1
|
Ma YN, Hu X, Karako K, Song P, Tang W, Xia Y. Agarwood as a potential therapeutic for Alzheimer's disease: Mechanistic insights and target identification. Drug Discov Ther 2025; 18:375-386. [PMID: 39710406 DOI: 10.5582/ddt.2024.01085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by cognitive decline, memory loss, and functional impairments. Despite extensive research, its pathogenesis remains incompletely understood, and effective treatments are limited. This study explored the therapeutic potential of agarwood in AD by integrating network pharmacology, protein-protein interaction (PPI) network analysis, and single-cell expression analysis. The results revealed that agarwood compounds may modulate key inflammatory genes such as NFKB1, STAT1, and TLR4, alleviating neuroinflammation; enhance the expression of HSP90 and regulate KDR signaling to improve blood-brain barrier (BBB) integrity; and promote the activity of PTPN11 and CXCR4 to support oligodendrocyte precursor cell (OPC) repair and remyelination. Single-cell expression analysis highlighted cell-type-specific expression patterns, particularly in OPCs and endothelial cells, underscoring their relevance in AD pathology. Agarwood's multi-dimensional therapeutic potential positions it as a promising candidate for the development of novel AD treatments.
Collapse
Affiliation(s)
- Ya-Nan Ma
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Xiqi Hu
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| | - Kenji Karako
- Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Peipei Song
- National Center for Global Health and Medicine, Tokyo, Japan
| | - Wei Tang
- Department of Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- National Center for Global Health and Medicine, Tokyo, Japan
| | - Ying Xia
- Department of Neurosurgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, China
| |
Collapse
|
2
|
Chaudhary R, Rehman M, Agarwal V, Kumar A, Kaushik AS, Srivastava S, Srivastava S, Verma R, Rajinikanth PS, Mishra V. Terra incognita of glial cell dynamics in the etiology of leukodystrophies: Broadening disease and therapeutic perspectives. Life Sci 2024; 354:122953. [PMID: 39122110 DOI: 10.1016/j.lfs.2024.122953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/09/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Neuroglial cells, also known as glia, are primarily characterized as auxiliary cells within the central nervous system (CNS). The recent findings have shed light on their significance in numerous physiological processes and their involvement in various neurological disorders. Leukodystrophies encompass an array of rare and hereditary neurodegenerative conditions that were initially characterized by the deficiency, aberration, or degradation of myelin sheath within CNS. The primary cellular populations that experience significant alterations are astrocytes, oligodendrocytes and microglia. These glial cells are either structurally or metabolically impaired due to inherent cellular dysfunction. Alternatively, they may fall victim to the accumulation of harmful by-products resulting from metabolic disturbances. In either situation, the possible replacement of glial cells through the utilization of implanted tissue or stem cell-derived human neural or glial progenitor cells hold great promise as a therapeutic strategy for both the restoration of structural integrity through remyelination and the amelioration of metabolic deficiencies. Various emerging treatment strategies like stem cell therapy, ex-vivo gene therapy, infusion of adeno-associated virus vectors, emerging RNA-based therapies as well as long-term therapies have demonstrated success in pre-clinical studies and show promise for rapid clinical translation. Here, we addressed various leukodystrophies in a comprehensive and detailed manner as well as provide prospective therapeutic interventions that are being considered for clinical trials. Further, we aim to emphasize the crucial role of different glial cells in the pathogenesis of leukodystrophies. By doing so, we hope to advance our understanding of the disease, elucidate underlying mechanisms, and facilitate the development of potential treatment interventions.
Collapse
Affiliation(s)
- Rishabh Chaudhary
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Mujeeba Rehman
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Vipul Agarwal
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Anand Kumar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Arjun Singh Kaushik
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Siddhi Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Sukriti Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Rajkumar Verma
- University of Connecticut School of Medicine, 200 Academic Way, Farmington, CT 06032, USA
| | - P S Rajinikanth
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Vikas Mishra
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India.
| |
Collapse
|
3
|
Chaves-Filho A, Eyres C, Blöbaum L, Landwehr A, Tremblay MÈ. The emerging neuroimmune hypothesis of bipolar disorder: An updated overview of neuroimmune and microglial findings. J Neurochem 2024; 168:1780-1816. [PMID: 38504593 DOI: 10.1111/jnc.16098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/21/2024]
Abstract
Bipolar disorder (BD) is a severe and multifactorial disease, with onset usually in young adulthood, which follows a progressive course throughout life. Replicated epidemiological studies have suggested inflammatory mechanisms and neuroimmune risk factors as primary contributors to the onset and development of BD. While not all patients display overt markers of inflammation, significant evidence suggests that aberrant immune signaling contributes to all stages of the disease and seems to be mood phase dependent, likely explaining the heterogeneity of findings observed in this population. As the brain's immune cells, microglia orchestrate the brain's immune response and play a critical role in maintaining the brain's health across the lifespan. Microglia are also highly sensitive to environmental changes and respond to physiological and pathological events by adapting their functions, structure, and molecular expression. Recently, it has been highlighted that instead of a single population of cells, microglia comprise a heterogeneous community with specialized states adjusted according to the local molecular cues and intercellular interactions. Early evidence has highlighted the contribution of microglia to BD neuropathology, notably for severe outcomes, such as suicidality. However, the roles and diversity of microglial states in this disease are still largely undermined. This review brings an updated overview of current literature on the contribution of neuroimmune risk factors for the onset and progression of BD, the most prominent neuroimmune abnormalities (including biomarker, neuroimaging, ex vivo studies) and the most recent findings of microglial involvement in BD neuropathology. Combining these different shreds of evidence, we aim to propose a unifying hypothesis for BD pathophysiology centered on neuroimmune abnormalities and microglia. Also, we highlight the urgent need to apply novel multi-system biology approaches to characterize the diversity of microglial states and functions involved in this enigmatic disorder, which can open bright perspectives for novel biomarkers and therapeutic discoveries.
Collapse
Affiliation(s)
- Adriano Chaves-Filho
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Women Health Research Institute, Vancouver, British Columbia, Canada
- Brain Health Cluster at the Institute on Aging & Lifelong Health (IALH), Victoria, British Columbia, Canada
| | - Capri Eyres
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Leonie Blöbaum
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Antonia Landwehr
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia, Canada
- Women Health Research Institute, Vancouver, British Columbia, Canada
- Brain Health Cluster at the Institute on Aging & Lifelong Health (IALH), Victoria, British Columbia, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, British Columbia, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
- Neurology and Neurosurgery Department, McGill University, Montréal, Quebec, Canada
- Department of Molecular Medicine, Université Laval, Québec City, Quebec, Canada
| |
Collapse
|
4
|
Munro DAD, Bestard-Cuche N, McQuaid C, Chagnot A, Shabestari SK, Chadarevian JP, Maheshwari U, Szymkowiak S, Morris K, Mohammad M, Corsinotti A, Bradford B, Mabbott N, Lennen RJ, Jansen MA, Pridans C, McColl BW, Keller A, Blurton-Jones M, Montagne A, Williams A, Priller J. Microglia protect against age-associated brain pathologies. Neuron 2024; 112:2732-2748.e8. [PMID: 38897208 DOI: 10.1016/j.neuron.2024.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 04/17/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024]
Abstract
Microglia are brain-resident macrophages that contribute to central nervous system (CNS) development, maturation, and preservation. Here, we examine the consequences of permanent microglial deficiencies on brain aging using the Csf1rΔFIRE/ΔFIRE mouse model. In juvenile Csf1rΔFIRE/ΔFIRE mice, we show that microglia are dispensable for the transcriptomic maturation of other brain cell types. By contrast, with advancing age, pathologies accumulate in Csf1rΔFIRE/ΔFIRE brains, macroglia become increasingly dysregulated, and white matter integrity declines, mimicking many pathological features of human CSF1R-related leukoencephalopathy. The thalamus is particularly vulnerable to neuropathological changes in the absence of microglia, with atrophy, neuron loss, vascular alterations, macroglial dysregulation, and severe tissue calcification. We show that populating Csf1rΔFIRE/ΔFIRE brains with wild-type microglia protects against many of these pathological changes. Together with the accompanying study by Chadarevian and colleagues1, our results indicate that the lifelong absence of microglia results in an age-related neurodegenerative condition that can be counteracted via transplantation of healthy microglia.
Collapse
Affiliation(s)
- David A D Munro
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, Chancellor's Building, Edinburgh EH16 4SB, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK.
| | - Nadine Bestard-Cuche
- Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Conor McQuaid
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, Chancellor's Building, Edinburgh EH16 4SB, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Audrey Chagnot
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, Chancellor's Building, Edinburgh EH16 4SB, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Sepideh Kiani Shabestari
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Jean Paul Chadarevian
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA
| | - Upasana Maheshwari
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Stefan Szymkowiak
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, Chancellor's Building, Edinburgh EH16 4SB, UK; Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Kim Morris
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, Chancellor's Building, Edinburgh EH16 4SB, UK
| | - Mehreen Mohammad
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, Chancellor's Building, Edinburgh EH16 4SB, UK
| | - Andrea Corsinotti
- Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Barry Bradford
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian, UK
| | - Neil Mabbott
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush Campus, Midlothian, UK
| | - Ross J Lennen
- Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK
| | - Maurits A Jansen
- Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh EH16 4TJ, UK; Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA, USA
| | - Clare Pridans
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Barry W McColl
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, Chancellor's Building, Edinburgh EH16 4SB, UK; Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh EH8 9XD, UK
| | - Annika Keller
- Department of Neurosurgery, Clinical Neuroscience Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Mathew Blurton-Jones
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA
| | - Axel Montagne
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, Chancellor's Building, Edinburgh EH16 4SB, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | - Anna Williams
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, Chancellor's Building, Edinburgh EH16 4SB, UK; Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Josef Priller
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, Chancellor's Building, Edinburgh EH16 4SB, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK; Department of Psychiatry and Psychotherapy, School of Medicine and Health, Klinikum rechts der Isar, Technical University Munich, and German Center for Mental Health (DZPG), 81675 Munich, Germany; Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité - Universitätsmedizin Berlin and DZNE, 10117 Berlin, Germany.
| |
Collapse
|
5
|
VanRyzin JW, Marquardt AE, McCarthy MM. Feminization of social play behavior depends on microglia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.19.608675. [PMID: 39229086 PMCID: PMC11370478 DOI: 10.1101/2024.08.19.608675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Many sex differences in brain and behavior are established developmentally by the opposing processes of feminization and masculinization, which manifest following differential steroid hormone exposure in early life. The cellular mechanisms underlying masculinization are well-documented, a result of the fact that it is steroid-mediated and can be easily induced in newborn female rodents via exogenous steroid treatment. However, the study of feminization of particular brain regions has largely been relegated to being "not masculinization" given the absence of an identified initiating trigger. As a result, the mechanisms of this key developmental process remain elusive. Here we describe a novel role for microglia, the brain's innate immune cell, in the feminization of the medial amygdala and a complex social behavior, juvenile play. In the developing amygdala, microglia promote proliferation of astrocytes equally in both sexes, with no apparent effect on rates of cell division, but support cell survival selectively in females through the trophic actions of Tumor Necrosis Factor α (TNFα). We demonstrate that disrupting TNFα signaling, either by depleting microglia or inhibiting the associated signaling pathways, prevents the feminization of astrocyte density and increases juvenile play levels to that seen in males. This data, combined with our previous finding that male-like patterns of astrocyte density are sculpted by developmental microglial phagocytosis, reveals that sexual differentiation of the medial amygdala involves opposing tensions between active masculinization and active feminization, both of which require microglia but are achieved via distinct processes.
Collapse
Affiliation(s)
- Jonathan W VanRyzin
- Department of Pharmacology, Physiology and Drug Development and University of Maryland Medicine – Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, MD 21201
| | - Ashley E Marquardt
- Department of Pharmacology, Physiology and Drug Development and University of Maryland Medicine – Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, MD 21201
| | - Margaret M McCarthy
- Department of Pharmacology, Physiology and Drug Development and University of Maryland Medicine – Institute for Neuroscience Discovery (UM-MIND), University of Maryland School of Medicine, Baltimore, MD 21201
| |
Collapse
|
6
|
Okechukwu NG, Klein C, Jamann H, Maitre M, Patte-Mensah C, Mensah-Nyagan AG. Monomeric Amyloid Peptide-induced Toxicity in Human Oligodendrocyte Cell Line and Mouse Brain Primary Mixed-glial Cell Cultures: Evidence for a Neuroprotective Effect of Neurosteroid 3α-O-allyl-allopregnanolone. Neurotox Res 2024; 42:37. [PMID: 39102123 DOI: 10.1007/s12640-024-00715-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 04/18/2024] [Accepted: 07/22/2024] [Indexed: 08/06/2024]
Abstract
Amyloid-peptide (Aβ) monomeric forms (ABM) occurring in presymptomatic Alzheimer's disease (AD) brain are thought to be devoid of neurotoxicity while the transition/aggregation of ABM into oligomers is determinant for Aβ-induced toxicity since Aβ is predominantly monomeric up to 3 µM and aggregates over this concentration. However, recent imaging and/or histopathological investigations revealed alterations of myelin in prodromal AD brain in absence of aggregated Aβ oligomers, suggesting that ABM may induce toxicity in myelin-producing cells in early AD-stages. To check this hypothesis, here we studied ABM effects on the viability of the Human oligodendrocyte cell line (HOG), a reliable oligodendrocyte model producing myelin proteins. Furthermore, to mimic closely interactions between oligodendrocytes and other glial cells regulating myelination, we investigated also ABM effects on mouse brain primary mixed-glial cell cultures. Various methods were combined to show that ABM concentrations (600 nM-1 µM), extremely lower than 3 µM, significantly decreased HOG cell and mouse brain primary mixed-glial cell survival. Interestingly, flow-cytometry studies using specific cell-type markers demonstrated that oligodendrocytes represent the most vulnerable glial cell population affected by ABM toxicity. Our work also shows that the neurosteroid 3α-O-allyl-allopregnanolone BR351 (250 and 500 nM) efficiently prevented ABM-induced HOG and brain primary glial cell toxicity. Bicuculline (50-100 nM), the GABA-A-receptor antagonist, was unable to block/reduce BR351 effect against ABM-induced HOG and primary glial cell toxicity, suggesting that BR351-evoked neuroprotection of these cells may not depend on GABA-A-receptor allosterically modulated by neurosteroids. Altogether, our results suggest that further exploration of BR351 therapeutic potential may offer interesting perspectives to develop effective neuroprotective strategies.
Collapse
Affiliation(s)
- Nwife Getrude Okechukwu
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, 67 000, Strasbourg, France
| | - Christian Klein
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, 67 000, Strasbourg, France
- Centre d'Investigation Clinique de Strasbourg (CIC), Equipe CIC-Recherche Translationnelle Neuro, INSERM 1434, Université de Strasbourg, Bâtiment CRBS, 1 rue Eugène Boeckel, 67000, Strasbourg, France
| | - Hélène Jamann
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, 67 000, Strasbourg, France
- Centre d'Investigation Clinique de Strasbourg (CIC), Equipe CIC-Recherche Translationnelle Neuro, INSERM 1434, Université de Strasbourg, Bâtiment CRBS, 1 rue Eugène Boeckel, 67000, Strasbourg, France
| | - Michel Maitre
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, 67 000, Strasbourg, France
- Centre d'Investigation Clinique de Strasbourg (CIC), Equipe CIC-Recherche Translationnelle Neuro, INSERM 1434, Université de Strasbourg, Bâtiment CRBS, 1 rue Eugène Boeckel, 67000, Strasbourg, France
| | - Christine Patte-Mensah
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, 67 000, Strasbourg, France
- Centre d'Investigation Clinique de Strasbourg (CIC), Equipe CIC-Recherche Translationnelle Neuro, INSERM 1434, Université de Strasbourg, Bâtiment CRBS, 1 rue Eugène Boeckel, 67000, Strasbourg, France
| | - Ayikoé-Guy Mensah-Nyagan
- Biopathologie de la Myéline, Neuroprotection et Stratégies Thérapeutiques, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Bâtiment CRBS de la Faculté de Médecine, 1 rue Eugène Boeckel, 67 000, Strasbourg, France.
- Centre d'Investigation Clinique de Strasbourg (CIC), Equipe CIC-Recherche Translationnelle Neuro, INSERM 1434, Université de Strasbourg, Bâtiment CRBS, 1 rue Eugène Boeckel, 67000, Strasbourg, France.
| |
Collapse
|
7
|
Anilkumar S, Wright-Jin E. NF-κB as an Inducible Regulator of Inflammation in the Central Nervous System. Cells 2024; 13:485. [PMID: 38534329 PMCID: PMC10968931 DOI: 10.3390/cells13060485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/01/2024] [Accepted: 03/09/2024] [Indexed: 03/28/2024] Open
Abstract
The NF-κB (nuclear factor K-light-chain-enhancer of activated B cells) transcription factor family is critical for modulating the immune proinflammatory response throughout the body. During the resting state, inactive NF-κB is sequestered by IκB in the cytoplasm. The proteasomal degradation of IκB activates NF-κB, mediating its translocation into the nucleus to act as a nuclear transcription factor in the upregulation of proinflammatory genes. Stimuli that initiate NF-κB activation are diverse but are canonically attributed to proinflammatory cytokines and chemokines. Downstream effects of NF-κB are cell type-specific and, in the majority of cases, result in the activation of pro-inflammatory cascades. Acting as the primary immune responders of the central nervous system, microglia exhibit upregulation of NF-κB upon activation in response to pathological conditions. Under such circumstances, microglial crosstalk with other cell types in the central nervous system can induce cell death, further exacerbating the disease pathology. In this review, we will emphasize the role of NF-κB in triggering neuroinflammation mediated by microglia.
Collapse
Affiliation(s)
- Sudha Anilkumar
- Neonatal Brain Injury Laboratory, Division of Biomedical Research, Nemours Children’s Health, Wilmington, DE 19803, USA
| | - Elizabeth Wright-Jin
- Neonatal Brain Injury Laboratory, Division of Biomedical Research, Nemours Children’s Health, Wilmington, DE 19803, USA
- Division of Neurology, Department of Pediatrics, Nemours Children’s Health, Wilmington, DE 19803, USA
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, USA
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
8
|
Xu P, Yu Y, Wu P. Role of microglia in brain development after viral infection. Front Cell Dev Biol 2024; 12:1340308. [PMID: 38298216 PMCID: PMC10825034 DOI: 10.3389/fcell.2024.1340308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/04/2024] [Indexed: 02/02/2024] Open
Abstract
Microglia are immune cells in the brain that originate from the yolk sac and enter the developing brain before birth. They play critical roles in brain development by supporting neural precursor proliferation, synaptic pruning, and circuit formation. However, microglia are also vulnerable to environmental factors, such as infection and stress that may alter their phenotype and function. Viral infection activates microglia to produce inflammatory cytokines and anti-viral responses that protect the brain from damage. However, excessive or prolonged microglial activation impairs brain development and leads to long-term consequences such as autism spectrum disorder and schizophrenia spectrum disorder. Moreover, certain viruses may attack microglia and deploy them as "Trojan horses" to infiltrate the brain. In this brief review, we describe the function of microglia during brain development and examine their roles after infection through microglia-neural crosstalk. We also identify limitations for current studies and highlight future investigated questions.
Collapse
Affiliation(s)
- Pei Xu
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, United States
| | - Yongjia Yu
- Department of Radiation Oncology, University of Texas Medical Branch, Galveston, TX, United States
| | - Ping Wu
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
9
|
Lei Z, Lin W. Mechanisms Governing Oligodendrocyte Viability in Multiple Sclerosis and Its Animal Models. Cells 2024; 13:116. [PMID: 38247808 PMCID: PMC10814231 DOI: 10.3390/cells13020116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/23/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune inflammatory demyelinating disease of the central nervous system (CNS), which is triggered by an autoimmune assault targeting oligodendrocytes and myelin. Recent research indicates that the demise of oligodendrocytes due to an autoimmune attack contributes significantly to the pathogenesis of MS and its animal model experimental autoimmune encephalomyelitis (EAE). A key challenge in MS research lies in comprehending the mechanisms governing oligodendrocyte viability and devising therapeutic approaches to enhance oligodendrocyte survival. Here, we provide an overview of recent findings that highlight the contributions of oligodendrocyte death to the development of MS and EAE and summarize the current literature on the mechanisms governing oligodendrocyte viability in these diseases.
Collapse
Affiliation(s)
- Zhixin Lei
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China;
| | - Wensheng Lin
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
10
|
Harry GJ. Microglia Colonization Associated with Angiogenesis and Neural Cell Development. ADVANCES IN NEUROBIOLOGY 2024; 37:163-178. [PMID: 39207692 DOI: 10.1007/978-3-031-55529-9_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The temporal and spatial pattern of microglia colonization of the nervous system implies a role in early stages of organ development including cell proliferation, differentiation, and neurovascularization. As microglia colonize and establish within the developing nervous system, they assume a neural-specific identity and contribute to key developmental events. Their association around blood vessels implicates them in development of the vascular system or vice versa. A similar association has been reported for neural cell proliferation and associated phenotypic shifts and for cell fate differentiation to neuronal or glial phenotypes. These processes are accomplished by phagocytic activities, cell-cell contact relationships, and secretion of various factors. This chapter will present data currently available from studies evaluating the dynamic and interactive nature of these processes throughout the progression of nervous system development.
Collapse
Affiliation(s)
- G Jean Harry
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute Environmental Health Sciences, Research Triangle Park, NC, USA.
| |
Collapse
|
11
|
Auguste YSS, Ferro A, Kahng JA, Xavier AM, Dixon JR, Vrudhula U, Nichitiu AS, Rosado D, Wee TL, Pedmale UV, Cheadle L. Oligodendrocyte precursor cells engulf synapses during circuit remodeling in mice. Nat Neurosci 2022; 25:1273-1278. [PMID: 36171430 PMCID: PMC9534756 DOI: 10.1038/s41593-022-01170-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 08/18/2022] [Indexed: 01/13/2023]
Abstract
Oligodendrocyte precursor cells (OPCs) give rise to myelinating oligodendrocytes throughout life, but the functions of OPCs are not limited to oligodendrogenesis. Here we show that OPCs contribute to thalamocortical presynapse elimination in the developing and adult mouse visual cortex. OPC-mediated synapse engulfment increases in response to sensory experience during neural circuit refinement. Our data suggest that OPCs may regulate synaptic connectivity in the brain independently of oligodendrogenesis.
Collapse
Affiliation(s)
| | - Austin Ferro
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Jessica A Kahng
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
- School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Andre M Xavier
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | - Uma Vrudhula
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | - Daniele Rosado
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Tse-Luen Wee
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | | | - Lucas Cheadle
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA.
| |
Collapse
|
12
|
Targeting microglia–oligodendrocyte crosstalk in neurodegenerative and psychiatric disorders. Drug Discov Today 2022; 27:2562-2573. [DOI: 10.1016/j.drudis.2022.06.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 06/09/2022] [Accepted: 06/29/2022] [Indexed: 02/07/2023]
|
13
|
Jansen MI, Thomas Broome S, Castorina A. Exploring the Pro-Phagocytic and Anti-Inflammatory Functions of PACAP and VIP in Microglia: Implications for Multiple Sclerosis. Int J Mol Sci 2022; 23:ijms23094788. [PMID: 35563181 PMCID: PMC9104531 DOI: 10.3390/ijms23094788] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 02/04/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic neuroinflammatory and demyelinating disease of the central nervous system (CNS), characterised by the infiltration of peripheral immune cells, multifocal white-matter lesions, and neurodegeneration. In recent years, microglia have emerged as key contributors to MS pathology, acting as scavengers of toxic myelin/cell debris and modulating the inflammatory microenvironment to promote myelin repair. In this review, we explore the role of two neuropeptides, pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP), as important regulators of microglial functioning during demyelination, myelin phagocytosis, and remyelination, emphasising the potential of these neuropeptides as therapeutic targets for the treatment of MS.
Collapse
|
14
|
Cathomas F, Holt LM, Parise EM, Liu J, Murrough JW, Casaccia P, Nestler EJ, Russo SJ. Beyond the neuron: Role of non-neuronal cells in stress disorders. Neuron 2022; 110:1116-1138. [PMID: 35182484 PMCID: PMC8989648 DOI: 10.1016/j.neuron.2022.01.033] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/15/2021] [Accepted: 01/24/2022] [Indexed: 12/11/2022]
Abstract
Stress disorders are leading causes of disease burden in the U.S. and worldwide, yet available therapies are fully effective in less than half of all individuals with these disorders. Although to date, much of the focus has been on neuron-intrinsic mechanisms, emerging evidence suggests that chronic stress can affect a wide range of cell types in the brain and periphery, which are linked to maladaptive behavioral outcomes. Here, we synthesize emerging literature and discuss mechanisms of how non-neuronal cells in limbic regions of brain interface at synapses, the neurovascular unit, and other sites of intercellular communication to mediate the deleterious, or adaptive (i.e., pro-resilient), effects of chronic stress in rodent models and in human stress-related disorders. We believe that such an approach may one day allow us to adopt a holistic "whole body" approach to stress disorder research, which could lead to more precise diagnostic tests and personalized treatment strategies. Stress is a major risk factor for many psychiatric disorders. Cathomas et al. review new insight into how non-neuronal cells mediate the deleterious effects, as well as the adaptive, protective effects, of stress in rodent models and human stress-related disorders.
Collapse
Affiliation(s)
- Flurin Cathomas
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leanne M Holt
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric M Parise
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jia Liu
- Neuroscience Initiative, Advanced Science Research Center, Program in Biology and Biochemistry at The Graduate Center of The City University of New York, New York, NY, USA
| | - James W Murrough
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Patrizia Casaccia
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Neuroscience Initiative, Advanced Science Research Center, Program in Biology and Biochemistry at The Graduate Center of The City University of New York, New York, NY, USA
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Scott J Russo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
15
|
Li SY, Johnson R, Smyth LC, Dragunow M. Platelet-derived growth factor signalling in neurovascular function and disease. Int J Biochem Cell Biol 2022; 145:106187. [PMID: 35217189 DOI: 10.1016/j.biocel.2022.106187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 02/08/2022] [Accepted: 02/21/2022] [Indexed: 11/25/2022]
Abstract
Platelet-derived growth factors are critical for cerebrovascular development and homeostasis. Abnormalities in this signalling pathway are implicated in neurological diseases, especially those where neurovascular dysfunction and neuroinflammation plays a prominent role in disease pathologies, such as stroke and Alzheimer's disease; the angiogenic nature of this pathway also draws its significance in brain malignancies such as glioblastoma where tumour angiogenesis is profuse. In this review, we provide an updated overview of the actions of the platelet-derived growth factors on neurovascular function, their role in the regulation of perivascular cell types expressing the cognate receptors, neurological diseases associated with aberrance in signalling, and highlight the clinical relevance and therapeutic potentials of this pathway for central nervous system diseases.
Collapse
Affiliation(s)
- Susan Ys Li
- Department of Pharmacology and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| | - Rebecca Johnson
- Department of Pharmacology and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| | - Leon Cd Smyth
- Center for Brain Immunology and Glia, Department of Pathology and Immunology, Washington University in St Louis, MO, USA.
| | - Mike Dragunow
- Department of Pharmacology and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
16
|
Gliovascular Mechanisms and White Matter Injury in Vascular Cognitive Impairment and Dementia. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00013-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
17
|
Lespay-Rebolledo C, Tapia-Bustos A, Perez-Lobos R, Vio V, Casanova-Ortiz E, Farfan-Troncoso N, Zamorano-Cataldo M, Redel-Villarroel M, Ezquer F, Quintanilla ME, Israel Y, Morales P, Herrera-Marschitz M. Sustained Energy Deficit Following Perinatal Asphyxia: A Shift towards the Fructose-2,6-bisphosphatase (TIGAR)-Dependent Pentose Phosphate Pathway and Postnatal Development. Antioxidants (Basel) 2021; 11:74. [PMID: 35052577 PMCID: PMC8773255 DOI: 10.3390/antiox11010074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022] Open
Abstract
Labor and delivery entail a complex and sequential metabolic and physiologic cascade, culminating in most circumstances in successful childbirth, although delivery can be a risky episode if oxygen supply is interrupted, resulting in perinatal asphyxia (PA). PA causes an energy failure, leading to cell dysfunction and death if re-oxygenation is not promptly restored. PA is associated with long-term effects, challenging the ability of the brain to cope with stressors occurring along with life. We review here relevant targets responsible for metabolic cascades linked to neurodevelopmental impairments, that we have identified with a model of global PA in rats. Severe PA induces a sustained effect on redox homeostasis, increasing oxidative stress, decreasing metabolic and tissue antioxidant capacity in vulnerable brain regions, which remains weeks after the insult. Catalase activity is decreased in mesencephalon and hippocampus from PA-exposed (AS), compared to control neonates (CS), in parallel with increased cleaved caspase-3 levels, associated with decreased glutathione reductase and glutathione peroxidase activity, a shift towards the TIGAR-dependent pentose phosphate pathway, and delayed calpain-dependent cell death. The brain damage continues long after the re-oxygenation period, extending for weeks after PA, affecting neurons and glial cells, including myelination in grey and white matter. The resulting vulnerability was investigated with organotypic cultures built from AS and CS rat newborns, showing that substantia nigra TH-dopamine-positive cells from AS were more vulnerable to 1 mM of H2O2 than those from CS animals. Several therapeutic strategies are discussed, including hypothermia; N-acetylcysteine; memantine; nicotinamide, and intranasally administered mesenchymal stem cell secretomes, promising clinical translation.
Collapse
Affiliation(s)
- Carolyne Lespay-Rebolledo
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Andrea Tapia-Bustos
- School of Pharmacy, Faculty of Medicine, Universidad Andres Bello, Santiago 8370149, Chile;
| | - Ronald Perez-Lobos
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Valentina Vio
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Emmanuel Casanova-Ortiz
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Nancy Farfan-Troncoso
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Marta Zamorano-Cataldo
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Martina Redel-Villarroel
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Fernando Ezquer
- Center for Regenerative Medicine, Faculty of Medicine-Clínica Alemana, Universidad del Desarrollo, Santiago 7710162, Chile;
| | - Maria Elena Quintanilla
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Yedy Israel
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
- Center for Regenerative Medicine, Faculty of Medicine-Clínica Alemana, Universidad del Desarrollo, Santiago 7710162, Chile;
| | - Paola Morales
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
- Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Mario Herrera-Marschitz
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| |
Collapse
|
18
|
Hughes EG, Stockton ME. Premyelinating Oligodendrocytes: Mechanisms Underlying Cell Survival and Integration. Front Cell Dev Biol 2021; 9:714169. [PMID: 34368163 PMCID: PMC8335399 DOI: 10.3389/fcell.2021.714169] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/30/2021] [Indexed: 12/31/2022] Open
Abstract
In the central nervous system, oligodendrocytes produce myelin sheaths that enwrap neuronal axons to provide trophic support and increase conduction velocity. New oligodendrocytes are produced throughout life through a process referred to as oligodendrogenesis. Oligodendrogenesis consists of three canonical stages: the oligodendrocyte precursor cell (OPC), the premyelinating oligodendrocyte (preOL), and the mature oligodendrocyte (OL). However, the generation of oligodendrocytes is inherently an inefficient process. Following precursor differentiation, a majority of premyelinating oligodendrocytes are lost, likely due to apoptosis. If premyelinating oligodendrocytes progress through this survival checkpoint, they generate new myelinating oligodendrocytes in a process we have termed integration. In this review, we will explore the intrinsic and extrinsic signaling pathways that influence preOL survival and integration by examining the intrinsic apoptotic pathways, metabolic demands, and the interactions between neurons, astrocytes, microglia, and premyelinating oligodendrocytes. Additionally, we will discuss similarities between the maturation of newly generated neurons and premyelinating oligodendrocytes. Finally, we will consider how increasing survival and integration of preOLs has the potential to increase remyelination in multiple sclerosis. Deepening our understanding of premyelinating oligodendrocyte biology may open the door for new treatments for demyelinating disease and will help paint a clearer picture of how new oligodendrocytes are produced throughout life to facilitate brain function.
Collapse
Affiliation(s)
- Ethan G Hughes
- Department of Cell and Developmental Biology, School of Medicine, University of Colorado, Aurora, CO, United States
| | - Michael E Stockton
- Department of Cell and Developmental Biology, School of Medicine, University of Colorado, Aurora, CO, United States
| |
Collapse
|
19
|
Kalafatakis I, Karagogeos D. Oligodendrocytes and Microglia: Key Players in Myelin Development, Damage and Repair. Biomolecules 2021; 11:1058. [PMID: 34356682 PMCID: PMC8301746 DOI: 10.3390/biom11071058] [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: 05/30/2021] [Revised: 07/16/2021] [Accepted: 07/16/2021] [Indexed: 12/13/2022] Open
Abstract
Oligodendrocytes, the myelin-making cells of the CNS, regulate the complex process of myelination under physiological and pathological conditions, significantly aided by other glial cell types such as microglia, the brain-resident, macrophage-like innate immune cells. In this review, we summarize how oligodendrocytes orchestrate myelination, and especially myelin repair after damage, and present novel aspects of oligodendroglial functions. We emphasize the contribution of microglia in the generation and regeneration of myelin by discussing their beneficial and detrimental roles, especially in remyelination, underlining the cellular and molecular components involved. Finally, we present recent findings towards human stem cell-derived preclinical models for the study of microglia in human pathologies and on the role of microbiome on glial cell functions.
Collapse
Affiliation(s)
- Ilias Kalafatakis
- Laboratory of Neuroscience, Department of Basic Science, University of Crete Medical School, 70013 Heraklion, Greece;
- IMBB FORTH, Nikolaou Plastira 100, Vassilika Vouton, 70013 Heraklion, Greece
| | - Domna Karagogeos
- Laboratory of Neuroscience, Department of Basic Science, University of Crete Medical School, 70013 Heraklion, Greece;
- IMBB FORTH, Nikolaou Plastira 100, Vassilika Vouton, 70013 Heraklion, Greece
| |
Collapse
|
20
|
De I, Maklakova V, Litscher S, Boyd MM, Klemm LC, Wang Z, Kendziorski C, Collier LS. Microglial responses to CSF1 overexpression do not promote the expansion of other glial lineages. J Neuroinflammation 2021; 18:162. [PMID: 34281564 PMCID: PMC8290555 DOI: 10.1186/s12974-021-02212-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 07/05/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Colony-stimulating factor 1 (CSF1) expression in the central nervous system (CNS) increases in response to a variety of stimuli, and CSF1 is overexpressed in many CNS diseases. In young adult mice, we previously showed that CSF1 overexpression in the CNS caused the proliferation of IBA1+ microglia without promoting the expression of M2 polarization markers. METHODS Immunohistochemical and molecular analyses were performed to further examine the impact of CSF1 overexpression on glia in both young and aged mice. RESULTS As CSF1 overexpressing mice age, IBA1+ cell numbers are constrained by a decline in proliferation rate. Compared to controls, there were no differences in expression of the M2 markers ARG1 and MRC1 (CD206) in CSF1 overexpressing mice of any age, indicating that even prolonged exposure to increased CSF1 does not impact M2 polarization status in vivo. Moreover, RNA-sequencing confirmed the lack of increased expression of markers of M2 polarization in microglia exposed to CSF1 overexpression but did reveal changes in expression of other immune-related genes. Although treatment with inhibitors of the CSF1 receptor, CSF1R, has been shown to impact other glia, no increased expression of oligodendrocyte lineage or astrocyte markers was observed in CSF1 overexpressing mice. CONCLUSIONS Our study indicates that microglia are the primary glial lineage impacted by CSF1 overexpression in the CNS and that microglia ultimately adapt to the presence of the CSF1 mitogenic signal.
Collapse
Affiliation(s)
- Ishani De
- Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin, Madison, USA
| | - Vilena Maklakova
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, USA
| | - Suzanne Litscher
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, USA
| | - Michelle M Boyd
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, USA
| | - Lucas C Klemm
- Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin, Madison, USA
| | - Ziyue Wang
- Department of Statistics, University of Wisconsin, Madison, USA
| | - Christina Kendziorski
- Department of BiostatisticsUniversity of Wisconsin, Madison, USA
- University of Wisconsin Carbone Comprehensive Cancer Center, Madison, USA
| | - Lara S Collier
- Molecular and Cellular Pharmacology Graduate Program, University of Wisconsin, Madison, USA.
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, USA.
- University of Wisconsin Carbone Comprehensive Cancer Center, Madison, USA.
| |
Collapse
|
21
|
Mockenhaupt K, Gonsiewski A, Kordula T. RelB and Neuroinflammation. Cells 2021; 10:1609. [PMID: 34198987 PMCID: PMC8307460 DOI: 10.3390/cells10071609] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/23/2021] [Accepted: 06/23/2021] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation within the central nervous system involves multiple cell types that coordinate their responses by secreting and responding to a plethora of inflammatory mediators. These factors activate multiple signaling cascades to orchestrate initial inflammatory response and subsequent resolution. Activation of NF-κB pathways in several cell types is critical during neuroinflammation. In contrast to the well-studied role of p65 NF-κB during neuroinflammation, the mechanisms of RelB activation in specific cell types and its roles during neuroinflammatory response are less understood. In this review, we summarize the mechanisms of RelB activation in specific cell types of the CNS and the specialized effects this transcription factor exerts during neuroinflammation.
Collapse
Affiliation(s)
| | | | - Tomasz Kordula
- Department of Biochemistry and Molecular Biology, School of Medicine and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VI 23298, USA; (K.M.); (A.G.)
| |
Collapse
|
22
|
Psenicka MW, Smith BC, Tinkey RA, Williams JL. Connecting Neuroinflammation and Neurodegeneration in Multiple Sclerosis: Are Oligodendrocyte Precursor Cells a Nexus of Disease? Front Cell Neurosci 2021; 15:654284. [PMID: 34234647 PMCID: PMC8255483 DOI: 10.3389/fncel.2021.654284] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 05/20/2021] [Indexed: 12/14/2022] Open
Abstract
The pathology in neurodegenerative diseases is often accompanied by inflammation. It is well-known that many cells within the central nervous system (CNS) also contribute to ongoing neuroinflammation, which can promote neurodegeneration. Multiple sclerosis (MS) is both an inflammatory and neurodegenerative disease in which there is a complex interplay between resident CNS cells to mediate myelin and axonal damage, and this communication network can vary depending on the subtype and chronicity of disease. Oligodendrocytes, the myelinating cell of the CNS, and their precursors, oligodendrocyte precursor cells (OPCs), are often thought of as the targets of autoimmune pathology during MS and in several animal models of MS; however, there is emerging evidence that OPCs actively contribute to inflammation that directly and indirectly contributes to neurodegeneration. Here we discuss several contributors to MS disease progression starting with lesion pathology and murine models amenable to studying particular aspects of disease. We then review how OPCs themselves can play an active role in promoting neuroinflammation and neurodegeneration, and how other resident CNS cells including microglia, astrocytes, and neurons can impact OPC function. Further, we outline the very complex and pleiotropic role(s) of several inflammatory cytokines and other secreted factors classically described as solely deleterious during MS and its animal models, but in fact, have many neuroprotective functions and promote a return to homeostasis, in part via modulation of OPC function. Finally, since MS affects patients from the onset of disease throughout their lifespan, we discuss the impact of aging on OPC function and CNS recovery. It is becoming clear that OPCs are not simply a bystander during MS progression and uncovering the active roles they play during different stages of disease will help uncover potential new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Morgan W. Psenicka
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Brandon C. Smith
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Biological, Geological, and Environmental Sciences, Cleveland State University, Cleveland, OH, United States
| | - Rachel A. Tinkey
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- School of Biomedical Sciences, Kent State University, Kent, OH, United States
| | - Jessica L. Williams
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Brain Health Research Institute, Kent State University, Kent, OH, United States
| |
Collapse
|
23
|
Sherafat A, Pfeiffer F, Reiss AM, Wood WM, Nishiyama A. Microglial neuropilin-1 promotes oligodendrocyte expansion during development and remyelination by trans-activating platelet-derived growth factor receptor. Nat Commun 2021; 12:2265. [PMID: 33859199 PMCID: PMC8050320 DOI: 10.1038/s41467-021-22532-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 03/08/2021] [Indexed: 02/02/2023] Open
Abstract
Nerve-glia (NG2) glia or oligodendrocyte precursor cells (OPCs) are distributed throughout the gray and white matter and generate myelinating cells. OPCs in white matter proliferate more than those in gray matter in response to platelet-derived growth factor AA (PDGF AA), despite similar levels of its alpha receptor (PDGFRα) on their surface. Here we show that the type 1 integral membrane protein neuropilin-1 (Nrp1) is expressed not on OPCs but on amoeboid and activated microglia in white but not gray matter in an age- and activity-dependent manner. Microglia-specific deletion of Nrp1 compromised developmental OPC proliferation in white matter as well as OPC expansion and subsequent myelin repair after acute demyelination. Exogenous Nrp1 increased PDGF AA-induced OPC proliferation and PDGFRα phosphorylation on dissociated OPCs, most prominently in the presence of suboptimum concentrations of PDGF AA. These findings uncover a mechanism of regulating oligodendrocyte lineage cell density that involves trans-activation of PDGFRα on OPCs via Nrp1 expressed by adjacent microglia.
Collapse
Affiliation(s)
- Amin Sherafat
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - Friederike Pfeiffer
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Alexander M Reiss
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - William M Wood
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - Akiko Nishiyama
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA.
- Institute for Systems Genomics, University of Connecticut, Storrs, CT, USA.
- The Connecticut Institute for the Brain and Cognitive Sciences, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
24
|
Tapia-Bustos A, Lespay-Rebolledo C, Vío V, Pérez-Lobos R, Casanova-Ortiz E, Ezquer F, Herrera-Marschitz M, Morales P. Neonatal Mesenchymal Stem Cell Treatment Improves Myelination Impaired by Global Perinatal Asphyxia in Rats. Int J Mol Sci 2021; 22:ijms22063275. [PMID: 33806988 PMCID: PMC8004671 DOI: 10.3390/ijms22063275] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/07/2021] [Accepted: 03/15/2021] [Indexed: 01/09/2023] Open
Abstract
The effect of perinatal asphyxia (PA) on oligodendrocyte (OL), neuroinflammation, and cell viability was evaluated in telencephalon of rats at postnatal day (P)1, 7, and 14, a period characterized by a spur of neuronal networking, evaluating the effect of mesenchymal stem cell (MSCs)-treatment. The issue was investigated with a rat model of global PA, mimicking a clinical risk occurring under labor. PA was induced by immersing fetus-containing uterine horns into a water bath for 21 min (AS), using sibling-caesarean-delivered fetuses (CS) as controls. Two hours after delivery, AS and CS neonates were injected with either 5 μL of vehicle (10% plasma) or 5 × 104 MSCs into the lateral ventricle. Samples were assayed for myelin-basic protein (MBP) levels; Olig-1/Olig-2 transcriptional factors; Gglial phenotype; neuroinflammation, and delayed cell death. The main effects were observed at P7, including: (i) A decrease of MBP-immunoreactivity in external capsule, corpus callosum, cingulum, but not in fimbriae of hippocampus; (ii) an increase of Olig-1-mRNA levels; (iii) an increase of IL-6-mRNA, but not in protein levels; (iv) an increase in cell death, including OLs; and (v) MSCs treatment prevented the effect of PA on myelination, OLs number, and cell death. The present findings show that PA induces regional- and developmental-dependent changes on myelination and OLs maturation. Neonatal MSCs treatment improves survival of mature OLs and myelination in telencephalic white matter.
Collapse
Affiliation(s)
- Andrea Tapia-Bustos
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (A.T.-B.); (C.L.-R.); (V.V.); (R.P.-L.); (E.C.-O.)
- Faculty of Medicine, School of Pharmacy, Universidad Andres Bello, Santiago 8370149, Chile
| | - Carolyne Lespay-Rebolledo
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (A.T.-B.); (C.L.-R.); (V.V.); (R.P.-L.); (E.C.-O.)
| | - Valentina Vío
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (A.T.-B.); (C.L.-R.); (V.V.); (R.P.-L.); (E.C.-O.)
| | - Ronald Pérez-Lobos
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (A.T.-B.); (C.L.-R.); (V.V.); (R.P.-L.); (E.C.-O.)
| | - Emmanuel Casanova-Ortiz
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (A.T.-B.); (C.L.-R.); (V.V.); (R.P.-L.); (E.C.-O.)
| | - Fernando Ezquer
- Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Av. Las Condes 12438, Lo Barnechea, Santiago 7710162, Chile;
| | - Mario Herrera-Marschitz
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (A.T.-B.); (C.L.-R.); (V.V.); (R.P.-L.); (E.C.-O.)
- Correspondence: (M.H.-M.); (P.M.); Tel.: +56-229786788 (M.H.-M. & P.M.)
| | - Paola Morales
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (A.T.-B.); (C.L.-R.); (V.V.); (R.P.-L.); (E.C.-O.)
- Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
- Correspondence: (M.H.-M.); (P.M.); Tel.: +56-229786788 (M.H.-M. & P.M.)
| |
Collapse
|
25
|
Yang EJ, Song KS. The ameliorative effects of capsidiol isolated from elicited Capsicum annuum on mouse splenocyte immune responses and neuroinflammation. Phytother Res 2020; 35:1597-1608. [PMID: 33124100 DOI: 10.1002/ptr.6927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 10/04/2020] [Accepted: 10/09/2020] [Indexed: 11/10/2022]
Abstract
Capsidiol, is an anti-fungal phytoalexin produced by plants of Solanaceae. Capsidiol was examined in cultures of primary splenocytes (SPLCs) isolated from healthy C57BL/6 mice and from those with induced experimental autoimmune encephalomyelitis (EAE) as a mouse model for autoimmune neurodegenerative multiple sclerosis (MS). We also examined the impact of capsidiol in IFN-γ-stimulated mouse BV2 microglial cells. Capsidiol resulted in a significant reduction in the anti-CD3/CD28 (αCD3/CD28)-induced IFN-γ+ CD4+ (Th1) and IFN-γ+ CD8+ (Tc1) populations as well as in the production of cytokines (IFN-γ, IL-17A, IL-6, IL-2, TNF-α, and IP-10). Specifically, the CD4+ and CD8+ populations (T-bet+ IFN-γ- , T-bet+ IFN-γ+ , and T-bet- IFN-γ+ ) and cytokine production mediated by Th1/Tc1 polarization were diminished by 25 μM capsidiol. MOG35-55 restimulation of SPLCs from EAE mice resulted in an increase in antigen-specific T cells, including Th1, IL-17A+ CD4+ (Th17), and IL-17A+ CD8+ (Tc17) populations. By contrast, capsidiol resulted in a decrease in the proportions of Th17 and Tc17 cells; MOG35-55 -specific cytokine production was also diminished by capsidiol. Capsidiol treatment resulted in diminished levels of IFN-γ-induced nitric oxide and IL-6; expression of iNOS and COX-2 were suppressed by 50 μM capsidiol in IFN-γ-stimulated BV2 cells. This is the first report of capsidiol-mediated immunomodulatory and antineuroinflammatory activities that may serve to prevent neurodegeneration.
Collapse
Affiliation(s)
- Eun-Ju Yang
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea
| | - Kyung-Sik Song
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
26
|
Nelson LH, Peketi P, Lenz KM. Microglia Regulate Cell Genesis in a Sex-dependent Manner in the Neonatal Hippocampus. Neuroscience 2020; 453:237-255. [PMID: 33129890 DOI: 10.1016/j.neuroscience.2020.10.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 10/03/2020] [Accepted: 10/06/2020] [Indexed: 01/15/2023]
Abstract
Microglia, the innate immune cells of the brain, regulate brain development through many processes such as synaptic pruning, supporting cell genesis and phagocytosing living and dying cells. There are sex differences in these same developmental processes throughout the brain, thus microglia may contribute to brain sex differences. We examined whether microglia support a known sex difference in neonatal hippocampal neurogenesis and whether juvenile hippocampal neurogenesis was impacted by the loss of neonatal microglia. We used central infusion of liposomal clodronate to selectively deplete microglia and found decreased cell genesis in the male, but not female, dentate gyrus and hippocampus. We found that loss of microglia decreased cell genesis in the cortex and amygdala of both males and females. We assessed the expression of several cytokines and growth factors that have previously been shown to support cell genesis. We found that expression of Il1b and Tnf were decreased in the hippocampus due to microglia depletion however, there were no sex differences in the expression of any immune genes. In adolescence, there was an increase in the number of mitotic cells in the subgranular zone of the dentate gyrus of previously microglia depleted rats however, the number of newly-born neurons was unchanged in the adolescent animals. We also sought to determine whether there was a sex difference in the number of progenitor cells in the dentate gyrus in the neonatal period. We found no sex differences in the number of progenitor cells. Overall, these studies show that microglia are important for regulating region-specific sex differences in cell genesis in the developing brain.
Collapse
Affiliation(s)
- Lars H Nelson
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH, United States.
| | - Pavan Peketi
- Department of Psychology, The Ohio State University, Columbus, OH, United States
| | - Kathryn M Lenz
- Department of Neuroscience, The Ohio State University, Columbus, OH, United States; Department of Psychology, The Ohio State University, Columbus, OH, United States; Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
27
|
Pinto MV, Fernandes A. Microglial Phagocytosis-Rational but Challenging Therapeutic Target in Multiple Sclerosis. Int J Mol Sci 2020; 21:ijms21175960. [PMID: 32825077 PMCID: PMC7504120 DOI: 10.3390/ijms21175960] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 08/12/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022] Open
Abstract
Multiple sclerosis (MS) is the most common autoimmune and demyelinating disease of the central nervous system (CNS), characterized, in the majority of cases, by initial relapses that later evolve into progressive neurodegeneration, severely impacting patients’ motor and cognitive functions. Despite the availability of immunomodulatory therapies effective to reduce relapse rate and slow disease progression, they all failed to restore CNS myelin that is necessary for MS full recovery. Microglia are the primary inflammatory cells present in MS lesions, therefore strongly contributing to demyelination and lesion extension. Thus, many microglial-based therapeutic strategies have been focused on the suppression of microglial pro-inflammatory phenotype and neurodegenerative state to reduce disease severity. On the other hand, the contribution of myelin phagocytosis advocating the neuroprotective role of microglia in MS has been less explored. Indeed, despite the presence of functional oligodendrocyte precursor cells (OPCs), within lesioned areas, MS plaques fail to remyelinate as a result of the over-accumulation of myelin-toxic debris that must be cleared away by microglia. Dysregulation of this process has been associated with the impaired neuronal recovery and deficient remyelination. In line with this, here we provide a comprehensive review of microglial myelin phagocytosis and its involvement in MS development and repair. Alongside, we discuss the potential of phagocytic-mediated therapeutic approaches and encourage their modulation as a novel and rational approach to ameliorate MS-associated pathology.
Collapse
Affiliation(s)
- Maria V. Pinto
- Neuron-Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal;
| | - Adelaide Fernandes
- Neuron-Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal;
- Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal
- Correspondence: ; Tel.: +351-217946400
| |
Collapse
|
28
|
NF-κB Activation Accounts for the Cytoprotective Effects of PERK Activation on Oligodendrocytes during EAE. J Neurosci 2020; 40:6444-6456. [PMID: 32661025 DOI: 10.1523/jneurosci.1156-20.2020] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/22/2020] [Accepted: 06/30/2020] [Indexed: 01/09/2023] Open
Abstract
Previous studies demonstrate that activation of pancreatic ER kinase (PERK) protects oligodendrocytes against inflammation in the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis (MS). Interestingly, data indicate that the cytoprotective effects of PERK activation on oligodendrocytes during EAE are not mediated by activating transcription factor 4 (ATF4) but are accompanied by activation of nuclear factor κB (NF-κB). NF-κB plays a critical role in MS and EAE; however, the effects of NF-κB activation on oligodendrocytes in these diseases remain elusive. Herein, we generated a mouse model that allow for activation of NF-κB specifically in oligodendrocytes and found that enhanced NF-κB activation in oligodendrocytes had a minimal effect on their viability and function under normal conditions (both male and female mice). Interestingly, we found that enhanced NF-κB activation in oligodendrocytes attenuated EAE disease severity and ameliorated EAE-induced oligodendrocyte loss, demyelination, and axon degeneration, without affecting inflammation (female mice). Moreover, we showed that the detrimental effects of PERK inactivation in oligodendrocytes in EAE were accompanied by impaired NF-κB activation in oligodendrocytes, and were completely rescued by enhanced NF-κB activation in oligodendrocytes (female mice). These findings suggest that NF-κB activation accounts for the cytoprotective effects of PERK activation on oligodendrocytes in MS and EAE.SIGNIFICANCE STATEMENT Nuclear factor κB (NF-κB) is activated in oligodendrocytes in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE); however, the role of NF-κB activation in oligodendrocytes in MS and EAE remains elusive. Herein, we generated a mouse model that allows for activation of NF-κB selectively in oligodendrocytes and demonstrated that NF-κB activation prevented oligodendrocyte death and myelin damage in the EAE model. We further demonstrated that NF-κB activation contributed to the protective effects of pancreatic ER kinase (PERK) activation on oligodendrocytes in the EAE model. As such, this work will facilitate the development of new treatments that enhance oligodendrocyte survival in MS patients by targeting the PERK-NF-κB pathway.
Collapse
|
29
|
Marsters CM, Nesan D, Far R, Klenin N, Pittman QJ, Kurrasch DM. Embryonic microglia influence developing hypothalamic glial populations. J Neuroinflammation 2020; 17:146. [PMID: 32375817 PMCID: PMC7201702 DOI: 10.1186/s12974-020-01811-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/13/2020] [Indexed: 11/15/2022] Open
Abstract
Background Although historically microglia were thought to be immature in the fetal brain, evidence of purposeful interactions between these immune cells and nearby neural progenitors is becoming established. Here, we examined the influence of embryonic microglia on gliogenesis within the developing tuberal hypothalamus, a region later important for energy balance, reproduction, and thermoregulation. Methods We used immunohistochemistry to quantify the location and numbers of glial cells in the embryonic brain (E13.5–E17.5), as well as a pharmacological approach (i.e., PLX5622) to knock down fetal microglia. We also conducted cytokine and chemokine analyses on embryonic brains in the presence or absence of microglia, and a neurosphere assay to test the effects of the altered cytokines on hypothalamic progenitor behaviors. Results We identified a subpopulation of activated microglia that congregated adjacent to the third ventricle alongside embryonic Olig2+ neural progenitor cells (NPCs) that are destined to give rise to oligodendrocyte and astrocyte populations. In the absence of microglia, we observed an increase in Olig2+ glial progenitor cells that remained at the ventricle by E17.5 and a concomitant decrease of these Olig2+ cells in the mantle zone, indicative of a delay in migration of these precursor cells. A further examination of maturing oligodendrocytes in the hypothalamic grey and white matter area in the absence of microglia revealed migrating oligodendrocyte progenitor cells (OPCs) within the grey matter at E17.5, a time point when OPCs begin to slow their migration. Finally, quantification of cytokine and chemokine signaling in ex vivo E15.5 hypothalamic cultures +/− microglia revealed decreases in the protein levels of several cytokines in the absence of microglia. We assayed the influence of two downregulated cytokines (CCL2 and CXCL10) on neurosphere-forming capacity and lineage commitment of hypothalamic NPCs in culture and showed an increase in NPC proliferation as well as neuronal and oligodendrocyte differentiation. Conclusion These data demonstrate that microglia influence gliogenesis in the developing tuberal hypothalamus.
Collapse
Affiliation(s)
- Candace M Marsters
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Dinushan Nesan
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Rena Far
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Natalia Klenin
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Quentin J Pittman
- Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Deborah M Kurrasch
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada. .,Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada. .,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
30
|
Traiffort E, Kassoussi A, Zahaf A, Laouarem Y. Astrocytes and Microglia as Major Players of Myelin Production in Normal and Pathological Conditions. Front Cell Neurosci 2020; 14:79. [PMID: 32317939 PMCID: PMC7155218 DOI: 10.3389/fncel.2020.00079] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 03/19/2020] [Indexed: 12/13/2022] Open
Abstract
Myelination is an essential process that consists of the ensheathment of axons by myelin. In the central nervous system (CNS), myelin is synthesized by oligodendrocytes. The proliferation, migration, and differentiation of oligodendrocyte precursor cells constitute a prerequisite before mature oligodendrocytes extend their processes around the axons and progressively generate a multilamellar lipidic sheath. Although myelination is predominately driven by oligodendrocytes, the other glial cells including astrocytes and microglia, also contribute to this process. The present review is an update of the most recent emerging mechanisms involving astrocyte and microglia in myelin production. The contribution of these cells will be first described during developmental myelination that occurs in the early postnatal period and is critical for the proper development of cognition and behavior. Then, we will report the novel findings regarding the beneficial or deleterious effects of astroglia and microglia, which respectively promote or impair the endogenous capacity of oligodendrocyte progenitor cells (OPCs) to induce spontaneous remyelination after myelin loss. Acute delineation of astrocyte and microglia activities and cross-talk should uncover the way towards novel therapeutic perspectives aimed at recovering proper myelination during development or at breaking down the barriers impeding the regeneration of the damaged myelin that occurs in CNS demyelinating diseases.
Collapse
Affiliation(s)
| | | | - Amina Zahaf
- U1195 Inserm, University Paris-Saclay, Kremlin-Bicêtre, France
| | - Yousra Laouarem
- U1195 Inserm, University Paris-Saclay, Kremlin-Bicêtre, France
| |
Collapse
|
31
|
Immune cell regulation of glia during CNS injury and disease. Nat Rev Neurosci 2020; 21:139-152. [PMID: 32042145 DOI: 10.1038/s41583-020-0263-9] [Citation(s) in RCA: 146] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2020] [Indexed: 12/13/2022]
|
32
|
Abstract
Microglia are resident macrophages of the CNS that are involved in its development, homeostasis and response to infection and damage. Microglial activation is a common feature of neurological disorders, and although in some instances this activation can be damaging, protective and regenerative functions of microglia have been revealed. The most prominent example of the regenerative functions is a role for microglia in supporting regeneration of myelin after injury, a process that is critical for axonal health and relevant to numerous disorders in which loss of myelin integrity is a prevalent feature, such as multiple sclerosis, Alzheimer disease and motor neuron disease. Although drugs that are intended to promote remyelination are entering clinical trials, the mechanisms by which remyelination is controlled and how microglia are involved are not completely understood. In this Review, we discuss work that has identified novel regulators of microglial activation - including molecular drivers, population heterogeneity and turnover - that might influence their pro-remyelination capacity. We also discuss therapeutic targeting of microglia as a potential approach to promoting remyelination.
Collapse
|
33
|
Pitman KA, Ricci R, Gasperini R, Beasley S, Pavez M, Charlesworth J, Foa L, Young KM. The voltage-gated calcium channel CaV1.2 promotes adult oligodendrocyte progenitor cell survival in the mouse corpus callosum but not motor cortex. Glia 2019; 68:376-392. [PMID: 31605513 PMCID: PMC6916379 DOI: 10.1002/glia.23723] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 09/02/2019] [Accepted: 09/10/2019] [Indexed: 12/21/2022]
Abstract
Throughout life, oligodendrocyte progenitor cells (OPCs) proliferate and differentiate into myelinating oligodendrocytes. OPCs express cell surface receptors and channels that allow them to detect and respond to neuronal activity, including voltage‐gated calcium channel (VGCC)s. The major L‐type VGCC expressed by developmental OPCs, CaV1.2, regulates their differentiation. However, it is unclear whether CaV1.2 similarly influences OPC behavior in the healthy adult central nervous system (CNS). To examine the role of CaV1.2 in adulthood, we conditionally deleted this channel from OPCs by administering tamoxifen to P60 Cacna1cfl/fl (control) and Pdgfrα‐CreER:: Cacna1cfl/fl (CaV1.2‐deleted) mice. Whole cell patch clamp analysis revealed that CaV1.2 deletion reduced L‐type voltage‐gated calcium entry into adult OPCs by ~60%, confirming that it remains the major L‐type VGCC expressed by OPCs in adulthood. The conditional deletion of CaV1.2 from adult OPCs significantly increased their proliferation but did not affect the number of new oligodendrocytes produced or influence the length or number of internodes they elaborated. Unexpectedly, CaV1.2 deletion resulted in the dramatic loss of OPCs from the corpus callosum, such that 7 days after tamoxifen administration CaV1.2‐deleted mice had an OPC density ~42% that of control mice. OPC density recovered within 2 weeks of CaV1.2 deletion, as the lost OPCs were replaced by surviving CaV1.2‐deleted OPCs. As OPC density was not affected in the motor cortex or spinal cord, we conclude that calcium entry through CaV1.2 is a critical survival signal for a subpopulation of callosal OPCs but not for all OPCs in the mature CNS.
Collapse
Affiliation(s)
- Kimberley A Pitman
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Raphael Ricci
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Robert Gasperini
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia.,School of Medicine, University of Tasmania, Hobart, Australia
| | - Shannon Beasley
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Macarena Pavez
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Jac Charlesworth
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| | - Lisa Foa
- School of Medicine, University of Tasmania, Hobart, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia
| |
Collapse
|
34
|
Rutherford HA, Hamilton N. Animal models of leukodystrophy: a new perspective for the development of therapies. FEBS J 2019; 286:4176-4191. [DOI: 10.1111/febs.15060] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/31/2019] [Accepted: 09/09/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Holly A. Rutherford
- The Bateson Centre, Department of Infection, Immunity and Cardiovascular Disease University of Sheffield UK
| | - Noémie Hamilton
- The Bateson Centre, Department of Infection, Immunity and Cardiovascular Disease University of Sheffield UK
| |
Collapse
|
35
|
Chen T, Noto D, Hoshino Y, Mizuno M, Miyake S. Butyrate suppresses demyelination and enhances remyelination. J Neuroinflammation 2019; 16:165. [PMID: 31399117 PMCID: PMC6688239 DOI: 10.1186/s12974-019-1552-y] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 07/24/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The association of gut microbiota and diseases of the central nervous system (CNS), including multiple sclerosis (MS), has attracted much attention. Although a previous analysis of MS gut microbiota revealed a reduction in species producing short-chain fatty acids (SCFAs), the influence of these metabolites on demyelination and remyelination, the critical factors of MS pathogenesis, remains unclear. METHODS To investigate the relationship between demyelination and gut microbiota, we administered a mixture of non-absorbing antibiotics or SCFAs to mice with cuprizone-induced demyelination and evaluated demyelination and the accumulation of microglia. To analyze the direct effect of SCFAs on demyelination or remyelination, we induced demyelination in an organotypic cerebellar slice culture using lysolecithin and analyzed the demyelination and maturation of oligodendrocyte precursor cells with or without SCFA treatment. RESULTS The oral administration of antibiotics significantly enhanced cuprizone-induced demyelination. The oral administration of butyrate significantly ameliorated demyelination, even though the accumulation of microglia into demyelinated lesions was not affected. Furthermore, we showed that butyrate treatment significantly suppressed lysolecithin-induced demyelination and enhanced remyelination in an organotypic slice culture in the presence or absence of microglia, suggesting that butyrate may affect oligodendrocytes directly. Butyrate treatment facilitated the differentiation of immature oligodendrocytes. CONCLUSIONS We revealed that treatment with butyrate suppressed demyelination and enhanced remyelination in an organotypic slice culture in association with facilitating oligodendrocyte differentiation. Our findings shed light on a novel mechanism of interaction between the metabolites of gut microbiota and the CNS and may provide a strategy to control demyelination and remyelination in MS.
Collapse
Affiliation(s)
- Tong Chen
- Department of Immunology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Daisuke Noto
- Department of Immunology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan.
| | - Yasunobu Hoshino
- Department of Immunology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
- Department of Neurology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Miho Mizuno
- Department of Immunology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Sachiko Miyake
- Department of Immunology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
36
|
Gupta AS, Biswas DD, Brown LSN, Mockenhaupt K, Marone M, Hoskins A, Siebenlist U, Kordula T. A detrimental role of RelB in mature oligodendrocytes during experimental acute encephalomyelitis. J Neuroinflammation 2019; 16:161. [PMID: 31362762 PMCID: PMC6664766 DOI: 10.1186/s12974-019-1548-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 07/17/2019] [Indexed: 12/31/2022] Open
Abstract
Background Multiple sclerosis (MS) is an autoimmune demyelinating disease of the central nervous system (CNS). It is firmly established that overactivation of the p65 (RelA) nuclear factor kappa B (NF-κB) transcription factor upregulates expression of inflammatory mediators in both immune and non-immune resident CNS cells and promotes inflammation during MS. In contrast to p65, NF-κB family member RelB regulates immune cell development and can limit inflammation. Although RelB expression is induced during inflammation in the CNS, its role in MS remains unknown. Methods To examine the role of RelB in non-immune CNS cells, we generated mice with RelB specifically deleted in astrocytes (RelBΔAST), oligodendrocytes (RelBΔOLIGO), or neural progenitor-derived cells (RelBΔNP). We used experimental autoimmune encephalomyelitis (EAE), an accepted mouse model of MS, to assess the effect of RelB deletion on disease outcomes and performed analysis on the histological, cellular, and molecular level. Results Despite being a negative regulator of inflammation, conditional knockout of RelB in non-immune resident CNS cells surprisingly decreased the severity of EAE. This protective effect was recapitulated by conditional deletion of RelB in oligodendrocytes but not astrocytes. Deletion of RelB in oligodendrocytes reduced disease severity, promoted survival of mature oligodendrocytes, and correlated with increased activation of p65 NF-κB. Conclusions These findings suggest that RelB fine tunes inflammation and cell death/survival during EAE. Importantly, our data points out the detrimental role RelB plays in controlling survival of mature oligodendrocytes, which could be explored as a viable option to treat MS in the future. Electronic supplementary material The online version of this article (10.1186/s12974-019-1548-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Angela S Gupta
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, School of Medicine and the Massey Cancer Center, Richmond, VA, 23298, USA
| | - Debolina D Biswas
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, School of Medicine and the Massey Cancer Center, Richmond, VA, 23298, USA
| | - La Shardai N Brown
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, School of Medicine and the Massey Cancer Center, Richmond, VA, 23298, USA
| | - Karli Mockenhaupt
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, School of Medicine and the Massey Cancer Center, Richmond, VA, 23298, USA
| | - Michael Marone
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, School of Medicine and the Massey Cancer Center, Richmond, VA, 23298, USA
| | - Andrew Hoskins
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, School of Medicine and the Massey Cancer Center, Richmond, VA, 23298, USA
| | - Ulrich Siebenlist
- Laboratory of Molecular Immunology, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Tomasz Kordula
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, School of Medicine and the Massey Cancer Center, Richmond, VA, 23298, USA.
| |
Collapse
|
37
|
Schoor C, Brocke-Ahmadinejad N, Gieselmann V, Winter D. Investigation of Oligodendrocyte Precursor Cell Differentiation by Quantitative Proteomics. Proteomics 2019; 19:e1900057. [PMID: 31216117 DOI: 10.1002/pmic.201900057] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 06/02/2019] [Indexed: 01/20/2023]
Abstract
Oligodendrocytes, the myelinating cells of the central nervous system, are essential for correct brain function. They originate from oligodendrocyte precursor cells through a differentiation process which is only incompletely understood and impaired in a variety of demyelinating diseases. Better knowledge of this differentiation holds the promise to develop novel therapies for these disorders. The differentiation of rat oligodendrocyte precursor cells to oligodendrocytes in vitro is investigated. After confirmation of differentiation by immunohistochemical analysis using cell type-specific marker proteins, a quantitative proteomics study using tandem mass tags (TMT) is conducted. Four time points of differentiation covering early, intermediate, and late stages are investigated. Data analysis by Mascot and MaxQuant identified 5259 protein groups of which 471 are not described in the context of cells of the oligodendroglial lineage before. Quantitative analysis of the dataset revealed distinct regulation patterns for proteins of different functional categories including metabolic processes, regulation of the cell cycle, and transcriptional control of protein expression. The present data confirm a significant number of proteins known to play a role in oligodendrocytes and myelination. Furthermore, novel candidate proteins are identified which may play an important role in this differentiation process providing a valuable resource for oligodendrocyte research.
Collapse
Affiliation(s)
- Carmen Schoor
- Institute for Biochemistry and Molecular Biology, University of Bonn, 53115, Bonn, Germany
| | | | - Volkmar Gieselmann
- Institute for Biochemistry and Molecular Biology, University of Bonn, 53115, Bonn, Germany
| | - Dominic Winter
- Institute for Biochemistry and Molecular Biology, University of Bonn, 53115, Bonn, Germany
| |
Collapse
|
38
|
Protective Role of NF-κB in Inflammatory Demyelination. J Neurosci 2019; 38:2416-2417. [PMID: 30995609 DOI: 10.1523/jneurosci.3286-17.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 01/23/2018] [Accepted: 01/26/2018] [Indexed: 01/21/2023] Open
|
39
|
Yang EJ, Song IS, Song KS. Ethanol extract of Glycyrrhizae Radix modulates the responses of antigen-specific splenocytes in experimental autoimmune encephalomyelitis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 54:56-65. [PMID: 30668383 DOI: 10.1016/j.phymed.2018.09.189] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/22/2018] [Accepted: 09/17/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) is an autoimmune disorder resulting in paralysis, and the responses of reactive T cells against self-antigens are hallmarks. Glycyrrhizae Radix (GR) has been used for detoxification and reducing inflammation. However, very few reports have described the effects of GR on MS. PURPOSE The immunomodulatory effects of GR extract on autoimmune responses were evaluated through in vitro, ex vivo, and in vivo assays using primary mouse splenocytes (SPLC), mouse microglia BV2 cell line, and a mouse model of experimental autoimmune encephalomyelitis (EAE). STUDY DESIGN Ethanol extract of GR was used in vitro with primary SPLC in the condition of anti-CD3/CD28 stimulation and interferon (IFN)-γ-producing CD4+ (TH1)/CD8+ (TC1) polarization as well as IFN-γ-stimulated BV2 cells. For EAE induction, female C57BL/6 mice were immunized with 200 μg of myelin oligodendrocyte glycoprotein (MOG)35-55 without pertussis toxin. EAE SPLC (ex vivo) and EAE mice (in vivo) were treated with GR extract to evaluate the changes in antigen-specific responses. SPLC media containing antigen-specific responses were used to stimulate BV2 cells. RESULTS GR extract effectively modulated the responses of reactive splenic T cells through the reduction in IFN-γ+ T cell populations, the expressions of cell adhesion molecules (CAMs), and secretions of cytokines containing IFN-γ and a chemokine IFN-γ-induced protein 10 (IP-10) in vitro. In addition, GR extract significantly decreased nitric oxide production and secretion of tumor necrosis factor (TNF)-α and IP-10 in IFN-γ-stimulated BV2 cells. The antigen-specific TH1 and TC1 populations were decreased following administration of 100 mg/kg of GR extract, whereas CD8+IL-17A+ (TC17) population was increased on day 36 after EAE induction. Moreover, IFN-γ, which showed the highest secretion among examined cytokines, and IP-10 decreased on day 36. SPLC media derived from 100 mg/kg GR extract-administered EAE mice revealed the ameliorative effects on BV2 cell stimulation. CONCLUSION This is the first report on the immunomodulatory effects of GR extract on antigen-specific SPLC responses in EAE. These results could be helpful for the discovery of drug candidates for MS by focusing on IFN-γ-related autoimmune responses.
Collapse
Affiliation(s)
- Eun-Ju Yang
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, 80 Daehak-ro, Daegu, 41566, Republic of Korea.
| | - Im-Sook Song
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, 80 Daehak-ro, Daegu, 41566, Republic of Korea
| | - Kyung-Sik Song
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, 80 Daehak-ro, Daegu, 41566, Republic of Korea.
| |
Collapse
|
40
|
Sekiya K, Nishihara T, Abe N, Konishi A, Nandate H, Hamada T, Ikemune K, Takasaki Y, Tanaka J, Asano M, Yorozuya T. Carbon monoxide poisoning-induced delayed encephalopathy accompanies decreased microglial cell numbers: Distinctive pathophysiological features from hypoxemia-induced brain damage. Brain Res 2018; 1710:22-32. [PMID: 30578768 DOI: 10.1016/j.brainres.2018.12.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/15/2018] [Accepted: 12/18/2018] [Indexed: 11/15/2022]
Abstract
Carbon monoxide (CO) causes not only acute fatal poisoning but also may cause a delayed neurologic syndrome called delayed encephalopathy (DE), which occasionally occurs after an interval of several days to several weeks post-exposure. However, the mechanisms of DE have not been fully elucidated. This study aimed to clarify the pathophysiology of CO-induced DE and its distinctive features compared with hypoxemic hypoxia. Rats were randomly assigned to three groups; the air group, the CO group (exposed to CO), and the low O2 group (exposed to low concentration of O2). Impairment of memory function was observed only in the CO group. The hippocampus tissues were collected and analyzed for assessment of CO-induced changes and microglial reaction. Demyelination was observed only in the CO group and it was more severe and persisted longer than that observed in the low O2 group. Moreover, in the CO group, decreased in microglial cell numbers were observed using flow cytometry, and microglia with detached branches were observed were observed using immunohistochemistry. Conversely, microglial cells with shortened branches and enlarged somata were observed in the low O2 group. Furthermore, mRNAs encoding several neurotrophic factors expressed by microglia were decreased in the CO group but were increased in the low O2 group. Thus, CO-induced DE displayed distinctive pathological features from those of simple hypoxic insults: prolonged demyelination accompanying a significant decrease in microglial cells. Decreased neurotrophic factor expression by microglial cells may be one of the causes of CO-induced DE.
Collapse
Affiliation(s)
- Keisuke Sekiya
- Department of Anesthesia and Perioperative Medicine, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan; Department of Legal Medicine, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Tasuku Nishihara
- Department of Anesthesia and Perioperative Medicine, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan.
| | - Naoki Abe
- Department of Anesthesia and Perioperative Medicine, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Amane Konishi
- Department of Anesthesia and Perioperative Medicine, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan.
| | - Hideyuki Nandate
- Department of Anesthesia and Perioperative Medicine, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Taisuke Hamada
- Department of Anesthesia and Perioperative Medicine, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Keizo Ikemune
- Department of Anesthesia and Perioperative Medicine, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan.
| | - Yasushi Takasaki
- Department of Anesthesia and Perioperative Medicine, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan
| | - Junya Tanaka
- Department of Molecular and Cellular Physiology, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan.
| | - Migiwa Asano
- Department of Legal Medicine, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan.
| | - Toshihiro Yorozuya
- Department of Anesthesia and Perioperative Medicine, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan.
| |
Collapse
|
41
|
Wang F, Cao Y, Ma L, Pei H, Rausch WD, Li H. Dysfunction of Cerebrovascular Endothelial Cells: Prelude to Vascular Dementia. Front Aging Neurosci 2018; 10:376. [PMID: 30505270 PMCID: PMC6250852 DOI: 10.3389/fnagi.2018.00376] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/29/2018] [Indexed: 12/19/2022] Open
Abstract
Vascular dementia (VaD) is the second most common type of dementia after Alzheimer's disease (AD), characterized by progressive cognitive impairment, memory loss, and thinking or speech problems. VaD is usually caused by cerebrovascular disease, during which, cerebrovascular endothelial cells (CECs) are vulnerable. CEC dysfunction occurs before the onset of VaD and can eventually lead to dysregulation of cerebral blood flow and blood-brain barrier damage, followed by the activation of glia and inflammatory environment in the brain. White matter, neuronal axons, and synapses are compromised in this process, leading to cognitive impairment. The present review summarizes the mechanisms underlying CEC impairment during hypoperfusion and pathological role of CECs in VaD. Through the comprehensive examination and summarization, endothelial nitric oxide synthase (eNOS)/nitric oxide (NO) signaling pathway, Ras homolog gene family member A (RhoA) signaling pathway, and CEC-derived caveolin-1 (CAV-1) are proposed to serve as targets of new drugs for the treatment of VaD.
Collapse
Affiliation(s)
- Feixue Wang
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Cao
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Lina Ma
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Hui Pei
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Wolf Dieter Rausch
- Department for Biomedical Sciences, Institute of Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Hao Li
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
42
|
Wellman SM, Cambi F, Kozai TD. The role of oligodendrocytes and their progenitors on neural interface technology: A novel perspective on tissue regeneration and repair. Biomaterials 2018; 183:200-217. [PMID: 30172245 PMCID: PMC6469877 DOI: 10.1016/j.biomaterials.2018.08.046] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 08/08/2018] [Accepted: 08/20/2018] [Indexed: 12/11/2022]
Abstract
Oligodendrocytes and their precursors are critical glial facilitators of neurophysiology, which is responsible for cognition and behavior. Devices that are used to interface with the brain allow for a more in-depth analysis of how neurons and these glia synergistically modulate brain activity. As projected by the BRAIN Initiative, technologies that acquire a high resolution and robust sampling of neural signals can provide a greater insight in both the healthy and diseased brain and support novel discoveries previously unobtainable with the current state of the art. However, a complex series of inflammatory events triggered during device insertion impede the potential applications of implanted biosensors. Characterizing the biological mechanisms responsible for the degradation of intracortical device performance will guide novel biomaterial and tissue regenerative approaches to rehabilitate the brain following injury. Glial subtypes which assist with neuronal survival and exchange of electrical signals, mainly oligodendrocytes, their precursors, and the insulating myelin membranes they produce, are sensitive to inflammation commonly induced from insults to the brain. This review explores essential physiological roles facilitated by oligodendroglia and their precursors and provides insight into their pathology following neurodegenerative injury and disease. From this knowledge, inferences can be made about the impact of device implantation on these supportive glia in order to engineer effective strategies that can attenuate their responses, enhance the efficacy of neural interfacing technology, and provide a greater understanding of the challenges that impede wound healing and tissue regeneration during pathology.
Collapse
Affiliation(s)
- Steven M Wellman
- Department of Bioengineering, University of Pittsburgh, USA; Center for the Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Franca Cambi
- Veterans Administration Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, USA
| | - Takashi Dy Kozai
- Department of Bioengineering, University of Pittsburgh, USA; Center for the Neural Basis of Cognition, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, USA; McGowan Institute of Regenerative Medicine, University of Pittsburgh, USA; NeuroTech Center, University of Pittsburgh Brain Institute, USA.
| |
Collapse
|
43
|
Hammond TR, Robinton D, Stevens B. Microglia and the Brain: Complementary Partners in Development and Disease. Annu Rev Cell Dev Biol 2018; 34:523-544. [PMID: 30089221 DOI: 10.1146/annurev-cellbio-100616-060509] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
An explosion of findings driven by powerful new technologies has expanded our understanding of microglia, the resident immune cells of the central nervous system (CNS). This wave of discoveries has fueled a growing interest in the roles that these cells play in the development of the CNS and in the neuropathology of a diverse array of disorders. In this review, we discuss the crucial roles that microglia play in shaping the brain-from their influence on neurons and glia within the developing CNS to their roles in synaptic maturation and brain wiring-as well as some of the obstacles to overcome when assessing their contributions to normal brain development. Furthermore, we examine how normal developmental functions of microglia are perturbed or remerge in neurodevelopmental and neurodegenerative disease.
Collapse
Affiliation(s)
- Timothy R Hammond
- FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA;
| | - Daisy Robinton
- FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA;
| | - Beth Stevens
- FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA; .,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
44
|
Singh DK, Ling EA, Kaur C. Hypoxia and myelination deficits in the developing brain. Int J Dev Neurosci 2018; 70:3-11. [PMID: 29964158 DOI: 10.1016/j.ijdevneu.2018.06.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 05/28/2018] [Accepted: 06/26/2018] [Indexed: 12/15/2022] Open
Abstract
Myelination is a complex and orderly process during brain development that is essential for normal motor, cognitive and sensory functions. Cellular and molecular interactions between myelin-forming oligodendrocytes and axons are required for normal myelination in the developing brain. Oligodendrocyte progenitor cells (OPCs) proliferate and differentiate into mature myelin-forming oligodendrocytes. In this connection, astrocytes and microglia are also involved in survival and proliferation of OPCs. Hypoxic insults during the perinatal period affect the normal development, differentiation and maturation of the OPCs or cause their death resulting in impaired myelination. Several factors such as augmented release of proinflammatory cytokines by activated microglia and astrocytes, extracellular accumulation of excess glutamate and increased levels of nitric oxide are some of the underlying factors for hypoxia induced damage to the OPCs. Additionally, hypoxia also leads to down-regulation of several genes involved in oligodendrocyte differentiation encoding proteolipid protein, platelet-derived growth factor receptor and myelin-associated glycoprotein in the developing brain. Furthermore, oligodendrocytes may also accumulate increased amounts of iron in hypoxic conditions that triggers endoplasmic reticulum stress, misfolding of proteins and generation of reactive oxygen species that ultimately would lead to myelination deficits. More in-depth studies to elucidate the pathophysiological mechanisms underlying the inability of oligodendrocytes to myelinate the developing brain in hypoxic insults are desirable to develop new therapeutic options or strategies for myelination deficits.
Collapse
Affiliation(s)
- Dhiraj Kumar Singh
- Department of Anatomy, Yong Loo Lin School of Medicine, MD10, 4 Medical drive, National University of Singapore, 117597, Singapore
| | - Eng-Ang Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, MD10, 4 Medical drive, National University of Singapore, 117597, Singapore
| | - Charanjit Kaur
- Department of Anatomy, Yong Loo Lin School of Medicine, MD10, 4 Medical drive, National University of Singapore, 117597, Singapore.
| |
Collapse
|
45
|
Molecular Mechanisms of Oligodendrocyte Regeneration in White Matter-Related Diseases. Int J Mol Sci 2018; 19:ijms19061743. [PMID: 29895784 PMCID: PMC6032201 DOI: 10.3390/ijms19061743] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/25/2018] [Accepted: 06/06/2018] [Indexed: 12/13/2022] Open
Abstract
Even in adult brains, restorative mechanisms are still retained to maintain the microenvironment. Under the pathological conditions of central nervous system (CNS) diseases, several immature cells in the brain would be activated as a compensative response. As the concept of the neurovascular unit emphasizes, cell-cell interactions play important roles in this restorative process. White matter damage and oligodendrocyte loss are representative characteristics for many neurodegenerative diseases. In response to oligodendrocyte damage, residual oligodendrocyte precursor cells (OPCs) initiate their proliferation and differentiation for the purpose of remyelination. Although mechanisms of oligodendrogenesis and remyelination in CNS diseases are still mostly unknown and understudied, accumulated evidence now suggests that support from neighboring cells is necessary for OPC proliferation and differentiation. In this review, we first overview basic mechanisms of interaction between oligodendrocyte lineage cells and neighboring cells, and then introduce how oligodendrogenesis occurs under the conditions of neurodegenerative diseases, focusing on vascular cognitive impairment syndrome, Alzheimer’s disease, and multiple sclerosis.
Collapse
|
46
|
Sil S, Periyasamy P, Thangaraj A, Chivero ET, Buch S. PDGF/PDGFR axis in the neural systems. Mol Aspects Med 2018; 62:63-74. [PMID: 29409855 DOI: 10.1016/j.mam.2018.01.006] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 12/08/2017] [Accepted: 01/22/2018] [Indexed: 12/14/2022]
Abstract
Platelet-derived growth factors (PDGFs) and their receptors (PDGFRs) are expressed in several cell types including the brain cells such as neuronal progenitors, neurons, astrocytes, and oligodendrocytes. Emerging evidence shows that PDGF-mediated signaling regulates diverse functions in the central nervous system (CNS) such as neurogenesis, cell survival, synaptogenesis, modulation of ligand-gated ion channels, and development of specific types of neurons. Interestingly, PDGF/PDFGR signaling can elicit paradoxical roles in the CNS, depending on the cell type and the activation stimuli and is implicated in the pathogenesis of various neurodegenerative diseases. This review summarizes the role of PDGFs/PDGFRs in several neurodegenerative diseases such as Alzheimer disease, Parkinson disease, amyotrophic lateral sclerosis, brain cancer, cerebral ischemia, HIV-1 and drug abuse. Understanding PDGF/PDGFR signaling may lead to novel approaches for the future development of therapeutic strategies for combating CNS pathologies.
Collapse
Affiliation(s)
- Susmita Sil
- Department of Pharmacology and Experimental Neuroscience, 985880 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Palsamy Periyasamy
- Department of Pharmacology and Experimental Neuroscience, 985880 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Annadurai Thangaraj
- Department of Pharmacology and Experimental Neuroscience, 985880 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ernest T Chivero
- Department of Pharmacology and Experimental Neuroscience, 985880 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, 985880 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
47
|
Yue Y, Stone S, Lin W. Role of nuclear factor κB in multiple sclerosis and experimental autoimmune encephalomyelitis. Neural Regen Res 2018; 13:1507-1515. [PMID: 30127103 PMCID: PMC6126134 DOI: 10.4103/1673-5374.237109] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The transcription factor nuclear factor κB (NF-κB) plays major roles in inflammatory diseases through regulation of inflammation and cell viability. Multiple sclerosis (MS) is a chronic inflammatory demyelinating and neurodegenerative disease of the central nervous system (CNS). It has been shown that NF-κB is activated in multiple cell types in the CNS of MS patients, including T cells, microglia/macrophages, astrocytes, oligodendrocytes, and neurons. Interestingly, data from animal model studies, particularly studies of experimental autoimmune encephalomyelitis, have suggested that NF-κB activation in these individual cell types has distinct effects on the development of MS. In this review, we will cover the current literature on NF-κB and the evidence for its role in the development of MS and its animal model experimental autoimmune encephalomyelitis.
Collapse
Affiliation(s)
- Yuan Yue
- Department of Neuroscience; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Sarrabeth Stone
- Department of Neuroscience; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Wensheng Lin
- Department of Neuroscience; Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
48
|
Kaur C, Rathnasamy G, Ling EA. Biology of Microglia in the Developing Brain. J Neuropathol Exp Neurol 2017; 76:736-753. [PMID: 28859332 DOI: 10.1093/jnen/nlx056] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Microglia exist in different morphological forms in the developing brain. They show a small cell body with scanty cytoplasm with many branching processes in the grey matter of the developing brain. However, in the white matter such as the corpus callosum where the unmyelinated axons are loosely organized, they appear in an amoeboid form having a round cell body endowed with copious cytoplasm rich in organelles. The amoeboid cells eventually transform into ramified microglia in the second postnatal week when the tissue becomes more compact with the onset of myelination. Microglia serve as immunocompetent macrophages that act as neuropathology sensors to detect and respond swiftly to subtle changes in the brain tissues in pathological conditions. Microglial functions are broadly considered as protective in the normal brain development as they phagocytose dead cells and sculpt neuronal connections by pruning excess axons and synapses. They also secrete a number of trophic factors such as insulin-like growth factor-1 and transforming growth factor-β among many others that are involved in neuronal and oligodendrocyte survival. On the other hand, microglial cells when activated produce a plethora of molecules such as proinflammatory cytokines, chemokines, reactive oxygen species, and nitric oxide that are implicated in the pathogenesis of many pathological conditions such as epilepsy, cerebral palsy, autism, and perinatal hypoxic-ischemic brain injury. Although many studies have investigated the origin and functions of the microglia in the developing brain, in-depth in vivo studies along with analysis of their transcriptome and epigenetic changes need to be undertaken to elucidate their full potential be it protective or neurotoxic. This would lead to a better understanding of their roles in the healthy and diseased developing brain and advancement of therapeutic strategies to target microglia-mediated neurotoxicity.
Collapse
Affiliation(s)
- Charanjit Kaur
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; and Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Gurugirijha Rathnasamy
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; and Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| | - Eng-Ang Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; and Department of Ophthalmology and Visual Sciences, School of Medicine and Public Health, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
49
|
van der Knaap MS, Bugiani M. Leukodystrophies: a proposed classification system based on pathological changes and pathogenetic mechanisms. Acta Neuropathol 2017; 134:351-382. [PMID: 28638987 PMCID: PMC5563342 DOI: 10.1007/s00401-017-1739-1] [Citation(s) in RCA: 241] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 06/06/2017] [Accepted: 06/06/2017] [Indexed: 12/29/2022]
Abstract
Leukodystrophies are genetically determined disorders characterized by the selective involvement of the central nervous system white matter. Onset may be at any age, from prenatal life to senescence. Many leukodystrophies are degenerative in nature, but some only impair white matter function. The clinical course is mostly progressive, but may also be static or even improving with time. Progressive leukodystrophies are often fatal, and no curative treatment is known. The last decade has witnessed a tremendous increase in the number of defined leukodystrophies also owing to a diagnostic approach combining magnetic resonance imaging pattern recognition and next generation sequencing. Knowledge on white matter physiology and pathology has also dramatically built up. This led to the recognition that only few leukodystrophies are due to mutations in myelin- or oligodendrocyte-specific genes, and many are rather caused by defects in other white matter structural components, including astrocytes, microglia, axons and blood vessels. We here propose a novel classification of leukodystrophies that takes into account the primary involvement of any white matter component. Categories in this classification are the myelin disorders due to a primary defect in oligodendrocytes or myelin (hypomyelinating and demyelinating leukodystrophies, leukodystrophies with myelin vacuolization); astrocytopathies; leuko-axonopathies; microgliopathies; and leuko-vasculopathies. Following this classification, we illustrate the neuropathology and disease mechanisms of some leukodystrophies taken as example for each category. Some leukodystrophies fall into more than one category. Given the complex molecular and cellular interplay underlying white matter pathology, recognition of the cellular pathology behind a disease becomes crucial in addressing possible treatment strategies.
Collapse
Affiliation(s)
- Marjo S van der Knaap
- Department of Pediatrics/Child Neurology, VU University Medical Centre, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Functional Genomics, Centre for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU University, Amsterdam, The Netherlands
| | - Marianna Bugiani
- Department of Pediatrics/Child Neurology, VU University Medical Centre, Amsterdam Neuroscience, Amsterdam, The Netherlands.
- Department of Pathology, VU University Medical Centre, Amsterdam Neuroscience, Amsterdam, The Netherlands.
| |
Collapse
|
50
|
NF-κB Activation Protects Oligodendrocytes against Inflammation. J Neurosci 2017; 37:9332-9344. [PMID: 28842413 PMCID: PMC5607472 DOI: 10.1523/jneurosci.1608-17.2017] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/07/2017] [Accepted: 08/16/2017] [Indexed: 01/13/2023] Open
Abstract
NF-κB is a key player in inflammatory diseases, including multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). However, the effects of NF-κB activation on oligodendrocytes in MS and EAE remain unknown. We generated a mouse model that expresses IκBαΔN, a super-suppressor of NF-κB, specifically in oligodendrocytes and demonstrated that IκBαΔN expression had no effect on oligodendrocytes under normal conditions (both sexes). Interestingly, we showed that oligodendrocyte-specific expression of IκBαΔN blocked NF-κB activation in oligodendrocytes and resulted in exacerbated oligodendrocyte death and hypomyelination in young, developing mice that express IFN-γ ectopically in the CNS (both sexes). We also showed that NF-κB inactivation in oligodendrocytes aggravated IFN-γ-induced remyelinating oligodendrocyte death and remyelination failure in the cuprizone model (male mice). Moreover, we found that NF-κB inactivation in oligodendrocytes increased the susceptibility of mice to EAE (female mice). These findings imply the cytoprotective effects of NF-κB activation on oligodendrocytes in MS and EAE.SIGNIFICANCE STATEMENT Multiple sclerosis (MS) is an inflammatory demyelinating disease of the CNS. NF-κB is a major player in inflammatory diseases that acts by regulating inflammation and cell viability. Data indicate that NF-κB activation in inflammatory cells facilitates the development of MS. However, to date, attempts to understand the role of NF-κB activation in oligodendrocytes in MS have been unsuccessful. Herein, we generated a mouse model that allows for inactivation of NF-κB specifically in oligodendrocytes and then used this model to determine the precise role of NF-κB activation in oligodendrocytes in models of MS. The results presented in this study represent the first demonstration that NF-κB activation acts cell autonomously to protect oligodendrocytes against inflammation in animal models of MS.
Collapse
|