151
|
Zhang Y, Chu JMT, Wong GTC. Cerebral Glutamate Regulation and Receptor Changes in Perioperative Neuroinflammation and Cognitive Dysfunction. Biomolecules 2022; 12:biom12040597. [PMID: 35454185 PMCID: PMC9029551 DOI: 10.3390/biom12040597] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/11/2022] [Accepted: 04/15/2022] [Indexed: 12/23/2022] Open
Abstract
Glutamate is the major excitatory neurotransmitter in the central nervous system and is intricately linked to learning and memory. Its activity depends on the expression of AMPA and NMDA receptors and excitatory amino transporters on neurons and glial cells. Glutamate transporters prevent the excess accumulation of glutamate in synapses, which can lead to aberrant synaptic signaling, excitotoxicity, or cell death. Neuroinflammation can occur acutely after surgical trauma and contributes to the development of perioperative neurocognitive disorders, which are characterized by impairment in multiple cognitive domains. In this review, we aim to examine how glutamate handling and glutamatergic function are affected by neuroinflammation and their contribution to cognitive impairment. We will first summarize the current data regarding glutamate in neurotransmission, its receptors, and their regulation and trafficking. We will then examine the impact of inflammation on glutamate handling and neurotransmission, focusing on changes in glial cells and the effect of cytokines. Finally, we will discuss these changes in the context of perioperative neuroinflammation and the implications they have for perioperative neurocognitive disorders.
Collapse
|
152
|
Ahmad S, Srivastava RK, Singh P, Naik UP, Srivastava AK. Role of Extracellular Vesicles in Glia-Neuron Intercellular Communication. Front Mol Neurosci 2022; 15:844194. [PMID: 35493327 PMCID: PMC9043804 DOI: 10.3389/fnmol.2022.844194] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
Cross talk between glia and neurons is crucial for a variety of biological functions, ranging from nervous system development, axonal conduction, synaptic transmission, neural circuit maturation, to homeostasis maintenance. Extracellular vesicles (EVs), which were initially described as cellular debris and were devoid of biological function, are now recognized as key components in cell-cell communication and play a critical role in glia-neuron communication. EVs transport the proteins, lipids, and nucleic acid cargo in intercellular communication, which alters target cells structurally and functionally. A better understanding of the roles of EVs in glia-neuron communication, both in physiological and pathological conditions, can aid in the discovery of novel therapeutic targets and the development of new biomarkers. This review aims to demonstrate that different types of glia and neuronal cells secrete various types of EVs, resulting in specific functions in intercellular communications.
Collapse
Affiliation(s)
- Shahzad Ahmad
- Department of Medical Elementology and Toxicology, Jamia Hamdard University, New Delhi, India
| | - Rohit K. Srivastava
- Department of Pediatric Surgery, Texas Children’s Hospital, Houston, TX, United States
- M.E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Pratibha Singh
- Department of Biochemistry and Cell Biology, Biosciences Research Collaborative, Rice University, Houston, TX, United States
| | - Ulhas P. Naik
- Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Cardeza Foundation for Hematologic Research, Philadelphia, PA, United States
| | - Amit K. Srivastava
- Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Cardeza Foundation for Hematologic Research, Philadelphia, PA, United States
- *Correspondence: Amit K. Srivastava,
| |
Collapse
|
153
|
The Pivotal Immunoregulatory Functions of Microglia and Macrophages in Glioma Pathogenesis and Therapy. JOURNAL OF ONCOLOGY 2022; 2022:8903482. [PMID: 35419058 PMCID: PMC9001141 DOI: 10.1155/2022/8903482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 03/24/2022] [Indexed: 12/16/2022]
Abstract
Gliomas are mixed solid tumors composed of both neoplastic and nonneoplastic cells. In glioma microenvironment, the most common nonneoplastic and infiltrating cells are macrophages and microglia. Microglia are the exact phagocytes of the central nervous system, whereas macrophages are myeloid immune cells that are depicted with ardent phagocytosis. Microglia are heterogeneously located in almost all nonoverlapping sections of the brain as well as the spinal cord, while macrophages are derived from circulating monocytes. Microglia and macrophages utilize a variety of receptors for the detection of molecules, particles, and cells that they engulf. Both microglia and peripheral macrophages interact directly with vessels both in the periphery of and within the tumor. In glioma milieu, normal human astrocytes, glioma cells, and microglia all exhibited the ability of phagocytosing glioma cells and precisely apoptotic tumor cells. Also, microglia and macrophages are robustly triggered by the glioma via the expression of chemoattractants such as monocyte chemoattractant protein, stromal-derived factor-1, and macrophage-colony stimulating factor. Glioma-associated microglia and/or macrophages positively correlated with glioma invasiveness, immunosuppression, and patients' poor outcome, making these cells a suitable target for immunotherapeutic schemes.
Collapse
|
154
|
Han J, Chitu V, Stanley ER, Wszolek ZK, Karrenbauer VD, Harris RA. Inhibition of colony stimulating factor-1 receptor (CSF-1R) as a potential therapeutic strategy for neurodegenerative diseases: opportunities and challenges. Cell Mol Life Sci 2022; 79:219. [PMID: 35366105 PMCID: PMC8976111 DOI: 10.1007/s00018-022-04225-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/06/2022] [Accepted: 02/26/2022] [Indexed: 12/12/2022]
Abstract
Microglia are specialized dynamic immune cells in the central nervous system (CNS) that plays a crucial role in brain homeostasis and in disease states. Persistent neuroinflammation is considered a hallmark of many neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson's disease (PD), Huntington’s disease (HD), amyotrophic lateral sclerosis (ALS) and primary progressive multiple sclerosis (MS). Colony stimulating factor 1-receptor (CSF-1R) is predominantly expressed on microglia and its expression is significantly increased in neurodegenerative diseases. Cumulative findings have indicated that CSF-1R inhibitors can have beneficial effects in preclinical neurodegenerative disease models. Research using CSF-1R inhibitors has now been extended into non-human primates and humans. This review article summarizes the most recent advances using CSF-1R inhibitors in different neurodegenerative conditions including AD, PD, HD, ALS and MS. Potential challenges for translating these findings into clinical practice are presented.
Collapse
Affiliation(s)
- Jinming Han
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Violeta Chitu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| | - E. Richard Stanley
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
| | | | - Virginija Danylaité Karrenbauer
- Department of Clinical Neuroscience, Center for Molecular Medicine L8:04, Karolinska Institutet, Karolinska University Hospital, 171 76 Stockholm, Sweden
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
| | - Robert A. Harris
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| |
Collapse
|
155
|
Zwergal A, Lindner M, Grosch M, Dieterich M. In vivo neuroplasticity in vestibular animal models. Mol Cell Neurosci 2022; 120:103721. [PMID: 35338004 DOI: 10.1016/j.mcn.2022.103721] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/09/2022] [Accepted: 03/14/2022] [Indexed: 12/21/2022] Open
Abstract
An acute unilateral vestibulopathy leads to symptoms of vestibular tone imbalance, which gradually decrease over days to weeks due to central vestibular compensation. Animal models of acute peripheral vestibular lesions are optimally suited to investigate the mechanisms underlying this lesion-induced adaptive neuroplasticity. Previous studies applied ex vivo histochemical techniques or local in vivo electrophysiological recordings mostly in the vestibular nucleus complex to delineate the mechanisms involved. Recently, the use of imaging methods, such as positron emission tomography (PET) or magnetic resonance imaging (MRI), in vestibular animal models have opened a complementary perspective by depicting whole-brain structure and network changes of neuronal activity over time and in correlation to behaviour. Here, we review recent multimodal imaging studies in vestibular animal models with a focus on PET-based measurements of glucose metabolism, glial activation and synaptic plasticity. [18F]-FDG-PET studies indicate dynamic alterations of regional glucose metabolism in brainstem-cerebellar, thalamic, cortical sensory and motor, as well as limbic areas starting early after unilateral labyrinthectomy (UL) in the rat. Sequential whole-brain analysis of the metabolic connectome during vestibular compensation shows a significant increase of connections mostly in the contralesional hemisphere after UL, which reaches a maximum at day 3 and thereby parallels the course of vestibular recovery. Glial activation in the ipsilesional vestibular nerve and nucleus peak between days 7 and 15 after UL. Synaptic density in brainstem-cerebellar circuits decreases until 8 weeks after UL, while it increases in frontal, motor and sensory cortical areas. We finally report how pharmacological compounds modulate the functional and structural plasticity mechanisms during vestibular compensation.
Collapse
Affiliation(s)
- Andreas Zwergal
- Department of Neurology, University Hospital, LMU Munich, Germany; German Center for Vertigo and Balance Disorders, DSGZ, LMU Munich, Germany.
| | - Magdalena Lindner
- German Center for Vertigo and Balance Disorders, DSGZ, LMU Munich, Germany; Department of Nuclear Medicine, LMU Munich, Germany
| | - Maximilian Grosch
- German Center for Vertigo and Balance Disorders, DSGZ, LMU Munich, Germany
| | - Marianne Dieterich
- Department of Neurology, University Hospital, LMU Munich, Germany; German Center for Vertigo and Balance Disorders, DSGZ, LMU Munich, Germany; Munich Cluster of Systems Neurology, SyNergy, Munich, Germany
| |
Collapse
|
156
|
Lim JY, Lee JE, Park SA, Park SI, Yon JM, Park JA, Jeun SS, Kim SJ, Lee HJ, Kim SW, Yang SH. Protective Effect of Human-Neural-Crest-Derived Nasal Turbinate Stem Cells against Amyloid-β Neurotoxicity through Inhibition of Osteopontin in a Human Cerebral Organoid Model of Alzheimer’s Disease. Cells 2022; 11:cells11061029. [PMID: 35326480 PMCID: PMC8947560 DOI: 10.3390/cells11061029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 11/16/2022] Open
Abstract
The aim of this study was to validate the use of human brain organoids (hBOs) to investigate the therapeutic potential and mechanism of human-neural-crest-derived nasal turbinate stem cells (hNTSCs) in models of Alzheimer’s disease (AD). We generated hBOs from human induced pluripotent stem cells, investigated their characteristics according to neuronal markers and electrophysiological features, and then evaluated the protective effect of hNTSCs against amyloid-β peptide (Aβ1–42) neurotoxic activity in vitro in hBOs and in vivo in a mouse model of AD. Treatment of hBOs with Aβ1–42 induced neuronal cell death concomitant with decreased expression of neuronal markers, which was suppressed by hNTSCs cocultured under Aβ1–42 exposure. Cytokine array showed a significantly decreased level of osteopontin (OPN) in hBOs with hNTSC coculture compared with hBOs only in the presence of Aβ1–42. Silencing OPN via siRNA suppressed Aβ-induced neuronal cell death in cell culture. Notably, compared with PBS, hNTSC transplantation significantly enhanced performance on the Morris water maze, with reduced levels of OPN after transplantation in a mouse model of AD. These findings reveal that hBO models are useful to evaluate the therapeutic effect and mechanism of stem cells for application in treating AD.
Collapse
Affiliation(s)
- Jung Yeon Lim
- Department of Otolaryngology-Head and Neck Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Jung Eun Lee
- Department of Neurosurgery, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon 16247, Kyonggi-do, Korea
| | - Soon A Park
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Sang In Park
- Institute of Catholic Integrative Medicine (ICIM), Incheon St. Mary's Hospital, The Catholic University of Korea, Seoul 06591, Korea
| | - Jung-Min Yon
- Department of Otolaryngology-Head and Neck Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Jeong-Ah Park
- Department of Otolaryngology-Head and Neck Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Sin-Soo Jeun
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Seung Joon Kim
- Division of Pulmonology, Critical Care and Allergy, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Hong Jun Lee
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju 28644, Korea
| | - Sung Won Kim
- Department of Otolaryngology-Head and Neck Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Seung Ho Yang
- Department of Neurosurgery, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon 16247, Kyonggi-do, Korea
| |
Collapse
|
157
|
Abstract
Chandelier cells (ChCs) are a unique type of GABAergic interneuron that form axo-axonic synapses exclusively on the axon initial segment (AIS) of neocortical pyramidal neurons (PyNs), allowing them to exert powerful yet precise control over PyN firing and population output. The importance of proper ChC function is further underscored by the association of ChC connectivity defects with various neurological conditions. Despite this, the cellular mechanisms governing ChC axo-axonic synapse formation remain poorly understood. Here, we identify microglia as key regulators of ChC axonal morphogenesis and AIS synaptogenesis, and show that disease-induced aberrant microglial activation perturbs proper ChC synaptic development/connectivity in the neocortex. In doing so, such findings highlight the therapeutic potential of manipulating microglia to ensure proper brain wiring. Microglia have emerged as critical regulators of synapse development and circuit formation in the healthy brain. To date, examination of microglia in such processes has largely been focused on excitatory synapses. Their roles, however, in the modulation of GABAergic interneuron synapses—particularly those targeting the axon initial segment (AIS)—during development remain enigmatic. Here, we identify a synaptogenic/growth-promoting role for microglia in regulating pyramidal neuron (PyN) AIS synapse formation by chandelier cells (ChCs), a unique interneuron subtype whose axonal terminals, called cartridges, selectively target the AIS. We show that a subset of microglia contacts PyN AISs and ChC cartridges and that such tripartite interactions, which rely on the unique AIS cytoskeleton and microglial GABAB1 receptors, are associated with increased ChC cartridge length and bouton number and AIS synaptogenesis. Conversely, microglia depletion or disease-induced aberrant microglia activation impairs the proper development and maintenance of ChC cartridges and boutons, as well as AIS synaptogenesis. These findings unveil key roles for homeostatic, AIS-associated microglia in regulating proper ChC axonal morphogenesis and synaptic connectivity in the neocortex.
Collapse
|
158
|
de Witte LD, Wang Z, Snijders GLJL, Mendelev N, Liu Q, Sneeboer MAM, Boks MPM, Ge Y, Haghighi F. Contribution of Age, Brain Region, Mood Disorder Pathology, and Interindividual Factors on the Methylome of Human Microglia. Biol Psychiatry 2022; 91:572-581. [PMID: 35027166 PMCID: PMC11181298 DOI: 10.1016/j.biopsych.2021.10.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Transcriptome studies have revealed age-, disease-, and region-associated microglial phenotypes reflecting changes in microglial function during development, aging, central nervous system homeostasis, and pathology. The molecular mechanisms that contribute to these transcriptomic changes are largely unknown. The aim of this study was to characterize the DNA methylation landscape of human microglia and the factors that contribute to variations in the microglia methylome. We hypothesized that both age and brain region would have a large impact on DNA methylation in microglia. METHODS Microglia from postmortem brain tissue of four different brain regions of 22 donors, encompassing 1 patient with schizophrenia, 13 patients with mood disorder pathology, and 8 control subjects, were isolated and assayed using a genome-wide methylation array. RESULTS We found that human microglial cells have a methylation profile distinct from bulk brain tissue and neurons, and age explained a considerable part of the variation. Additionally, we showed that interindividual factors had a much larger effect on the methylation landscape of microglia than brain region, which was also seen at the transcriptome level. In our exploratory analysis, we found various differentially methylated regions that were related to disease status (mood disorder vs. control). This included differentially methylated regions that are linked to gene expression in microglia, as well as to myeloid cell function or neuropsychiatric disorders. CONCLUSIONS Although based on relatively small samples, these findings suggest that the methylation profile of microglia is responsive to interindividual variations and thereby plays an important role in the heterogeneity of microglia observed at the transcriptome level.
Collapse
Affiliation(s)
- Lot D de Witte
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, Bronx, New York; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Zhaoyu Wang
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, Bronx, New York; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Gijsje L J L Snijders
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, Bronx, New York; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Natalia Mendelev
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, Bronx, New York; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Qingkun Liu
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, Bronx, New York; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Marjolein A M Sneeboer
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Psychiatry, University Medical Center Utrecht, UMC Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands; Department of Translational Neuroscience, University Medical Center Utrecht, UMC Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Marco P M Boks
- Department of Psychiatry, University Medical Center Utrecht, UMC Utrecht Brain Center, Utrecht University, Utrecht, the Netherlands
| | - Yongchao Ge
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Fatemeh Haghighi
- Mental Illness Research, Education and Clinical Center, James J Peters VA Medical Center, Bronx, New York; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
159
|
Hösli L, Binini N, Ferrari KD, Thieren L, Looser ZJ, Zuend M, Zanker HS, Berry S, Holub M, Möbius W, Ruhwedel T, Nave KA, Giaume C, Weber B, Saab AS. Decoupling astrocytes in adult mice impairs synaptic plasticity and spatial learning. Cell Rep 2022; 38:110484. [PMID: 35263595 DOI: 10.1016/j.celrep.2022.110484] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/20/2021] [Accepted: 02/14/2022] [Indexed: 12/16/2022] Open
Abstract
The mechanisms by which astrocytes modulate neural homeostasis, synaptic plasticity, and memory are still poorly explored. Astrocytes form large intercellular networks by gap junction coupling, mainly composed of two gap junction channel proteins, connexin 30 (Cx30) and connexin 43 (Cx43). To circumvent developmental perturbations and to test whether astrocytic gap junction coupling is required for hippocampal neural circuit function and behavior, we generate and study inducible, astrocyte-specific Cx30 and Cx43 double knockouts. Surprisingly, disrupting astrocytic coupling in adult mice results in broad activation of astrocytes and microglia, without obvious signs of pathology. We show that hippocampal CA1 neuron excitability, excitatory synaptic transmission, and long-term potentiation are significantly affected. Moreover, behavioral inspection reveals deficits in sensorimotor performance and a complete lack of spatial learning and memory. Together, our findings establish that astrocytic connexins and an intact astroglial network in the adult brain are vital for neural homeostasis, plasticity, and spatial cognition.
Collapse
Affiliation(s)
- Ladina Hösli
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland; Neuroscience Center Zurich, University and ETH Zurich, 8057 Zurich, Switzerland
| | - Noemi Binini
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland; Neuroscience Center Zurich, University and ETH Zurich, 8057 Zurich, Switzerland
| | - Kim David Ferrari
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland; Neuroscience Center Zurich, University and ETH Zurich, 8057 Zurich, Switzerland
| | - Laetitia Thieren
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland; Neuroscience Center Zurich, University and ETH Zurich, 8057 Zurich, Switzerland
| | - Zoe J Looser
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland; Neuroscience Center Zurich, University and ETH Zurich, 8057 Zurich, Switzerland
| | - Marc Zuend
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland; Neuroscience Center Zurich, University and ETH Zurich, 8057 Zurich, Switzerland
| | - Henri S Zanker
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland; Neuroscience Center Zurich, University and ETH Zurich, 8057 Zurich, Switzerland
| | - Stewart Berry
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Martin Holub
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland; Neuroscience Center Zurich, University and ETH Zurich, 8057 Zurich, Switzerland
| | - Wiebke Möbius
- Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Torben Ruhwedel
- Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Klaus-Armin Nave
- Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Christian Giaume
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, 75231 Paris Cedex 05, France
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland; Neuroscience Center Zurich, University and ETH Zurich, 8057 Zurich, Switzerland.
| | - Aiman S Saab
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland; Neuroscience Center Zurich, University and ETH Zurich, 8057 Zurich, Switzerland.
| |
Collapse
|
160
|
Gaspar R, Soares-Cunha C, Domingues AV, Coimbra B, Baptista FI, Pinto L, Ambrósio AF, Rodrigues AJ, Gomes CA. The Duration of Stress Determines Sex Specificities in the Vulnerability to Depression and in the Morphologic Remodeling of Neurons and Microglia. Front Behav Neurosci 2022; 16:834821. [PMID: 35330844 PMCID: PMC8940280 DOI: 10.3389/fnbeh.2022.834821] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/31/2022] [Indexed: 11/24/2022] Open
Abstract
Stress exposure has been shown to induce a variety of molecular and functional alterations associated with anxiety and depression. Some studies suggest that microglia, the immune cells of the brain, play a significant role in determining neuronal and behavioral responses to chronic stress and also contribute to the development of stress-related psychopathologies. However, little is known about the impact of the duration of stress exposure upon microglia and neurons morphology, particularly considering sex differences. This issue deserves particular investigation, considering that the process of morphologic remodeling of neurons and microglia is usually accompanied by functional changes with behavioral expression. Here, we examine the effects of short and long unpredictable chronic mild stress (uCMS) protocols on behavior, evaluating in parallel microglia and neurons morphology in the dorsal hippocampus (dHIP) and in the nucleus accumbens (NAc), two brain regions involved in the etiology of depression. We report that long-term uCMS induced more behavioral alterations in males, which present anxiety and depression-like phenotypes (anhedonia and helplessness behavior), while females only display anxiety-like behavior. After short-term uCMS, both sexes presented anxiety-like behavior. Microglia cells undergo a process of morphologic adaptation to short-term uCMS, dependent on sex, in the NAc: we observed a hypertrophy in males and an atrophy in females, transient effects that do not persist after long-term uCMS. In the dHIP, the morphologic adaptation of microglia is only observed in females (hypertrophy) and after the protocol of long uCMS. Interestingly, males are more vulnerable to neuronal morphological alterations in a region-specific manner: dendritic atrophy in granule neurons of the dHIP and hypertrophy in the medium spiny neurons of the NAc, both after short- or long-term uCMS. The morphology of neurons in these brain regions were not affected in females. These findings raise the possibility that, by differentially affecting neurons and microglia in dHIP and NAc, chronic stress may contribute for differences in the clinical presentation of stress-related disorders under the control of sex-specific mechanisms.
Collapse
Affiliation(s)
- Rita Gaspar
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Carina Soares-Cunha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s –PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ana Verónica Domingues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s –PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Bárbara Coimbra
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s –PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Filipa I. Baptista
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Luísa Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s –PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - António F. Ambrósio
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Ana João Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s –PT Government Associate Laboratory, Braga/Guimarães, Portugal
- *Correspondence: Ana João Rodrigues,
| | - Catarina A. Gomes
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
- Catarina A. Gomes,
| |
Collapse
|
161
|
Cortical axon sub-population maintains density, but not turnover, of en passant boutons in the aged APP/PS1 amyloidosis model. Neurobiol Aging 2022; 115:29-38. [DOI: 10.1016/j.neurobiolaging.2022.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/10/2022] [Accepted: 03/12/2022] [Indexed: 11/21/2022]
|
162
|
Jin Y, Kang Y, Wang M, Wu B, Su B, Yin H, Tang Y, Li Q, Wei W, Mei Q, Hu G, Lukacs-Kornek V, Li J, Wu K, Yuan X, Wang W. Targeting polarized phenotype of microglia via IL6/JAK2/STAT3 signaling to reduce NSCLC brain metastasis. Signal Transduct Target Ther 2022; 7:52. [PMID: 35194016 PMCID: PMC8864012 DOI: 10.1038/s41392-022-00872-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 12/29/2021] [Accepted: 12/31/2021] [Indexed: 12/27/2022] Open
Abstract
Tumor-associated macrophages have emerged as crucial factors for metastases. Microglia are indispensable components of the brain microenvironment and play vital roles in brain metastasis (BM). However, the underlying mechanism of how activated microglia promote brain metastasis of non-small cell lung cancer (NSCLC) remains elusive. Here, we purified cell lines with brain-metastatic tropism and employed a co-culture system to reveal their communication with microglia. By single-cell RNA-sequencing and transcriptome difference analysis, we identified IL6 as the key regulator in brain-metastatic cells (A549-F3) to induce anti-inflammatory microglia via JAK2/STAT3 signaling, which in turn promoted the colonization process in metastatic A549-F3 cells. In our clinical samples, patients with higher levels of IL6 in serum showed higher propensity for brain metastasis. Additionally, the TCGA (The Cancer Genome Atlas) data revealed that NSCLC patients with a lower level of IL6 had a longer overall survival time compared to those with a higher level of IL6. Overall, our data indicate that the targeting of IL6/JAK2/STAT3 signaling in activated microglia may be a promising new approach for inhibiting brain metastasis in NSCLC patients.
Collapse
Affiliation(s)
- Yu Jin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yalin Kang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Minghuan Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Bili Wu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Beibei Su
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Han Yin
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Yang Tang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Qianxia Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Wenjie Wei
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Qi Mei
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Guangyuan Hu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Veronika Lukacs-Kornek
- Institute of Experimental Immunology, University Clinic of Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | - Jian Li
- Institute of Experimental Immunology, University Clinic of Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | - Kongming Wu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei Province, China.
| |
Collapse
|
163
|
Sarkar T, Patro N, Patro IK. Perinatal exposure to synergistic multiple stressors lead to cellular and behavioral deficits mimicking Schizophrenia like pathology. Biol Open 2022; 11:274201. [PMID: 35107124 PMCID: PMC8918990 DOI: 10.1242/bio.058870] [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: 06/01/2021] [Accepted: 01/24/2022] [Indexed: 11/24/2022] Open
Abstract
Protein malnourishment and immune stress are potent perinatal stressors, encountered by children born under poor socioeconomic conditions. Thus, it is necessary to investigate how such stressors synergistically contribute towards developing neurological disorders in affected individuals. Pups from Wistar females, maintained on normal (high-protein, HP:20%) and low-protein (LP:8%) diets were used. Single and combined exposures of Poly I:C (viral mimetic: 5 mg/kg body weight) and Lipopolysaccharide (LPS; bacterial endotoxin: 0.3 mg/kg body weight) were injected to both HP and LP pups at postnatal days (PND) 3 and 9 respectively, creating eight groups: HP (control); HP+Poly I:C; HP+LPS; HP+Poly I:C+LPS; LP; LP+Poly I:C; LP+LPS; LP+Poly I:C+LPS (multi-hit). The effects of stressors on hippocampal cytoarchitecture and behavioral abilities were studied at PND 180. LP animals were found to be more vulnerable to immune stressors than HP animals and symptoms like neuronal damage, spine loss, downregulation of Egr 1 and Arc proteins, gliosis and behavioral deficits were maximum in the multi-hit group. Thus, from these findings it is outlined that cellular and behavioral changes that occur following multi-hit exposure may predispose individuals to developing Schizophrenia-like pathologies during adulthood. Summary: This study reports that exposure to perinatal multi-hit stress (protein malnourishment and immune stress) causes changes in the hippocampal cells alongside behavioral deficits which are also observed in Schizophrenic condition.
Collapse
Affiliation(s)
- Tiyasha Sarkar
- School of Studies in Neuroscience, Jiwaji University, Gwalior-474011, India
| | - Nisha Patro
- School of Studies in Neuroscience, Jiwaji University, Gwalior-474011, India
| | - Ishan Kumar Patro
- School of Studies in Neuroscience, Jiwaji University, Gwalior-474011, India
| |
Collapse
|
164
|
Kim CK, Sachdev PS, Braidy N. Recent Neurotherapeutic Strategies to Promote Healthy Brain Aging: Are we there yet? Aging Dis 2022; 13:175-214. [PMID: 35111369 PMCID: PMC8782556 DOI: 10.14336/ad.2021.0705] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/05/2021] [Indexed: 12/21/2022] Open
Abstract
Owing to the global exponential increase in population ageing, there is an urgent unmet need to develop reliable strategies to slow down and delay the ageing process. Age-related neurodegenerative diseases are among the main causes of morbidity and mortality in our contemporary society and represent a major socio-economic burden. There are several controversial factors that are thought to play a causal role in brain ageing which are continuously being examined in experimental models. Among them are oxidative stress and brain inflammation which are empirical to brain ageing. Although some candidate drugs have been developed which reduce the ageing phenotype, their clinical translation is limited. There are several strategies currently in development to improve brain ageing. These include strategies such as caloric restriction, ketogenic diet, promotion of cellular nicotinamide adenine dinucleotide (NAD+) levels, removal of senescent cells, 'young blood' transfusions, enhancement of adult neurogenesis, stem cell therapy, vascular risk reduction, and non-pharmacological lifestyle strategies. Several studies have shown that these strategies can not only improve brain ageing by attenuating age-related neurodegenerative disease mechanisms, but also maintain cognitive function in a variety of pre-clinical experimental murine models. However, clinical evidence is limited and many of these strategies are awaiting findings from large-scale clinical trials which are nascent in the current literature. Further studies are needed to determine their long-term efficacy and lack of adverse effects in various tissues and organs to gain a greater understanding of their potential beneficial effects on brain ageing and health span in humans.
Collapse
Affiliation(s)
- Chul-Kyu Kim
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
| | - Perminder S Sachdev
- Neuropsychiatric Institute, Euroa Centre, Prince of Wales Hospital, Sydney, Australia
| | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, Australia
| |
Collapse
|
165
|
Paasila PJ, Aramideh JA, Sutherland GT, Graeber MB. Synapses, Microglia, and Lipids in Alzheimer's Disease. Front Neurosci 2022; 15:778822. [PMID: 35095394 PMCID: PMC8789683 DOI: 10.3389/fnins.2021.778822] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/06/2021] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is characterised by synaptic dysfunction accompanied by the microscopically visible accumulation of pathological protein deposits and cellular dystrophy involving both neurons and glia. Late-stage AD shows pronounced loss of synapses and neurons across several differentially affected brain regions. Recent studies of advanced AD using post-mortem brain samples have demonstrated the direct involvement of microglia in synaptic changes. Variants of the Apolipoprotein E and Triggering Receptors Expressed on Myeloid Cells gene represent important determinants of microglial activity but also of lipid metabolism in cells of the central nervous system. Here we review evidence that may help to explain how abnormal lipid metabolism, microglial activation, and synaptic pathophysiology are inter-related in AD.
Collapse
Affiliation(s)
- Patrick J. Paasila
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Jason A. Aramideh
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Greg T. Sutherland
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Manuel B. Graeber
- Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
166
|
Lupori L, Cornuti S, Mazziotti R, Borghi E, Ottaviano E, Cas MD, Sagona G, Pizzorusso T, Tognini P. The gut microbiota of environmentally enriched mice regulates visual cortical plasticity. Cell Rep 2022; 38:110212. [PMID: 35021093 DOI: 10.1016/j.celrep.2021.110212] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 11/08/2021] [Accepted: 12/14/2021] [Indexed: 12/24/2022] Open
Abstract
Exposing animals to an enriched environment (EE) has dramatic effects on brain structure, function, and plasticity. The poorly known "EE-derived signals'' mediating the EE effects are thought to be generated within the central nervous system. Here, we shift the focus to the body periphery, revealing that gut microbiota signals are crucial for EE-driven plasticity. Developmental analysis reveals striking differences in intestinal bacteria composition between EE and standard rearing (ST) mice, as well as enhanced levels of short-chain fatty acids (SCFA) in EE mice. Depleting the microbiota of EE mice with antibiotics strongly decreases SCFA and prevents activation of adult ocular dominance plasticity, spine dynamics, and microglia rearrangement. SCFA treatment in ST mice mimics EE induction of ocular dominance plasticity and microglial remodeling. Remarkably, transferring the microbiota of EE mice to ST recipients activates adult ocular dominance plasticity. Thus, experience-dependent changes in gut microbiota regulate brain plasticity.
Collapse
Affiliation(s)
| | - Sara Cornuti
- BIO@SNS Lab, Scuola Normale Superiore, 56126 Pisa, Italy
| | - Raffaele Mazziotti
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy
| | - Elisa Borghi
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy
| | | | - Michele Dei Cas
- Department of Health Sciences, Università degli Studi di Milano, 20142 Milan, Italy
| | - Giulia Sagona
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy
| | - Tommaso Pizzorusso
- BIO@SNS Lab, Scuola Normale Superiore, 56126 Pisa, Italy; Department of Neuroscience, Psychology, Drug Research and Child Health NEUROFARBA University of Florence, 50100 Florence, Italy; Institute of Neuroscience, National Research Council, 56124 Pisa, Italy
| | - Paola Tognini
- BIO@SNS Lab, Scuola Normale Superiore, 56126 Pisa, Italy; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy.
| |
Collapse
|
167
|
Abstract
The last century was characterized by a significant scientific effort aimed at unveiling the neurobiological basis of learning and memory. Thanks to the characterization of the mechanisms regulating the long-term changes of neuronal synaptic connections, it was possible to understand how specific neural networks shape themselves during the acquisition of memory traces or complex motor tasks. In this chapter, we will summarize the mechanisms underlying the main forms of synaptic plasticity taking advantage of the studies performed in the hippocampus and in the nucleus striatum, key brain structures that play a crucial role in cognition. Moreover, we will discuss how the molecular pathways involved in the induction of physiologic synaptic long-term changes could be disrupted during neurodegenerative and neuroinflammatory disorders, highlighting the translational relevance of this intriguing research field.
Collapse
Affiliation(s)
- Andrea Mancini
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| | - Antonio de Iure
- IRCCS San Raffaele Roma, Laboratory of Experimental Neurophysiology, Rome, Italy
| | - Barbara Picconi
- IRCCS San Raffaele Roma, Laboratory of Experimental Neurophysiology, Rome, Italy; University San Raffaele, Rome, Italy.
| |
Collapse
|
168
|
Chen HR, Chen CW, Kuo YM, Chen B, Kuan IS, Huang H, Lee J, Anthony N, Kuan CY, Sun YY. Monocytes promote acute neuroinflammation and become pathological microglia in neonatal hypoxic-ischemic brain injury. Theranostics 2022; 12:512-529. [PMID: 34976198 PMCID: PMC8692901 DOI: 10.7150/thno.64033] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 11/08/2021] [Indexed: 12/17/2022] Open
Abstract
Rationale: Monocytes belong to the mononuclear phagocyte system and are immune responders to tissue injury and infection. There were also reports of monocytes transforming to microglia-like cells. Here we explore the roles of monocytes in microglia ontogeny and the pathogenesis of neonatal cerebral hypoxic-ischemic (HI) brain injury in mice. Methods: We used three genetic methods to track the development of monocytes, including CX3CR1GFP/+; CCR2RFP/+ reporter mice, adoptive transfer of GFP+ monocytes, and fate-mapping with CCR2-CreER mice, in neonatal mouse brains with or without lipopolysaccharide (LPS, 0.3 mg/kg)-sensitized Vannucci HI. We also used genetic (CCR2RFP/ RFP, CCR2 knockout) and pharmacological methods (RS102895, a CCR2 antagonist) to test the roles of monocytic influx in LPS/HI brain injury. Results: CCR2+ monocytes entered the late-embryonic brains via choroid plexus, but rapidly became CX3CR1+ amoeboid microglial cells (AMCs). The influx of CCR2+ monocytes declined after birth, but recurred after HI or LPS-sensitized HI (LPS/HI) brain injury, particularly in the hippocampus. The CCR2-CreER-based fate-mapping showed that CCR2+ monocytes became CD68+ TNFα+ macrophages within 4 d after LPS/HI, and maintained as TNFα+ MHCII+ macrophages or persisted as Tmem119+ Sall1+ P2RY12+ ramified microglia for at least five months after injury. Genetic deletion of the chemokine receptor CCR2 markedly diminished monocytic influx, the expression of pro- and anti-inflammatory cytokines, and brain damage. Post-LPS/HI application of RS102895 also reduced inflammatory responses and brain damage, leading to better cognitive functions. Conclusion: These results suggest that monocytes promote acute inflammatory responses and may become pathological microglia long after the neonatal LPS/HI insult. Further, blocking the influx of monocytes may be a potential therapy for neonatal brain injury.
Collapse
Affiliation(s)
- Hong-Ru Chen
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Ching-Wen Chen
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Yi-Min Kuo
- Department of Anesthesiology, Taipei Veterans General Hospital and National Yang Ming Chiao Tung University School of Medicine, Taipei, Taiwan
| | - Brandon Chen
- University of Louisville School of Medicine, Louisville, KY, USA
| | - Irena S. Kuan
- St. Louis University School of Medicine, St. Louis, MO, USA
| | - Henry Huang
- Department of Anesthesiology, Rhode Island Hospital, Providence, RI, USA
| | - Jolly Lee
- Emory University School of Medicine, Atlanta, GA, USA
| | - Neil Anthony
- Emory Integrated Cellular Imaging, Atlanta, GA, USA
| | - Chia-Yi Kuan
- Department of Neuroscience, Center for Brain Immunology and Glia (BIG), University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Yu-Yo Sun
- Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung, Taiwan
| |
Collapse
|
169
|
Bayat M, Khalili A, Bayat G, Akbari S, Yousefi Nejad A, Borhani Haghighi A, Haghani M. Effects of platelet-rich plasma on the memory impairment, apoptosis, and hippocampal synaptic plasticity in a rat model of hepatic encephalopathy. Brain Behav 2022; 12:e2447. [PMID: 34855284 PMCID: PMC8785608 DOI: 10.1002/brb3.2447] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/22/2021] [Accepted: 10/30/2021] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES In the present study, we aimed to determine whether intraperitoneal injection of platelet-rich plasma (PRP) could have a neuroprotective effect on learning, memory, and synaptic plasticity impairment as well as hippocampal apoptosis in rats with hepatic encephalopathy induced by bile duct ligated (BDL). METHODS The rats were divided into four groups: the control, sham, BDL+ V (vehicle), and BDL+ PRP. The BDL rats were treated with PRP immediately after the surgery, and the injection was done every 3 days for 30 days. The passive avoidance and Morris water maze tests were used for the evaluation of learning and memory. The long-term potentiation (LTP), basal-synaptic transmission, and paired-pulse ratio, as an index for measurement of neurotransmitter release probability, were evaluated by field-potential recording. After taking a blood sample for assessment of the liver enzymes, the animals were sacrificed and their hippocampus was removed for evaluation of cleaved caspase-3 by Western blot. RESULTS Serological assessment of the liver function showed that BDL severely impaired the liver function. Also, PRP treatment could partially improve the liver dysfunction along with recovery in fear memory and spatial learning memory performance, LTP, basal-synaptic transmission, and neurotransmitter release probability. PRP-treated rats also showed a significant reduction in neuronal apoptosis in the CA1 area. CONCLUSIONS The results of this study suggest that PRP improves cognitive performance and synaptic plasticity in BDL rats via direct neuroprotective property and/or indirectly by improvement of hepatic dysfunction.
Collapse
Affiliation(s)
- Mahnaz Bayat
- Clinical Neurology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Azadeh Khalili
- Department of Physiology-Pharmacology-Medical Physic, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Gholamreza Bayat
- Department of Physiology-Pharmacology-Medical Physic, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.,Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Somayeh Akbari
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amirhossein Yousefi Nejad
- Faculty of Veterinary Medicine, Department of Veterinary Medicine, Islamic Azad University of Kazeroon, Shiraz, Iran
| | | | - Masoud Haghani
- Department of Physiology, The Medical School, Shiraz University of Medical Sciences, Shiraz, Iran.,Histomorphometry and Stereology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
170
|
Francelle L, Mazzulli JR. Neuroinflammation in aucher disease, neuronal ceroid lipofuscinosis, and commonalities with Parkinson’s disease. Brain Res 2022; 1780:147798. [PMID: 35063468 PMCID: PMC9126024 DOI: 10.1016/j.brainres.2022.147798] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 01/05/2022] [Accepted: 01/13/2022] [Indexed: 12/13/2022]
Abstract
Lysosomal storage diseases (LSDs) are rare genetic disorders caused by a disruption in cellular clearance, resulting in pathological storage of undegraded lysosomal substrates. Recent clinical and genetic studies have uncovered links between multiple LSDs and common neurodegenerative diseases such as Parkinson's disease (PD). Here, we review recent literature describing the role of glia cells and neuroinflammation in PD and LSDs, including Gaucher disease (GD) and neuronal ceroid lipofuscinosis (NCL), and highlight converging inflammation pathways that lead to neuron loss. Recent data indicates that lysosomal dysfunction and accumulation of storage materials can initiate the activation of glial cells, through interaction with cell surface or cytosolic pattern recognition receptors that detect pathogenic aggregates of cellular debris. Activated glia cells could act to protect neurons through the elimination of toxic protein or lipid aggregates early in the disease process. However prolonged glial activation that occurs over several decades in chronic-age related neurodegeneration could induce the inappropriate elimination of synapses, leading to neuron loss. These studies provide mechanistic insight into the relationship between lysosomal dysfunction and glial activation, and offer novel therapeutic pathways for the treatment of PD and LSDs focused on reducing neuroinflammation and mitigating cell loss.
Collapse
|
171
|
Systemic LPS-induced microglial activation results in increased GABAergic tone: A mechanism of protection against neuroinflammation in the medial prefrontal cortex in mice. Brain Behav Immun 2022; 99:53-69. [PMID: 34582995 DOI: 10.1016/j.bbi.2021.09.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/19/2021] [Accepted: 09/21/2021] [Indexed: 02/01/2023] Open
Abstract
Neuroinflammation with excess microglial activation and synaptic dysfunction are early symptoms of most neurological diseases. However, how microglia-associated neuroinflammation regulates synaptic activity remains obscure. We report here that acute neuroinflammation induced by intraperitoneal injection of lipopolysaccharide (LPS) results in cell-type-specific increases in inhibitory postsynaptic currents in the glutamatergic, but not the GABAergic, neurons of medial prefrontal cortex (mPFC), coinciding with excessive microglial activation. LPS causes upregulation in levels of GABAAR subunits, glutamine synthetase and vesicular GABA transporter, and downregulation in brain-derived neurotrophic factor (BDNF) and its receptor, pTrkB. Blockage of microglial activation by minocycline ameliorates LPS-induced abnormal expression of GABA signaling-related proteins and activity of synaptic and network. Moreover, minocycline prevents the mice from LPS-induced aberrant behavior, such as a reduction in total distance and time spent in the centre in the open field test; decreases in entries into the open arm of elevated-plus maze and in consumption of sucrose; increased immobility in the tail suspension test. Furthermore, upregulation of GABA signaling by tiagabine also prevents LPS-induced microglial activation and aberrant behavior. This study illustrates a mode of bidirectional constitutive signaling between the neural and immune compartments of the brain, and suggests that the mPFC is an important area for brain-immune system communication. Moreover, the present study highlights GABAergic signaling as a key therapeutic target for mitigating neuroinflammation-induced abnormal synaptic activity in the mPFC, together with the associated behavioral abnormalities.
Collapse
|
172
|
Haidar MA, Ibeh S, Shakkour Z, Reslan MA, Nwaiwu J, Moqidem YA, Sader G, Nickles RG, Babale I, Jaffa AA, Salama M, Shaito A, Kobeissy F. Crosstalk between Microglia and Neurons in Neurotrauma: An Overview of the Underlying Mechanisms. Curr Neuropharmacol 2022; 20:2050-2065. [PMID: 34856905 PMCID: PMC9886840 DOI: 10.2174/1570159x19666211202123322] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 11/22/2022] Open
Abstract
Microglia are the resident immune cells of the brain and play a crucial role in housekeeping and maintaining homeostasis of the brain microenvironment. Upon injury or disease, microglial cells become activated, at least partly, via signals initiated by injured neurons. Activated microglia, thereby, contribute to both neuroprotection and neuroinflammation. However, sustained microglial activation initiates a chronic neuroinflammatory response which can disturb neuronal health and disrupt communications between neurons and microglia. Thus, microglia-neuron crosstalk is critical in a healthy brain as well as during states of injury or disease. As most studies focus on how neurons and microglia act in isolation during neurotrauma, there is a need to understand the interplay between these cells in brain pathophysiology. This review highlights how neurons and microglia reciprocally communicate under physiological conditions and during brain injury and disease. Furthermore, the modes of microglia-neuron communication are exposed, focusing on cell-contact dependent signaling and communication by the secretion of soluble factors like cytokines and growth factors. In addition, it has been discussed that how microglia-neuron interactions could exert either beneficial neurotrophic effects or pathologic proinflammatory responses. We further explore how aberrations in microglia-neuron crosstalk may be involved in central nervous system (CNS) anomalies, namely traumatic brain injury (TBI), neurodegeneration, and ischemic stroke. A clear understanding of how the microglia-neuron crosstalk contributes to the pathogenesis of brain pathologies may offer novel therapeutic avenues of brain trauma treatment.
Collapse
Affiliation(s)
- Muhammad Ali Haidar
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Stanley Ibeh
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Zaynab Shakkour
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Mohammad Amine Reslan
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Judith Nwaiwu
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Yomna Adel Moqidem
- Biotechnology Program, School of Science and Engineering, The American University in Cairo, Cairo, Egypt
| | - Georgio Sader
- Faculty of Medicine, University of Balamand, Balamand, Lebanon
| | - Rachel G. Nickles
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Ismail Babale
- Department of Biomedical Engineering, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Aneese A. Jaffa
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Beirut, Lebanon
| | - Mohamed Salama
- Institute of Global Health and Human Ecology (I-GHHE), The American University in Cairo, New Cairo 11835, Egypt
- Global Brain Health Institute (GBHI), Trinity College Dublin, Dublin, Ireland
| | - Abdullah Shaito
- Biomedical Research Center, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Biomedical Engineering, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
173
|
Frintrop L, Trinh S, Seitz J, Kipp M. The Role of Glial Cells in Regulating Feeding Behavior: Potential Relevance to Anorexia Nervosa. J Clin Med 2021; 11:jcm11010186. [PMID: 35011927 PMCID: PMC8745326 DOI: 10.3390/jcm11010186] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/17/2021] [Accepted: 12/25/2021] [Indexed: 12/16/2022] Open
Abstract
Eating behavior is controlled by hypothalamic circuits in which agouti-related peptide-expressing neurons when activated in the arcuate nucleus, promote food intake while pro-opiomelanocortin-producing neurons promote satiety. The respective neurotransmitters signal to other parts of the hypothalamus such as the paraventricular nucleus as well as several extra-hypothalamic brain regions to orchestrate eating behavior. This complex process of food intake may be influenced by glia cells, in particular astrocytes and microglia. Recent studies showed that GFAP+ astrocyte cell density is reduced in the central nervous system of an experimental anorexia nervosa model. Anorexia nervosa is an eating disorder that causes, among the well-known somatic symptoms, brain volume loss which was associated with neuropsychological deficits while the underlying pathophysiology is unknown. In this review article, we summarize the findings of glia cells in anorexia nervosa animal models and try to deduce which role glia cells might play in the pathophysiology of eating disorders, including anorexia nervosa. A better understanding of glia cell function in the regulation of food intake and eating behavior might lead to the identification of new drug targets.
Collapse
Affiliation(s)
- Linda Frintrop
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany;
- Correspondence: ; Tel.: +49-(0)-381-494-8406
| | - Stefanie Trinh
- Institute of Neuroanatomy, RWTH Aachen University, 52074 Aachen, Germany;
| | - Jochen Seitz
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, RWTH Aachen University, 52074 Aachen, Germany;
| | - Markus Kipp
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany;
| |
Collapse
|
174
|
Var SR, Shetty AV, Grande AW, Low WC, Cheeran MC. Microglia and Macrophages in Neuroprotection, Neurogenesis, and Emerging Therapies for Stroke. Cells 2021; 10:3555. [PMID: 34944064 PMCID: PMC8700390 DOI: 10.3390/cells10123555] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/12/2021] [Accepted: 12/15/2021] [Indexed: 12/20/2022] Open
Abstract
Stroke remains the number one cause of morbidity in the United States. Within weeks to months after an ischemic event, there is a resolution of inflammation and evidence of neurogenesis; however, years following a stroke, there is evidence of chronic inflammation in the central nervous system, possibly by the persistence of an autoimmune response to brain antigens as a result of ischemia. The mechanisms underlying the involvement of macrophage and microglial activation after stroke are widely acknowledged as having a role in ischemic stroke pathology; thus, modulating inflammation and neurological recovery is a hopeful strategy for treating the long-term outcomes after ischemic injury. Current treatments fail to provide neuroprotective or neurorestorative benefits after stroke; therefore, to ameliorate brain injury-induced deficits, therapies must alter both the initial response to injury and the subsequent inflammatory process. This review will address differences in macrophage and microglia nomenclature and summarize recent work in elucidating the mechanisms of macrophage and microglial participation in antigen presentation, neuroprotection, angiogenesis, neurogenesis, synaptic remodeling, and immune modulating strategies for treating the long-term outcomes after ischemic injury.
Collapse
Affiliation(s)
- Susanna R. Var
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (S.R.V.); (A.W.G.)
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN 55108, USA
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Anala V. Shetty
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
- Department of Biological Sciences, University of Minnesota Medical School, Minneapolis, MN 55108, USA
| | - Andrew W. Grande
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (S.R.V.); (A.W.G.)
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Walter C. Low
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA; (S.R.V.); (A.W.G.)
- Stem Cell Institute, University of Minnesota Medical School, Minneapolis, MN 55455, USA;
| | - Maxim C. Cheeran
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN 55108, USA
| |
Collapse
|
175
|
Levi H, Bar E, Cohen-Adiv S, Sweitat S, Kanner S, Galron R, Mitiagin Y, Barzilai A. Dysfunction of cerebellar microglia in Ataxia-telangiectasia. Glia 2021; 70:536-557. [PMID: 34854502 DOI: 10.1002/glia.24122] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 12/13/2022]
Abstract
Ataxia-telangiectasia (A-T) is a multisystem autosomal recessive disease caused by mutations in the ATM gene and characterized by cerebellar atrophy, progressive ataxia, immunodeficiency, male and female sterility, radiosensitivity, cancer predisposition, growth retardation, insulin-resistant diabetes, and premature aging. ATM phosphorylates more than 1500 target proteins, which are involved in cell cycle control, DNA repair, apoptosis, modulation of chromatin structure, and other cytoplasmic as well as mitochondrial processes. In our quest to better understand the mechanisms by which ATM deficiency causes cerebellar degeneration, we hypothesized that specific vulnerabilities of cerebellar microglia underlie the etiology of A-T. Our hypothesis is based on the recent finding that dysfunction of glial cells affect a variety of process leading to impaired neuronal functionality (Song et al., 2019). Whereas astrocytes and neurons descend from the neural tube, microglia originate from the hematopoietic system, invade the brain at early embryonic stage, and become the innate immune cells of the central nervous system and important participants in development of synaptic plasticity. Here we demonstrate that microglia derived from Atm-/- mouse cerebellum display accelerated cell migration and are severely impaired in phagocytosis, secretion of neurotrophic factors, and mitochondrial activity, suggestive of apoptotic processes. Interestingly, no microglial impairment was detected in Atm-deficient cerebral cortex, and Atm deficiency had less impact on astroglia than microglia. Collectively, our findings validate the roles of glial cells in cerebellar attrition in A-T.
Collapse
Affiliation(s)
- Hadar Levi
- Department of Neurobiology, George S. Wise, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ela Bar
- Department of Neurobiology, George S. Wise, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Stav Cohen-Adiv
- Department of Neurobiology, George S. Wise, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Suzan Sweitat
- Department of Neurobiology, George S. Wise, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Sivan Kanner
- Department of Neurobiology, George S. Wise, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ronit Galron
- Department of Neurobiology, George S. Wise, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yulia Mitiagin
- Department of Neurobiology, George S. Wise, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ari Barzilai
- Department of Neurobiology, George S. Wise, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
176
|
Mossad O, Nent E, Woltemate S, Folschweiller S, Buescher JM, Schnepf D, Erny D, Staeheli P, Bartos M, Szalay A, Stecher B, Vital M, Sauer JF, Lämmermann T, Prinz M, Blank T. Microbiota-dependent increase in δ-valerobetaine alters neuronal function and is responsible for age-related cognitive decline. NATURE AGING 2021; 1:1127-1136. [PMID: 37117525 DOI: 10.1038/s43587-021-00141-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 10/25/2021] [Indexed: 04/30/2023]
Abstract
Understanding the physiological origins of age-related cognitive decline is of critical importance given the rising age of the world's population1. Previous work in animal models has established a strong link between cognitive performance and the microbiota2-5, and it is known that the microbiome undergoes profound remodeling in older adults6. Despite growing evidence for the association between age-related cognitive decline and changes in the gut microbiome, the mechanisms underlying such interactions between the brain and the gut are poorly understood. Here, using fecal microbiota transplantation (FMT), we demonstrate that age-related remodeling of the gut microbiota leads to decline in cognitive function in mice and that this impairment can be rescued by transplantation of microbiota from young animals. Moreover, using a metabolomic approach, we found elevated concentrations of δ-valerobetaine, a gut microbiota-derived metabolite, in the blood and brain of aged mice and older adults. We then demonstrated that δ-valerobetaine is deleterious to learning and memory processes in mice. At the neuronal level, we showed that δ-valerobetaine modulates inhibitory synaptic transmission and neuronal network activity. Finally, we identified specific bacterial taxa that significantly correlate with δ-valerobetaine levels in the brain. Based on our findings, we propose that δ-valerobetaine contributes to microbiota-driven brain aging and that the associated mechanisms represent a promising target for countering age-related cognitive decline.
Collapse
Affiliation(s)
- Omar Mossad
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Elisa Nent
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Sabrina Woltemate
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Shani Folschweiller
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Institute of Physiology I, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Joerg M Buescher
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Daniel Schnepf
- Institute of Virology, Medical Center University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs University Freiburg, Freiburg, Germany
| | - Daniel Erny
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Berta-Ottenstein-Programme, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Peter Staeheli
- Institute of Virology, Medical Center University of Freiburg, Freiburg, Germany
- Berta-Ottenstein-Programme, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marlene Bartos
- Institute of Physiology I, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Bärbel Stecher
- Max-von-Pettenkofer Institute, LMU Munich, Munich, Germany
- German Center for Infection Research (DZIF), partner site LMU Munich, Munich, Germany
| | - Marius Vital
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Jonas F Sauer
- Institute of Physiology I, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tim Lämmermann
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Center for NeuroModulation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Thomas Blank
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
177
|
Abstract
Bio-memristors constitute candidates for the next generation of non-volatile storage and bionic synapses due to their biocompatibility, environmental benignity, sustainability, flexibility, degradability, and impressive memristive performance. Silk fibroin (SF), a natural and abundant biomaterial with excellent mechanical, optical, electrical, and structure-adjustable properties as well as being easy to process, has been utilized and shown to have potential in the construction of bio-memristors. Here, we first summarize the fundamental mechanisms of bio-memristors based on SF. Then, the latest achievements and developments of pristine and composited SF-based memristors are highlighted, followed by the integration of memristive devices. Finally, the challenges and insights associated with SF-based bio-memristors are presented. Advances in SF-based bio-memristors will open new avenues in the design and integration of high-performance bio-integrated systems and facilitate their application in logic operations, complex circuits, and neural networks.
Collapse
Affiliation(s)
- Yi Zhang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, P. R. China.
| | - Suna Fan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, P. R. China.
| | - Yaopeng Zhang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai 201620, P. R. China.
| |
Collapse
|
178
|
Doremus-Fitzwater TL, Deak T. Adolescent neuroimmune function and its interaction with alcohol. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 161:167-208. [PMID: 34801169 DOI: 10.1016/bs.irn.2021.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Adolescence is an evolutionarily conserved developmental period associated with behavioral change, including increased risk-taking and alcohol use. Experimentation with alcohol typically begins in adolescence and transitions to binge-like patterns of consumption. Alcohol exposure during adolescence can alter normative changes in brain structure and function. Understanding mechanisms by which ethanol impacts neurodevelopmental processes is important for preventing and ameliorating the deleterious consequences of adolescent alcohol abuse. This review focuses on the neuroimmune system as a key contributor to ethanol-induced changes in adolescent brain and behavior. After brief review of neuroimmune system development, acute and chronic effects of ethanol on adolescent neuroimmune functioning are addressed. Comparisons between stress/immunological challenges and ethanol on adolescent neuroimmunity are reviewed, as cross-sensitization is relevant during adolescence. The mechanisms by which ethanol alters neuroimmune functioning are then discussed, as they may portend development of neuropathological consequences and thus increase vulnerability to subsequent challenges and potentiate addictive behaviors.
Collapse
Affiliation(s)
- T L Doremus-Fitzwater
- Department of Psychology, Ithaca College, Ithaca, NY, United States; Developmental Exposure Alcohol Research Center (DEARC), Binghamton, NY, United States.
| | - T Deak
- Developmental Exposure Alcohol Research Center (DEARC), Binghamton, NY, United States; Binghamton University-SUNY, Binghamton, NY, United States
| |
Collapse
|
179
|
Rahman S, Rahman ZI, Ronan PJ, Lutfy K, Bell RL. Adolescent opioid abuse: Role of glial and neuroimmune mechanisms. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2021; 161:147-165. [PMID: 34801168 DOI: 10.1016/bs.irn.2021.07.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Opioids are widely prescribed for pain management, and prescription opioid misuse in adolescents has become a major epidemic in the United States and worldwide. Emerging data indicate that adolescence represents a critical period of brain development, and exposure to opioids during adolescence may increase the risk of addiction in adulthood. There is growing evidence that disruptions in brain glial function may be implicated in numerous chronic neuropathologies. Evidence suggests that glial mechanisms have an important role in the development and maintenance of opioid abuse and the risk for addiction. This review will describe glial and neuroimmune mechanisms involved in opioid use disorders during adolescence, which may increase substance use disorder liability later in life. Moreover, this review will identify some important neuro-glial targets, involved in opioid abuse and addiction, to develop future preventions and treatment strategies.
Collapse
Affiliation(s)
- S Rahman
- Department of Pharmaceutical Sciences, South Dakota State University, Brookings, SD, United States.
| | - Z I Rahman
- University of Minnesota Medical School, Minneapolis, MN, United States
| | - P J Ronan
- Department of Psychiatry and Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Sioux Falls, SD, United States; Research Service, Sioux Falls VA Healthcare System, Sioux Falls, SD, United States
| | - K Lutfy
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| | - R L Bell
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
180
|
Yan Y, Aierken A, Wang C, Song D, Ni J, Wang Z, Quan Z, Qing H. A potential biomarker of preclinical Alzheimer's disease: The olfactory dysfunction and its pathogenesis-based neural circuitry impairments. Neurosci Biobehav Rev 2021; 132:857-869. [PMID: 34810025 DOI: 10.1016/j.neubiorev.2021.11.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/26/2021] [Accepted: 11/07/2021] [Indexed: 01/24/2023]
Abstract
The olfactory dysfunction can signal and act as a potential biomarker of preclinical AD. However, the precise regulatory mechanism of olfactory function on the neural pathogenesis of AD is still unclear. The impairment of neural networks in olfaction system has been shown to be tightly associated with AD. As key brain regions of the olfactory system, the olfactory bulb (OB) and the piriform cortex (PCx) have a profound influence on the olfactory function. Therefore, this review will explore the mechanism of olfactory dysfunction in preclinical AD in the perspective of abnormal neural networks in the OB and PCx and their associated brain regions, especially from two aspects of aberrant oscillations and synaptic plasticity damages, which help better understand the underlying mechanism of olfactory neural network damages related to AD.
Collapse
Affiliation(s)
- Yan Yan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Ailikemu Aierken
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Chunjian Wang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Da Song
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Zhe Wang
- Advanced Innovation Center for Human Brain Protection, Capital Medical University, The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhenzhen Quan
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
| |
Collapse
|
181
|
Parrott JM, Oster T, Lee HY. Altered inflammatory response in FMRP-deficient microglia. iScience 2021; 24:103293. [PMID: 34820601 PMCID: PMC8602000 DOI: 10.1016/j.isci.2021.103293] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 02/08/2021] [Accepted: 10/14/2021] [Indexed: 02/04/2023] Open
Abstract
Fragile X syndrome (FXS) is an inherited intellectual disability with a high risk for comorbid autism spectrum disorders. Since FXS is a genetic disease, patients are more susceptible to environmental factors aggravating symptomatology. However, this confounding interaction between FXS environmental and genetic risk factors is under-investigated. Here, Fmr1 knock-out (KO) mice and the immune stimulus lipopolysaccharide (LPS) were used to explore this interaction between FXS development and inflammation in microglia, the brain’s primary immune cell. Our results demonstrate that Fmr1 KO and wild-type (WT) microglia are not different in inflammatory outcomes without LPS. However, Fmr1 KO microglia produces an elevated pro-inflammatory and phagocytic response following LPS treatment when compared to WT microglia. Our experiments also revealed baseline differences in mitochondrial function and morphology between WT and Fmr1 KO microglia, which LPS treatment exaggerated. Our data suggest an altered inflammatory mechanism in Fmr1 KO microglia implicating a gene and environment interaction. Fmr1 KO microglia display elevated LPS-induced pro-inflammatory gene expressions Fmr1 KO microglia display elevated LPS-induced pro-inflammatory cytokine releases Fmr1 KO microglia demonstrate increased LPS-induced phagocytic responses Fmr1 KO microglial mitochondria have altered properties and LPS-stimulated responses
Collapse
Affiliation(s)
- Jennifer M Parrott
- The Department of Cellular and Integrative Physiology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Thomas Oster
- The Department of Cellular and Integrative Physiology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Hye Young Lee
- The Department of Cellular and Integrative Physiology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
182
|
Lipopolysaccharide-induced neuroinflammation disrupts functional connectivity and community structure in primary cortical microtissues. Sci Rep 2021; 11:22303. [PMID: 34785714 PMCID: PMC8595892 DOI: 10.1038/s41598-021-01616-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 10/29/2021] [Indexed: 12/15/2022] Open
Abstract
Three-dimensional (3D) neural microtissues are a powerful in vitro paradigm for studying brain development and disease under controlled conditions, while maintaining many key attributes of the in vivo environment. Here, we used primary cortical microtissues to study the effects of neuroinflammation on neural microcircuits. We demonstrated the use of a genetically encoded calcium indicator combined with a novel live-imaging platform to record spontaneous calcium transients in microtissues from day 14-34 in vitro. We implemented graph theory analysis of calcium activity to characterize underlying functional connectivity and community structure of microcircuits, which are capable of capturing subtle changes in network dynamics during early disease states. We found that microtissues cultured for 34 days displayed functional remodeling of microcircuits and that community structure strengthened over time. Lipopolysaccharide, a neuroinflammatory agent, significantly increased functional connectivity and disrupted community structure 5-9 days after exposure. These microcircuit-level changes have broad implications for the role of neuroinflammation in functional dysregulation of neural networks.
Collapse
|
183
|
Zhang L, Wei W, Ai X, Kilic E, Hermann DM, Venkataramani V, Bähr M, Doeppner TR. Extracellular vesicles from hypoxia-preconditioned microglia promote angiogenesis and repress apoptosis in stroke mice via the TGF-β/Smad2/3 pathway. Cell Death Dis 2021; 12:1068. [PMID: 34753919 PMCID: PMC8578653 DOI: 10.1038/s41419-021-04363-7] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 09/15/2021] [Accepted: 10/20/2021] [Indexed: 12/17/2022]
Abstract
Systemic transplantation of oxygen-glucose deprivation (OGD)-preconditioned primary microglia enhances neurological recovery in rodent stroke models, albeit the underlying mechanisms have not been sufficiently addressed. Herein, we analyzed whether or not extracellular vesicles (EVs) derived from such microglia are the biological mediators of these observations and which signaling pathways are involved in the process. Exposing bEnd.3 endothelial cells (ECs) and primary cortical neurons to OGD, the impact of EVs from OGD-preconditioned microglia on angiogenesis and neuronal apoptosis by the tube formation assay and TUNEL staining was assessed. Under these conditions, EV treatment stimulated both angiogenesis and tube formation in ECs and repressed neuronal cell injury. Characterizing microglia EVs by means of Western blot analysis and other techniques revealed these EVs to be rich in TGF-β1. The latter turned out to be a key compound for the therapeutic potential of microglia EVs, affecting the Smad2/3 pathway in both ECs and neurons. EV infusion in stroke mice confirmed the aforementioned in vitro results, demonstrating an activation of the TGF-β/Smad2/3 signaling pathway within the ischemic brain. Furthermore, enriched TGF-β1 in EVs secreted from OGD-preconditioned microglia stimulated M2 polarization of residing microglia within the ischemic cerebral environment, which may contribute to a regulation of an early inflammatory response in postischemic hemispheres. These observations are not only interesting from the mechanistic point of view but have an immediate therapeutic implication as well, since stroke mice treated with such EVs displayed a better functional recovery in the behavioral test analyses. Hence, the present findings suggest a new way of action of EVs derived from OGD-preconditioned microglia by regulating the TGF-β/Smad2/3 pathway in order to promote tissue regeneration and neurological recovery in stroke mice.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Wei Wei
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Xiaoyu Ai
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Ertugrul Kilic
- Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Istanbul, Turkey
| | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Vivek Venkataramani
- Department of Medicine II, University Hospital Frankfurt, Frankfurt, Germany
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Mathias Bähr
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Thorsten R Doeppner
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany.
- Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Istanbul, Turkey.
| |
Collapse
|
184
|
Wang XL, Li L. Microglia Regulate Neuronal Circuits in Homeostatic and High-Fat Diet-Induced Inflammatory Conditions. Front Cell Neurosci 2021; 15:722028. [PMID: 34720877 PMCID: PMC8549960 DOI: 10.3389/fncel.2021.722028] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
Microglia are brain resident macrophages, which actively survey the surrounding microenvironment and promote tissue homeostasis under physiological conditions. During this process, microglia participate in synaptic remodeling, neurogenesis, elimination of unwanted neurons and cellular debris. The complex interplay between microglia and neurons drives the formation of functional neuronal connections and maintains an optimal neural network. However, activation of microglia induced by chronic inflammation increases synaptic phagocytosis and leads to neuronal impairment or death. Microglial dysfunction is implicated in almost all brain diseases and leads to long-lasting functional deficiency, such as hippocampus-related cognitive decline and hypothalamus-associated energy imbalance (i.e., obesity). High-fat diet (HFD) consumption triggers mediobasal hypothalamic microglial activation and inflammation. Moreover, HFD-induced inflammation results in cognitive deficits by triggering hippocampal microglial activation. Here, we have summarized the current knowledge of microglial characteristics and biological functions and also reviewed the molecular mechanism of microglia in shaping neural circuitries mainly related to cognition and energy balance in homeostatic and diet-induced inflammatory conditions.
Collapse
Affiliation(s)
- Xiao-Lan Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lianjian Li
- Department of Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China.,Hubei Province Academy of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
185
|
Scabia G, Testa G, Scali M, Del Turco S, Desiato G, Berardi N, Sale A, Matteoli M, Maffei L, Maffei M, Mainardi M. Reduced ccl11/eotaxin mediates the beneficial effects of environmental stimulation on the aged hippocampus. Brain Behav Immun 2021; 98:234-244. [PMID: 34418501 DOI: 10.1016/j.bbi.2021.08.222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 07/15/2021] [Accepted: 08/13/2021] [Indexed: 02/08/2023] Open
Abstract
A deterioration in cognitive performance accompanies brain aging, even in the absence of neurodegenerative pathologies. However, the rate of cognitive decline can be slowed down by enhanced cognitive and sensorimotor stimulation protocols, such as environmental enrichment (EE). Understanding how EE exerts its beneficial effects on the aged brain pathophysiology can help in identifying new therapeutic targets. In this regard, the inflammatory chemokine ccl11/eotaxin-1 is a marker of aging with a strong relevance for neurodegenerative processes. Here, we demonstrate that EE in both elderly humans and aged mice decreases circulating levels of ccl11. Interfering, in mice, with the ccl11 decrease induced by EE ablated the beneficial effects on long-term memory retention, hippocampal neurogenesis, activation of local microglia and of ribosomal protein S6. On the other hand, treatment of standard-reared aged mice with an anti-ccl11 antibody resulted in EE-like improvements in spatial memory, hippocampal neurogenesis, and microglial activation. Taken together, our findings point to a decrease in circulating ccl11 concentration as a key mediator of the enhanced hippocampal function resulting from exposure to EE.
Collapse
Affiliation(s)
- Gaia Scabia
- Institute of Clinical Physiology, National Research Council (IFC-CNR), Pisa, Italy; Obesity and Lipodystrophies Center at Pisa University Hospital, Pisa, Italy
| | - Giovanna Testa
- Laboratory of Biology "Bio@SNS", Scuola Normale Superiore, Pisa, Italy
| | - Manuela Scali
- Institute of Neuroscience, National Research Council (IN-CNR), Pisa, Italy
| | - Serena Del Turco
- Institute of Clinical Physiology, National Research Council (IFC-CNR), Pisa, Italy
| | - Genni Desiato
- Institute of Neuroscience, National Research Council (IN-CNR), Milan, Italy; Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| | - Nicoletta Berardi
- Institute of Neuroscience, National Research Council (IN-CNR), Pisa, Italy; Department of Neuroscience, Psychology, Drug Research and Child Health, NEUROFARBA University of Florence, Florence, Italy
| | - Alessandro Sale
- Institute of Neuroscience, National Research Council (IN-CNR), Pisa, Italy
| | - Michela Matteoli
- Institute of Neuroscience, National Research Council (IN-CNR), Milan, Italy; Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| | - Lamberto Maffei
- Laboratory of Biology "Bio@SNS", Scuola Normale Superiore, Pisa, Italy; Institute of Neuroscience, National Research Council (IN-CNR), Pisa, Italy
| | - Margherita Maffei
- Institute of Clinical Physiology, National Research Council (IFC-CNR), Pisa, Italy; Obesity and Lipodystrophies Center at Pisa University Hospital, Pisa, Italy.
| | - Marco Mainardi
- Laboratory of Biology "Bio@SNS", Scuola Normale Superiore, Pisa, Italy; Institute of Neuroscience, National Research Council (IN-CNR), Pisa, Italy.
| | | |
Collapse
|
186
|
Shao Y, Chen C, Zhu T, Sun Z, Li S, Gong L, Dong X, Shen W, Zeng L, Xie Y, Jiang P. TRPM2 contributes to neuroinflammation and cognitive deficits in a cuprizone-induced multiple sclerosis model via NLRP3 inflammasome. Neurobiol Dis 2021; 160:105534. [PMID: 34673151 DOI: 10.1016/j.nbd.2021.105534] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/23/2021] [Accepted: 10/17/2021] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) is a disease of the central nervous system (CNS) that is characterized by demyelination, axonal injury and neurological deterioration. Few medications are available for progressive MS, which is associated with neuroinflammation confined to the CNS compartment. Transient receptor potential melastatin 2 (TRPM2) is a calcium-permeable, non-selective cation channel that plays pathological roles in a wide range of neuroinflammatory diseases; however, the underlying molecular mechanisms of TRPM2 remain elusive. Here, we established a cuprizone model that presents hallmark MS pathologies to investigate the role of TRPM2 in progressive MS. We demonstrated that genetic deletion of TRPM2 yields protection from the cuprizone-induced demyelination, synapse loss, microglial activation, NLRP3 inflammasome activation and proinflammatory cytokines production and ultimately leads to an improvement in cognitive decline. Furthermore, we showed that the pharmacological inhibition of NLRP3 ameliorated the demyelination, neuroinflammation and cognitive impairment in the model with no additive effects on the TRPM2 KO mice. Taken together, these results indicated that TRPM2 plays important roles in regulating neuroinflammation in progressive MS via NLRP3 inflammasome, and the results shed light on TRPM2's potential role as a therapeutic target for MS.
Collapse
Affiliation(s)
- Yu Shao
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China; Department of Pediatrics, Wenling First People's Hospital, Wenling 317500, China
| | - Chen Chen
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China
| | - Tao Zhu
- Department of Critical Care Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310052, China
| | - Zengxian Sun
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China; Department of Pediatrics, Lishui Central Hospital, Lishui 323000, China
| | - Shufen Li
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China; Department of Pediatrics, Lishui Central Hospital, Lishui 323000, China
| | - Lifen Gong
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China
| | - Xinyan Dong
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China
| | - Weida Shen
- Department of Pharmacy, Zhejiang University City College School of Medicine, Hangzhou 310015, China
| | - Linghui Zeng
- Department of Pharmacy, Zhejiang University City College School of Medicine, Hangzhou 310015, China
| | - Yicheng Xie
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China.
| | - Peifang Jiang
- Department of Neurology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child Health, Hangzhou 310052, China.
| |
Collapse
|
187
|
Silva NJ, Dorman LC, Vainchtein ID, Horneck NC, Molofsky AV. In situ and transcriptomic identification of microglia in synapse-rich regions of the developing zebrafish brain. Nat Commun 2021; 12:5916. [PMID: 34625548 PMCID: PMC8501082 DOI: 10.1038/s41467-021-26206-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 09/16/2021] [Indexed: 01/02/2023] Open
Abstract
Microglia are brain resident macrophages that play vital roles in central nervous system (CNS) development, homeostasis, and pathology. Microglia both remodel synapses and engulf apoptotic cell corpses during development, but whether unique molecular programs regulate these distinct phagocytic functions is unknown. Here we identify a molecularly distinct microglial subset in the synapse rich regions of the zebrafish (Danio rerio) brain. We found that ramified microglia increased in synaptic regions of the midbrain and hindbrain between 7 and 28 days post fertilization. In contrast, microglia in the optic tectum were ameboid and clustered around neurogenic zones. Using single-cell mRNA sequencing combined with metadata from regional bulk sequencing, we identified synaptic-region associated microglia (SAMs) that were highly enriched in the hindbrain and expressed multiple candidate synapse modulating genes, including genes in the complement pathway. In contrast, neurogenic associated microglia (NAMs) were enriched in the optic tectum, had active cathepsin activity, and preferentially engulfed neuronal corpses. These data reveal that molecularly distinct phagocytic programs mediate synaptic remodeling and cell engulfment, and establish the zebrafish hindbrain as a model for investigating microglial-synapse interactions. Microglia remodel synapses and engulf apoptotic cells. The molecular program underlying these distinct functions are unclear. Here, the authors identify distinct microglial subsets associated with synaptic vs. neurogenic regions of the developing zebrafish brain.
Collapse
Affiliation(s)
- Nicholas J Silva
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Leah C Dorman
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.,Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Ilia D Vainchtein
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Nadine C Horneck
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Anna V Molofsky
- Department of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA. .,Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
188
|
Sapio MR, Kim JJ, Loydpierson AJ, Maric D, Goto T, Vazquez FA, Dougherty MK, Narasimhan R, Muhly WT, Iadarola MJ, Mannes AJ. The Persistent Pain Transcriptome: Identification of Cells and Molecules Activated by Hyperalgesia. THE JOURNAL OF PAIN 2021; 22:1146-1179. [PMID: 33892151 PMCID: PMC9441406 DOI: 10.1016/j.jpain.2021.03.155] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 12/21/2022]
Abstract
During persistent pain, the dorsal spinal cord responds to painful inputs from the site of injury, but the molecular modulatory processes have not been comprehensively examined. Using transcriptomics and multiplex in situ hybridization, we identified the most highly regulated receptors and signaling molecules in rat dorsal spinal cord in peripheral inflammatory and post-surgical incisional pain models. We examined a time course of the response including acute (2 hours) and longer term (2 day) time points after peripheral injury representing the early onset and instantiation of hyperalgesic processes. From this analysis, we identify a key population of superficial dorsal spinal cord neurons marked by somatotopic upregulation of the opioid neuropeptide precursor prodynorphin, and 2 receptors: the neurokinin 1 receptor, and anaplastic lymphoma kinase. These alterations occur specifically in the glutamatergic subpopulation of superficial dynorphinergic neurons. In addition to specific neuronal gene regulation, both models showed induction of broad transcriptional signatures for tissue remodeling, synaptic rearrangement, and immune signaling defined by complement and interferon induction. These signatures were predominantly induced ipsilateral to tissue injury, implying linkage to primary afferent drive. We present a comprehensive set of gene regulatory events across 2 models that can be targeted for the development of non-opioid analgesics. PERSPECTIVE: The deadly impact of the opioid crisis and the need to replace morphine and other opioids in clinical practice is well recognized. Embedded within this research is an overarching goal of obtaining foundational knowledge from transcriptomics to search for non-opioid analgesic targets. Developing such analgesics would address unmet clinical needs.
Collapse
Affiliation(s)
- Matthew R Sapio
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, NIH, Bethesda, Maryland
| | - Jenny J Kim
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, NIH, Bethesda, Maryland
| | - Amelia J Loydpierson
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, NIH, Bethesda, Maryland
| | - Dragan Maric
- National Institute of Neurological Disorders and Stroke, Flow and Imaging Cytometry Core Facility, NIH, Bethesda, Maryland
| | - Taichi Goto
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, NIH, Bethesda, Maryland; National Institute of Nursing Research, Symptom Management Branch, NIH, Bethesda, Maryland; Japan Society for the Promotion of Science Overseas Research Fellowship, Tokyo, Japan
| | - Fernando A Vazquez
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, NIH, Bethesda, Maryland
| | - Mary K Dougherty
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, NIH, Bethesda, Maryland
| | - Radhika Narasimhan
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, NIH, Bethesda, Maryland
| | - Wallis T Muhly
- National Institute of Nursing Research, Symptom Management Branch, NIH, Bethesda, Maryland; Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael J Iadarola
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, NIH, Bethesda, Maryland.
| | - Andrew J Mannes
- Department of Perioperative Medicine, National Institutes of Health Clinical Center, NIH, Bethesda, Maryland
| |
Collapse
|
189
|
Melbourne JK, Chandler CM, Van Doorn CE, Bardo MT, Pauly JR, Peng H, Nixon K. Primed for addiction: A critical review of the role of microglia in the neurodevelopmental consequences of adolescent alcohol drinking. Alcohol Clin Exp Res 2021; 45:1908-1926. [PMID: 34486128 PMCID: PMC8793635 DOI: 10.1111/acer.14694] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 07/22/2021] [Accepted: 08/03/2021] [Indexed: 12/15/2022]
Abstract
Alcohol is one of the most widely used recreational substances worldwide, with drinking frequently initiated during adolescence. The developmental state of the adolescent brain makes it vulnerable to initiating alcohol use, often in high doses, and particularly susceptible to alcohol-induced brain changes. Microglia, the brain parenchymal macrophages, have been implicated in mediating some of these effects, though the role that these cells play in the progression from alcohol drinking to dependence remains unclear. Microglia are uniquely positioned to sense and respond to central nervous system insult, and are now understood to exhibit innate immune memory, or "priming," altering their future functional responses based on prior exposures. In alcohol use disorders (AUDs), the role of microglia is debated. Whereas microglial activation can be pathogenic, contributing to neuroinflammation, tissue damage, and behavioral changes, or protective, it can also engage protective functions, providing support and mediating the resolution of damage. Understanding the role of microglia in adolescent AUDs is complicated by the fact that microglia are thought to be involved in developmental processes such as synaptic refinement and myelination, which underlie the functional maturation of multiple brain systems in adolescence. Thus, the role microglia play in the impact of alcohol use in adolescence is likely multifaceted. Long-term sequelae may be due to a failure to recover from EtOH-induced tissue damage, altered neurodevelopmental trajectories, and/or persistent changes to microglial responsivity and function. Here, we review critically the literature surrounding the effects of alcohol on microglia in models of adolescent alcohol misuse. We attempt to disentangle what is known about microglia from other neuroimmune effectors, to which we apply recent discoveries on the role of microglia in development and plasticity. Considered altogether, these studies challenge assumptions that proinflammatory microglia drive addiction. Alcohol priming microglia and thereby perturbing their homeostatic roles in neurodevelopment, especially during critical periods of plasticity such as adolescence, may have more serious implications for the neuropathogenesis of AUDs in adolescents.
Collapse
Affiliation(s)
- Jennifer K. Melbourne
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas, USA
| | - Cassie M. Chandler
- Department of Psychology, University of Kentucky, Lexington, Kentucky, USA
| | | | - Michael T. Bardo
- Department of Psychology, University of Kentucky, Lexington, Kentucky, USA
| | - James R. Pauly
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Hui Peng
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Kimberly Nixon
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
190
|
Song P, Yi Z, Fu Y, Song D, Chen K, Zheng J, Sun Y, Diao Y. Reversing Postcardiopulmonary Bypass Associated Cognitive Dysfunction Using k-Opioid Receptor Agonists to Regulate Microglial Polarization via the NLRP3/Caspase-1 Pathway. JOURNAL OF HEALTHCARE ENGINEERING 2021; 2021:3048383. [PMID: 34630980 PMCID: PMC8500742 DOI: 10.1155/2021/3048383] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/14/2021] [Indexed: 12/21/2022]
Abstract
Cardiopulmonary bypass (CPB) is mainly used during cardiac surgeries that treat ischemic, valvular, or congenital heart disease and aortic dissections. The disorders of central nervous system (CNS) that occur after cardiopulmonary bypass are attracting considerable interest. Postoperative neurocognitive disorders (PND) have been reported as the leading cause of patients' disability and death following CPB. The k-opioid receptor (KOR) agonists (U50488H) have been suggested to be vital in the treatment of surgically induced CNS neuroinflammatory responses. In this article, the transitions between the M1 and M2 microglial polarization state phenotypes were hypothesized to significantly affect the regulatory mechanisms of KOR agonists on postcardiopulmonary bypass (post-CPB) neuroinflammation. We investigated the effects of U50488H on neuroinflammation and microglia polarization in rats exposed to CPB and explored the method of the NLRP3/caspase-1 pathway. Thirty SD rats were randomly divided into three groups: sham operation group, cardiopulmonary bypass model group, and CPB+ k-opioid receptor agonist (U50488H) group, with ten rats in each group. The Morris water maze was used to evaluate the changes in the cognitive function of CPB rats. Hematoxylin and eosin (HE) staining and TUNEL were performed to assess the rats' hippocampal damage. Enzyme-Linked Immunosorbent Assay (ELISA) was used to detect changes in brain injury markers and inflammatory factors. Furthermore, immunofluorescence was used to observe the expression of microglia polarization and NLRP3 followed by Western blots to detect the expression of the NLRP3/caspase-1 pathway and microglia polarization-related proteins. Rat microglia were cultured in vitro, with LPS stimulation, and treated with U50488H and a caspase-1 antagonist to evaluate the effects and mechanism of action of U50488H. KORs alleviated hippocampal damage caused by CPB and improved PND. CPB activated the NLRP3 inflammasome and upregulated pro-caspase-1 expression which promoted the expression of pro-IL-lβ and pro-IL-18 and resulted in increased inflammation. However, KORs also inhibited NLRP3 and transformed microglia from the M1 to the M2 state. Caspase-1 inhibitor treatment reduced the microglial polarization induced by KORs. The κ-opioid receptor agonists inhibited the inflammation mediated by microglia and improved PND through the NLRP3/caspase-1 signaling pathway.
Collapse
Affiliation(s)
- Pei Song
- Department of Anesthesia, Postgraduate Training Base of Jinzhou Medical University in the General Hospital of Northern Theater Command, Shenyang 110016, China
- Department of Anesthesia, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning, China
| | - Zhuo Yi
- Department of Anesthesia, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning, China
| | - Yiji Fu
- Department of Anesthesiology, Anshan Central Hospital, Anshan 114002, Liaoning, China
| | - Dandan Song
- Department of Anesthesia, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning, China
| | - Keyan Chen
- Department of Laboratory Animal Science, China Medical University, Shenyang 110000, Liaoning, China
| | - Jingjing Zheng
- Department of Anesthesia, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning, China
| | - Yingjie Sun
- Department of Anesthesia, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning, China
| | - Yugang Diao
- Department of Anesthesia, General Hospital of Northern Theater Command, Shenyang 110016, Liaoning, China
| |
Collapse
|
191
|
Augusto-Oliveira M, Arrifano GP, Delage CI, Tremblay MÈ, Crespo-Lopez ME, Verkhratsky A. Plasticity of microglia. Biol Rev Camb Philos Soc 2021; 97:217-250. [PMID: 34549510 DOI: 10.1111/brv.12797] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 09/02/2021] [Accepted: 09/06/2021] [Indexed: 02/06/2023]
Abstract
Microglial cells are the scions of foetal macrophages which invade the neural tube early during embryogenesis. The nervous tissue environment instigates the phenotypic metamorphosis of foetal macrophages into idiosyncratic surveilling microglia, which are generally characterised by a small cell body and highly ramified motile processes that constantly scan the nervous tissue for signs of changes in homeostasis and allow microglia to perform crucial homeostatic functions. The surveilling microglial phenotype is evolutionarily conserved from early invertebrates to humans. Despite this evolutionary conservation, microglia show substantial heterogeneity in their gene and protein expression, as well as morphological appearance. These differences are age, region and context specific and reflect a high degree of plasticity underlying the life-long adaptation of microglia, supporting the exceptional adaptive capacity of the central nervous system. Microgliocytes are essential elements of cellular network formation and refinement in the developing nervous tissue. Several distinct patrolling modes of microglial processes contribute to the formation, modification, and pruning of synapses; to the support and protection of neurones through microglial-somatic junctions; and to the control of neuronal and axonal excitability by specific microglia-axonal contacts. In pathology, microglia undergo proliferation and reactive remodelling known as microgliosis, which is context dependent, yet represents an evolutionarily conserved defence response. Microgliosis results in the emergence of multiple disease and context-specific reactive states; in addition, neuropathology is associated with the appearance of specific protective or recovery microglial forms. In summary, the plasticity of microglia supports the development and functional activity of healthy nervous tissue and provides highly sophisticated defences against disease.
Collapse
Affiliation(s)
- Marcus Augusto-Oliveira
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, 66075-110, Belém, Brazil
| | - Gabriela P Arrifano
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, 66075-110, Belém, Brazil
| | - Charlotte Isabelle Delage
- Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, V8P 5C2, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, V8P 5C2, Canada.,Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, 2705 Boulevard Laurier, Québec City, QC, G1V 4G2, Canada.,Neurology and Neurosurgery Department, McGill University, 3801 University Street, Montreal, QC, H3A 2B4, Canada.,Department of Molecular Medicine, Université Laval, Pavillon Ferdinand-Vandry, Bureau 4835, 1050 Avenue de la Médecine, Québec City, QC, G1V 0A6, Canada.,Department of Biochemistry and Molecular Biology, The University of British Columbia, Life Sciences Center, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Maria Elena Crespo-Lopez
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, 66075-110, Belém, Brazil
| | - Alexei Verkhratsky
- Faculty of Life Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PT, U.K.,Achucarro Center for Neuroscience, IKERBASQUE, 48011, Bilbao, Spain.,Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102, Vilnius, Lithuania
| |
Collapse
|
192
|
Alshelh Z, Brusaferri L, Saha A, Morrissey E, Knight P, Kim M, Zhang Y, Hooker JM, Albrecht D, Torrado-Carvajal A, Placzek MS, Akeju O, Price J, Edwards RR, Lee J, Sclocco R, Catana C, Napadow V, Loggia ML. Neuro-immune signatures in chronic low back pain subtypes. Brain 2021; 145:1098-1110. [PMID: 34528069 DOI: 10.1093/brain/awab336] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/11/2021] [Accepted: 08/24/2021] [Indexed: 11/14/2022] Open
Abstract
We recently showed that patients with different chronic pain conditions (such as chronic low back pain, fibromyalgia, migraine, and Gulf War Illness) demonstrated elevated brain and/or spinal cord levels of the glial marker 18 kDa translocator protein, which suggests that neuroinflammation might be a pervasive phenomenon observable across multiple etiologically heterogeneous pain disorders. Interestingly, the spatial distribution of this neuroinflammatory signal appears to exhibit a degree of disease specificity (e.g. with respect to the involvement of the primary somatosensory cortex), suggesting that different pain conditions may exhibit distinct "neuroinflammatory signatures". To further explore this hypothesis, we tested whether neuroinflammatory signal can characterize putative etiological subtypes of chronic low back pain patients based on clinical presentation. Specifically, we explored neuroinflammation in patients whose chronic low back pain either did or did not radiate to the leg (i.e. "radicular" vs. "axial" back pain). Fifty-four chronic low back pain patients, twenty-six with axial back pain (43.7 ± 16.6 y.o. [mean±SD]) and twenty-eight with radicular back pain (48.3 ± 13.2 y.o.), underwent PET/MRI with [11C]PBR28, a second-generation radioligand for the 18 kDa translocator protein. [11C]PBR28 signal was quantified using standardized uptake values ratio (validated against volume of distribution ratio; n = 23). Functional MRI data were collected simultaneously to the [11C]PBR28 data 1) to functionally localize the primary somatosensory cortex back and leg subregions and 2) to perform functional connectivity analyses (in order to investigate possible neurophysiological correlations of the neuroinflammatory signal). PET and functional MRI measures were compared across groups, cross-correlated with one another and with the severity of "fibromyalgianess" (i.e. the degree of pain centralization, or "nociplastic pain"). Furthermore, statistical mediation models were employed to explore possible causal relationships between these three variables. For the primary somatosensory cortex representation of back/leg, [11C]PBR28 PET signal and functional connectivity to the thalamus were: 1) higher in radicular compared to axial back pain patients, 2) positively correlated with each other and 3) positively correlated with fibromyalgianess scores, across groups. Finally, 4) fibromyalgianess mediated the association between [11C]PBR28 PET signal and primary somatosensory cortex-thalamus connectivity across groups. Our findings support the existence of "neuroinflammatory signatures" that are accompanied by neurophysiological changes, and correlate with clinical presentation (in particular, with the degree of nociplastic pain) in chronic pain patients. These signatures may contribute to the subtyping of distinct pain syndromes and also provide information about inter-individual variability in neuro-immune brain signals, within diagnostic groups, that could eventually serve as targets for mechanism-based precision medicine approaches.
Collapse
Affiliation(s)
- Zeynab Alshelh
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Ludovica Brusaferri
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Atreyi Saha
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Erin Morrissey
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Paulina Knight
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Minhae Kim
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Yi Zhang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jacob M Hooker
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Daniel Albrecht
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Angel Torrado-Carvajal
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.,Medical Image Analysis and Biometry Laboratory, Universidad Rey Juan Carlos, Madrid, Spain
| | - Michael S Placzek
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Oluwaseun Akeju
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Julie Price
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Robert R Edwards
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jeungchan Lee
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Roberta Sclocco
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.,Department of Radiology, Logan University, Chesterfield, MO, USA
| | - Ciprian Catana
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Vitaly Napadow
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.,Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Marco L Loggia
- Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
193
|
Palomino-Segura M, Hidalgo A. Circadian immune circuits. J Exp Med 2021; 218:211639. [PMID: 33372990 PMCID: PMC7774593 DOI: 10.1084/jem.20200798] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 12/30/2022] Open
Abstract
Immune responses are gated to protect the host against specific antigens and microbes, a task that is achieved through antigen- and pattern-specific receptors. Less appreciated is that in order to optimize responses and to avoid collateral damage to the host, immune responses must be additionally gated in intensity and time. An evolutionary solution to this challenge is provided by the circadian clock, an ancient time-keeping mechanism that anticipates environmental changes and represents a fundamental property of immunity. Immune responses, however, are not exclusive to immune cells and demand the coordinated action of nonhematopoietic cells interspersed within the architecture of tissues. Here, we review the circadian features of innate immunity as they encompass effector immune cells as well as structural cells that orchestrate their responses in space and time. We finally propose models in which the central clock, structural elements, and immune cells establish multidirectional circadian circuits that may shape the efficacy and strength of immune responses and other physiological processes.
Collapse
Affiliation(s)
- Miguel Palomino-Segura
- Area of Cell and Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Andrés Hidalgo
- Area of Cell and Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| |
Collapse
|
194
|
Li J, Zeng Q, Su W, Song M, Xie M, Mao L. FBXO10 prevents chronic unpredictable stress-induced behavioral despair and cognitive impairment through promoting RAGE degradation. CNS Neurosci Ther 2021; 27:1504-1517. [PMID: 34492157 PMCID: PMC8611766 DOI: 10.1111/cns.13727] [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: 04/17/2021] [Revised: 08/13/2021] [Accepted: 08/27/2021] [Indexed: 12/15/2022] Open
Abstract
Aims Depression is one of the leading causes of disability worldwide. The receptor for advanced glycosylation end products (RAGE) is closely related to chronic stress and is a target of F‐box protein O10 (FBXO10) which promotes the degradation of RAGE by ubiquitination. Here, we explored the role of FBXO10 and RAGE in chronic unpredictable stress (CUS)‐induced behavioral despair, cognitive impairment, neuroinflammation, and the polarization microglia. Methods Male C57BL/6 mice with or without infusion of viral in the medial prefrontal cortex (PFC) were subjected to CUS. Then the mice were exposed to forced swim test, sucrose consumption test, novelty‐suppressed feeding test, and temporal object recognition task to assess the behavioral despair and cognitive impairment. Inflammatory cytokines and the neurotrophic factor brain‐derived neurotrophic factor (BDNF) levels in PFC were assessed by enzyme‐linked immunosorbent assay. Immunofluorescence and immunohistochemistry staining were performed to observe the activation and phenotypic transformation of microglia in PFC. LPS‐induced cell model was constructed to explore the effect of FBXO10/RAGE axis in the polarization of microglia in vitro. Results FBXO10 promoted RAGE degradation by ubiquitination in BV2 cells. FBXO10 protein levels were reduced whereas RAGE protein levels were enhanced in CUS mice. FBXO10 overexpression or RAGE knockdown inhibited proinflammatory cytokine release, promoted BDNF expression, mitigated the depressive‐like and cognitive impairment behaviors, and affected the polarization of microglia induced by CUS exposure. FBXO10/RAGE axis promoted the polarization of microglia from the M1 to the M2 phenotype in vitro. Moreover, p38 MAPK and NF‐κΒ were identified to be the downstream effect factors for FBXO10/RAGE axis. Conclusions FBXO10 administration prevents CUS‐induced behavioral despair, cognitive impairment, neuroinflammation, and the polarization of microglia through decreasing the accumulation of RAGE, p38 MAPK, and NF‐κΒ, suggesting potential therapeutic strategies for the prevention and treatment of depression.
Collapse
Affiliation(s)
- Jiacen Li
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Qingcui Zeng
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China.,Geriatric Intensive Care Unit, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Wenjie Su
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Menglong Song
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China.,Emergency Intensive Care Unit, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Min Xie
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Lei Mao
- Department of Anesthesiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| |
Collapse
|
195
|
Magham SV, Thaggikuppe Krishnamurthy P, Shaji N, Mani L, Balasubramanian S. Cannabinoid receptor 2 selective agonists and Alzheimer's disease: An insight into the therapeutic potentials. J Neurosci Res 2021; 99:2888-2905. [PMID: 34486749 DOI: 10.1002/jnr.24933] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/04/2021] [Accepted: 07/14/2021] [Indexed: 12/19/2022]
Abstract
Endocannabinoid system has been extensively studied in recent decades, particularly the cannabinoid receptors CB1 and CB2, due to their important role in neuroinflammation. Among these, CB2 has gained prominence due to its selective overexpression in glial cells during neuroinflammation. In contrast to CB1 agonists, CB2 agonists have no side effects such as ataxia, hypothermia, euphoria, psychological, or addiction liabilities. CB2 and its selective agonists' above-mentioned unique properties have become a research focus in neurodegenerative disorders such as Alzheimer's disease (AD). The review discusses the neuroprotective role of CB receptors, particularly CB2, in AD, as well as the significance and limitations of this research.
Collapse
Affiliation(s)
- Sai Varshini Magham
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, India
| | | | - Neenu Shaji
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, India
| | - Lalithkumar Mani
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, India
| | | |
Collapse
|
196
|
Zhang Y, Li Y, Fu X, Wang P, Wang Q, Meng W, Wang T, Yang J, Chai R. The Detrimental and Beneficial Functions of Macrophages After Cochlear Injury. Front Cell Dev Biol 2021; 9:631904. [PMID: 34458249 PMCID: PMC8385413 DOI: 10.3389/fcell.2021.631904] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 07/14/2021] [Indexed: 12/20/2022] Open
Abstract
Macrophages are the main intrinsic immune cells in the cochlea; they can be activated and play a complicated role after cochlear injury. Many studies have shown that the number of macrophages and their morphological characteristics within the major cochlear partitions undergo significant changes under various pathological conditions including acoustic trauma, ototoxic drug treatment, age-related cochlear degeneration, selective hair cell (HC) and spiral ganglion neuron (SGN) elimination, and surgery. However, the exact role of these macrophages after cochlear injury is still unclear. Regulating the migration and activity of macrophages may be a therapeutic approach to reduce the risk or magnitude of trauma-induced hearing loss, and this review highlights the role of macrophages on the peripheral auditory structures of the cochlea and elucidate the mechanisms of macrophage injury and the strategies to reduce the injury by regulating macrophage.
Collapse
Affiliation(s)
- Yuan Zhang
- MOE Key Laboratory for Developmental Genes and Human Disease, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Sciences and Technology, Southeast University, Nanjing, China.,Department of Otolaryngology Head and Neck, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yiyuan Li
- MOE Key Laboratory for Developmental Genes and Human Disease, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Sciences and Technology, Southeast University, Nanjing, China
| | - Xiaolong Fu
- MOE Key Laboratory for Developmental Genes and Human Disease, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Sciences and Technology, Southeast University, Nanjing, China
| | - Pengjun Wang
- Department of Otorhinolaryngology, Affiliated Sixth People's Hospital of Shanghai Jiao Tong University, Shanghai, China
| | - Qin Wang
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wei Meng
- Department of Otolaryngology Head and Neck, Nanjing Tongren Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Tian Wang
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jianming Yang
- Department of Otorhinolaryngology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Renjie Chai
- MOE Key Laboratory for Developmental Genes and Human Disease, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, School of Life Sciences and Technology, Southeast University, Nanjing, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
197
|
Cao P, Chen C, Liu A, Shan Q, Zhu X, Jia C, Peng X, Zhang M, Farzinpour Z, Zhou W, Wang H, Zhou JN, Song X, Wang L, Tao W, Zheng C, Zhang Y, Ding YQ, Jin Y, Xu L, Zhang Z. Early-life inflammation promotes depressive symptoms in adolescence via microglial engulfment of dendritic spines. Neuron 2021; 109:2573-2589.e9. [PMID: 34233151 DOI: 10.1016/j.neuron.2021.06.012] [Citation(s) in RCA: 174] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 05/02/2021] [Accepted: 06/10/2021] [Indexed: 02/07/2023]
Abstract
Early-life inflammation increases the risk for depression in later life. Here, we demonstrate how early-life inflammation causes adolescent depressive-like symptoms: by altering the long-term neuronal spine engulfment capacity of microglia. For mice exposed to lipopolysaccharide (LPS)-induced inflammation via the Toll-like receptor 4/NF-κB signaling pathway at postnatal day (P) 14, ongoing longitudinal imaging of the living brain revealed that later stress (delivered during adolescence on P45) increases the extent of microglial engulfment around anterior cingulate cortex (ACC) glutamatergic neuronal (ACCGlu) spines. When the ACC microglia of LPS-treated mice were deleted or chemically inhibited, the mice did not exhibit depressive-like behaviors during adolescence. Moreover, we show that the fractalkine receptor CX3CR1 mediates stress-induced engulfment of ACCGlu neuronal spines. Together, our findings establish that early-life inflammation causes dysregulation of microglial engulfment capacity, which encodes long-lasting maladaptation of ACCGlu neurons to stress, thus promoting development of depression-like symptoms during adolescence.
Collapse
Affiliation(s)
- Peng Cao
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China
| | - Changmao Chen
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China
| | - An Liu
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230022, China
| | - Qinghong Shan
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China
| | - Xia Zhu
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China
| | - Chunhui Jia
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China
| | - Xiaoqi Peng
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China
| | - Mingjun Zhang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China
| | - Zahra Farzinpour
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China
| | - Wenjie Zhou
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China
| | - Haitao Wang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China
| | - Jiang-Ning Zhou
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China
| | - Xiaoyuan Song
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China
| | - Liecheng Wang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230022, China
| | - Wenjuan Tao
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230022, China
| | - Changjian Zheng
- Department of Anesthesiology, the First Affiliated Hospital of Wannan Medical College, Wuhu 241002, China
| | - Yan Zhang
- Stroke Center & Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China
| | - Yu-Qiang Ding
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Department of Laboratory Animal Science, Fudan University, Shanghai 200032, China
| | - Yan Jin
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China.
| | - Lin Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms, and Laboratory of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming 650223, China.
| | - Zhi Zhang
- Department of Anesthesiology, The First Affiliated Hospital of USTC, Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230036, China.
| |
Collapse
|
198
|
da Silva MCM, Gomes GF, de Barros Fernandes H, da Silva AM, Teixeira AL, Moreira FA, de Miranda AS, de Oliveira ACP. Inhibition of CSF1R, a receptor involved in microglia viability, alters behavioral and molecular changes induced by cocaine. Sci Rep 2021; 11:15989. [PMID: 34362959 PMCID: PMC8346567 DOI: 10.1038/s41598-021-95059-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/13/2021] [Indexed: 02/07/2023] Open
Abstract
Different data suggest that microglia may participate in the drug addiction process as these cells respond to neurochemical changes induced by the administration of these substances. In order to study the role of microglia in drug abuse, Swiss mice aged 8-9 weeks were treated with the CSF1R inhibitor PLX3397 (40 mg/kg, p.o.) and submitted to behavioral sensitization or conditioned place preference (CPP) induced by cocaine (15 mg/kg, i.p.). Thereafter, brains were used to evaluate the effects of CSF1R inhibition and cocaine administration on morphological, biochemical and molecular changes. CSF1R inhibition attenuated behavioral sensitization, reduced the number of Iba-1+ cells and increased ramification and lengths of the branches in the remaining microglia. Additionally, both cocaine and PLX3397 increased the cell body to total cell size ratio of Iba-1+ cells, as well as CD68+ and GFAP+ stained areas, suggesting an activated pattern of the glial cells. Besides, CSF1R inhibition increased CX3CL1 levels in the striatum, prefrontal cortex and hippocampus, as well as reduced CX3CR1 expression in the hippocampus. In this region, cocaine also reduced BDNF levels, an effect that was enhanced by CSF1R inhibition. In summary, our results suggest that microglia participate in the behavioral and molecular changes induced by cocaine. This study contributes to the understanding of the role of microglia in cocaine addiction.
Collapse
Affiliation(s)
- Maria Carolina Machado da Silva
- Neuropharmacology Laboratory, Department of Pharmacology, Universidade Federal de Minas Gerais, Av. Antonio Carlos 6627, Belo Horizonte, MG, 31270-901, Brazil
| | - Giovanni Freitas Gomes
- Neuropharmacology Laboratory, Department of Pharmacology, Universidade Federal de Minas Gerais, Av. Antonio Carlos 6627, Belo Horizonte, MG, 31270-901, Brazil
| | - Heliana de Barros Fernandes
- Neurobiology Laboratory Conceição Machado, Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Laboratory of Inflammatory Genes, Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Aristóbolo Mendes da Silva
- Laboratory of Inflammatory Genes, Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Antônio Lúcio Teixeira
- Department of Psychiatry and Behavioral Science McGovern School, The University of Texas Health Science Center at Houston, Houston, USA
| | - Fabrício A Moreira
- Neuropsychopharmacology Laboratory, Department of Pharmacology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Aline Silva de Miranda
- Neurobiology Laboratory Conceição Machado, Department of Morphology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Antônio Carlos Pinheiro de Oliveira
- Neuropharmacology Laboratory, Department of Pharmacology, Universidade Federal de Minas Gerais, Av. Antonio Carlos 6627, Belo Horizonte, MG, 31270-901, Brazil.
| |
Collapse
|
199
|
Connecting the Neurobiology of Developmental Brain Injury: Neuronal Arborisation as a Regulator of Dysfunction and Potential Therapeutic Target. Int J Mol Sci 2021; 22:ijms22158220. [PMID: 34360985 PMCID: PMC8348801 DOI: 10.3390/ijms22158220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 11/17/2022] Open
Abstract
Neurodevelopmental disorders can derive from a complex combination of genetic variation and environmental pressures on key developmental processes. Despite this complex aetiology, and the equally complex array of syndromes and conditions diagnosed under the heading of neurodevelopmental disorder, there are parallels in the neuropathology of these conditions that suggest overlapping mechanisms of cellular injury and dysfunction. Neuronal arborisation is a process of dendrite and axon extension that is essential for the connectivity between neurons that underlies normal brain function. Disrupted arborisation and synapse formation are commonly reported in neurodevelopmental disorders. Here, we summarise the evidence for disrupted neuronal arborisation in these conditions, focusing primarily on the cortex and hippocampus. In addition, we explore the developmentally specific mechanisms by which neuronal arborisation is regulated. Finally, we discuss key regulators of neuronal arborisation that could link to neurodevelopmental disease and the potential for pharmacological modification of arborisation and the formation of synaptic connections that may provide therapeutic benefit in the future.
Collapse
|
200
|
Activation of the Hippocampal LXRβ Improves Sleep-Deprived Cognitive Impairment by Inhibiting Neuroinflammation. Mol Neurobiol 2021; 58:5272-5288. [PMID: 34278533 DOI: 10.1007/s12035-021-02446-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 06/06/2021] [Indexed: 10/20/2022]
Abstract
Sleep deprivation (SD) leads to cognitive impairment due to neuroinflammation associated with impaired hippocampal neuronal plasticity and memory processes. Liver X receptors (LXRs), including LXRα and LXRβ isoforms, are crucial for synaptic plasticity and neuroinflammation. However, the potential roles of LXRs in the pathogenesis of cognitive impairment induced by SD remain unclear. We revealed that SD resulted in LXRβ reduction in the hippocampus, which was associated with upregulated expression of high mobility group box 1 (HMGB1)/toll-like receptor 4 (TLR4)/NF-κB p65, and knockdown of hippocampal LXRβ by shRNA (shLXRβ) led to cognitive impairment. GW3965, a dual agonist for both LXRα and LXRβ, ameliorated SD-induced cognitive impairment by inhibiting microglia activation, suppressing HMGB1/TLR4/NF-κB p65 pathway, and ultimately affecting the hippocampal expression of inflammatory cytokines in SD mice. LXRβ knockdown by shLXRβ abrogated the GW3965-mediated inhibition of the HMGB1/TLR4/NF-κB p65 pathway, therefore, abolishing the cognitive improvement. Moreover, inhibition of HMGB1 by glycyrrhizin (GLY) synergistic promoted GW3965-mediated anti-inflammation in activated microglia after lipopolysaccharide (LPS)/ATP stimulation and facilitated the cognitive improvement after GW administration by activating LXRβ. All the data suggested that GW3965 ameliorated impaired cognition in SD mice by suppressing the HMGB1/TLR4/NF-κB p65 pathway followed LXRβ activation. This study correlates a deficit of LXRβ in cognitive dysfunction in SD associated with HMGB1 inflammatory pathway in hippocampus, and LXRs may serve as a potential therapeutic target for cognitive impairment with anti-inflammation.
Collapse
|