampus CA1 neurons was increased in the P2Y12R knockout mice. The results suggest that microglial P2Y12R regulates neuronal excitability and innate fear behaviors in developing and adult mice.
Collapse
Affiliation(s)
- Jiyun Peng
- Institute of Life Science, Nanchang University, Nanchang, 330031, China.,Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Yong Liu
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Anthony D Umpierre
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Manling Xie
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Dai-Shi Tian
- Department of Neurology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jason R Richardson
- Departments of Environmental Health Sciences, Florida International University, Miami, FL, 33199, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA. .,Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA. .,Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
232
|
Eyo UB, Wu LJ. Microglia: Lifelong patrolling immune cells of the brain. Prog Neurobiol 2019; 179:101614. [PMID: 31075285 PMCID: PMC6599472 DOI: 10.1016/j.pneurobio.2019.04.003] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/11/2019] [Accepted: 04/19/2019] [Indexed: 02/02/2023]
Abstract
Microglial cells are the predominant parenchymal immune cell of the brain. Recent evidence suggests that like peripheral immune cells, microglia patrol the brain in health and disease. Reviewing these data, we first examine the evidence that microglia invade the brain mesenchyme early in embryonic development, establish residence therein, proliferate and subsequently maintain their numbers throughout life. We, then, summarize established and novel evidence for microglial process surveillance in the healthy and injured brain. Finally, we discuss emerging evidence for microglial cell body dynamics that challenge existing assumptions of their sessile nature. We conclude that microglia are long-lived immune cells that patrol the brain through both cell body and process movements. This recognition has significant implications for neuroimmune interactions throughout the animal lifespan.
Collapse
Affiliation(s)
- Ukpong B Eyo
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, USA; Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA; Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
233
|
Chia K, Keatinge M, Mazzolini J, Sieger D. Brain tumours repurpose endogenous neuron to microglia signalling mechanisms to promote their own proliferation. eLife 2019; 8:e46912. [PMID: 31313988 PMCID: PMC6685703 DOI: 10.7554/elife.46912] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 07/16/2019] [Indexed: 12/30/2022] Open
Abstract
Previously we described direct cellular interactions between microglia and AKT1+ brain tumour cells in zebrafish (Chia et al., 2018). However, it was unclear how these interactions were initiated: it was also not clear if they had an impact on the growth of tumour cells. Here, we show that neoplastic cells hijack mechanisms that are usually employed to direct microglial processes towards highly active neurons and injuries in the brain. We show that AKT1+ cells possess dynamically regulated high intracellular Ca2+ levels. Using a combination of live imaging, genetic and pharmacological tools, we show that these Ca2+ transients stimulate ATP-mediated interactions with microglia. Interfering with Ca2+ levels, inhibiting ATP release and CRISPR-mediated mutation of the p2ry12 locus abolishes these interactions. Finally, we show that reducing the number of microglial interactions significantly impairs the proliferation of neoplastic AKT1 cells. In conclusion, neoplastic cells repurpose the endogenous neuron to microglia signalling mechanism via P2ry12 activation to promote their own proliferation.
Collapse
Affiliation(s)
- Kelda Chia
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
| | - Marcus Keatinge
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
| | - Julie Mazzolini
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
| | - Dirk Sieger
- Centre for Discovery Brain SciencesUniversity of EdinburghEdinburghUnited Kingdom
| |
Collapse
|
234
|
Vogels T, Murgoci AN, Hromádka T. Intersection of pathological tau and microglia at the synapse. Acta Neuropathol Commun 2019; 7:109. [PMID: 31277708 PMCID: PMC6612163 DOI: 10.1186/s40478-019-0754-y] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 06/19/2019] [Indexed: 02/07/2023] Open
Abstract
Tauopathies are a heterogenous class of diseases characterized by cellular accumulation of aggregated tau and include diseases such as Alzheimer’s disease (AD), progressive supranuclear palsy and chronic traumatic encephalopathy. Tau pathology is strongly linked to neurodegeneration and clinical symptoms in tauopathy patients. Furthermore, synapse loss is an early pathological event in tauopathies and is the strongest correlate of cognitive decline. Tau pathology is additionally associated with chronic neuroinflammatory processes, such as reactive microglia, astrocytes, and increased levels of pro-inflammatory molecules (e.g. complement proteins, cytokines). Recent studies show that as the principal immune cells of the brain, microglia play a particularly important role in the initiation and progression of tau pathology and associated neurodegeneration. Furthermore, AD risk genes such as Triggering receptor expressed on myeloid cells 2 (TREM2) and Apolipoprotein E (APOE) are enriched in the innate immune system and modulate the neuroinflammatory response of microglia to tau pathology. Microglia can play an active role in synaptic dysfunction by abnormally phagocytosing synaptic compartments of neurons with tau pathology. Furthermore, microglia are involved in synaptic spreading of tau – a process which is thought to underlie the progressive nature of tau pathology propagation through the brain. Spreading of pathological tau is also the predominant target for tau-based immunotherapy. Active tau vaccines, therapeutic tau antibodies and other approaches targeting the immune system are actively explored as treatment options for AD and other tauopathies. This review describes the role of microglia in the pathobiology of tauopathies and the mechanism of action of potential therapeutics targeting the immune system in tauopathies.
Collapse
|
235
|
Frere S, Slutsky I. Alzheimer's Disease: From Firing Instability to Homeostasis Network Collapse. Neuron 2019; 97:32-58. [PMID: 29301104 DOI: 10.1016/j.neuron.2017.11.028] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/14/2017] [Accepted: 11/17/2017] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) starts from pure cognitive impairments and gradually progresses into degeneration of specific brain circuits. Although numerous factors initiating AD have been extensively studied, the common principles underlying the transition from cognitive deficits to neuronal loss remain unknown. Here we describe an evolutionarily conserved, integrated homeostatic network (IHN) that enables functional stability of central neural circuits and safeguards from neurodegeneration. We identify the critical modules comprising the IHN and propose a central role of neural firing in controlling the complex homeostatic network at different spatial scales. We hypothesize that firing instability and impaired synaptic plasticity at early AD stages trigger a vicious cycle, leading to dysregulation of the whole IHN. According to this hypothesis, the IHN collapse represents the major driving force of the transition from early memory impairments to neurodegeneration. Understanding the core elements of homeostatic control machinery, the reciprocal connections between distinct IHN modules, and the role of firing homeostasis in this hierarchy has important implications for physiology and should offer novel conceptual approaches for AD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Samuel Frere
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel.
| |
Collapse
|
236
|
Zhang Y, Zhu W, Yu H, Yu J, Zhang M, Pan X, Gao X, Wang Q, Sun H. P2Y4/TSP-1/TGF-β1/pSmad2/3 pathway contributes to acute generalized seizures induced by kainic acid. Brain Res Bull 2019; 149:106-119. [PMID: 31005663 DOI: 10.1016/j.brainresbull.2019.04.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/07/2019] [Accepted: 04/08/2019] [Indexed: 01/23/2023]
Abstract
Epilepsy is accompanied by angiogenesis and blood-brain barrier (BBB) disruption. The transforming growth factor-β1 (TGF-β1)/phosphorylated small mothers against decapentaplegic 2 and 3 (pSmad2/3)/vascular endothelial growth factor (VEGF) pathway, activated by thrombospondin-1 (TSP-1), which is further regulated by Y type P2 purinergic receptor activity, may participate in angiogenesis. We sought to investigate the relationship between the P2R/TSP-1/TGF-β1/pSmad2/3/VEGF pathway, angiogenesis, and BBB damage in a kainic acid (KA) model of acute generalized seizure. Our results demonstrated that KA-induced seizures were accompanied by angiogenesis and BBB damage. In addition, expression of TSP-1, TGF-β1, and pSmad2/3 was increased. Rats treated with pyridoxal phosphate-6-azophenyl-2', 4'-disulfonic acid, a broad P2 receptor antagonist, or Reactive Blue 2, a potent P2Y4 receptor antagonist, showed significant attenuation of TSP-1 expression and Smad2/3 phosphorylation levels. Furthermore, angiogenesis, BBB damage, and acute seizure severity were also reduced. The inhibition of TSP-1 expression by siRNA or TGF-β1 activation by Leu-Ser-Lys-Leu (LSKL) treatment prevented KA-induced phosphorylation of Smad2/3, angiogenesis, BBB damage, and acute seizures. Our results strongly indicate that the P2Y4/TSP-1/TGF-β1/pSmad2/3/VEGF pathway plays an essential role in seizure pathophysiology and angiogenesis. Therapeutic interventions targeting this pathway may offer new treatment options for acute seizures.
Collapse
Affiliation(s)
- Yurong Zhang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, China
| | - Wei Zhu
- Shandong Academy of Medical Sciences, Jinan, China
| | - Haiying Yu
- Shandong Academy of Medical Sciences, Jinan, China
| | - Jie Yu
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, China
| | - Mengdi Zhang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, China
| | - Xiaohong Pan
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, China
| | - Xue Gao
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, China
| | - Qiaoyun Wang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, China
| | - Hongliu Sun
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, China.
| |
Collapse
|
237
|
Microglia along sex lines: From brain colonization, maturation and function, to implication in neurodevelopmental disorders. Semin Cell Dev Biol 2019; 94:152-163. [PMID: 31201858 DOI: 10.1016/j.semcdb.2019.06.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 06/10/2019] [Accepted: 06/11/2019] [Indexed: 12/30/2022]
Abstract
In addition to their traditional role as immune sentinels, recent discoveries over the last decade have shown that microglial functions now include regulation of neuronal/glial cell migration, differentiation and maturation, as well as neuronal network formation. It was thus proposed that disruption of these microglial roles, during critical periods of brain development, could lead to the pathological onset of several neurodevelopmental disorders, including autism spectrum disorder, attention deficit hyperactivity disorder, epilepsy, schizophrenia, and major depressive disorder. The prevalence of these disorders exhibits a clear distinction along sex lines with very little known about the mechanisms underlying this difference. One of the fundamental discoveries that arose from recent research into the physiological roles of microglia in neurodevelopment is their sexual dimorphism, raising the intriguing possibility that sex differences in microglial colonization, maturation and/or function in the developing brain could underlie the emergence of various neurodevelopmental disorders. This review discusses the physiological roles of microglia across neurodevelopment, these roles in the two sexes, and the recent evidence that microglial sexually dimorphic nature may contribute, at least partially, to neurodevelopmental disorders.
Collapse
|
238
|
Köles L, Szepesy J, Berekméri E, Zelles T. Purinergic Signaling and Cochlear Injury-Targeting the Immune System? Int J Mol Sci 2019; 20:ijms20122979. [PMID: 31216722 PMCID: PMC6627352 DOI: 10.3390/ijms20122979] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/14/2019] [Accepted: 06/14/2019] [Indexed: 02/06/2023] Open
Abstract
Hearing impairment is the most common sensory deficit, affecting more than 400 million people worldwide. Sensorineural hearing losses currently lack any specific or efficient pharmacotherapy largely due to the insufficient knowledge of the pathomechanism. Purinergic signaling plays a substantial role in cochlear (patho)physiology. P2 (ionotropic P2X and the metabotropic P2Y) as well as adenosine receptors expressed on cochlear sensory and non-sensory cells are involved mostly in protective mechanisms of the cochlea. They are implicated in the sensitivity adjustment of the receptor cells by a K+ shunt and can attenuate the cochlear amplification by modifying cochlear micromechanics. Cochlear blood flow is also regulated by purines. Here, we propose to comprehend this field with the purine-immune interactions in the cochlea. The role of harmful immune mechanisms in sensorineural hearing losses has been emerging in the horizon of cochlear pathologies. In addition to decreasing hearing sensitivity and increasing cochlear blood supply, influencing the immune system can be the additional avenue for pharmacological targeting of purinergic signaling in the cochlea. Elucidating this complexity of purinergic effects on cochlear functions is necessary and it can result in development of new therapeutic approaches in hearing disabilities, especially in the noise-induced ones.
Collapse
Affiliation(s)
- László Köles
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary.
| | - Judit Szepesy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary.
| | - Eszter Berekméri
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary.
- Department of Ecology, University of Veterinary Medicine, H-1078 Budapest, Hungary.
| | - Tibor Zelles
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, H-1089 Budapest, Hungary.
- Department of Pharmacology, Institute of Experimental Medicine, Hungarian Academy of Sciences, H-1083 Budapest, Hungary.
| |
Collapse
|
239
|
Kaur T, Clayman AC, Nash AJ, Schrader AD, Warchol ME, Ohlemiller KK. Lack of Fractalkine Receptor on Macrophages Impairs Spontaneous Recovery of Ribbon Synapses After Moderate Noise Trauma in C57BL/6 Mice. Front Neurosci 2019; 13:620. [PMID: 31263398 PMCID: PMC6585312 DOI: 10.3389/fnins.2019.00620] [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: 04/02/2019] [Accepted: 05/29/2019] [Indexed: 01/08/2023] Open
Abstract
Noise trauma causes loss of synaptic connections between cochlear inner hair cells (IHCs) and the spiral ganglion neurons (SGNs). Such synaptic loss can trigger slow and progressive degeneration of SGNs. Macrophage fractalkine signaling is critical for neuron survival in the injured cochlea, but its role in cochlear synaptopathy is unknown. Fractalkine, a chemokine, is constitutively expressed by SGNs and signals via its receptor CX3CR1 that is expressed on macrophages. The present study characterized the immune response and examined the function of fractalkine signaling in degeneration and repair of cochlear synapses following noise trauma. Adult mice wild type, heterozygous and knockout for CX3CR1 on a C57BL/6 background were exposed for 2 h to an octave band noise at 90 dB SPL. Noise exposure caused temporary shifts in hearing thresholds without any evident loss of hair cells in CX3CR1 heterozygous mice that have intact fractalkine signaling. Enhanced macrophage migration toward the IHC-synaptic region was observed immediately after exposure in all genotypes. Synaptic immunolabeling revealed a rapid loss of ribbon synapses throughout the basal turn of the cochlea of all genotypes. The damaged synapses spontaneously recovered in mice with intact CX3CR1. However, CX3CR1 knockout (KO) animals displayed enhanced synaptic degeneration that correlated with attenuated suprathreshold neural responses at higher frequencies. Exposed CX3CR1 KO mice also exhibited increased loss of IHCs and SGN cell bodies compared to exposed heterozygous mice. These results indicate that macrophages can promote repair of damaged synapses after moderate noise trauma and that repair requires fractalkine signaling.
Collapse
Affiliation(s)
- Tejbeer Kaur
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States
| | - Anna C Clayman
- Program in Audiology and Communication Sciences, Washington University School of Medicine, St. Louis, MO, United States
| | - Andrew J Nash
- Program in Audiology and Communication Sciences, Washington University School of Medicine, St. Louis, MO, United States
| | - Angela D Schrader
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States
| | - Mark E Warchol
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States
| | - Kevin K Ohlemiller
- Department of Otolaryngology, Washington University School of Medicine, St. Louis, MO, United States.,Program in Audiology and Communication Sciences, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
240
|
Leclercq K, Liefferinge JV, Albertini G, Neveux M, Dardenne S, Mairet‐Coello G, Vandenplas C, Deprez T, Chong S, Foerch P, Bentea E, Sato H, Maher P, Massie A, Smolders I, Kaminski RM. Anticonvulsant and antiepileptogenic effects of system xc− inactivation in chronic epilepsy models. Epilepsia 2019; 60:1412-1423. [DOI: 10.1111/epi.16055] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 05/10/2019] [Accepted: 05/10/2019] [Indexed: 12/14/2022]
Affiliation(s)
| | - Joeri Van Liefferinge
- Department of Pharmaceutical Chemistry Drug Analysis and Drug Information Center for Neurosciences C4N Vrije Universiteit Brussel Brussels Belgium
| | - Giulia Albertini
- Department of Pharmaceutical Chemistry Drug Analysis and Drug Information Center for Neurosciences C4N Vrije Universiteit Brussel Brussels Belgium
| | | | | | | | | | | | | | | | - Eduard Bentea
- Department of Pharmaceutical Biotechnology and Molecular Biology Center for Neurosciences C4N Vrije Universiteit Brussel Brussels Belgium
| | - Hideyo Sato
- Faculty of Medicine Niigata University Niigata Japan
| | - Pamela Maher
- Cellular Neurobiology Laboratory The Salk Institute for Biological Studies La Jolla California
| | - Ann Massie
- Department of Pharmaceutical Biotechnology and Molecular Biology Center for Neurosciences C4N Vrije Universiteit Brussel Brussels Belgium
| | - Ilse Smolders
- Department of Pharmaceutical Chemistry Drug Analysis and Drug Information Center for Neurosciences C4N Vrije Universiteit Brussel Brussels Belgium
| | | |
Collapse
|
241
|
Microglia-neuron crosstalk: Signaling mechanism and control of synaptic transmission. Semin Cell Dev Biol 2019; 94:138-151. [PMID: 31112798 DOI: 10.1016/j.semcdb.2019.05.017] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/17/2019] [Accepted: 05/16/2019] [Indexed: 12/13/2022]
Abstract
The continuous crosstalk between microglia and neurons is required for microglia housekeeping functions and contributes to brain homeostasis. Through these exchanges, microglia take part in crucial brain functions, including development and plasticity. The alteration of neuron-microglia communication contributes to brain disease states with consequences, ranging from synaptic function to neuronal survival. This review focuses on the signaling pathways responsible for neuron-microglia crosstalk, highlighting their physiological roles and their alteration or specific involvement in disease. In particular, we discuss studies, establishing how these signaling allow microglial cells to control relevant physiological functions during brain development, including synaptic formation and circuit refinement. In addition, we highlight how microglia and neurons interact functionally to regulate highly dynamical synaptic functions. Microglia are able to release several signaling molecules involved in the regulation of synaptic activity and plasticity. On the other side, molecules of neuronal origin control microglial processes motility in an activity-dependent manner. Indeed, the continuous crosstalk between microglia and neurons is required for the sensing and housekeeping functions of microglia and contributes to the maintenance of brain homeostasis and, particularly, to the sculpting of neuronal connections during development. These interactions lay on the delicate edge between physiological processes and homeostasis alteration in pathology and are themselves altered during neuroinflammation. The full description of these processes could be fundamental for understanding brain functioning in health and disease.
Collapse
|
242
|
Chaves-Filho AJM, Macedo DS, de Lucena DF, Maes M. Shared microglial mechanisms underpinning depression and chronic fatigue syndrome and their comorbidities. Behav Brain Res 2019; 372:111975. [PMID: 31136774 DOI: 10.1016/j.bbr.2019.111975] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/09/2019] [Accepted: 05/23/2019] [Indexed: 12/24/2022]
Abstract
In 2011, it was reviewed that a) there is a strong co-occurrence between major depression and chronic fatigue syndrome (CFS), with fatigue and physio-somatic symptoms being key symptoms of depression, and depressive symptoms appearing during the course of CFS; and b) the comorbidity between both disorders may in part be explained by activated immune-inflammatory pathways, including increased translocation of Gram-negative bacteria and increased levels of pro-inflammatory cytokines, such as interleukin (IL)-1. Nevertheless, the possible involvement of activated microglia in this comorbidity has remained unclear. This paper aims to review microglial disturbances in major depression, CFS and their comorbidity. A comprehensive literature search was conducted using the PubMed / MEDLINE database to identify studies, which are relevant to this current review. Depressed patients present neuroinflammatory alterations, probably related to microglial activation, while animal models show that a microglial response to immune challenges including lipopolysaccharides is accompanied by depressive-like behaviors. Recent evidence from preclinical studies indicates that activated microglia have a key role in the onset of fatigue. In chronic inflammatory conditions, such as infections and senescence, microglia orchestrate an inflammatory microenvironment thereby causing fatigue. In conclusion, based on our review we may posit that shared immune-inflammatory pathways and especially activated microglia underpin comorbid depression and CFS. As such, microglial activation and neuro-inflammation may be promising targets to treat the overlapping manifestations of both depression and CFS.
Collapse
Affiliation(s)
- Adriano José Maia Chaves-Filho
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil.
| | - Danielle S Macedo
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil; National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, Brazil.
| | - David Freitas de Lucena
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil.
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; IMPACT Strategic Research Center, Deakin University, Geelong, Australia.
| |
Collapse
|
243
|
Dual Functions of Microglia in Ischemic Stroke. Neurosci Bull 2019; 35:921-933. [PMID: 31062335 DOI: 10.1007/s12264-019-00388-3] [Citation(s) in RCA: 332] [Impact Index Per Article: 66.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 12/30/2018] [Indexed: 12/16/2022] Open
Abstract
Ischemic stroke is a leading cause of morbidity and mortality worldwide. Resident microglia are the principal immune cells of the brain, and the first to respond to the pathophysiological changes induced by ischemic stroke. Traditionally, it has been thought that microglial activation is deleterious in ischemic stroke, and therapies to suppress it have been intensively explored. However, increasing evidence suggests that microglial activation is also critical for neurogenesis, angiogenesis, and synaptic remodeling, thereby promoting functional recovery after cerebral ischemia. Here, we comprehensively review the dual role of microglia during the different phases of ischemic stroke, and the possible mechanisms controlling the post-ischemic activity of microglia. In addition, we discuss the dynamic interactions between microglia and other cells, such as neurons, astrocytes, oligodendrocytes, and endothelial cells within the brain parenchyma and the neurovascular unit.
Collapse
|
244
|
Context-Specific Switch from Anti- to Pro-epileptogenic Function of the P2Y 1 Receptor in Experimental Epilepsy. J Neurosci 2019; 39:5377-5392. [PMID: 31048325 DOI: 10.1523/jneurosci.0089-19.2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/21/2019] [Accepted: 03/23/2019] [Indexed: 12/15/2022] Open
Abstract
Extracellular ATP activates inflammatory responses to tissue injury. It is also implicated in establishing lasting network hyperexcitability in the brain by acting upon independent receptor systems. Whereas the fast-acting P2X channels have well-established roles driving neuroinflammation and increasing hyperexcitability, the slower-acting metabotropic P2Y receptors have received much less attention. Recent studies of P2Y1 receptor function in seizures and epilepsy have produced contradictory results, suggesting that the role of this receptor during seizure pathology may be highly sensitive to context. Here, by using male mice, we demonstrate that the metabotropic P2Y1 receptor mediates either proconvulsive or anticonvulsive responses, dependent on the time point of activation in relation to the induction of status epilepticus. P2Y1 deficiency or a P2Y1 antagonist (MRS2500) administered before a chemoconvulsant, exacerbates epileptiform activity, whereas a P2Y1 agonist (MRS2365) administered at this time point is anticonvulsant. When these drugs are administered after the onset of status epilepticus, however, their effect on seizure severity is reversed, with the antagonist now anticonvulsant and the agonist proconvulsant. This result was consistent across two different mouse models of status epilepticus (intra-amygdala kainic acid and intraperitoneal pilocarpine). Pharmacologic P2Y1 blockade during status epilepticus reduces also associated brain damage, delays the development of epilepsy and, when applied during epilepsy, suppresses spontaneous seizures, in mice. Our data show a context-specific role for P2Y1 during seizure pathology and demonstrate that blocking P2Y1 after status epilepticus and during epilepsy has potent anticonvulsive effects, suggesting that P2Y1 may be a novel candidate for the treatment of drug-refractory status epilepticus and epilepsy.SIGNIFICANCE STATEMENT This is the first study to fully characterize the contribution of a metabotropic purinergic P2Y receptor during acute seizures and epilepsy. The findings suggest that targeting P2Y1 may offer a potential novel treatment strategy for drug-refractory status epilepticus and epilepsy. Our data demonstrate a context-specific role of P2Y1 activation during seizures, switching from a proconvulsive to an anticonvulsive role depending on physiopathological context. Thus, our study provides a possible explanation for seemingly conflicting results obtained between studies of different brain diseases where P2Y1 targeting has been proposed as a potential treatment strategy and highlights that the timing of pharmacological interventions is of critical importance to the understanding of how receptors contribute to the generation of seizures and the development of epilepsy.
Collapse
|
245
|
Abstract
Human microglia regional heterogeneity and phenotypes determined by multiplexed single-cell mass cytometry Böttcher C, Schlickeiser S, Sneeboer MAM, et al. Nat Neurosci. 2019;22(1):78-90. doi:10.1038/s41593-018-0290-2 Microglia, the specialized innate immune cells of the central nervous system, play crucial roles in neural development and function. Different phenotypes and functions have been ascribed to rodent microglia, but little is known about human microglia (huMG) heterogeneity. Difficulties in procuring huMG and their susceptibility to cryopreservation damage have limited large-scale studies. Here we applied multiplexed mass cytometry for a comprehensive characterization of postmortem huMG (103-104 cells). We determined expression levels of 57 markers on huMG isolated from up to 5 different brain regions of 9 donors. We identified the phenotypic signature of huMG, which was distinct from peripheral myeloid cells but was comparable to fresh huMG. We detected microglia regional heterogeneity using a hybrid workflow combining Cytobank and R/Bioconductor for multidimensional data analysis. Together, these methodologies allowed us to perform high-dimensional, large-scale Immunophenotyping of huMG at the single-cell level, which facilitates their unambiguous profiling in health and disease.
Collapse
|
246
|
Uweru JO, Eyo UB. A decade of diverse microglial-neuronal physical interactions in the brain (2008-2018). Neurosci Lett 2019; 698:33-38. [PMID: 30625349 PMCID: PMC6435396 DOI: 10.1016/j.neulet.2019.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/29/2018] [Accepted: 01/01/2019] [Indexed: 12/17/2022]
Abstract
Microglia are unique cells of the central nervous system (CNS) with a distinct ontogeny and molecular profile. They are the predominant immune resident cell in the CNS. Recent studies have revealed a diversity of transient and terminal physical interactions between microglia and neurons in the vertebrate brain. In this review, we follow the historical trail of the discovery of these interactions, summarize their notable features, provide implications of these discoveries to CNS function, emphasize emerging themes along the way and peak into the future of what outstanding questions remain to move the field forward.
Collapse
Affiliation(s)
- Joseph O Uweru
- Department of Neuroscience, University of Virginia, Charlottesville, VA, United States; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, United States; Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, United States
| | - Ukpong B Eyo
- Department of Neuroscience, University of Virginia, Charlottesville, VA, United States; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, United States; Center for Brain Immunology and Glia (BIG), University of Virginia, Charlottesville, VA, United States.
| |
Collapse
|
247
|
Feng L, Murugan M, Bosco DB, Liu Y, Peng J, Worrell GA, Wang HL, Ta LE, Richardson JR, Shen Y, Wu LJ. Microglial proliferation and monocyte infiltration contribute to microgliosis following status epilepticus. Glia 2019; 67:1434-1448. [PMID: 31179602 DOI: 10.1002/glia.23616] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 12/15/2022]
Abstract
Microglial activation has been recognized as a major contributor to inflammation of the epileptic brain. Seizures are commonly accompanied by remarkable microgliosis and loss of neurons. In this study, we utilize the CX3CR1GFP/+ CCR2RFP/+ genetic mouse model, in which CX3CR1+ resident microglia and CCR2+ monocytes are labeled with GFP and RFP, respectively. Using a combination of time-lapse two-photon imaging and whole-cell patch clamp recording, we determined the distinct morphological, dynamic, and electrophysiological characteristics of infiltrated monocytes and resident microglia, and the evolution of their behavior at different time points following kainic acid-induced seizures. Seizure activated microglia presented enlarged somas with less ramified processes, whereas, infiltrated monocytes were smaller, highly motile cells that lacked processes. Moreover, resident microglia, but not infiltrated monocytes, proliferate locally in the hippocampus after seizure. Microglial proliferation was dependent on the colony-stimulating factor 1 receptor (CSF-1R) pathway. Pharmacological inhibition of CSF-1R reduced seizure-induced microglial proliferation, which correlated with attenuation of neuronal death without altering acute seizure behaviors. Taken together, we demonstrated that proliferation of activated resident microglia contributes to neuronal death in the hippocampus via CSF-1R after status epilepticus, providing potential therapeutic targets for neuroprotection in epilepsy.
Collapse
Affiliation(s)
- Lijie Feng
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China.,Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers University, Piscataway, New Jersey
| | - Madhuvika Murugan
- Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers University, Piscataway, New Jersey.,Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Dale B Bosco
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Yong Liu
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Jiyun Peng
- Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers University, Piscataway, New Jersey.,Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | | | - Hai-Long Wang
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Lauren E Ta
- Department of Neurology, Mayo Clinic, Rochester, Minnesota
| | - Jason R Richardson
- Department of Environmental Health Sciences, Robert Stempel School of Public Health and Social Work, Florida International University, Miami, Florida
| | - Yuxian Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Long-Jun Wu
- Department of Cell Biology and Neuroscience, School of Arts and Sciences, Rutgers University, Piscataway, New Jersey.,Department of Neurology, Mayo Clinic, Rochester, Minnesota.,Department of Neuroscience, Mayo Clinic, Jacksonville, Florida.,Department of Immunology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
248
|
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
|
249
|
Sepulveda-Rodriguez A, Li P, Khan T, Ma JD, Carlone CA, Bozzelli PL, Conant KE, Forcelli PA, Vicini S. Electroconvulsive Shock Enhances Responsive Motility and Purinergic Currents in Microglia in the Mouse Hippocampus. eNeuro 2019; 6:ENEURO.0056-19.2019. [PMID: 31058213 PMCID: PMC6498419 DOI: 10.1523/eneuro.0056-19.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/09/2019] [Indexed: 12/24/2022] Open
Abstract
Microglia are in a privileged position to both affect and be affected by neuroinflammation, neuronal activity and injury, which are all hallmarks of seizures and the epilepsies. Hippocampal microglia become activated after prolonged, damaging seizures known as status epilepticus (SE). However, since SE causes both hyperactivity and injury of neurons, the mechanisms triggering this activation remain unclear, as does the relevance of the microglial activation to the ensuing epileptogenic processes. In this study, we use electroconvulsive shock (ECS) to study the effect of neuronal hyperactivity without neuronal degeneration on mouse hippocampal microglia. Unlike SE, ECS did not alter hippocampal CA1 microglial density, morphology, or baseline motility. In contrast, both ECS and SE produced a similar increase in ATP-directed microglial process motility in acute slices, and similarly upregulated expression of the chemokine C-C motif chemokine ligand 2 (CCL2). Whole-cell patch-clamp recordings of hippocampal CA1sr microglia showed that ECS enhanced purinergic currents mediated by P2X7 receptors in the absence of changes in passive properties or voltage-gated currents, or changes in receptor expression. This differs from previously described alterations in intrinsic characteristics which coincided with enhanced purinergic currents following SE. These ECS-induced effects point to a "seizure signature" in hippocampal microglia characterized by altered purinergic signaling. These data demonstrate that ictal activity per se can drive alterations in microglial physiology without neuronal injury. These physiological changes, which up until now have been associated with prolonged and damaging seizures, are of added interest as they may be relevant to electroconvulsive therapy (ECT), which remains a gold-standard treatment for depression.
Collapse
Affiliation(s)
- Alberto Sepulveda-Rodriguez
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC 20007
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC 20007
| | - Pinggan Li
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC 20007
- Department of Pediatrics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Tahiyana Khan
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC 20007
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC 20007
| | - James D Ma
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC 20007
| | - Colby A Carlone
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC 20007
| | - P Lorenzo Bozzelli
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC 20007
- Department of Neuroscience, Georgetown University, Washington, DC 20007
| | - Katherine E Conant
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC 20007
- Department of Neuroscience, Georgetown University, Washington, DC 20007
| | - Patrick A Forcelli
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC 20007
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC 20007
| | - Stefano Vicini
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC 20007
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC 20007
| |
Collapse
|
250
|
Abstract
Cognitive impairments reported across psychiatric conditions (ie, major depressive disorder, bipolar disorder, schizophrenia, and posttraumatic stress disorder) strongly impair the quality of life of patients and the recovery of those conditions. There is therefore a great need for consideration for cognitive dysfunction in the management of psychiatric disorders. The redundant pattern of cognitive impairments across such conditions suggests possible shared mechanisms potentially leading to their development. Here, we review for the first time the possible role of inflammation in cognitive dysfunctions across psychiatric disorders. Raised inflammatory processes (microglia activation and elevated cytokine levels) across diagnoses could therefore disrupt neurobiological mechanisms regulating cognition, including Hebbian and homeostatic plasticity, neurogenesis, neurotrophic factor, the HPA axis, and the kynurenine pathway. This redundant association between elevated inflammation and cognitive alterations across psychiatric disorders hence suggests that a cross-disorder approach using pharmacological and nonpharmacological (ie, physical activity and nutrition) anti-inflammatory/immunomodulatory strategies should be considered in the management of cognition in psychiatry.
Collapse
|