1
|
Arizanovska D, Dallera CA, Folorunso OO, Bush GF, Frye JB, Doyle KP, Jagid JR, Wolosker H, Monaco BA, Cordeiro JG, Atkins CM, Griswold AJ, Liebl DJ. Cognitive dysfunction following brain trauma results from sex-specific reactivation of the developmental pruning processes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.13.607610. [PMID: 39211262 PMCID: PMC11360988 DOI: 10.1101/2024.08.13.607610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Cognitive losses resulting from severe brain trauma have long been associated with the focal region of tissue damage, leading to devastating functional impairment. For decades, researchers have focused on the sequelae of cellular alterations that exist within the perilesional tissues; however, few clinical trials have been successful. Here, we employed a mouse brain injury model that resulted in expansive synaptic damage to regions outside the focal injury. Our findings demonstrate that synaptic damage results from the prolonged increase in D-serine release from activated microglia and astrocytes, which leads to hyperactivation of perisynaptic NMDARs, tagging of damaged synapses by complement components, and the reactivation of developmental pruning processes. We show that this mechanistic pathway is reversible at several stages within a prolonged and progressive period of synaptic loss. Importantly, these key factors are present in acutely injured brain tissue acquired from patients with brain injury, which supports a therapeutic neuroprotective strategy.
Collapse
|
2
|
Hafez SMNA, Abdelhafez ESMN. The possible protective effect of l-cysteine in a rat model of sciatic nerve ischemia-reperfusion: A possible role for NRF1 and Caspase 3; Biochemical, Histological, and Immunohistochemical study. J Chem Neuroanat 2024; 137:102412. [PMID: 38460773 DOI: 10.1016/j.jchemneu.2024.102412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 03/11/2024]
Abstract
Organ damage brought on by ischemia is exacerbated by the reperfusion process. L-cysteine is a semi-essential amino acid that acts as a substrate for cystathionine-β-synthase in the central nervous system. The aim of this study was to investigate the possible protective effects of L- cysteine against the structural and biochemical changes that occur in the rat sciatic nerve after ischemia reperfusion (I/R) and to address some of the underlying mechanisms of these effects. Rats were divided into 4 groups: sham, l-cysteine, I/R, and l-cysteine- I/R groups. Specimens of sciatic nerve were processed for biochemical, histological, and immunohistochemical assessment. The results showed in I/R group, a significant increase in malondialdehyde with a significant decrease in both Nuclear respiratory factor-1 (NRF1) and superoxide dismutase levels. Moreover, with histological alteration. There was a significant increase in the mean surface area fraction of anti-caspase immunopositive cells as well as a significantdecrease in mean surface area fraction of anti-CD 34 immunopositive cells. In contrast, the l-cysteine- I/R group showed amelioration of these biochemical, structural, and immunohistochemical changes. To the best of our knowledge, this is the first study showed the protective effects of l-cysteine in sciatic nerve I/R via NRF1and caspase 3 modulation as well as telocyte activation.
Collapse
Affiliation(s)
| | - El-Shimaa M N Abdelhafez
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Faculty of Medicine, Minia 61111, Egypt.
| |
Collapse
|
3
|
Koole L, Martinez-Martinez P, Amelsvoort TV, Evelo CT, Ehrhart F. Interactive neuroinflammation pathways and transcriptomics-based identification of drugs and chemical compounds for schizophrenia. World J Biol Psychiatry 2024; 25:116-129. [PMID: 37961844 DOI: 10.1080/15622975.2023.2281514] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 11/06/2023] [Indexed: 11/15/2023]
Abstract
OBJECTIVES Schizophrenia is a psychiatric disorder affecting 1% of the population. Accumulating evidence indicates that neuroinflammation is involved in the pathology of these disorders by altering neurodevelopmental processes and specifically affecting glutamatergic signalling and astrocytic functioning. The aim of this study was to curate interactive biological pathways involved in schizophrenia for the identification of novel pharmacological targets implementing pathway, gene ontology, and network analysis. METHODS Neuroinflammatory pathways were created using PathVisio and published in WikiPathways. A transcriptomics dataset, originally created by Narla et al. was selected for data visualisation and analysis. Transcriptomics data was visualised within pathways and networks, extended with transcription factors, pathways, and drugs. Network hubs were determined based on degrees of connectivity. RESULTS Glutamatergic, immune, and astrocytic signalling as well as extracellular matrix reorganisation were altered in schizophrenia while we did not find an effect on the complement system. Pharmacological agents that target the glutamate receptor subunits, inflammatory mediators, and metabolic enzymes were identified. CONCLUSIONS New neuroinflammatory pathways incorporating the extracellular matrix, glutamatergic neurons, and astrocytes in the aetiology of schizophrenia were established. Transcriptomics based network analysis provided novel targets, including extra-synaptic glutamate receptors, glutamate transporters and extracellular matrix molecules that can be evaluated for therapeutic strategies.
Collapse
Affiliation(s)
- Lisa Koole
- Department of Bioinformatics - BiGCaT, NUTRIM, FHML, Maastricht University, Maastricht, The Netherlands
- Department of Psychiatry and Neuropsychology, MHeNs, FHML, Maastricht University, Maastricht, The Netherlands
| | - Pilar Martinez-Martinez
- Department of Psychiatry and Neuropsychology, MHeNs, FHML, Maastricht University, Maastricht, The Netherlands
| | - Therese van Amelsvoort
- Department of Psychiatry and Neuropsychology, MHeNs, FHML, Maastricht University, Maastricht, The Netherlands
| | - Chris T Evelo
- Department of Bioinformatics - BiGCaT, NUTRIM, FHML, Maastricht University, Maastricht, The Netherlands
| | - Friederike Ehrhart
- Department of Bioinformatics - BiGCaT, NUTRIM, FHML, Maastricht University, Maastricht, The Netherlands
- Department of Psychiatry and Neuropsychology, MHeNs, FHML, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
4
|
Hu Y, Tao W. Current perspectives on microglia-neuron communication in the central nervous system: Direct and indirect modes of interaction. J Adv Res 2024:S2090-1232(24)00006-7. [PMID: 38195039 DOI: 10.1016/j.jare.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 10/05/2023] [Accepted: 01/06/2024] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND The incessant communication that takes place between microglia and neurons is essential the development, maintenance, and pathogenesis of the central nervous system (CNS). As mobile phagocytic cells, microglia serve a critical role in surveilling and scavenging the neuronal milieu to uphold homeostasis. AIM OF REVIEW This review aims to discuss the various mechanisms that govern the interaction between microglia and neurons, from the molecular to the organ system level, and to highlight the importance of these interactions in the development, maintenance, and pathogenesis of the CNS. KEY SCIENTIFIC CONCEPTS OF REVIEW Recent research has revealed that microglia-neuron interaction is vital for regulating fundamental neuronal functions, such as synaptic pruning, axonal remodeling, and neurogenesis. The review will elucidate the intricate signaling pathways involved in these interactions, both direct and indirect, to provide a better understanding of the fundamental mechanisms of brain function. Furthermore, gaining insights into these signals could lead to the development of innovative therapies for neural disorders.
Collapse
Affiliation(s)
- Yue Hu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 220023, China; School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Weiwei Tao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing 220023, China; School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
5
|
Cserép C, Pósfai B, Szabadits E, Dénes Á. Contactomics of Microglia and Intercellular Communication. ADVANCES IN NEUROBIOLOGY 2024; 37:135-149. [PMID: 39207690 DOI: 10.1007/978-3-031-55529-9_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Microglia represent the main immunocompetent cell type in the parenchyma of the brain and the spinal cord, with roles extending way beyond their immune functions. While emerging data show the pivotal role of microglia in brain development, brain health and brain diseases, the exact mechanisms through which microglia contribute to complex neuroimmune interactions are still largely unclear. Understanding the communication between microglia and other cells represents an important cornerstone of these interactions, which may provide novel opportunities for therapeutic interventions in neurological or psychiatric disorders. As such, in line with studying the effects of the numerous soluble mediators that influence neuroimmune processes, attention on physical interactions between microglia and other cells in the CNS has increased substantially in recent years. In this chapter, we briefly summarize the latest literature on "microglial contactomics" and its functional implications in health and disease.
Collapse
Affiliation(s)
- Csaba Cserép
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Balázs Pósfai
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Eszter Szabadits
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Ádám Dénes
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary.
| |
Collapse
|
6
|
Le L, Miyanishi K, Tanaka J, Majewska AK. Microglial Regulation of Sleep and Wakefulness. ADVANCES IN NEUROBIOLOGY 2024; 37:243-260. [PMID: 39207696 DOI: 10.1007/978-3-031-55529-9_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Sleep serves a multitude of roles in brain maturation and function. Although the neural networks involved in sleep regulation have been extensively characterized, it is increasingly recognized that neurons are not the sole conductor orchestrating the rhythmic cycle of sleep and wakefulness. In the central nervous system, microglia have emerged as an important player in sleep regulation. Within the last two decades, microglia have gained substantial attention for carrying out numerous nonimmune tasks that are crucial for brain development and function by co-opting similar mechanisms used in their conventional immune functions. Here, we highlight the importance of microglia in sleep regulation with recent findings reporting an arrhythmic sleep/wake cycle in the absence of microglia. Although the underlying mechanisms for such regulation are still being uncovered, it is likely that microglial contributions to the rhythmic control of the sleep/wake cycle come from their influence on synaptic strength and neuronal activity. We review the current literature to provide speculative signaling pathways and suggest key questions for future research. Advancing our knowledge of the mechanistic contribution of microglia to sleep regulation will not only further our insight into this critical biological process but also be instrumental in providing novel therapeutic strategies for sleep disorders.
Collapse
Affiliation(s)
- Linh Le
- Department of Neuroscience, Del Monte Institute of Neuroscience, Center for Visual Science, Neuroscience Graduate Program, University of Rochester, Rochester, NY, USA
| | - Kazuya Miyanishi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Junya Tanaka
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, Ehime University, Toon, Japan.
| | - Ania K Majewska
- Department of Neuroscience, Del Monte Institute of Neuroscience, Center for Visual Science, Neuroscience Graduate Program, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
7
|
Anzovino A, Canepa E, Alves M, Lemon NL, Carare RO, Fossati S. Amyloid Beta Oligomers Activate Death Receptors and Mitochondria-Mediated Apoptotic Pathways in Cerebral Vascular Smooth Muscle Cells; Protective Effects of Carbonic Anhydrase Inhibitors. Cells 2023; 12:2840. [PMID: 38132159 PMCID: PMC10741628 DOI: 10.3390/cells12242840] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/08/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023] Open
Abstract
Amyloid beta (Aβ) deposition within the brain vasculature is an early hallmark of Alzheimer's disease (AD), which triggers loss of brain vascular smooth muscle cells (BVSMCs) in cerebral arteries, via poorly understood mechanisms, altering cerebral blood flow, brain waste clearance, and promoting cognitive impairment. We have previously shown that, in brain endothelial cells (ECs), vasculotropic Aβ species induce apoptosis through death receptors (DRs) DR4 and DR5 and mitochondria-mediated mechanisms, while FDA-approved carbonic anhydrase inhibitors (CAIs) prevent mitochondria-mediated EC apoptosis in vitro and in vivo. In this study, we analyzed Aβ-induced extrinsic and intrinsic (DR- and mitochondria-mediated) apoptotic pathways in BVSMC, aiming to unveil new therapeutic targets to prevent BVSMC stress and death. We show that both apoptotic pathways are activated in BVSMCs by oligomeric Aβ42 and Aβ40-Q22 (AβQ22) and mitochondrial respiration is severely impaired. Importantly, the CAIs methazolamide (MTZ) and acetazolamide (ATZ) prevent the pro-apoptotic effects in BVSMCs, while reducing caspase 3 activation and Aβ deposition in the arterial walls of TgSwDI animals, a murine model of cerebral amyloid angiopathy (CAA). This study reveals new molecular targets and a promising therapeutic strategy against BVSMC dysfunction in AD, CAA, and ARIA (amyloid-related imaging abnormalities) complications of recently FDA-approved anti-Aβ antibodies.
Collapse
Affiliation(s)
- Amy Anzovino
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, 3500 N Broad St, Philadelphia, PA 19140, USA; (A.A.); (E.C.); (M.A.); (N.L.L.)
| | - Elisa Canepa
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, 3500 N Broad St, Philadelphia, PA 19140, USA; (A.A.); (E.C.); (M.A.); (N.L.L.)
| | - Micaelly Alves
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, 3500 N Broad St, Philadelphia, PA 19140, USA; (A.A.); (E.C.); (M.A.); (N.L.L.)
| | - Nicole L. Lemon
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, 3500 N Broad St, Philadelphia, PA 19140, USA; (A.A.); (E.C.); (M.A.); (N.L.L.)
| | - Roxana O. Carare
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK;
| | - Silvia Fossati
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, 3500 N Broad St, Philadelphia, PA 19140, USA; (A.A.); (E.C.); (M.A.); (N.L.L.)
| |
Collapse
|
8
|
Guan PP, Ge TQ, Wang P. As a Potential Therapeutic Target, C1q Induces Synapse Loss Via Inflammasome-activating Apoptotic and Mitochondria Impairment Mechanisms in Alzheimer's Disease. J Neuroimmune Pharmacol 2023; 18:267-284. [PMID: 37386257 DOI: 10.1007/s11481-023-10076-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 06/16/2023] [Indexed: 07/01/2023]
Abstract
C1q, the initiator of the classical pathway of the complement system, is activated during Alzheimer's disease (AD) development and progression and is especially associated with the production and deposition of β-amyloid protein (Aβ) and phosphorylated tau in β-amyloid plaques (APs) and neurofibrillary tangles (NFTs). Activation of C1q is responsible for induction of synapse loss, leading to neurodegeneration in AD. Mechanistically, C1q could activate glial cells, which results in the loss of synapses via regulation of synapse pruning and phagocytosis in AD. In addition, C1q induces neuroinflammation by inducing proinflammatory cytokine secretion, which is partially mediated by inflammasome activation. Activation of inflammasomes might mediate the effects of C1q on induction of synapse apoptosis. On the other hand, activation of C1q impairs mitochondria, which hinders the renovation and regeneration of synapses. All these actions of C1q contribute to the loss of synapses during neurodegeneration in AD. Therefore, pharmacological, or genetic interventions targeting C1q may provide potential therapeutic strategies for combating AD.
Collapse
Affiliation(s)
- Pei-Pei Guan
- College of Life and Health Sciences, Northeastern University, 110819, Shenyang, People's Republic of China
| | - Tong-Qi Ge
- College of Life and Health Sciences, Northeastern University, 110819, Shenyang, People's Republic of China
| | - Pu Wang
- College of Life and Health Sciences, Northeastern University, 110819, Shenyang, People's Republic of China.
| |
Collapse
|
9
|
Datta D. Interrogating the Etiology of Sporadic Alzheimer's Disease Using Aging Rhesus Macaques: Cellular, Molecular, and Cortical Circuitry Perspectives. J Gerontol A Biol Sci Med Sci 2023; 78:1523-1534. [PMID: 37279946 PMCID: PMC10460555 DOI: 10.1093/gerona/glad134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Indexed: 06/08/2023] Open
Abstract
Aging is the most significant risk factor for neurodegenerative disorders such as Alzheimer's disease (AD) associated with profound socioeconomic and personal costs. Consequently, there is an urgent need for animal models that recapitulate the age-related spatial and temporal complexity and patterns of pathology identical to human AD. Our research in aging nonhuman primate models involving rhesus macaques has revealed naturally occurring amyloid and tau pathology, including the formation of amyloid plaques and neurofibrillary tangles comprising hyperphosphorylated tau. Moreover, rhesus macaques exhibit synaptic dysfunction in association cortices and cognitive impairments with advancing age, and thus can be used to interrogate the etiological mechanisms that generate neuropathological cascades in sporadic AD. Particularly, unique molecular mechanisms (eg, feedforward cyclic adenosine 3',5'-monophosphate [cAMP]-Protein kinase A (PKA)-calcium signaling) in the newly evolved primate dorsolateral prefrontal cortex are critical for persistent firing required for subserving higher-order cognition. For example, dendritic spines in primate dorsolateral prefrontal cortex contain a specialized repertoire of proteins to magnify feedforward cAMP-PKA-calcium signaling such as N-methyl-d-aspartic acid receptors and calcium channels on the smooth endoplasmic reticulum (eg, ryanodine receptors). This process is constrained by phosphodiesterases (eg, PDE4) that hydrolyze cAMP and calcium-buffering proteins (eg, calbindin) in the cytosol. However, genetic predispositions and age-related insults exacerbate feedforward cAMP-Protein kinase A-calcium signaling pathways that induce a myriad of downstream effects, including the opening of K+ channels to weaken network connectivity, calcium-mediated dysregulation of mitochondria, and activation of inflammatory cascades to eliminate synapses, thereby increasing susceptibility to atrophy. Therefore, aging rhesus macaques provide an invaluable model to explore novel therapeutic strategies in sporadic AD.
Collapse
Affiliation(s)
- Dibyadeep Datta
- Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
10
|
Michailidou I, Fluiter K, Boziki M, Grigoriadis N, Baas F. Editorial: Complement in nervous system disease. Front Cell Neurosci 2023; 17:1268023. [PMID: 37614913 PMCID: PMC10442514 DOI: 10.3389/fncel.2023.1268023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/25/2023] Open
Affiliation(s)
- Iliana Michailidou
- Laboratory of Experimental Neurology and Neuroimmunology and the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kees Fluiter
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology and the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology and the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Frank Baas
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
11
|
Cangalaya C, Wegmann S, Sun W, Diez L, Gottfried A, Richter K, Stoyanov S, Pakan J, Fischer KD, Dityatev A. Real-time mechanisms of exacerbated synaptic remodeling by microglia in acute models of systemic inflammation and tauopathy. Brain Behav Immun 2023; 110:245-259. [PMID: 36906076 DOI: 10.1016/j.bbi.2023.02.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/13/2023] Open
Abstract
Remodeling of synapses by microglia is essential for synaptic plasticity in the brain. However, during neuroinflammation and neurodegenerative diseases, microglia can induce excessive synaptic loss, although the precise underlying mechanisms are unknown. To directly observe microglia-synapse interactions under inflammatory conditions, we performed in vivo two-photon time-lapse imaging of microglia-synapse interactions after bacterial lipopolysaccharide administration to model systemic inflammation, or after inoculation of Alzheimer's disease (AD) brain extracts to model disease-associated neuroinflammatory microglial response. Both treatments prolonged microglia-neuron contacts, decreased basal surveillance of synapses and promoted synaptic remodeling in response to synaptic stress induced by focal single-synapse photodamage. Spine elimination correlated with the expression of microglial complement system/phagocytic proteins and the occurrence of synaptic filopodia. Microglia were observed contacting spines, then stretching and phagocytosing spine head filopodia. Thus, in response to inflammatory stimuli microglia exacerbated spine remodeling through prolonged microglial contact and elimination of spines 'tagged' by synaptic filopodia.
Collapse
Affiliation(s)
- Carla Cangalaya
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany; Institut für Biochemie und Zellbiologie, Otto-von-Guericke-University, Medical Faculty, Magdeburg, Germany; ESF International Graduate School on Analysis, Imaging and Modelling of Neuronal and Inflammatory Processes, Magdeburg, Germany
| | - Susanne Wegmann
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Weilun Sun
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Lisa Diez
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Anna Gottfried
- Institut für Biochemie und Zellbiologie, Otto-von-Guericke-University, Medical Faculty, Magdeburg, Germany
| | - Karin Richter
- Institut für Biochemie und Zellbiologie, Otto-von-Guericke-University, Medical Faculty, Magdeburg, Germany
| | - Stoyan Stoyanov
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Janelle Pakan
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany; Institute of Cognitive Neurology and Dementia Research, Otto-von-Guericke University, Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Klaus-Dieter Fischer
- Institut für Biochemie und Zellbiologie, Otto-von-Guericke-University, Medical Faculty, Magdeburg, Germany
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany; Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany.
| |
Collapse
|
12
|
Ma Y, Liu Z, Jiang L, Wang L, Li Y, Liu Y, Wang Y, Yang GY, Ding J, Zhang Z. Endothelial progenitor cell transplantation attenuates synaptic loss associated with enhancing complement receptor 3-dependent microglial/macrophage phagocytosis in ischemic mice. J Cereb Blood Flow Metab 2023; 43:379-392. [PMID: 36457150 PMCID: PMC9941864 DOI: 10.1177/0271678x221135841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 09/01/2022] [Accepted: 09/26/2022] [Indexed: 12/04/2022]
Abstract
Endothelial progenitor cell (EPC) transplantation has therapeutic effects in cerebral ischemia. However, how EPCs modulate microglial activity remains unclear. In the study, we explored whether EPCs modulated microglial/macrophage activity and facilitated injured brain repair. Adult male mice (n = 184) underwent transient middle cerebral artery occlusion, and EPCs were transplanted into the brain immediately after ischemia. Microglial/macrophage activity and complement receptor 3 (CR3) expression were evaluated in ischemic brains and cultured microglia. CR3 agonist leukadherin-1 was administrated into mice immediately after ischemia to imitate the effects of EPCs. Synaptophysin and postsynaptic density protein 95 (PSD-95) expressions were detected in EPC- and leukadherin-1 treated mice. We found that EPC transplantation increased the number of M2 microglia/macrophage-phagocytizing apoptotic cells and CR3 expression in ischemic brains at 3 days after ischemia (p < 0.05). EPC-conditional medium or cultured EPCs increased microglial migration and phagocytosis and upregulated CR3 expression in cultured microglia under oxygen-glucose deprivation condition (p < 0.05). Leukadherin-1 reduced brain atrophy volume and neurological deficits at 14 days after ischemia (p < 0.05). Both EPC transplantation and leukadherin-1 increased synaptophysin and PSD-95 expression at 14 days after ischemia (p < 0.05). EPC transplantation promoted CR3-mediated microglial/macrophage phagocytosis and subsequently attenuated synaptic loss. Our study provided a novel therapeutic mechanism for EPCs.
Collapse
Affiliation(s)
- Yuanyuan Ma
- Department of Neurology, Zhongshan Hospital, Fudan University,
Shanghai, China
- Department of Neurology, Ruijin Hospital, School of Medicine and
School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai,
China
| | - Ze Liu
- Department of Neurology, Ruijin Hospital, School of Medicine and
School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai,
China
| | - Lu Jiang
- Department of Neurology, Ruijin Hospital, School of Medicine and
School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai,
China
| | - Liping Wang
- Department of Neurology, Renji Hospital, School of Medicine,
Shanghai Jiao Tong University, Shanghai, China
| | - Yongfang Li
- Department of Neurology, Ruijin Hospital, School of Medicine and
School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai,
China
| | - Yanqun Liu
- Department of Neurology, Changhai Hospital, Second Military
Medical University, Shanghai, China
| | - Yongting Wang
- Department of Neurology, Ruijin Hospital, School of Medicine and
School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai,
China
| | - Guo-Yuan Yang
- Department of Neurology, Ruijin Hospital, School of Medicine and
School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai,
China
| | - Jing Ding
- Department of Neurology, Zhongshan Hospital, Fudan University,
Shanghai, China
| | - Zhijun Zhang
- Department of Neurology, Ruijin Hospital, School of Medicine and
School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai,
China
| |
Collapse
|
13
|
Enomoto S, Ohgidani M, Sagata N, Inamine S, Kato TA. Preliminary analysis of hippocampus synaptic apoptosis and microglial phagocytosis induced by severe restraint stress. Neuropsychopharmacol Rep 2023; 43:120-125. [PMID: 36419367 PMCID: PMC10009418 DOI: 10.1002/npr2.12298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/11/2022] [Accepted: 10/14/2022] [Indexed: 11/25/2022] Open
Abstract
AIM Several studies reported stress-induced microglial phagocytosis, but the biochemical mechanisms by which stress alters microglial synaptic phagocytosis are not fully uncovered. Local or limited apoptosis without cell death was observed at neuronal synapses in previous studies, and proposed as an upstream mechanism for microglial synapse elimination. In this micro-report, we aimed to preliminary examine local synaptic apoptosis in the mouse hippocampus following severe restraint stress, and its effect on microglial phagocytosis. METHODS Mice were exposed to 10-day water immersion restraint stress (WIRS). Brain sections including stratum lucidum in the hippocampal CA3 subfield were stained with antibodies against cleaved caspase 3, ionized calcium-binding adapter molecule1 (Iba1), lysosomal-associated membrane protein1 (LAMP1), vesicular glutamate transporter1 (VGLUT1). Co-localization among these proteins were calculated. RESULTS Our image analysis revealed that synaptic apoptosis was increased while there were no significant changes in microglial phagocytic activity and synaptic phagocytosis following 10-day WIRS. CONCLUSION Severe restraint stress enhanced pre-synaptic apoptosis in mouse CA3 stratum lucidum region, but did not promote microglial phagocytosis. The phenomenon microglia fail to phagocytose weakened and unnecessary synapses may be related to pathology of stress.
Collapse
Affiliation(s)
- Shingo Enomoto
- Self Defense Force, Fukuoka Hospital, Fukuoka, Japan.,Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masahiro Ohgidani
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Hokkaido, Japan
| | - Noriaki Sagata
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shogo Inamine
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takahiro A Kato
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
14
|
Rasia-Filho AA, Calcagnotto ME, von Bohlen Und Halbach O. Glial Cell Modulation of Dendritic Spine Structure and Synaptic Function. ADVANCES IN NEUROBIOLOGY 2023; 34:255-310. [PMID: 37962798 DOI: 10.1007/978-3-031-36159-3_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Glia comprise a heterogeneous group of cells involved in the structure and function of the central and peripheral nervous system. Glial cells are found from invertebrates to humans with morphological specializations related to the neural circuits in which they are embedded. Glial cells modulate neuronal functions, brain wiring and myelination, and information processing. For example, astrocytes send processes to the synaptic cleft, actively participate in the metabolism of neurotransmitters, and release gliotransmitters, whose multiple effects depend on the targeting cells. Human astrocytes are larger and more complex than their mice and rats counterparts. Astrocytes and microglia participate in the development and plasticity of neural circuits by modulating dendritic spines. Spines enhance neuronal connectivity, integrate most postsynaptic excitatory potentials, and balance the strength of each input. Not all central synapses are engulfed by astrocytic processes. When that relationship occurs, a different pattern for thin and large spines reflects an activity-dependent remodeling of motile astrocytic processes around presynaptic and postsynaptic elements. Microglia are equally relevant for synaptic processing, and both glial cells modulate the switch of neuroendocrine secretion and behavioral display needed for reproduction. In this chapter, we provide an overview of the structure, function, and plasticity of glial cells and relate them to synaptic maturation and modulation, also involving neurotrophic factors. Together, neurons and glia coordinate synaptic transmission in both normal and abnormal conditions. Neglected over decades, this exciting research field can unravel the complexity of species-specific neural cytoarchitecture as well as the dynamic region-specific functional interactions between diverse neurons and glial subtypes.
Collapse
Affiliation(s)
- Alberto A Rasia-Filho
- Department of Basic Sciences/Physiology and Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, RS, Brazil
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Maria Elisa Calcagnotto
- Graduate Program in Neuroscience, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Department of Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Graduate Program in Biochemistry, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Graduate Program in Psychiatry and Behavioral Science, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | |
Collapse
|
15
|
Lala T, Doan JK, Takatsu H, Hartzell HC, Shin HW, Hall RA. Phosphatidylserine exposure modulates adhesion GPCR BAI1 (ADGRB1) signaling activity. J Biol Chem 2022; 298:102685. [PMID: 36370845 PMCID: PMC9723945 DOI: 10.1016/j.jbc.2022.102685] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 10/28/2022] [Accepted: 10/31/2022] [Indexed: 11/10/2022] Open
Abstract
Brain-specific angiogenesis inhibitor 1 (BAI1; also called ADGRB1 or B1) is an adhesion G protein-coupled receptor known from studies on macrophages to bind to phosphatidylserine (PS) on apoptotic cells via its N-terminal thrombospondin repeats. A separate body of work has shown that B1 regulates postsynaptic function and dendritic spine morphology via signaling pathways involving Rac and Rho. However, it is unknown if PS binding by B1 has any effect on the receptor's signaling activity. To shed light on this subject, we studied G protein-dependent signaling by B1 in the absence and presence of coexpression with the PS flippase ATP11A in human embryonic kidney 293T cells. ATP11A expression reduced the amount of PS exposed extracellularly and also strikingly reduced the signaling activity of coexpressed full-length B1 but not a truncated version of the receptor lacking the thrombospondin repeats. Further experiments with an inactive mutant of ATP11A showed that the PS flippase function of ATP11A was required for modulation of B1 signaling. In coimmunoprecipitation experiments, we made the surprising finding that ATP11A not only modulates B1 signaling but also forms complexes with B1. Parallel studies in which PS in the outer leaflet was reduced by an independent method, deletion of the gene encoding the endogenous lipid scramblase anoctamin 6 (ANO6), revealed that this manipulation also markedly reduced B1 signaling. These findings demonstrate that B1 signaling is modulated by PS exposure and suggest a model in which B1 serves as a PS sensor at synapses and in other cellular contexts.
Collapse
Affiliation(s)
- Trisha Lala
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Juleva K Doan
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Hiroyuki Takatsu
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - H Criss Hartzell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Hye-Won Shin
- Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Randy A Hall
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, Georgia, USA.
| |
Collapse
|
16
|
Romero-Molina C, Garretti F, Andrews SJ, Marcora E, Goate AM. Microglial efferocytosis: Diving into the Alzheimer's disease gene pool. Neuron 2022; 110:3513-3533. [PMID: 36327897 DOI: 10.1016/j.neuron.2022.10.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/04/2022] [Accepted: 10/07/2022] [Indexed: 11/07/2022]
Abstract
Genome-wide association studies and functional genomics studies have linked specific cell types, genes, and pathways to Alzheimer's disease (AD) risk. In particular, AD risk alleles primarily affect the abundance or structure, and thus the activity, of genes expressed in macrophages, strongly implicating microglia (the brain-resident macrophages) in the etiology of AD. These genes converge on pathways (endocytosis/phagocytosis, cholesterol metabolism, and immune response) with critical roles in core macrophage functions such as efferocytosis. Here, we review these pathways, highlighting relevant genes identified in the latest AD genetics and genomics studies, and describe how they may contribute to AD pathogenesis. Investigating the functional impact of AD-associated variants and genes in microglia is essential for elucidating disease risk mechanisms and developing effective therapeutic approaches.
Collapse
Affiliation(s)
- Carmen Romero-Molina
- Ronald M. Loeb Center for Alzheimer's Disease, 1 Gustave L. Levy Place, New York, NY 10029-6574, USA; Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Francesca Garretti
- Ronald M. Loeb Center for Alzheimer's Disease, 1 Gustave L. Levy Place, New York, NY 10029-6574, USA; Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shea J Andrews
- Ronald M. Loeb Center for Alzheimer's Disease, 1 Gustave L. Levy Place, New York, NY 10029-6574, USA; Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
| | - Edoardo Marcora
- Ronald M. Loeb Center for Alzheimer's Disease, 1 Gustave L. Levy Place, New York, NY 10029-6574, USA; Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Alison M Goate
- Ronald M. Loeb Center for Alzheimer's Disease, 1 Gustave L. Levy Place, New York, NY 10029-6574, USA; Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
17
|
Devinney MJ, VanDusen KW, Kfouri JM, Avasarala P, Spector AR, Mathew JP, Berger M. The potential link between obstructive sleep apnea and postoperative neurocognitive disorders: current knowledge and possible mechanisms. Can J Anaesth 2022; 69:1272-1287. [PMID: 35982354 PMCID: PMC9924301 DOI: 10.1007/s12630-022-02302-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 04/08/2022] [Accepted: 04/08/2022] [Indexed: 01/12/2023] Open
Abstract
PURPOSE This narrative review examines the current evidence on whether obstructive sleep apnea (OSA) is associated with postoperative delirium (POD) and postoperative cognitive dysfunction (POCD). The mechanisms that could predispose OSA patients to these disorders are also explored. SOURCE Relevant literature was identified by searching for pertinent terms in Medline®, Pubmed, ScopusTM, and Google scholar databases. Case reports, abstracts, review articles, original research articles, and meta-analyses were reviewed. The bibliographies of retrieved sources were also searched to identify relevant papers. PRINCIPAL FINDINGS Seven studies have investigated the association between OSA and POD, with mixed results. No studies have examined the potential link between OSA and POCD. If these relationships exist, they could be mediated by several mechanisms, including increased neuroinflammation, blood-brain barrier breakdown, cerebrovascular disease, Alzheimer's disease neuropathology, disrupted cerebral autoregulation, sleep disruption, sympathovagal imbalance, and/or disrupted brain bioenergetics. CONCLUSION There is very limited evidence that OSA plays a role in postoperative neurocognitive disorders because few studies have been conducted in the perioperative setting. Additional perioperative prospective observational cohort studies and randomized controlled trials of sleep apnea treatment are needed. These investigations should also assess potential underlying mechanisms that could predispose patients with OSA to postoperative neurocognitive disorders. This review highlights the need for more research to improve postoperative neurocognitive outcomes for patients with OSA.
Collapse
Affiliation(s)
- Michael J Devinney
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA.
- Duke Hospital South, 3094 MS 01, 40 Medicine Circle, Rm 4324, Orange Zone, Durham, NC, 27710, USA.
| | - Keith W VanDusen
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Jad M Kfouri
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Pallavi Avasarala
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Andrew R Spector
- Department of Neurology, Duke University Medical Center, Durham, NC, USA
| | - Joseph P Mathew
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| | - Miles Berger
- Department of Anesthesiology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
18
|
Zheng Y, Fan L, Xia S, Yang Q, Zhang Z, Chen H, Zeng H, Fu X, Peng Y, Xu C, Yu K, Liu F, Cao S. Role of complement C1q/C3-CR3 signaling in brain injury after experimental intracerebral hemorrhage and the effect of minocycline treatment. Front Immunol 2022; 13:919444. [PMID: 36189326 PMCID: PMC9520460 DOI: 10.3389/fimmu.2022.919444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
AimThe complement cascade is activated and may play an important pathophysiologic role in brain injury after experimental intracerebral hemorrhage (ICH). However, the exact mechanism of specific complement components has not been well studied. This study determined the role of complement C1q/C3-CR3 signaling in brain injury after ICH in mice. The effect of minocycline on C1q/C3-CR3 signaling-induced brain damage was also examined.MethodsThere were three parts to the study. First, the natural time course of C1q and CR3 expression was determined within 7 days after ICH. Second, mice had an ICH with CR3 agonists, LA-1 or vehicle. Behavioral score, neuronal cell death, hematoma volume, and oxidative stress response were assessed at 7 days after ICH. Third, the effect of minocycline on C1q/C3-CR3 signaling and brain damage was examined.ResultsThere were increased numbers of C1q-positive and CR3-positive cells after ICH. Almost all perihematomal C1q-positive and CR3-positive cells were microglia/macrophages. CR3 agonist LA-1 aggravated neurological dysfunction, neuronal cell death, and oxidative stress response on day 7 after ICH, as well as enhancing the expression of the CD163/HO-1 pathway and accelerating hematoma resolution. Minocycline treatment exerted neuroprotective effects on brain injury following ICH, partly due to the inhibition of C1q/C3-CR3 signaling, and that could be reversed by LA-1.ConclusionsThe complement C1q/C3-CR3 signaling is upregulated after ICH. The activation of C1q/C3-CR3 signaling by LA-1 aggravates brain injury following ICH. The neuroprotection of minocycline, at least partly, is involved with the repression of the C1q/C3-CR3 signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Fuyi Liu
- *Correspondence: Fuyi Liu, ; Shenglong Cao,
| | | |
Collapse
|
19
|
Jiang L, Hao J, Yang XL, Zhu JX, Wang Y, Huang YL, Sun YE, Mao YT, Ni K, Gu XP, Ma ZL. Basolateral Amygdala Reactive Microglia May Contribute to Synaptic Impairment and Depressive-Like Behavior in Mice with Bone Cancer Pain. Neurochem Res 2022; 47:3454-3463. [PMID: 36002639 DOI: 10.1007/s11064-022-03731-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 08/07/2022] [Accepted: 08/18/2022] [Indexed: 12/24/2022]
Abstract
Anxiety and depression induced by cancer-related pain disturb quality of life and willingness to survive. As a component of the limbic system, the basolateral amygdala (BLA) is critical for processing negative emotions. The reactive microglial engulfment of synapses may promote depression during adolescence. However, whether microglia phagocytose synapses to mediate cancer pain-induced depression remains unclear. The present study established a bone cancer-pain model to investigate the association between dendritic spine synapses and depressive-like behavior and explore the phagocytic function of microglia in the BLA. We found that tumor-bearing mice experienced postoperative pain-related depression, and their BLAs exhibited reactive microglia, as well as phagocytic synapses. The microglial inhibitor minocycline effectively mitigated depressive behavior, synaptic damage, and the phagocytic function of microglia. Our study implicates microglia-mediated synaptic loss in the BLA may act as the pathological basis of depressive-like behavior in bone cancer pain model.
Collapse
Affiliation(s)
- Li Jiang
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Jing Hao
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Xu-Li Yang
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Ji-Xiang Zhu
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Yu Wang
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Yu-Lin Huang
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Yu-E Sun
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Yan-Ting Mao
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Kun Ni
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China.
| | - Xiao-Ping Gu
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China.
| | - Zheng-Liang Ma
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, 321 Zhongshan Road, Nanjing, 210008, China. .,Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China.
| |
Collapse
|
20
|
Ball JB, Green-Fulgham SM, Watkins LR. Mechanisms of Microglia-Mediated Synapse Turnover and Synaptogenesis. Prog Neurobiol 2022; 218:102336. [DOI: 10.1016/j.pneurobio.2022.102336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/30/2022] [Accepted: 08/02/2022] [Indexed: 10/31/2022]
|
21
|
Gonçalves de Andrade E, González Ibáñez F, Tremblay MÈ. Microglia as a Hub for Suicide Neuropathology: Future Investigation and Prevention Targets. Front Cell Neurosci 2022; 16:839396. [PMID: 35663424 PMCID: PMC9158339 DOI: 10.3389/fncel.2022.839396] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 02/22/2022] [Indexed: 12/27/2022] Open
Abstract
Suicide is a complex public health challenge associated worldwide with one death every 40 s. Research advances in the neuropathology of suicidal behaviors (SB) have defined discrete brain changes which may hold the key to suicide prevention. Physiological differences in microglia, the resident immune cells of the brain, are present in post-mortem tissue samples of individuals who died by suicide. Furthermore, microglia are mechanistically implicated in the outcomes of important risk factors for SB, including early-life adversity, stressful life events, and psychiatric disorders. SB risk factors result in inflammatory and oxidative stress activities which could converge to microglial synaptic remodeling affecting susceptibility or resistance to SB. To push further this perspective, in this Review we summarize current areas of opportunity that could untangle the functional participation of microglia in the context of suicide. Our discussion centers around microglial state diversity in respect to morphology, gene and protein expression, as well as function, depending on various factors, namely brain region, age, and sex.
Collapse
Affiliation(s)
- Elisa Gonçalves de Andrade
- Neuroscience Graduate Program, Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Fernando González Ibáñez
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
- *Correspondence: Marie-Ève Tremblay,
| |
Collapse
|
22
|
Wang J, Yu C, Zhuang J, Qi W, Jiang J, Liu X, Zhao W, Cao Y, Wu H, Qi J, Zhao RC. The role of phosphatidylserine on the membrane in immunity and blood coagulation. Biomark Res 2022; 10:4. [PMID: 35033201 PMCID: PMC8760663 DOI: 10.1186/s40364-021-00346-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/01/2021] [Indexed: 12/17/2022] Open
Abstract
The negatively charged aminophospholipid, phosphatidylserine (PtdSer), is located in the inner leaflet of the plasma membrane in normal cells, and may be exposed to the outer leaflet under some immune and blood coagulation processes. Meanwhile, Ptdser exposed to apoptotic cells can be recognized and eliminated by various immune cells, whereas on the surface of activated platelets Ptdser interacts with coagulation factors prompting enhanced production of thrombin which significantly facilitates blood coagulation. In the case where PtdSer fails in exposure or mistakenly occurs, there are occurrences of certain immunological and haematological diseases, such as the Scott syndrome and Systemic lupus erythematosus. Besides, viruses (e.g., Human Immunodeficiency Virus (HIV), Ebola virus (EBOV)) can invade host cells through binding the exposed PtdSer. Most recently, the Corona Virus Disease 2019 (COVID-19) has been similarly linked to PtdSer or its receptors. Therefore, it is essential to comprehensively understand PtdSer and its functional characteristics. Therefore, this review summarizes Ptdser, its eversion mechanism; interaction mechanism, particularly with its immune receptors and coagulation factors; recognition sites; and its function in immune and blood processes. This review illustrates the potential aspects for the underlying pathogenic mechanism of PtdSer-related diseases, and the discovery of new therapeutic strategies as well.
Collapse
Affiliation(s)
- Jiao Wang
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444, China.
| | - Changxin Yu
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
| | - Junyi Zhuang
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
| | - Wenxin Qi
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
| | - Jiawen Jiang
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
| | - Xuanting Liu
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
| | - Wanwei Zhao
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
| | - Yiyang Cao
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
| | - Hao Wu
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
| | - Jingxuan Qi
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
| | - Robert Chunhua Zhao
- School of Life Sciences, Shanghai University, 99 Shangda Road, Shanghai, 200444, China.
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, No. 5 Dongdansantiao, Beijing, 100005, China.
- Centre of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, China.
- Beijing Key Laboratory of New Drug Development and Clinical Trial of Stem Cell Therapy (BZ0381), Beijing, China.
| |
Collapse
|
23
|
Yoshimura A, Ohyagi M, Ito M. T cells in the brain inflammation. Adv Immunol 2022; 157:29-58. [PMID: 37061287 DOI: 10.1016/bs.ai.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The immune system is deeply involved in autoimmune diseases of the central nervous system (CNS), such as multiple sclerosis, N-methyl-d-aspartate (NMDA) receptor encephalitis, and narcolepsy. Additionally, the immune system is involved in various brain diseases including cerebral infarction and neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). In particular, reports related to T cells are increasing. T cells may also play important roles in brain deterioration and dementia that occur with aging. Our understanding of the role of immune cells in the context of the brain has been greatly improved by the use of acute ischemic brain injury models. Additionally, similar neural damage and repair events are shown to occur in more chronic brain neurodegenerative brain diseases. In this review, we focus on the role of T cells, including CD4+ T cells, CD8+ T cells and regulatory T cells (Tregs) in cerebral infarction and neurodegenerative diseases.
Collapse
|
24
|
Choudhury ME, Mikami K, Nakanishi Y, Matsuura T, Utsunomiya R, Yano H, Kubo M, Ando R, Iwanami J, Yamashita M, Nagai M, Tanaka J. Insomnia and depressive behavior of MyD88-deficient mice: Relationships with altered microglial functions. J Neuroimmunol 2021; 363:577794. [PMID: 34971898 DOI: 10.1016/j.jneuroim.2021.577794] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/16/2021] [Accepted: 12/19/2021] [Indexed: 12/27/2022]
Abstract
Myeloid differentiation primary response gene 88 (MyD88) is essential for microglial activation. Despite the significant role of microglia in regulating sleep homeostasis, the contribution of MyD88 to sleep is yet to be determined. To address this, we performed electroencephalographic and electromyographic recordings on MyD88-KO mice and wild-type mice to investigate their sleep/wake cycles. In the daytime, MyD88-KO mice exhibited prolonged wakefulness and shorter non-rapid eye movement sleep duration. Tail suspension and sucrose preference tests revealed that MyD88-KO mice displayed a depressive-like phenotype. We determined monoamines in the prefrontal cortex (PFC) using high-performance liquid chromatography and observed a decreased content of serotonin in the PFC of MyD88-KO mice. Flow cytometry revealed that CD11b, CD45, and F4/80 expressions were elevated at Zeitgeber time (ZT) 1 compared to at ZT13 only in wild-type mice. Furthermore, MFG-E8 and C1qB-tagged synapses were enhanced at ZT1 in the PFC of wild-type mice but not in MyD88-KO mice. Primary cultured microglia from MyD88-KO mice revealed decreased phagocytic ability. These findings indicate that genetic deletion of MyD88 induces insomnia and depressive behavior, at least in part, by affecting microglial homeostasis functions and lowering the serotonergic neuronal output.
Collapse
Affiliation(s)
- Mohammed E Choudhury
- Department of Molecular and Cellular Physiology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan.
| | - Kanta Mikami
- Department of Molecular and Cellular Physiology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Yuiko Nakanishi
- Department of Molecular and Cellular Physiology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Taisei Matsuura
- Department of Molecular and Cellular Physiology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Ryo Utsunomiya
- Department of Molecular and Cellular Physiology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Hajime Yano
- Department of Molecular and Cellular Physiology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Madoka Kubo
- Department of Clinical Pharmacology and Therapeutics, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Rina Ando
- Department of Clinical Pharmacology and Therapeutics, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Jun Iwanami
- Department of Cell Biology and Molecular Medicine, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Masakatsu Yamashita
- Department of Immunology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Masahiro Nagai
- Department of Clinical Pharmacology and Therapeutics, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Junya Tanaka
- Department of Molecular and Cellular Physiology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| |
Collapse
|
25
|
Tau aggregation and its relation to selected forms of neuronal cell death. Essays Biochem 2021; 65:847-857. [PMID: 34897457 PMCID: PMC8709892 DOI: 10.1042/ebc20210030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/17/2021] [Accepted: 11/17/2021] [Indexed: 12/12/2022]
Abstract
How neurons die in neurodegenerative diseases is still unknown. The distinction between apoptosis as a genetically controlled mechanism, and necrosis, which was viewed as an unregulated process, has blurred with the ever-increasing number of necrotic-like death subroutines underpinned by genetically defined pathways. It is therefore pertinent to ask whether any of them apply to neuronal cell death in tauopathies. Although Alzheimer's disease (AD) is the most prevalent tauopathy, tauopathies comprise an array of over 30 diseases in which the cytoplasmic protein tau aggregates in neurons, and also, in some diseases, in glia. Animal models have sought to distil the contribution of tau aggregation to the cell death process but despite intensive research, no one mechanism of cell death has been unequivocally defined. The process of tau aggregation, and the fibrillar structures that form, touch on so many cellular functions that there is unlikely to be a simple linear pathway of death; as one is blocked another is likely to take the lead. It is timely to ask how far we have advanced into defining whether any of the molecular players in the new death subroutines participate in the death process. Here we briefly review the currently known cell death routines and explore what is known about their participation in tau aggregation-related cell death. We highlight the involvement of cell autonomous and the more recent non-cell autonomous pathways that may enhance tau-aggregate toxicity, and discuss recent findings that implicate microglial phagocytosis of live neurons with tau aggregates as a mechanism of death.
Collapse
|
26
|
Busquets O, Parcerisas A, Verdaguer E, Ettcheto M, Camins A, Beas-Zarate C, Castro-Torres RD, Auladell C. c-Jun N-Terminal Kinases in Alzheimer's Disease: A Possible Target for the Modulation of the Earliest Alterations. J Alzheimers Dis 2021; 82:S127-S139. [PMID: 33216036 DOI: 10.3233/jad-201053] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Given the highly multifactorial origin of Alzheimer's disease (AD) neuropathology, disentangling and orderly knowing mechanisms involved in sporadic onset are arduous. Nevertheless, when the elements involved are dissected into smaller pieces, the task becomes more accessible. This review aimed to describe the link between c-Jun N-terminal Kinases (JNKs), master regulators of many cellular functions, and the early alterations of AD: synaptic loss and dysregulation of neuronal transport. Both processes have a role in the posterior cognitive decline observed in AD. The manuscript focuses on the molecular mechanisms of glutamatergic, GABA, and cholinergic synapses altered by the presence of amyloid-β aggregates and hyperphosphorylated tau, as well as on several consequences of the disruption of cellular processes linked to neuronal transport that is controlled by the JNK-JIP (c-jun NH2-terminal kinase (JNK)-interacting proteins (JIPs) complex, including the transport of AβPP or autophagosomes.
Collapse
Affiliation(s)
- Oriol Busquets
- Department of Pharmacology, Toxicology and Therapeutic Chemistry; Pharmacy and Food Sciences Faculty, Universitat de Barcelona, Barcelona, Spain.,Department of Biochemistry and Biotechnology, Medicine and Health Sciences Faculty, Universitat Rovira i Virgili, Reus, Spain.,Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Dominick P. Purpura Department of Neurosciences, Albert Einstein College of Medicine, New York City, NY, USA
| | - Antoni Parcerisas
- Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Biology Faculty, Universitat de Barcelona, Barcelona, Spain
| | - Ester Verdaguer
- Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Biology Faculty, Universitat de Barcelona, Barcelona, Spain
| | - Miren Ettcheto
- Department of Pharmacology, Toxicology and Therapeutic Chemistry; Pharmacy and Food Sciences Faculty, Universitat de Barcelona, Barcelona, Spain.,Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Antoni Camins
- Department of Pharmacology, Toxicology and Therapeutic Chemistry; Pharmacy and Food Sciences Faculty, Universitat de Barcelona, Barcelona, Spain.,Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Carlos Beas-Zarate
- Department of Cell and Molecular Biology, Laboratory of Neural Regeneration, C.U.C.B.A., Universidad de Guadalajara, Jalisco, Mexico
| | - Rubén Darío Castro-Torres
- Department of Cell and Molecular Biology, Laboratory of Biology of Neurotransmission, C.U.C.B.A., Universidad de Guadalajara, Jalisco, Mexico
| | - Carme Auladell
- Centre for Biomedical Research of Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Department of Cell Biology, Physiology and Immunology, Biology Faculty, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
27
|
Park J, Choi Y, Jung E, Lee S, Sohn J, Chung W. Microglial MERTK eliminates phosphatidylserine-displaying inhibitory post-synapses. EMBO J 2021; 40:e107121. [PMID: 34013588 PMCID: PMC8327958 DOI: 10.15252/embj.2020107121] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 04/09/2021] [Accepted: 04/16/2021] [Indexed: 01/11/2023] Open
Abstract
Glia contribute to synapse elimination through phagocytosis in the central nervous system. Despite the important roles of this process in development and neurological disorders, the identity and regulation of the "eat-me" signal that initiates glia-mediated phagocytosis of synapses has remained incompletely understood. Here, we generated conditional knockout mice with neuronal-specific deletion of the flippase chaperone Cdc50a, to induce stable exposure of phosphatidylserine, a well-known "eat-me" signal for apoptotic cells, on the neuronal outer membrane. Surprisingly, acute Cdc50a deletion in mature neurons causes preferential phosphatidylserine exposure in neuronal somas and specific loss of inhibitory post-synapses without effects on other synapses, resulting in abnormal excitability and seizures. Ablation of microglia or the deletion of microglial phagocytic receptor Mertk prevents the loss of inhibitory post-synapses and the seizure phenotype, indicating that microglial phagocytosis is responsible for inhibitory post-synapse elimination. Moreover, we found that phosphatidylserine is used for microglia-mediated pruning of inhibitory post-synapses in normal brains, suggesting that phosphatidylserine serves as a general "eat-me" signal for inhibitory post-synapse elimination.
Collapse
Affiliation(s)
- Jungjoo Park
- Department of Biological SciencesKorea Advanced Institute of Science and TechnologyDaejeonKorea
| | - Yeeun Choi
- Department of Biological SciencesKorea Advanced Institute of Science and TechnologyDaejeonKorea
| | - Eunji Jung
- Department of Biological SciencesKorea Advanced Institute of Science and TechnologyDaejeonKorea
| | - Seung‐Hee Lee
- Department of Biological SciencesKorea Advanced Institute of Science and TechnologyDaejeonKorea
| | - Jong‐Woo Sohn
- Department of Biological SciencesKorea Advanced Institute of Science and TechnologyDaejeonKorea
| | - Won‐Suk Chung
- Department of Biological SciencesKorea Advanced Institute of Science and TechnologyDaejeonKorea
| |
Collapse
|
28
|
Milinkeviciute G, Chokr SM, Castro EM, Cramer KS. CX3CR1 mutation alters synaptic and astrocytic protein expression, topographic gradients, and response latencies in the auditory brainstem. J Comp Neurol 2021; 529:3076-3097. [PMID: 33797066 DOI: 10.1002/cne.25150] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 03/04/2021] [Accepted: 03/27/2021] [Indexed: 01/14/2023]
Abstract
The precise and specialized circuitry in the auditory brainstem develops through adaptations of cellular and molecular signaling. We previously showed that elimination of microglia during development impairs synaptic pruning that leads to maturation of the calyx of Held, a large encapsulating synapse that terminates on neurons of the medial nucleus of the trapezoid body (MNTB). Microglia depletion also led to a decrease in glial fibrillary acidic protein (GFAP), a marker for mature astrocytes. Here, we investigated the role of signaling through the fractalkine receptor (CX3CR1), which is expressed by microglia and mediates communication with neurons. CX3CR1-/- and wild-type mice were studied before and after hearing onset and at 9 weeks of age. Levels of GFAP were significantly increased in the MNTB in mutants at 9 weeks. Pruning was unaffected at the calyx of Held, but we found an increase in expression of glycinergic synaptic marker in mutant mice at P14, suggesting an effect on maturation of inhibitory inputs. We observed disrupted tonotopic gradients of neuron and calyx size in MNTB in mutant mice. Auditory brainstem recording (ABR) revealed that CX3CR1-/- mice had normal thresholds and amplitudes but decreased latencies and interpeak latencies, particularly for the highest frequencies. These results demonstrate that disruption of fractalkine signaling has a significant effect on auditory brainstem development. Our findings highlight the importance of neuron-microglia-astrocyte communication in pruning of inhibitory synapses and establishment of tonotopic gradients early in postnatal development.
Collapse
Affiliation(s)
- Giedre Milinkeviciute
- Department of Neurobiology and Behavior, University of California, Irvine, California, USA
| | - Sima M Chokr
- Department of Neurobiology and Behavior, University of California, Irvine, California, USA
| | - Emily M Castro
- Department of Neurobiology and Behavior, University of California, Irvine, California, USA
| | - Karina S Cramer
- Department of Neurobiology and Behavior, University of California, Irvine, California, USA
| |
Collapse
|
29
|
Kono R, Ikegaya Y, Koyama R. Phagocytic Glial Cells in Brain Homeostasis. Cells 2021; 10:1348. [PMID: 34072424 PMCID: PMC8229427 DOI: 10.3390/cells10061348] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/22/2021] [Accepted: 05/25/2021] [Indexed: 11/16/2022] Open
Abstract
Phagocytosis by glial cells has been shown to play an important role in maintaining brain homeostasis. Microglia are currently considered to be the major phagocytes in the brain parenchyma, and these cells phagocytose a variety of materials, including dead cell debris, abnormally aggregated proteins, and, interestingly, the functional synapses of living neurons. The intracellular signaling mechanisms that regulate microglial phagocytosis have been studied extensively, and several important factors, including molecules known as "find me" signals and "eat me" signals and receptors on microglia that are involved in phagocytosis, have been identified. In addition, recent studies have revealed that astrocytes, which are another major glial cell in the brain parenchyma, also have phagocytic abilities. In this review, we will discuss the roles of microglia and astrocytes in phagocytosis-mediated brain homeostasis, focusing on the characteristics and differences of their phagocytic abilities.
Collapse
Affiliation(s)
- Rena Kono
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan; (R.K.); (Y.I.)
| | - Yuji Ikegaya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan; (R.K.); (Y.I.)
- Institute for AI and Beyond, The University of Tokyo, Tokyo 113-0033, Japan
- Center for Information and Neural Networks, National Institute of Information and Communications Technology, Suita City 565-0871, Japan
| | - Ryuta Koyama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan; (R.K.); (Y.I.)
| |
Collapse
|
30
|
Auditory Brainstem Deficits from Early Treatment with a CSF1R Inhibitor Largely Recover with Microglial Repopulation. eNeuro 2021; 8:ENEURO.0318-20.2021. [PMID: 33558268 PMCID: PMC8009669 DOI: 10.1523/eneuro.0318-20.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 12/10/2020] [Accepted: 01/13/2021] [Indexed: 12/20/2022] Open
Abstract
Signaling between neurons and glia is necessary for the formation of functional neural circuits. A role for microglia in the maturation of connections in the medial nucleus of the trapezoid body (MNTB) was previously demonstrated by postnatal microglial elimination using a colony stimulating factor 1 receptor (CSF1R). Defective pruning of calyces of Held and significant reduction of the mature astrocyte marker glial fibrillary acidic protein (GFAP) were observed after hearing onset. Here, we investigated the time course required for microglia to populate the mouse MNTB after cessation of CSF1R inhibitor treatment. We then examined whether defects seen after microglial depletion were rectified by microglial repopulation. We found that microglia returned to control levels at four weeks of age (18 d postcessation of treatment). Calyceal innervation of MNTB neurons was comparable to control levels at four weeks and GFAP expression recovered by seven weeks. We further investigated the effects of microglia elimination and repopulation on auditory function using auditory brainstem recordings (ABRs). Temporary microglial depletion significantly elevated auditory thresholds in response to 4. 8, and 12 kHz at four weeks. Treatment significantly affected latencies, interpeak latencies, and amplitudes of all the ABR peaks in response to many of the frequencies tested. These effects largely recovered by seven weeks. These findings highlight the functions of microglia in the formation of auditory neural circuits early in development. Further, the results suggest that microglia retain their developmental functions beyond the period of circuit refinement.
Collapse
|
31
|
Morini R, Bizzotto M, Perrucci F, Filipello F, Matteoli M. Strategies and Tools for Studying Microglial-Mediated Synapse Elimination and Refinement. Front Immunol 2021; 12:640937. [PMID: 33708226 PMCID: PMC7940197 DOI: 10.3389/fimmu.2021.640937] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 02/01/2021] [Indexed: 01/14/2023] Open
Abstract
The role of microglia in controlling synapse homeostasis is becoming increasingly recognized by the scientific community. In particular, the microglia-mediated elimination of supernumerary synapses during development lays the basis for the correct formation of neuronal circuits in adulthood, while the possible reactivation of this process in pathological conditions, such as schizophrenia or Alzheimer's Disease, provides a promising target for future therapeutic strategies. The methodological approaches to investigate microglial synaptic engulfment include different in vitro and in vivo settings. Basic in vitro assays, employing isolated microglia and microbeads, apoptotic membranes, liposomes or synaptosomes allow the quantification of the microglia phagocytic abilities, while co-cultures of microglia and neurons, deriving from either WT or genetically modified mice models, provide a relatively manageable setting to investigate the involvement of specific molecular pathways. Further detailed analysis in mice brain is then mandatory to validate the in vitro assays as representative for the in vivo situation. The present review aims to dissect the main technical approaches to investigate microglia-mediated phagocytosis of neuronal and synaptic substrates in critical developmental time windows.
Collapse
Affiliation(s)
- Raffaella Morini
- Laboratory of Pharmacology and Brain Pathology, Neurocenter, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy
| | - Matteo Bizzotto
- Laboratory of Pharmacology and Brain Pathology, Neurocenter, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Fabio Perrucci
- Laboratory of Pharmacology and Brain Pathology, Neurocenter, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Fabia Filipello
- Laboratory of Pharmacology and Brain Pathology, Neurocenter, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy.,Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States
| | - Michela Matteoli
- Laboratory of Pharmacology and Brain Pathology, Neurocenter, Humanitas Clinical and Research Center - IRCCS, Rozzano, Italy.,Consiglio Nazionale Delle Ricerche (CNR), Institute of Neuroscience - URT Humanitas, Rozzano, Italy
| |
Collapse
|
32
|
Rombaut B, Kessels S, Schepers M, Tiane A, Paes D, Solomina Y, Piccart E, Hove DVD, Brône B, Prickaerts J, Vanmierlo T. PDE inhibition in distinct cell types to reclaim the balance of synaptic plasticity. Theranostics 2021; 11:2080-2097. [PMID: 33500712 PMCID: PMC7797685 DOI: 10.7150/thno.50701] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023] Open
Abstract
Synapses are the functional units of the brain. They form specific contact points that drive neuronal communication and are highly plastic in their strength, density, and shape. A carefully orchestrated balance between synaptogenesis and synaptic pruning, i.e., the elimination of weak or redundant synapses, ensures adequate synaptic density. An imbalance between these two processes lies at the basis of multiple neuropathologies. Recent evidence has highlighted the importance of glia-neuron interactions in the synaptic unit, emphasized by glial phagocytosis of synapses and local excretion of inflammatory mediators. These findings warrant a closer look into the molecular basis of cell-signaling pathways in the different brain cells that are related to synaptic plasticity. In neurons, intracellular second messengers, such as cyclic guanosine or adenosine monophosphate (cGMP and cAMP, respectively), are known mediators of synaptic homeostasis and plasticity. Increased levels of these second messengers in glial cells slow down inflammation and neurodegenerative processes. These multi-faceted effects provide the opportunity to counteract excessive synapse loss by targeting cGMP and cAMP pathways in multiple cell types. Phosphodiesterases (PDEs) are specialized degraders of these second messengers, rendering them attractive targets to combat the detrimental effects of neurological disorders. Cellular and subcellular compartmentalization of the specific isoforms of PDEs leads to divergent downstream effects for these enzymes in the various central nervous system resident cell types. This review provides a detailed overview on the role of PDEs and their inhibition in the context of glia-neuron interactions in different neuropathologies characterized by synapse loss. In doing so, it provides a framework to support future research towards finding combinational therapy for specific neuropathologies.
Collapse
|
33
|
Zhang J, Li L, Gong J, Li H, Zhou M, Tan Y. The gastroprotective effect of alpinia officinarum extract on indomethacin-induced topical injuries in RGM-1 Cells: Involvement of H +/K +-ATPase- and mitochondrial-mediated apoptosis. Pharmacogn Mag 2021. [DOI: 10.4103/pm.pm_65_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
34
|
Cserép C, Pósfai B, Dénes Á. Shaping Neuronal Fate: Functional Heterogeneity of Direct Microglia-Neuron Interactions. Neuron 2020; 109:222-240. [PMID: 33271068 DOI: 10.1016/j.neuron.2020.11.007] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/13/2020] [Accepted: 11/06/2020] [Indexed: 12/11/2022]
Abstract
The functional contribution of microglia to normal brain development, healthy brain function, and neurological disorders is increasingly recognized. However, until recently, the nature of intercellular interactions mediating these effects remained largely unclear. Recent findings show microglia establishing direct contact with different compartments of neurons. Although communication between microglia and neurons involves intermediate cells and soluble factors, direct membrane contacts enable a more precisely regulated, dynamic, and highly effective form of interaction for fine-tuning neuronal responses and fate. Here, we summarize the known ultrastructural, molecular, and functional features of direct microglia-neuron interactions and their roles in brain disease.
Collapse
Affiliation(s)
- Csaba Cserép
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Szigony u. 43, 1083 Budapest, Hungary
| | - Balázs Pósfai
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Szigony u. 43, 1083 Budapest, Hungary; Szentágothai János Doctoral School of Neurosciences, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary
| | - Ádám Dénes
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Szigony u. 43, 1083 Budapest, Hungary.
| |
Collapse
|
35
|
Danger-Sensing/Patten Recognition Receptors and Neuroinflammation in Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21239036. [PMID: 33261147 PMCID: PMC7731137 DOI: 10.3390/ijms21239036] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/24/2020] [Accepted: 11/25/2020] [Indexed: 02/06/2023] Open
Abstract
Fibrillar aggregates and soluble oligomers of both Amyloid-β peptides (Aβs) and hyperphosphorylated Tau proteins (p-Tau-es), as well as a chronic neuroinflammation are the main drivers causing progressive neuronal losses and dementia in Alzheimer’s disease (AD). However, the underlying pathogenetic mechanisms are still much disputed. Several endogenous neurotoxic ligands, including Aβs, and/or p-Tau-es activate innate immunity-related danger-sensing/pattern recognition receptors (PPRs) thereby advancing AD’s neuroinflammation and progression. The major PRR families involved include scavenger, Toll-like, NOD-like, AIM2-like, RIG-like, and CLEC-2 receptors, plus the calcium-sensing receptor (CaSR). This quite intricate picture stresses the need to identify the pathogenetically topmost Aβ-activated PRR, whose signaling would trigger AD’s three main drivers and their intra-brain spread. In theory, the candidate might belong to any PRR family. However, results of preclinical studies using in vitro nontumorigenic human cortical neurons and astrocytes and in vivo AD-model animals have started converging on the CaSR as the pathogenetically upmost PRR candidate. In fact, the CaSR binds both Ca2+ and Aβs and promotes the spread of both Ca2+ dyshomeostasis and AD’s three main drivers, causing a progressive neurons’ death. Since CaSR’s negative allosteric modulators block all these effects, CaSR’s candidacy for topmost pathogenetic PRR has assumed a growing therapeutic potential worth clinical testing.
Collapse
|
36
|
Nakanishi H, Ni J, Nonaka S, Hayashi Y. Microglial circadian clock regulation of microglial structural complexity, dendritic spine density and inflammatory response. Neurochem Int 2020; 142:104905. [PMID: 33217515 DOI: 10.1016/j.neuint.2020.104905] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 02/08/2023]
Abstract
Cortical microglia exhibit a ramified shape during sleep, while they have a hyper-ramified shape during wakefulness, which is characterized by their longer processes with increased branching points. The microglial molecular circadian clock regulates expressions of both cathepsin S (CatS) and P2Y12 receptors in the brain with a peak at zeitgeber time 14 (2 h after beginning of the dark phase). We postulated that these two microglia-specific molecules contribute to diurnal alterations of microglial shapes and neuronal activities in the cerebral cortex. During wakefulness, CatS secreted from cortical microglia may be involved in P2Y12 receptor-dependent process extension. Secreted CatS subsequently degrades the perineuronal nets, initiating the downscaling of both spine density and synaptic strength of cortical neurons toward the beginning of sleep. The downscaling of both spine density and synaptic strength of cortical neurons during sleep could improve signal-to-noise, which would benefit memory consolidation, or allow for new learning to occur during subsequent waking. Furthermore, disruption of CatS induces the sleep disturbance and impaired social interaction in mice. Moreover, the microglial clock system disruption may also play a role in the early pathogenesis of Alzheimer's disease. The reduced expression of BMAL1 in cortical microglia caused by oligomeric amyloid β may induce the increased presence of inflammatory phenotype through a reduction in RORα, which in turn reduced IκBα and enhanced NF-κB activation. These observations suggest that the microglial clock system disruption contribute to pathogeneses of sleep disturbance, impaired social interaction and cognitive impairment. Therefore, the growing understanding of the microglial circadian molecular clock might aid in the development of novel pharmacological interventions against both neuropsychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- Hiroshi Nakanishi
- Department of Pharmacology, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, 731-0153, Japan.
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Saori Nonaka
- Department of Pharmacology, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, 731-0153, Japan
| | - Yoshinori Hayashi
- Department of Physiology, Nihon University School of Dentistry, 1-8-13 Kandasurugadai, Chiyoda-ku, Tokyo, 101-8310, Japan
| |
Collapse
|
37
|
Caspase inhibition rescues F1Fo ATP synthase dysfunction-mediated dendritic spine elimination. Sci Rep 2020; 10:17589. [PMID: 33067541 PMCID: PMC7568535 DOI: 10.1038/s41598-020-74613-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 09/23/2020] [Indexed: 12/26/2022] Open
Abstract
Dendritic spine injury underlies synaptic failure in many neurological disorders. Mounting evidence suggests a mitochondrial pathway of local nonapoptotic caspase signaling in mediating spine pruning. However, it remains unclear whether this caspase signaling plays a key role in spine loss when severe mitochondrial functional defects are present. The answer to this question is critical especially for some pathological states, in which mitochondrial deficits are prominent and difficult to fix. F1Fo ATP synthase is a pivotal mitochondrial enzyme and the dysfunction of this enzyme involves in diseases with spinopathy. Here, we inhibited F1Fo ATP synthase function in primary cultured hippocampal neurons by using non-lethal oligomycin A treatment. Oligomycin A induced mitochondrial defects including collapsed mitochondrial membrane potential, dissipated ATP production, and elevated reactive oxygen species (ROS) production. In addition, dendritic mitochondria underwent increased fragmentation and reduced positioning to dendritic spines along with increased caspase 3 cleavage in dendritic shaft and spines in response to oligomycin A. Concurring with these dendritic mitochondrial changes, oligomycin A-insulted neurons displayed spine loss and altered spine architecture. Such oligomycin A-mediated changes in dendritic spines were substantially prevented by the inhibition of caspase activation by using a pan-caspase inhibitor, quinolyl-valyl-O-methylaspartyl-[-2,6-difluorophenoxy]-methyl ketone (Q-VD-OPh). Of note, the administration of Q-VD-OPh showed no protective effect on oligomycin A-induced mitochondrial dysfunction. Our findings suggest a pivotal role of caspase 3 signaling in mediating spine injury and the modulation of caspase 3 activation may benefit neurons from spine loss in diseases, at least, in those with F1Fo ATP synthase defects.
Collapse
|
38
|
Dalakas MC, Alexopoulos H, Spaeth PJ. Complement in neurological disorders and emerging complement-targeted therapeutics. Nat Rev Neurol 2020; 16:601-617. [PMID: 33005040 PMCID: PMC7528717 DOI: 10.1038/s41582-020-0400-0] [Citation(s) in RCA: 184] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/07/2020] [Indexed: 12/30/2022]
Abstract
The complement system consists of a network of plasma and membrane proteins that modulate tissue homeostasis and contribute to immune surveillance by interacting with the innate and adaptive immune systems. Dysregulation, impairment or inadvertent activation of complement components contribute to the pathogenesis of some autoimmune neurological disorders and could even contribute to neurodegenerative diseases. In this Review, we summarize current knowledge about the main functions of the complement pathways and the involvement of complement in neurological disorders. We describe the complex network of complement proteins that target muscle, the neuromuscular junction, peripheral nerves, the spinal cord or the brain and discuss the autoimmune mechanisms of complement-mediated myopathies, myasthenia, peripheral neuropathies, neuromyelitis and other CNS disorders. We also consider the emerging role of complement in some neurodegenerative diseases, such as Alzheimer disease, amyotrophic lateral sclerosis and even schizophrenia. Finally, we provide an overview of the latest complement-targeted immunotherapies including monoclonal antibodies, fusion proteins and peptidomimetics that have been approved, that are undergoing phase I–III clinical trials or that show promise for the treatment of neurological conditions that respond poorly to existing immunotherapies. In this Review, Dalakas et al. discuss the complement system, the role it plays in autoimmune neurological disease and neurodegenerative disease, and provide an overview of the latest therapeutics that target complement and that can be used for or have potential in neurological disorders. Complement has an important physiological role in host immune defences and tissue remodelling. The physiological role of complement extends to the regulation of synaptic development. Complement has a key pathophysiological role in autoimmune neurological diseases and mediates the actions of pathogenic autoantibodies, such as acetylcholine receptor antibodies and aquaporin 4 antibodies. For some autoimmune neurological diseases, such as myasthenia gravis and neuromyelitis optica spectrum disorders, approved complement-targeted treatments are now available. Complement also seems to be of pathogenic relevance in neurodegenerative diseases such as Alzheimer disease, in which innate immune-driven inflammation is receiving increasing attention. The field of complement-targeted therapeutics is rapidly expanding, with several FDA-approved agents and others currently in phase II and phase III clinical trials.
Collapse
Affiliation(s)
- Marinos C Dalakas
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, USA. .,Neuroimmunology Unit, Department of Pathophysiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| | - Harry Alexopoulos
- Neuroimmunology Unit, Department of Pathophysiology, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Peter J Spaeth
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| |
Collapse
|
39
|
Leal AF, Benincore-Flórez E, Solano-Galarza D, Garzón Jaramillo RG, Echeverri-Peña OY, Suarez DA, Alméciga-Díaz CJ, Espejo-Mojica AJ. GM2 Gangliosidoses: Clinical Features, Pathophysiological Aspects, and Current Therapies. Int J Mol Sci 2020; 21:ijms21176213. [PMID: 32867370 PMCID: PMC7503724 DOI: 10.3390/ijms21176213] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/03/2020] [Accepted: 08/07/2020] [Indexed: 12/16/2022] Open
Abstract
GM2 gangliosidoses are a group of pathologies characterized by GM2 ganglioside accumulation into the lysosome due to mutations on the genes encoding for the β-hexosaminidases subunits or the GM2 activator protein. Three GM2 gangliosidoses have been described: Tay-Sachs disease, Sandhoff disease, and the AB variant. Central nervous system dysfunction is the main characteristic of GM2 gangliosidoses patients that include neurodevelopment alterations, neuroinflammation, and neuronal apoptosis. Currently, there is not approved therapy for GM2 gangliosidoses, but different therapeutic strategies have been studied including hematopoietic stem cell transplantation, enzyme replacement therapy, substrate reduction therapy, pharmacological chaperones, and gene therapy. The blood-brain barrier represents a challenge for the development of therapeutic agents for these disorders. In this sense, alternative routes of administration (e.g., intrathecal or intracerebroventricular) have been evaluated, as well as the design of fusion peptides that allow the protein transport from the brain capillaries to the central nervous system. In this review, we outline the current knowledge about clinical and physiopathological findings of GM2 gangliosidoses, as well as the ongoing proposals to overcome some limitations of the traditional alternatives by using novel strategies such as molecular Trojan horses or advanced tools of genome editing.
Collapse
Affiliation(s)
- Andrés Felipe Leal
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
| | - Eliana Benincore-Flórez
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
| | - Daniela Solano-Galarza
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
| | - Rafael Guillermo Garzón Jaramillo
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
| | - Olga Yaneth Echeverri-Peña
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
| | - Diego A. Suarez
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
- Faculty of Medicine, Universidad Nacional de Colombia, Bogotá 110231, Colombia
| | - Carlos Javier Alméciga-Díaz
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
- Correspondence: (C.J.A.-D.); (A.J.E.-M.); Tel.: +57-1-3208320 (ext. 4140) (C.J.A.-D.); +57-1-3208320 (ext. 4099) (A.J.E.-M.)
| | - Angela Johana Espejo-Mojica
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (A.F.L.); (E.B.-F); (D.S.-G.); (R.G.G.J.); (O.Y.E.-P.); (D.A.S.)
- Correspondence: (C.J.A.-D.); (A.J.E.-M.); Tel.: +57-1-3208320 (ext. 4140) (C.J.A.-D.); +57-1-3208320 (ext. 4099) (A.J.E.-M.)
| |
Collapse
|
40
|
IL-4 and IL-10 promotes phagocytic activity of microglia by up-regulation of TREM2. Cytotechnology 2020; 72:589-602. [PMID: 32623621 DOI: 10.1007/s10616-020-00409-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 06/26/2020] [Indexed: 02/06/2023] Open
Abstract
Triggering receptor expressed on myeloid cells-2 (TREM2) is an innate immune receptor that promotes phagocytosis by microglia. However, whether TREM2 is related to the stimulus-dependent phagocytic activity of microglia is unclear. In this study, the primary cultured microglia were stimulated with interferon (IFN)-γ, interleukin (IL)-4, and interleukin (IL)-10, respectively, and their phagocytic activity against microbeads and apoptotic neural stem cells (NSCs) was measured. TREM2 of microglia was detected by qPCR and western blotting. The TREM2 signal was blocked in microglia using the siRNA technique. The results showed that IL-4 or IL-10 treatment significantly increased the number of microglia gathered around the apoptotic neurosphere. IL-4 and IL-10 treatment also promoted phagocytosis of microbeads and apoptotic NSCs by primary cultured microglia. The TREM2 expression was up-regulated in IL-4- or IL-10- treated microglia. TREM2 siRNA treatment blocked the phagocytic activity of IL-4- or IL-10-treated microglia. In conclusion, these results indicated that IL-4 and IL-10 promote the phagocytic activity of microglia by the up-regulation of TREM2, which suggested a new potential therapeutic target for neurodegenerative disease.
Collapse
|
41
|
Nakanishi H. Cathepsin regulation on microglial function. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1868:140465. [PMID: 32526473 DOI: 10.1016/j.bbapap.2020.140465] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 05/25/2020] [Accepted: 05/28/2020] [Indexed: 12/15/2022]
Abstract
Microglia, the resident mononuclear phagocyte population in the brain, have long been implicated in the pathology of neurodegenerative age-associated disorders. However, activated microglia have now been identified as homeostatic keepers in the brain, because they are involved in the initiation and resolution of neuropathology. The complex roles of activated microglia appear to be linked to change from inflammatory and neurotoxic to anti-inflammatory and neuroprotective phenotypes. Increased expression and secretion of various cathepsins support roles of activated microglia in chronic neuroinflammation, the neurotoxic M1-like polarization and neuronal death. Moreover, changes in expression and localization of microglial cathepsin B play a critical role in the acceleration of the brain aging. Beyond the role as brain-resident macrophages, many lines of evidence have shown that microglia have essential roles in the maturation and maintenance of neuronal circuits in the developing and adult brain. Cathepsin S secreted from microglia induces the diurnal variation of spine density of cortical neurons though proteolytic modification of peri-synaptic extracellular matrix molecules. In this review, I highlight the emerging roles of cathepsins that support the roles of microglia in both normal healthy and pathological brains. In addition, I discuss cathepsin inhibitors as potential therapeutic targets for brain disorders.
Collapse
Affiliation(s)
- Hiroshi Nakanishi
- Department of Pharmacology, Faculty of Pharmacy, Yasuda Women's University, Hiroshima 731-0153, Japan.
| |
Collapse
|
42
|
Microglia: Multiple states and multiple roles in the normal and diseased brain and spinal cord. Neurosci Lett 2020; 729:135019. [DOI: 10.1016/j.neulet.2020.135019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
43
|
Marsters CM, Nesan D, Far R, Klenin N, Pittman QJ, Kurrasch DM. Embryonic microglia influence developing hypothalamic glial populations. J Neuroinflammation 2020; 17:146. [PMID: 32375817 PMCID: PMC7201702 DOI: 10.1186/s12974-020-01811-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/13/2020] [Indexed: 11/15/2022] Open
Abstract
Background Although historically microglia were thought to be immature in the fetal brain, evidence of purposeful interactions between these immune cells and nearby neural progenitors is becoming established. Here, we examined the influence of embryonic microglia on gliogenesis within the developing tuberal hypothalamus, a region later important for energy balance, reproduction, and thermoregulation. Methods We used immunohistochemistry to quantify the location and numbers of glial cells in the embryonic brain (E13.5–E17.5), as well as a pharmacological approach (i.e., PLX5622) to knock down fetal microglia. We also conducted cytokine and chemokine analyses on embryonic brains in the presence or absence of microglia, and a neurosphere assay to test the effects of the altered cytokines on hypothalamic progenitor behaviors. Results We identified a subpopulation of activated microglia that congregated adjacent to the third ventricle alongside embryonic Olig2+ neural progenitor cells (NPCs) that are destined to give rise to oligodendrocyte and astrocyte populations. In the absence of microglia, we observed an increase in Olig2+ glial progenitor cells that remained at the ventricle by E17.5 and a concomitant decrease of these Olig2+ cells in the mantle zone, indicative of a delay in migration of these precursor cells. A further examination of maturing oligodendrocytes in the hypothalamic grey and white matter area in the absence of microglia revealed migrating oligodendrocyte progenitor cells (OPCs) within the grey matter at E17.5, a time point when OPCs begin to slow their migration. Finally, quantification of cytokine and chemokine signaling in ex vivo E15.5 hypothalamic cultures +/− microglia revealed decreases in the protein levels of several cytokines in the absence of microglia. We assayed the influence of two downregulated cytokines (CCL2 and CXCL10) on neurosphere-forming capacity and lineage commitment of hypothalamic NPCs in culture and showed an increase in NPC proliferation as well as neuronal and oligodendrocyte differentiation. Conclusion These data demonstrate that microglia influence gliogenesis in the developing tuberal hypothalamus.
Collapse
Affiliation(s)
- Candace M Marsters
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Dinushan Nesan
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Rena Far
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Natalia Klenin
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Quentin J Pittman
- Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Deborah M Kurrasch
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada. .,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada. .,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
44
|
Yuan C, Aierken A, Xie Z, Li N, Zhao J, Qing H. The age-related microglial transformation in Alzheimer's disease pathogenesis. Neurobiol Aging 2020; 92:82-91. [PMID: 32408056 DOI: 10.1016/j.neurobiolaging.2020.03.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/11/2020] [Accepted: 03/31/2020] [Indexed: 12/15/2022]
Abstract
Neuroinflammatory responses mediated by microglia, the resident immune cells of the central nervous system, have long been a subject of study in the field of Alzheimer's disease (AD). Microglia express a wide range of receptors that act as molecular sensors, through which they can fulfill their various functions. In this review, we first analyzed the changes in the expression levels of microglial membrane receptors SR-A, TREM2, CD36, CD33, and CR3 in aging and AD and described the different roles of these receptors in amyloid-beta clearance and inflammatory responses. Two classical hallmarks of AD are extracellular amyloid-beta deposits and intracellular aggregated phosphorylated tau. In AD, microglia reaction was initially thought to be triggered by amyloid deposits. New evidence showed it also associated with increased phosphorylation of tau. However, which first appeared and induced activated microglia is not clear. Then we summarized diverse opinions on it. Besides, as AD is tightly linked to aging, and microglia changes dramatically on aging, yet the relative impacts of both aging and microglia are less frequently considered, so at last, we discussed the roles of aging microglia in AD. We hope to provide a reference for subsequent research.
Collapse
Affiliation(s)
- Chunxu Yuan
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biological Sciences, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Ailikemu Aierken
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biological Sciences, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Zhen Xie
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biological Sciences, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Nuomin Li
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biological Sciences, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Juan Zhao
- School of Materials Science and Engineering, Department of Materials Processing Engineering, Beijing Institute of Technology, Beijing, China.
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biological Sciences, School of Life Science, Beijing Institute of Technology, Beijing, China.
| |
Collapse
|
45
|
Datta D, Leslie SN, Morozov YM, Duque A, Rakic P, van Dyck CH, Nairn AC, Arnsten AFT. Classical complement cascade initiating C1q protein within neurons in the aged rhesus macaque dorsolateral prefrontal cortex. J Neuroinflammation 2020; 17:8. [PMID: 31906973 PMCID: PMC6945481 DOI: 10.1186/s12974-019-1683-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 12/17/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Cognitive impairment in schizophrenia, aging, and Alzheimer's disease is associated with spine and synapse loss from the dorsolateral prefrontal cortex (dlPFC) layer III. Complement cascade signaling is critical in driving spine loss and disease pathogenesis. Complement signaling is initiated by C1q, which tags synapses for elimination. C1q is thought to be expressed predominately by microglia, but its expression in primate dlPFC has never been examined. The current study assayed C1q levels in aging primate dlPFC and rat medial PFC (mPFC) and used immunoelectron microscopy (immunoEM), immunoblotting, and co-immunoprecipitation (co-IP) to reveal the precise anatomical distribution and interactions of C1q. METHODS Age-related changes in C1q levels in rhesus macaque dlPFC and rat mPFC were examined using immunoblotting. High-spatial resolution immunoEM was used to interrogate the subcellular localization of C1q in aged macaque layer III dlPFC and aged rat layer III mPFC. co-IP techniques quantified protein-protein interactions for C1q and proteins associated with excitatory and inhibitory synapses in macaque dlPFC. RESULTS C1q levels were markedly increased in the aged macaque dlPFC. Ultrastructural localization found the expected C1q localization in glia, including those ensheathing synapses, but also revealed extensive localization within neurons. C1q was found near synapses, within terminals and in spines, but was also observed in dendrites, often near abnormal mitochondria. Similar analyses in aging rat mPFC corroborated the findings in rhesus macaques. C1q protein increasingly associated with PSD95 with age in macaque, consistent with its synaptic localization as evidenced by EM. CONCLUSIONS These findings reveal novel, intra-neuronal distribution patterns for C1q in the aging primate cortex, including evidence of C1q in dendrites. They suggest that age-related changes in the dlPFC may increase C1q expression and synaptic tagging for glial phagocytosis, a possible mechanism for age-related degeneration.
Collapse
Affiliation(s)
- Dibyadeep Datta
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06511, USA.
| | - Shannon N Leslie
- Department of Psychiatry, Yale University School of Medicine, New Haven, USA
- Interdepartmental Neuroscience Program, Yale University School of Medicine, New Haven, USA
| | - Yury M Morozov
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06511, USA
| | - Alvaro Duque
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06511, USA
| | - Pasko Rakic
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06511, USA
| | - Christopher H van Dyck
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06511, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, USA
| | - Angus C Nairn
- Department of Psychiatry, Yale University School of Medicine, New Haven, USA
| | - Amy F T Arnsten
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar St., New Haven, CT, 06511, USA.
| |
Collapse
|
46
|
Olcum M, Tastan B, Kiser C, Genc S, Genc K. Microglial NLRP3 inflammasome activation in multiple sclerosis. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2019; 119:247-308. [PMID: 31997770 DOI: 10.1016/bs.apcsb.2019.08.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Multiple sclerosis (MS) is a chronic, autoimmune and neuroinflammatory disease of the central nervous system (CNS) mediated by autoreactive T cells directed against myelin antigens. Although the crucial role of adaptive immunity is well established in MS, the contribution of innate immunity has only recently been appreciated. Microglia are the main innate immune cells of the CNS. Similar to other myeloid cells, microglia recognize both exogenous and host-derived endogenous danger signals through pattern recognition receptors (PRRs) localized on their cell surface such as Toll Like receptor 4, or in the cytosol such as NLRP3. The second one is the sensor protein of the multi-molecular NLRP3 inflammasome complex in activated microglia that promotes the maturation and secretion of proinflammatory cytokines, interleukin-1β and interleukin-18. Overactivation of microglia and aberrant activation of the NLRP3 inflammasome have been implicated in the pathogenesis of MS. Indeed, experimental data, together with post-mortem and clinical studies have revealed an increased expression of NLRP3 inflammasome complex elements in microglia and other immune cells. In this review, we focus on microglial NLRP3 inflammasome activation in MS. First, we overview the basic knowledge about MS, microglia and the NLRP3 inflammasome. Then, we summarize studies about microglial NLRP3 inflammasome activation in MS and its animal models. We also highlight experimental therapeutic approaches that target different steps of NLRP inflammasome activation. Finally, we discuss future research avenues and new methods in this rapidly evolving area.
Collapse
Affiliation(s)
- Melis Olcum
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus Balcova, Izmir, Turkey
| | - Bora Tastan
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus Balcova, Izmir, Turkey; Izmir International Biomedicine and Genome Institute (iBG-Izmir), Dokuz Eylul University Health Campus, Balcova, Izmir, Turkey
| | - Cagla Kiser
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus Balcova, Izmir, Turkey; Izmir International Biomedicine and Genome Institute (iBG-Izmir), Dokuz Eylul University Health Campus, Balcova, Izmir, Turkey
| | - Sermin Genc
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus Balcova, Izmir, Turkey; Izmir International Biomedicine and Genome Institute (iBG-Izmir), Dokuz Eylul University Health Campus, Balcova, Izmir, Turkey; Department of Neuroscience, Institute of Health and Science, Dokuz Eylul University Health Campus, Balcova, Izmir, Turkey
| | - Kursad Genc
- Department of Neuroscience, Institute of Health and Science, Dokuz Eylul University Health Campus, Balcova, Izmir, Turkey
| |
Collapse
|