1
|
Van Hove H, De Feo D, Greter M, Becher B. Central Nervous System Macrophages in Health and Disease. Annu Rev Immunol 2025; 43:589-613. [PMID: 40036702 DOI: 10.1146/annurev-immunol-082423-041334] [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: 03/06/2025]
Abstract
The central nervous system (CNS) has a unique set of macrophages that seed the tissue early during embryonic development. Microglia reside in the parenchyma, and border-associated macrophages are present in border regions, including the meninges, perivascular spaces, and choroid plexus. CNS-resident macrophages support brain homeostasis during development and steady state. In the diseased brain, however, the immune landscape is altered, with phenotypic and transcriptional changes in resident macrophages and the invasion of blood-borne monocytes, which differentiate into monocyte-derived macrophages upon entering the CNS. In this review, we focus on the fate and function of the macrophage compartment in health, neurodegenerative conditions such as amyloidosis, and neuroinflammation as observed in multiple sclerosis and infection. We discuss our current understanding that monocyte-derived macrophages contribute to neuropathology whereas native macrophages play a neuroprotective role in disease.
Collapse
Affiliation(s)
- Hannah Van Hove
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland;
| | - Donatella De Feo
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland;
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland;
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland;
| |
Collapse
|
2
|
Montilla A, Zabala A, Calvo I, Bosch-Juan M, Tomé-Velasco I, Mata P, Koster M, Sierra A, Kooistra SM, Soria FN, Eggen BJL, Fresnedo O, Fernández JA, Tepavcevic V, Matute C, Domercq M. Microglia regulate myelin clearance and cholesterol metabolism after demyelination via interferon regulatory factor 5. Cell Mol Life Sci 2025; 82:131. [PMID: 40137979 PMCID: PMC11947375 DOI: 10.1007/s00018-025-05648-2] [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: 07/05/2024] [Revised: 12/16/2024] [Accepted: 01/08/2025] [Indexed: 03/29/2025]
Abstract
Interferon regulatory factor 5 (IRF5) is a transcription factor that plays a role in orchestrating innate immune responses, particularly in response to viral infections. Notably, IRF5 has been identified as a microglia risk gene linked to multiple sclerosis (MS), but its specific role in MS pathogenesis remains unclear. Through the use of Irf5-/- mice, our study uncovers a non-canonical function of IRF5 in MS recovery. Irf5-/- mice exhibited increased damage in an experimental autoimmune encephalomyelitis (EAE) model and demonstrated impaired oligodendrocyte recruitment into the lesion core following lysolecithin-induced demyelination. Transcriptomic and lipidomic analyses revealed that IRF5 has a role in microglia-mediated myelin phagocytosis, lipid metabolism, and cholesterol homeostasis. Indeed, Irf5-/- microglia phagocytose myelin, but myelin debris is not adequately degraded, leading to an accumulation of lipid droplets, cholesterol esters, and cholesterol crystals within demyelinating lesions. This abnormal buildup can hinder remyelination processes. Importantly, treatments that promote cholesterol transport were found to reduce lipid droplet accumulation and mitigate the exacerbated damage in Irf5-/- mice with EAE. Altogether, our study identified the antiviral transcription factor IRF5 as a key transcriptional regulator of lipid degradation and cholesterol homeostasis and suggest that loss of IRF5 function leads to pathogenic lipid accumulation in microglia, thereby obstructing remyelination. These data and the fact that Irf5 polymorphisms are significantly associated with MS, highlight IRF5 as a potential therapeutic target to promote regenerative responses.
Collapse
Affiliation(s)
- Alejandro Montilla
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain.
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain.
| | - Alazne Zabala
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Ibai Calvo
- Department of Physical Chemistry, Faculty of Sciences, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Marina Bosch-Juan
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Irene Tomé-Velasco
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Paloma Mata
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Mirjam Koster
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Amanda Sierra
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Ikerbasque Foundation, E-48009, Bilbao, Spain
- Department of Biochemistry and Molecular Biology, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Susanne M Kooistra
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Federico N Soria
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Ikerbasque Foundation, E-48009, Bilbao, Spain
| | - Bart J L Eggen
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Olatz Fresnedo
- Lipids & Liver Research Group, Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - José Andrés Fernández
- Department of Physical Chemistry, Faculty of Sciences, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Vanja Tepavcevic
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Carlos Matute
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain.
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain.
| | - María Domercq
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain.
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain.
| |
Collapse
|
3
|
Wu Y, Park J, Le QV, Byun J, Choi J, Xu E, Lee J, Oh YK. NET formation-mediated in situ protein delivery to the inflamed central nervous system. Nat Commun 2024; 15:10747. [PMID: 39737919 PMCID: PMC11686318 DOI: 10.1038/s41467-024-54817-7] [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: 04/08/2024] [Accepted: 11/18/2024] [Indexed: 01/01/2025] Open
Abstract
Delivering protein drugs to the central nervous system (CNS) is challenging due to the blood-brain and blood-spinal cord barrier. Here we show that neutrophils, which naturally migrate through these barriers to inflamed CNS sites and release neutrophil extracellular traps (NETs), can be leveraged for therapeutic delivery. Tannic acid nanoparticles tethered with anti-Ly6G antibody and interferon-β (aLy6G-IFNβ@TLP) are constructed for targeted neutrophil delivery. These nanoparticles protect interferon-β from reactive oxygen species and preferentially accumulate in neutrophils over other immune cells. Upon encountering inflammation, neutrophils release the nanoparticles during NET formation. In the female mouse model of experimental autoimmune encephalomyelitis, intravenous administration of aLy6G-IFNβ@TLP reduce disease progression and restore motor function. Although this study focuses on IFNβ and autoimmune encephalomyelitis, the concept of hitchhiking neutrophils for CNS delivery and employing NET formation for inflamed site-specific nanoparticle release can be further applied for delivery of other protein drugs in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yina Wu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jinwon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Quoc-Viet Le
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Junho Byun
- College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Jaehyun Choi
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Enzhen Xu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jaiwoo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
- College of Pharmacy, Korea University, Sejong, Republic of Korea.
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Nan Y, Ni S, Liu M, Hu K. The emerging role of microglia in the development and therapy of multiple sclerosis. Int Immunopharmacol 2024; 143:113476. [PMID: 39476566 DOI: 10.1016/j.intimp.2024.113476] [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: 06/28/2024] [Revised: 10/13/2024] [Accepted: 10/20/2024] [Indexed: 11/28/2024]
Abstract
Microglia are innate immune cells that maintain homeostasis of the central nervous system (CNS) and affect various neurodegenerative diseases, especially multiple sclerosis (MS). MS is an autoimmune disease of the CNS characterized by persistent inflammation, diffuse axonal damage, and microglia activation. Recent studies have shown that microglia are extremely related to the pathological state of MS and play an important role in the development of MS. This article reviews the multiple roles of microglia in the progression of MS, including the regulatory role of microglia in inflammation, remyelination, oxidative stress, the influence of phagocytosis and antigen-presenting capacity of microglia, and the recent progress by using microglia as a target for MS therapy. Microglia modulation may be a potential way for better MS therapy.
Collapse
Affiliation(s)
- Yunrong Nan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Industrial Development Center of Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shuting Ni
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mei Liu
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Industrial Development Center of Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Kaili Hu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
5
|
Escoubas CC, Dorman LC, Nguyen PT, Lagares-Linares C, Nakajo H, Anderson SR, Barron JJ, Wade SD, Cuevas B, Vainchtein ID, Silva NJ, Guajardo R, Xiao Y, Lidsky PV, Wang EY, Rivera BM, Taloma SE, Kim DK, Kaminskaya E, Nakao-Inoue H, Schwer B, Arnold TD, Molofsky AB, Condello C, Andino R, Nowakowski TJ, Molofsky AV. Type-I-interferon-responsive microglia shape cortical development and behavior. Cell 2024; 187:1936-1954.e24. [PMID: 38490196 PMCID: PMC11015974 DOI: 10.1016/j.cell.2024.02.020] [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: 07/14/2023] [Revised: 12/31/2023] [Accepted: 02/19/2024] [Indexed: 03/17/2024]
Abstract
Microglia are brain-resident macrophages that shape neural circuit development and are implicated in neurodevelopmental diseases. Multiple microglial transcriptional states have been defined, but their functional significance is unclear. Here, we identify a type I interferon (IFN-I)-responsive microglial state in the developing somatosensory cortex (postnatal day 5) that is actively engulfing whole neurons. This population expands during cortical remodeling induced by partial whisker deprivation. Global or microglial-specific loss of the IFN-I receptor resulted in microglia with phagolysosomal dysfunction and an accumulation of neurons with nuclear DNA damage. IFN-I gain of function increased neuronal engulfment by microglia in both mouse and zebrafish and restricted the accumulation of DNA-damaged neurons. Finally, IFN-I deficiency resulted in excess cortical excitatory neurons and tactile hypersensitivity. These data define a role for neuron-engulfing microglia during a critical window of brain development and reveal homeostatic functions of a canonical antiviral signaling pathway in the brain.
Collapse
Affiliation(s)
- Caroline C Escoubas
- Departments of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Leah C Dorman
- Departments of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Phi T Nguyen
- Departments of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Christian Lagares-Linares
- Departments of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Haruna Nakajo
- Departments of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sarah R Anderson
- Departments of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jerika J Barron
- Departments of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sarah D Wade
- Departments of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Beatriz Cuevas
- Departments of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ilia D Vainchtein
- Departments of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Nicholas J Silva
- Departments of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ricardo Guajardo
- Departments of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA; Medical Scientist Training Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yinghong Xiao
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Peter V Lidsky
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ellen Y Wang
- Departments of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA; UCSF SRTP program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Brianna M Rivera
- Institute for Neurodegenerative Diseases/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sunrae E Taloma
- Departments of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA; Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Dong Kyu Kim
- Departments of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Elizaveta Kaminskaya
- Departments of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Hiromi Nakao-Inoue
- Departments of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Bjoern Schwer
- Departments of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA; Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94158, USA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA; Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Thomas D Arnold
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ari B Molofsky
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Carlo Condello
- Institute for Neurodegenerative Diseases/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Raul Andino
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Tomasz J Nowakowski
- Departments of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA; Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA 94158, USA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA; Chan-Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Anna V Molofsky
- Departments of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA; Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA 94158, USA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
6
|
Gadhave DG, Sugandhi VV, Kokare CR. Potential biomaterials and experimental animal models for inventing new drug delivery approaches in the neurodegenerative disorder: Multiple sclerosis. Brain Res 2024; 1822:148674. [PMID: 37952871 DOI: 10.1016/j.brainres.2023.148674] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 09/14/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023]
Abstract
The tight junction of endothelial cells in the central nervous system (CNS) has an ideal characteristic, acting as a biological barrier that can securely regulate the movement of molecules in the brain. Tightly closed astrocyte cell junctions on blood capillaries are the blood-brain barrier (BBB). This biological barrier prohibits the entry of polar drugs, cells, and ions, which protect the brain from harmful toxins. However, delivering any therapeutic agent to the brain in neurodegenerative disorders (i.e., schizophrenia, multiple sclerosis, etc.) is extremely difficult. Active immune responses such as microglia, astrocytes, and lymphocytes cross the BBB and attack the nerve cells, which causes the demyelination of neurons. Therefore, there is a hindrance in transmitting electrical signals properly, resulting in blindness, paralysis, and neuropsychiatric problems. The main objective of this article is to shed light on the performance of biomaterials, which will help researchers to create nanocarriers that can cross the blood-brain barrier and achieve a therapeutic concentration of drugs in the CNS of patients with multiple sclerosis (MS). The present review focuses on the importance of biomaterials with diagnostic and therapeutic efficacy that can help enhance multiple sclerosis therapeutic potential. Currently, the development of MS in animal models is limited by immune responses, which prevent MS induction in healthy animals. Therefore, this article also showcases animal models currently used for treating MS. A future advance in developing a novel effective strategy for treating MS is now a potential area of research.
Collapse
Affiliation(s)
- Dnyandev G Gadhave
- Department of Pharmaceutics, Sinhgad Technical Education Society's, Sinhgad Institute of Pharmacy (Affiliated to Savitribai Phule Pune University), Narhe, Pune 411041, Maharashtra, India; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA; Department of Pharmaceutics, Dattakala Shikshan Sanstha's, Dattakala College of Pharmacy (Affiliated to Savitribai Phule Pune University), Swami Chincholi, Daund, Pune 413130, Maharashtra, India.
| | - Vrashabh V Sugandhi
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Chandrakant R Kokare
- Department of Pharmaceutics, Sinhgad Technical Education Society's, Sinhgad Institute of Pharmacy (Affiliated to Savitribai Phule Pune University), Narhe, Pune 411041, Maharashtra, India
| |
Collapse
|
7
|
Wies Mancini VSB, Mattera VS, Pasquini JM, Pasquini LA, Correale JD. Microglia-derived extracellular vesicles in homeostasis and demyelination/remyelination processes. J Neurochem 2024; 168:3-25. [PMID: 38055776 DOI: 10.1111/jnc.16011] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 10/10/2023] [Accepted: 11/03/2023] [Indexed: 12/08/2023]
Abstract
Microglia (MG) play a crucial role as the predominant myeloid cells in the central nervous system and are commonly activated in multiple sclerosis. They perform essential functions under normal conditions, such as actively surveying the surrounding parenchyma, facilitating synaptic remodeling, engulfing dead cells and debris, and protecting the brain against infectious pathogens and harmful self-proteins. Extracellular vesicles (EVs) are diverse structures enclosed by a lipid bilayer that originate from intracellular endocytic trafficking or the plasma membrane. They are released by cells into the extracellular space and can be found in various bodily fluids. EVs have recently emerged as a communication mechanism between cells, enabling the transfer of functional proteins, lipids, different RNA species, and even fragments of DNA from donor cells. MG act as both source and recipient of EVs. Consequently, MG-derived EVs are involved in regulating synapse development and maintaining homeostasis. These EVs also directly influence astrocytes, significantly increasing the release of inflammatory cytokines like IL-1β, IL-6, and TNF-α, resulting in a robust inflammatory response. Furthermore, EVs derived from inflammatory MG have been found to inhibit remyelination, whereas Evs produced by pro-regenerative MG effectively promote myelin repair. This review aims to provide an overview of the current understanding of MG-derived Evs, their impact on neighboring cells, and the cellular microenvironment in normal conditions and pathological states, specifically focusing on demyelination and remyelination processes.
Collapse
Affiliation(s)
- V S B Wies Mancini
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Cátedra de Química Biológica Patológica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - V S Mattera
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Cátedra de Química Biológica Patológica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - J M Pasquini
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Cátedra de Química Biológica Patológica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - L A Pasquini
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Cátedra de Química Biológica Patológica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - J D Correale
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Cátedra de Química Biológica Patológica, Universidad de Buenos Aires, Buenos Aires, Argentina
- Departamento de Neurología, Fleni, Buenos Aires, Argentina
| |
Collapse
|
8
|
Jonnalagadda D, Kihara Y, Groves A, Ray M, Saha A, Ellington C, Lee-Okada HC, Furihata T, Yokomizo T, Quadros EV, Rivera R, Chun J. FTY720 requires vitamin B 12-TCN2-CD320 signaling in astrocytes to reduce disease in an animal model of multiple sclerosis. Cell Rep 2023; 42:113545. [PMID: 38064339 PMCID: PMC11066976 DOI: 10.1016/j.celrep.2023.113545] [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: 10/04/2021] [Revised: 10/24/2023] [Accepted: 11/20/2023] [Indexed: 12/30/2023] Open
Abstract
Vitamin B12 (B12) deficiency causes neurological manifestations resembling multiple sclerosis (MS); however, a molecular explanation for the similarity is unknown. FTY720 (fingolimod) is a sphingosine 1-phosphate (S1P) receptor modulator and sphingosine analog approved for MS therapy that can functionally antagonize S1P1. Here, we report that FTY720 suppresses neuroinflammation by functionally and physically regulating the B12 pathways. Genetic and pharmacological S1P1 inhibition upregulates a transcobalamin 2 (TCN2)-B12 receptor, CD320, in immediate-early astrocytes (ieAstrocytes; a c-Fos-activated astrocyte subset that tracks with experimental autoimmune encephalomyelitis [EAE] severity). CD320 is also reduced in MS plaques. Deficiency of CD320 or dietary B12 restriction worsens EAE and eliminates FTY720's efficacy while concomitantly downregulating type I interferon signaling. TCN2 functions as a chaperone for FTY720 and sphingosine, whose complex induces astrocytic CD320 internalization, suggesting a delivery mechanism of FTY720/sphingosine via the TCN2-CD320 pathway. Taken together, the B12-TCN2-CD320 pathway is essential for the mechanism of action of FTY720.
Collapse
Affiliation(s)
- Deepa Jonnalagadda
- Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Yasuyuki Kihara
- Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA.
| | - Aran Groves
- Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA; Neuroscience Graduate Program, School of Medicine, University of California San Diego, 9500 Gilman Dr, La Jolla, CA 92093, USA
| | - Manisha Ray
- Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Arjun Saha
- Department of Chemistry, University of Southern California, Los Angeles, CA 90089, USA
| | - Clayton Ellington
- Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Hyeon-Cheol Lee-Okada
- Department of Biochemistry, Graduate School of Medicine, Juntendo University, Hongo 2-1-1, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Tomomi Furihata
- Laboratory of Clinical Pharmacy and Experimental Therapeutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Takehiko Yokomizo
- Department of Biochemistry, Graduate School of Medicine, Juntendo University, Hongo 2-1-1, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Edward V Quadros
- Department of Medicine, SUNY-Downstate Medical Center, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | - Richard Rivera
- Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
9
|
Thougaard E, Carney B, Wlodarczyk A, Brambilla R, Lambertsen KL. Peripherally derived myeloid cells induce disease-dependent phenotypic changes in microglia. Front Cell Neurosci 2023; 17:1295840. [PMID: 38155863 PMCID: PMC10752942 DOI: 10.3389/fncel.2023.1295840] [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: 09/17/2023] [Accepted: 11/23/2023] [Indexed: 12/30/2023] Open
Abstract
In central nervous system (CNS) injury and disease, peripherally derived myeloid cells infiltrate the CNS parenchyma and interact with resident cells, propagating the neuroinflammatory response. Because peripheral myeloid populations differ profoundly depending on the type and phase of injury, their crosstalk with CNS resident cells, particularly microglia, will lead to different functional outcomes. Thus, understanding how peripheral myeloid cells affect the phenotype and function of microglia in different disease conditions and phases may lead to a better understanding of disease-specific targetable pathways for neuroprotection and neurorepair. To this end, we set out to develop an in vitro system to investigate the communication between peripheral myeloid cells and microglia, with the goal of uncovering potential differences due to disease type and timing. We isolated peripheral myeloid cells from mice undergoing experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis, or acute cerebral ischemia by permanent middle cerebral artery occlusion (pMCAO) at different times after disease and probed their ability to change the phenotype of primary microglia isolated from the brain of adult mice. We identified changes not only dependent on the disease model, but also on the timepoint after disease onset from which the myeloid cells were isolated. Peripheral myeloid cells from acute EAE induced morphological changes in microglia, followed by increases in expression of genes involved in inflammatory signaling. Conversely, it was the peripheral myeloid cells from the chronic phase of pMCAO that induced gene expression changes in genes involved in inflammatory signaling and phagocytosis, which was not followed by a change in morphology. This underscores the importance of understanding the role of infiltrating myeloid cells in different disease contexts and phases. Furthermore, we showed that our assay is a valuable tool for investigating myeloid cell interactions in a range of CNS neuroinflammatory conditions.
Collapse
Affiliation(s)
- Estrid Thougaard
- Neurobiology Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- BRIDGE - Brain Research - Inter Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Brianna Carney
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Agnieszka Wlodarczyk
- Neurobiology Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- BRIDGE - Brain Research - Inter Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Roberta Brambilla
- Neurobiology Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- BRIDGE - Brain Research - Inter Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Kate Lykke Lambertsen
- Neurobiology Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- BRIDGE - Brain Research - Inter Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Neurology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
10
|
Distéfano-Gagné F, Bitarafan S, Lacroix S, Gosselin D. Roles and regulation of microglia activity in multiple sclerosis: insights from animal models. Nat Rev Neurosci 2023:10.1038/s41583-023-00709-6. [PMID: 37268822 DOI: 10.1038/s41583-023-00709-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2023] [Indexed: 06/04/2023]
Abstract
As resident macrophages of the CNS, microglia are critical immune effectors of inflammatory lesions and associated neural dysfunctions. In multiple sclerosis (MS) and its animal models, chronic microglial inflammatory activity damages myelin and disrupts axonal and synaptic activity. In contrast to these detrimental effects, the potent phagocytic and tissue-remodelling capabilities of microglia support critical endogenous repair mechanisms. Although these opposing capabilities have long been appreciated, a precise understanding of their underlying molecular effectors is only beginning to emerge. Here, we review recent advances in our understanding of the roles of microglia in animal models of MS and demyelinating lesions and the mechanisms that underlie their damaging and repairing activities. We also discuss how the structured organization and regulation of the genome enables complex transcriptional heterogeneity within the microglial cell population at demyelinating lesions.
Collapse
Affiliation(s)
- Félix Distéfano-Gagné
- Axe Neuroscience, Centre de Recherche du CHU de Québec - Université Laval, Québec, Québec, Canada
- Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Québec, Québec, Canada
| | - Sara Bitarafan
- Axe Neuroscience, Centre de Recherche du CHU de Québec - Université Laval, Québec, Québec, Canada
- Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Québec, Québec, Canada
| | - Steve Lacroix
- Axe Neuroscience, Centre de Recherche du CHU de Québec - Université Laval, Québec, Québec, Canada
- Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Québec, Québec, Canada
| | - David Gosselin
- Axe Neuroscience, Centre de Recherche du CHU de Québec - Université Laval, Québec, Québec, Canada.
- Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Québec, Québec, Canada.
| |
Collapse
|
11
|
Kihara Y, Chun J. Molecular and neuroimmune pharmacology of S1P receptor modulators and other disease-modifying therapies for multiple sclerosis. Pharmacol Ther 2023; 246:108432. [PMID: 37149155 DOI: 10.1016/j.pharmthera.2023.108432] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/25/2023] [Accepted: 05/02/2023] [Indexed: 05/08/2023]
Abstract
Multiple sclerosis (MS) is a neurological, immune-mediated demyelinating disease that affects people in the prime of life. Environmental, infectious, and genetic factors have been implicated in its etiology, although a definitive cause has yet to be determined. Nevertheless, multiple disease-modifying therapies (DMTs: including interferons, glatiramer acetate, fumarates, cladribine, teriflunomide, fingolimod, siponimod, ozanimod, ponesimod, and monoclonal antibodies targeting ITGA4, CD20, and CD52) have been developed and approved for the treatment of MS. All the DMTs approved to date target immunomodulation as their mechanism of action (MOA); however, the direct effects of some DMTs on the central nervous system (CNS), particularly sphingosine 1-phosphate (S1P) receptor (S1PR) modulators, implicate a parallel MOA that may also reduce neurodegenerative sequelae. This review summarizes the currently approved DMTs for the treatment of MS and provides details and recent advances in the molecular pharmacology, immunopharmacology, and neuropharmacology of S1PR modulators, with a special focus on the CNS-oriented, astrocyte-centric MOA of fingolimod.
Collapse
Affiliation(s)
- Yasuyuki Kihara
- Sanford Burnham Prebys Medical Discovery Institute, United States of America.
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, United States of America
| |
Collapse
|
12
|
Kapate N, Dunne M, Kumbhojkar N, Prakash S, Wang LLW, Graveline A, Park KS, Chandran Suja V, Goyal J, Clegg JR, Mitragotri S. A backpack-based myeloid cell therapy for multiple sclerosis. Proc Natl Acad Sci U S A 2023; 120:e2221535120. [PMID: 37075071 PMCID: PMC10151518 DOI: 10.1073/pnas.2221535120] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/20/2023] [Indexed: 04/20/2023] Open
Abstract
Multiple sclerosis (MS) is an incurable autoimmune disease and is currently treated by systemic immunosuppressants with off-target side effects. Although aberrant myeloid function is often observed in MS plaques in the central nervous system (CNS), the role of myeloid cells in therapeutic intervention is currently overlooked. Here, we developed a myeloid cell-based strategy to reduce the disease burden in experimental autoimmune encephalomyelitis (EAE), a mouse model of progressive MS. We developed monocyte-adhered microparticles ("backpacks") for activating myeloid cell phenotype to an anti-inflammatory state through localized interleukin-4 and dexamethasone signals. We demonstrate that backpack-laden monocytes infiltrated into the inflamed CNS and modulated both the local and systemic immune responses. Within the CNS, backpack-carrying monocytes regulated both the infiltrating and tissue-resident myeloid cell compartments in the spinal cord for functions related to antigen presentation and reactive species production. Treatment with backpack-monocytes also decreased the level of systemic pro-inflammatory cytokines. Additionally, backpack-laden monocytes induced modulatory effects on TH1 and TH17 populations in the spinal cord and blood, demonstrating cross talk between the myeloid and lymphoid arms of disease. Backpack-carrying monocytes conferred therapeutic benefit in EAE mice, as quantified by improved motor function. The use of backpack-laden monocytes offers an antigen-free, biomaterial-based approach to precisely tune cell phenotype in vivo, demonstrating the utility of myeloid cells as a therapeutic modality and target.
Collapse
Affiliation(s)
- Neha Kapate
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Allston, MA02134
- Wyss Institute for Biologically Inspired Engineering, Boston, MA02115
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Michael Dunne
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Allston, MA02134
- Wyss Institute for Biologically Inspired Engineering, Boston, MA02115
| | - Ninad Kumbhojkar
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Allston, MA02134
- Wyss Institute for Biologically Inspired Engineering, Boston, MA02115
| | - Supriya Prakash
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Allston, MA02134
- Wyss Institute for Biologically Inspired Engineering, Boston, MA02115
| | - Lily Li-Wen Wang
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Allston, MA02134
- Wyss Institute for Biologically Inspired Engineering, Boston, MA02115
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Amanda Graveline
- Wyss Institute for Biologically Inspired Engineering, Boston, MA02115
| | - Kyung Soo Park
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Allston, MA02134
- Wyss Institute for Biologically Inspired Engineering, Boston, MA02115
| | - Vineeth Chandran Suja
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Allston, MA02134
- Wyss Institute for Biologically Inspired Engineering, Boston, MA02115
| | - Juhee Goyal
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Allston, MA02134
| | - John R. Clegg
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Allston, MA02134
- Wyss Institute for Biologically Inspired Engineering, Boston, MA02115
| | - Samir Mitragotri
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Allston, MA02134
- Wyss Institute for Biologically Inspired Engineering, Boston, MA02115
| |
Collapse
|
13
|
Rao M, Huang YK, Liu CC, Meadows C, Cheng HC, Zhou M, Chen YC, Xia X, Goldberg JL, Williams AM, Kuwajima T, Chang KC. Aldose reductase inhibition decelerates optic nerve degeneration by alleviating retinal microglia activation. Sci Rep 2023; 13:5592. [PMID: 37019993 PMCID: PMC10076364 DOI: 10.1038/s41598-023-32702-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
As part of the central nervous system (CNS), retinal ganglion cells (RGCs) and their axons are the only neurons in the retina that transmit visual signals from the eye to the brain via the optic nerve (ON). Unfortunately, they do not regenerate upon injury in mammals. In ON trauma, retinal microglia (RMG) become activated, inducing inflammatory responses and resulting in axon degeneration and RGC loss. Since aldose reductase (AR) is an inflammatory response mediator highly expressed in RMG, we investigated if pharmacological inhibition of AR can attenuate ocular inflammation and thereby promote RGC survival and axon regeneration after ON crush (ONC). In vitro, we discovered that Sorbinil, an AR inhibitor, attenuates BV2 microglia activation and migration in the lipopolysaccharide (LPS) and monocyte chemoattractant protein-1 (MCP-1) treatments. In vivo, Sorbinil suppressed ONC-induced Iba1 + microglia/macrophage infiltration in the retina and ON and promoted RGC survival. Moreover, Sorbinil restored RGC function and delayed axon degeneration one week after ONC. RNA sequencing data revealed that Sorbinil protects the retina from ONC-induced degeneration by suppressing inflammatory signaling. In summary, we report the first study demonstrating that AR inhibition transiently protects RGC and axon from degeneration, providing a potential therapeutic strategy for optic neuropathies.
Collapse
Affiliation(s)
- Mishal Rao
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, 203 Lothrop, Pittsburgh, PA, 15213, USA
| | - Yu-Kai Huang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung, 80708, Taiwan
- Department of Surgery, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, 80145, Taiwan
| | - Chia-Chun Liu
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, 203 Lothrop, Pittsburgh, PA, 15213, USA
| | - Chandler Meadows
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, 203 Lothrop, Pittsburgh, PA, 15213, USA
| | - Hui-Chun Cheng
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, 203 Lothrop, Pittsburgh, PA, 15213, USA
| | - Mengli Zhou
- Department of Computational and Systems Biology, Hillman Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15260, USA
| | - Yu-Chih Chen
- Department of Computational and Systems Biology, Hillman Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15260, USA
| | - Xin Xia
- Spencer Center for Vision Research, Byers Eye Institute, School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| | - Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| | - Andrew M Williams
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, 203 Lothrop, Pittsburgh, PA, 15213, USA
| | - Takaaki Kuwajima
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, 203 Lothrop, Pittsburgh, PA, 15213, USA
| | - Kun-Che Chang
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, 203 Lothrop, Pittsburgh, PA, 15213, USA.
- Department of Neurobiology, Center of Neuroscience, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
14
|
Zhang C, Raveney B, Takahashi F, Yeh TW, Hohjoh H, Yamamura T, Oki S. Pathogenic Microglia Orchestrate Neurotoxic Properties of Eomes-Expressing Helper T Cells. Cells 2023; 12:cells12060868. [PMID: 36980209 PMCID: PMC10047905 DOI: 10.3390/cells12060868] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/08/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
In addition to disease-associated microglia (DAM), microglia with MHC-II and/or IFN-I signatures may form additional pathogenic subsets that are relevant to neurodegeneration. However, the significance of such MHC-II and IFN-I signatures remains elusive. We demonstrate here that these microglial subsets play intrinsic roles in orchestrating neurotoxic properties of neurotoxic Eomes+ Th cells under the neurodegeneration-associated phase of experimental autoimmune encephalomyelitis (EAE) that corresponds to progressive multiple sclerosis (MS). Microglia acquire IFN-signature after sensing ectopically expressed long interspersed nuclear element-1 (L1) gene. Furthermore, ORF1, an L1-encoded protein aberrantly expressed in the diseased central nervous system (CNS), stimulated Eomes+ Th cells after Trem2-dependent ingestion and presentation in MHC-II context by microglia. Interestingly, administration of an L1 inhibitor significantly ameliorated neurodegenerative symptoms of EAE concomitant with reduced accumulation of Eomes+ Th cells in the CNS. Collectively, our data highlight a critical contribution of new microglia subsets as a neuroinflammatory hub in immune-mediated neurodegeneration.
Collapse
Affiliation(s)
- Chenyang Zhang
- Department of Immunology, National Institute of Neuroscience, NCNP, Tokyo 187-8502, Japan
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8549, Japan
| | - Ben Raveney
- Department of Immunology, National Institute of Neuroscience, NCNP, Tokyo 187-8502, Japan
| | - Fumio Takahashi
- Department of Immunology, National Institute of Neuroscience, NCNP, Tokyo 187-8502, Japan
| | - Tzu-wen Yeh
- Department of Immunology, National Institute of Neuroscience, NCNP, Tokyo 187-8502, Japan
| | - Hirohiko Hohjoh
- Department of Molecular Pharmacology, National Institute of Neuroscience, NCNP, Tokyo 187-8502, Japan
| | - Takashi Yamamura
- Department of Immunology, National Institute of Neuroscience, NCNP, Tokyo 187-8502, Japan
- Correspondence: (T.Y.); (S.O.); Tel.: +81-42-341-2711 (T.Y. & S.O.)
| | - Shinji Oki
- Department of Immunology, National Institute of Neuroscience, NCNP, Tokyo 187-8502, Japan
- Correspondence: (T.Y.); (S.O.); Tel.: +81-42-341-2711 (T.Y. & S.O.)
| |
Collapse
|
15
|
Bühler M, Li D, Li L, Runft S, Waltl I, Pavlou A, Kalinke U, Ciurkiewicz M, Huehn J, Floess S, Beineke A, Baumgärtner W, Gerhauser I. IFNAR signaling of neuroectodermal cells is essential for the survival of C57BL/6 mice infected with Theiler's murine encephalomyelitis virus. J Neuroinflammation 2023; 20:58. [PMID: 36872323 PMCID: PMC9985866 DOI: 10.1186/s12974-023-02737-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 02/16/2023] [Indexed: 03/07/2023] Open
Abstract
BACKGROUND Theiler's murine encephalomyelitis virus (TMEV) is a single-stranded RNA virus that causes encephalitis followed by chronic demyelination in SJL mice and spontaneous seizures in C57BL/6 mice. Since earlier studies indicated a critical role of type I interferon (IFN-I) signaling in the control of viral replication in the central nervous system (CNS), mouse strain-specific differences in pathways induced by the IFN-I receptor (IFNAR) might determine the outcome of TMEV infection. METHODS Data of RNA-seq analysis and immunohistochemistry were used to compare the gene and protein expression of IFN-I signaling pathway members between mock- and TMEV-infected SJL and C57BL/6 mice at 4, 7 and 14 days post-infection (dpi). To address the impact of IFNAR signaling in selected brain-resident cell types, conditional knockout mice with an IFNAR deficiency in cells of the neuroectodermal lineage (NesCre±IFNARfl/fl), neurons (Syn1Cre±IFNARfl/fl), astrocytes (GFAPCre±IFNARfl/fl), and microglia (Sall1CreER±IFNARfl/fl) on a C57BL/6 background were tested. PCR and an immunoassay were used to quantify TMEV RNA and cytokine and chemokine expression in their brain at 4 dpi. RESULTS RNA-seq analysis revealed upregulation of most ISGs in SJL and C57BL/6 mice, but Ifi202b mRNA transcripts were only increased in SJL and Trim12a only in C57BL/6 mice. Immunohistochemistry showed minor differences in ISG expression (ISG15, OAS, PKR) between both mouse strains. While all immunocompetent Cre-negative control mice and the majority of mice with IFNAR deficiency in neurons or microglia survived until 14 dpi, lack of IFNAR expression in all cells (IFNAR-/-), neuroectodermal cells, or astrocytes induced lethal disease in most of the analyzed mice, which was associated with unrestricted viral replication. NesCre±IFNARfl/fl mice showed more Ifnb1, Tnfa, Il6, Il10, Il12b and Ifng mRNA transcripts than Cre-/-IFNARfl/fl mice. IFNAR-/- mice also demonstrated increased IFN-α, IFN-β, IL1-β, IL-6, and CXCL-1 protein levels, which highly correlated with viral load. CONCLUSIONS Ifi202b and Trim12a expression levels likely contribute to mouse strain-specific susceptibility to TMEV-induced CNS lesions. Restriction of viral replication is strongly dependent on IFNAR signaling of neuroectodermal cells, which also controls the expression of key pro- and anti-inflammatory cytokines during viral brain infection.
Collapse
Affiliation(s)
- Melanie Bühler
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
| | - Dandan Li
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
- Centre for Systems Neuroscience (ZSN), Hannover, Germany
| | - Lin Li
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
- Centre for Systems Neuroscience (ZSN), Hannover, Germany
- c/o School of Basic Medical Sciences, Shanxi Medical University, Shanxi, China
| | - Sandra Runft
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
- Centre for Systems Neuroscience (ZSN), Hannover, Germany
| | - Inken Waltl
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Andreas Pavlou
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Ulrich Kalinke
- Centre for Systems Neuroscience (ZSN), Hannover, Germany
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Malgorzata Ciurkiewicz
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124, Brunswick, Germany
| | - Stefan Floess
- Experimental Immunology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124, Brunswick, Germany
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
- Centre for Systems Neuroscience (ZSN), Hannover, Germany
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany
- Centre for Systems Neuroscience (ZSN), Hannover, Germany
| | - Ingo Gerhauser
- Department of Pathology, University of Veterinary Medicine Hannover, Foundation, Bünteweg 17, 30559, Hannover, Germany.
| |
Collapse
|
16
|
Peradinovic J, Mohovic N, Bulic K, Markovinovic A, Cimbro R, Munitic I. Ageing-Induced Decline in Primary Myeloid Cell Phagocytosis Is Unaffected by Optineurin Insufficiency. BIOLOGY 2023; 12:biology12020240. [PMID: 36829517 PMCID: PMC9953198 DOI: 10.3390/biology12020240] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
Optineurin is a ubiquitin-binding adaptor protein involved in multiple cellular processes, including innate inflammatory signalling. Mutations in optineurin were found in amyotrophic lateral sclerosis, an adult-onset fatal neurodegenerative disease that targets motor neurons. Neurodegeneration results in generation of neuronal debris, which is primarily cleared by myeloid cells. To assess the role of optineurin in phagocytosis, we performed a flow cytometry-based phagocytic assay of apoptotic neuronal debris and E. coli bioparticles in bone marrow-derived macrophages (BMDMs), and primary neonatal microglia from wild-type (WT) and optineurin-insufficient (Optn470T) mice. We found no difference in phagocytosis efficiency and the accompanying cytokine secretion in WT and Optn470T BMDMs and microglia. This was true at both steady state and upon proinflammatory polarization with lipopolysaccharide. When we analysed the effect of ageing as a major risk factor for neurodegeneration, we found a substantial decrease in the percentage of phagocytic cells and proinflammatory cytokine secretion in BMDMs from 2-year-old mice. However, this ageing-induced phagocytic decline was unaffected by optineurin insufficiency. All together, these results indicate that ageing is the factor that perturbs normal phagocytosis and proinflammatory cytokine secretion, but that optineurin is dispensable for these processes.
Collapse
Affiliation(s)
- Josip Peradinovic
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, R. Matejcic 2, 51000 Rijeka, Croatia
| | - Nikolina Mohovic
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, R. Matejcic 2, 51000 Rijeka, Croatia
| | - Katarina Bulic
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, R. Matejcic 2, 51000 Rijeka, Croatia
| | - Andrea Markovinovic
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 9RT, UK
| | - Raffaello Cimbro
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, R. Matejcic 2, 51000 Rijeka, Croatia
- Correspondence: (R.C.); (I.M.)
| | - Ivana Munitic
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, R. Matejcic 2, 51000 Rijeka, Croatia
- Correspondence: (R.C.); (I.M.)
| |
Collapse
|
17
|
Viengkhou B, Hofer MJ. Breaking down the cellular responses to type I interferon neurotoxicity in the brain. Front Immunol 2023; 14:1110593. [PMID: 36817430 PMCID: PMC9936317 DOI: 10.3389/fimmu.2023.1110593] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 01/25/2023] [Indexed: 02/05/2023] Open
Abstract
Since their original discovery, type I interferons (IFN-Is) have been closely associated with antiviral immune responses. However, their biological functions go far beyond this role, with balanced IFN-I activity being critical to maintain cellular and tissue homeostasis. Recent findings have uncovered a darker side of IFN-Is whereby chronically elevated levels induce devastating neuroinflammatory and neurodegenerative pathologies. The underlying causes of these 'interferonopathies' are diverse and include monogenetic syndromes, autoimmune disorders, as well as chronic infections. The prominent involvement of the CNS in these disorders indicates a particular susceptibility of brain cells to IFN-I toxicity. Here we will discuss the current knowledge of how IFN-Is mediate neurotoxicity in the brain by analyzing the cell-type specific responses to IFN-Is in the CNS, and secondly, by exploring the spectrum of neurological disorders arising from increased IFN-Is. Understanding the nature of IFN-I neurotoxicity is a crucial and fundamental step towards development of new therapeutic strategies for interferonopathies.
Collapse
Affiliation(s)
- Barney Viengkhou
- School of Life and Environmental Sciences and the Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
| | | |
Collapse
|
18
|
Intranasal interferon-beta alleviates anxiety and depressive-like behaviors by modulating microglia polarization in an Alzheimer's disease model. Neurosci Lett 2023; 792:136968. [PMID: 36396023 DOI: 10.1016/j.neulet.2022.136968] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/30/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022]
Abstract
Alzheimer's disease (AD) patients frequently experience neuropsychiatric symptoms (NPS), which are linked to a lower quality of life and a faster rate of disease progression. A growing body of research indicates that several microglial phenotypes control the inflammatory response and are crucial in the pathophysiology of AD-related NPS. Given the crucial role played by inflammatory mediators produced by microglia in developing of NPS, interferon-beta (IFNβ), a cytokine with anti-inflammatory capabilities, maybe a successful treatment for NPS caused by AD. In this investigation, using a rat model of AD, we examined the impact of intranasal treatment of IFNβ on anxious/depressive-like behavior and microglial M1/M2 polarization. The rat hippocampus was bilaterally injected with lentiviruses harboring mutant human amyloid precursor protein. Rats were given recombinant IFNβ1a (68,000 IU/rat) via the intranasal route, starting on day 23 following viral infection and continuing until day 49. On days 47-49, the elevated plus maze, forced swim, and tail suspension tests were applied to measure anxiety- and depressive-like behavior. Additionally, qPCR was utilized to quantify the expression of M1 markers (CD68, CD86, and CD40) and M2 markers (Ym1, CD206, Arg1, GDNF, BDNF, and SOCS1). Our findings demonstrated that decreased M2 marker expression is accompanied by anxious/depressive-like behavior when the mutant human APP gene is overexpressed in the hippocampus. In the rat model of AD, IFNβ therapy reduces anxious/depressive-like behaviors, at least in part by polarizing microglia towards M2. Therefore, IFNβ may be a viable therapeutic drug for reducing NPS in the context of AD.
Collapse
|
19
|
Drug-induced microglial phagocytosis in multiple sclerosis and experimental autoimmune encephalomyelitis and the underlying mechanisms. Mol Biol Rep 2023; 50:749-759. [PMID: 36309614 DOI: 10.1007/s11033-022-07968-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 09/21/2022] [Indexed: 02/01/2023]
Abstract
Microglia are resident macrophages of the central nervous system (CNS). It plays a significant role in immune surveillance under physiological conditions. On stimulation by pathogens, microglia change their phenotypes, phagocytize toxic molecules, secrete pro-inflammatory/anti-inflammatory factors, promotes tissue repair, and maintain the homeostasis in CNS. Accumulation of myelin debris in multiple sclerosis (MS)/experimental autoimmune encephalomyelitis (EAE) inhibits remyelination by decreasing the phagocytosis by microglia and prevent the recovery of MS/EAE. Drug induced microglia phagocytosis could be a novel therapeutic intervention for the treatment of MS/EAE. But the abnormal phagocytosis of neurons and synapses by activated microglia will lead to neuronal damage and degeneration. It indicates that the phagocytosis of microglia has many beneficial and harmful effects in central neurodegenerative diseases. Therefore, simply promoting or inhibiting the phagocytic activity of microglia may not achieve ideal therapeutic results. However, limited reports are available to elucidate the microglia mediated phagocytosis and its underlying molecular mechanisms. On this basis, the present review describes microglia-mediated phagocytosis, drug-induced microglia phagocytosis, molecular mechanism, and novel approach for MS/EAE treatment.
Collapse
|
20
|
Single-cell microglial transcriptomics during demyelination defines a microglial state required for lytic carcass clearance. Mol Neurodegener 2022; 17:82. [PMID: 36514132 PMCID: PMC9746011 DOI: 10.1186/s13024-022-00584-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 11/21/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Microglia regulate the response to injury and disease in the brain and spinal cord. In white matter diseases microglia may cause demyelination. However, how microglia respond and regulate demyelination is not fully understood. METHODS To understand how microglia respond during demyelination, we fed mice cuprizone-a potent demyelinating agent-and assessed the dynamics of genetically fate-mapped microglia. We then used single-cell RNA sequencing to identify and track the microglial subpopulations that arise during demyelination. To understand how microglia contribute to the clearance of dead oligodendrocytes, we ablated microglia starting at the peak of cuprizone-induced cell death and used the viability dye acridine orange to monitor apoptotic and lytic cell morphologies after microglial ablation. Lastly, we treated serum-free primary microglial cultures to model distinct aspects of cuprizone-induced demyelination and assessed the response. RESULTS The cuprizone diet generated a robust microglial response by week 4 of the diet. Single-cell RNA sequencing at this time point revealed the presence of several cuprizone-associated microglia (CAM) clusters. These clusters expressed a transcriptomic signature indicative of cytokine regulation and reactive oxygen species production with altered lysosomal and metabolic changes consistent with ongoing phagocytosis. Using acridine orange to monitor apoptotic and lytic cell death after microglial ablation, we found that microglia preferentially phagocytose lytic carcasses. In culture, microglia exposed to lytic carcasses partially recapitulated the CAM state, suggesting that phagocytosis contributes to this distinct microglial state during cuprizone demyelination. CONCLUSIONS Microglia serve multiple roles during demyelination, yet their transcriptomic state resembles other neurodegenerative conditions. The phagocytosis of cellular debris is likely a universal cause for a common neurodegenerative microglial state.
Collapse
|
21
|
Raftopoulou S, Rapti A, Karathanasis D, Evangelopoulos ME, Mavragani CP. The role of type I IFN in autoimmune and autoinflammatory diseases with CNS involvement. Front Neurol 2022; 13:1026449. [PMID: 36438941 PMCID: PMC9685560 DOI: 10.3389/fneur.2022.1026449] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/17/2022] [Indexed: 07/30/2023] Open
Abstract
Type I interferons (IFNs) are major mediators of innate immunity, with well-known antiviral, antiproliferative, and immunomodulatory properties. A growing body of evidence suggests the involvement of type I IFNs in the pathogenesis of central nervous system (CNS) manifestations in the setting of chronic autoimmune and autoinflammatory disorders, while IFN-β has been for years, a well-established therapeutic modality for multiple sclerosis (MS). In the present review, we summarize the current evidence on the mechanisms of type I IFN production by CNS cellular populations as well as its local effects on the CNS. Additionally, the beneficial effects of IFN-β in the pathophysiology of MS are discussed, along with the contributory role of type I IFNs in the pathogenesis of neuropsychiatric lupus erythematosus and type I interferonopathies.
Collapse
Affiliation(s)
- Sylvia Raftopoulou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Anna Rapti
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitris Karathanasis
- First Department of Neurology, National and Kapodistrian University of Athens, Aeginition Hospital, Athens, Greece
| | | | - Clio P. Mavragani
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
22
|
Mundt S, Greter M, Becher B. The CNS mononuclear phagocyte system in health and disease. Neuron 2022; 110:3497-3512. [PMID: 36327896 DOI: 10.1016/j.neuron.2022.10.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/21/2022] [Accepted: 10/04/2022] [Indexed: 11/05/2022]
Abstract
CNS-resident macrophages-including parenchymal microglia and border-associated macrophages (BAMs)-contribute to neuronal development and health, vascularization, and tissue integrity at steady state. Border-patrolling mononuclear phagocytes such as dendritic cells and monocytes confer important immune functions to the CNS, protecting it from pathogenic threats including aberrant cell growth and brain malignancies. Even though we have learned much about the contribution of lymphocytes to CNS pathologies, a better understanding of differential roles of tissue-resident and -invading phagocytes is slowly emerging. In this perspective, we propose that in CNS neuroinflammatory diseases, tissue-resident macrophages (TRMs) contribute to the clearing of debris and resolution of inflammation, whereas blood-borne phagocytes are drivers of immunopathology. We discuss the remaining challenges to resolve which specialized mononuclear phagocyte populations are driving or suppressing immune effector function, thereby potentially dictating the outcome of autoimmunity or brain cancer.
Collapse
Affiliation(s)
- Sarah Mundt
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
23
|
Oxidative Stress as a Potential Mechanism Underlying Membrane Hyperexcitability in Neurodegenerative Diseases. Antioxidants (Basel) 2022; 11:antiox11081511. [PMID: 36009230 PMCID: PMC9405356 DOI: 10.3390/antiox11081511] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 02/01/2023] Open
Abstract
Neurodegenerative diseases are characterized by gradually progressive, selective loss of anatomically or physiologically related neuronal systems that produce brain damage from which there is no recovery. Despite the differences in clinical manifestations and neuronal vulnerability, the pathological processes appear to be similar, suggesting common neurodegenerative pathways. It is well known that oxidative stress and the production of reactive oxygen radicals plays a key role in neuronal cell damage. It has been proposed that this stress, among other mechanisms, could contribute to neuronal degeneration and might be one of the factors triggering the development of these pathologies. Another common feature in most neurodegenerative diseases is neuron hyperexcitability, an aberrant electrical activity. This review, focusing mainly on primary motor cortex pyramidal neurons, critically evaluates the idea that oxidative stress and inflammation may be involved in neurodegeneration via their capacity to increase membrane excitability.
Collapse
|
24
|
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
|
25
|
Duan N, Zhang Y, Tan S, Sun J, Ye M, Gao H, Pu K, Wu M, Wang Q, Zhai Q. Therapeutic targeting of STING-TBK1-IRF3 signalling ameliorates chronic stress induced depression-like behaviours by modulating neuroinflammation and microglia phagocytosis. Neurobiol Dis 2022; 169:105739. [DOI: 10.1016/j.nbd.2022.105739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/13/2022] [Accepted: 04/16/2022] [Indexed: 12/19/2022] Open
|
26
|
Au NPB, Ma CHE. Neuroinflammation, Microglia and Implications for Retinal Ganglion Cell Survival and Axon Regeneration in Traumatic Optic Neuropathy. Front Immunol 2022; 13:860070. [PMID: 35309305 PMCID: PMC8931466 DOI: 10.3389/fimmu.2022.860070] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 02/14/2022] [Indexed: 12/12/2022] Open
Abstract
Traumatic optic neuropathy (TON) refers to a pathological condition caused by a direct or indirect insult to the optic nerves, which often leads to a partial or permanent vision deficit due to the massive loss of retinal ganglion cells (RGCs) and their axonal fibers. Retinal microglia are immune-competent cells residing in the retina. In rodent models of optic nerve crush (ONC) injury, resident retinal microglia gradually become activated, form end-to-end alignments in the vicinity of degenerating RGC axons, and actively internalized them. Some activated microglia adopt an amoeboid morphology that engulf dying RGCs after ONC. In the injured optic nerve, the activated microglia contribute to the myelin debris clearance at the lesion site. However, phagocytic capacity of resident retinal microglia is extremely poor and therefore the clearance of cellular and myelin debris is largely ineffective. The presence of growth-inhibitory myelin debris and glial scar formed by reactive astrocytes inhibit the regeneration of RGC axons, which accounts for the poor visual function recovery in patients with TON. In this Review, we summarize the current understanding of resident retinal microglia in RGC survival and axon regeneration after ONC. Resident retinal microglia play a key role in facilitating Wallerian degeneration and the subsequent axon regeneration after ONC. However, they are also responsible for producing pro-inflammatory cytokines, chemokines, and reactive oxygen species that possess neurotoxic effects on RGCs. Intraocular inflammation triggers a massive influx of blood-borne myeloid cells which produce oncomodulin to promote RGC survival and axon regeneration. However, intraocular inflammation induces chronic neuroinflammation which exacerbates secondary tissue damages and limits visual function recovery after ONC. Activated retinal microglia is required for the proliferation of oligodendrocyte precursor cells (OPCs); however, sustained activation of retinal microglia suppress the differentiation of OPCs into mature oligodendrocytes for remyelination after injury. Collectively, controlled activation of retinal microglia and infiltrating myeloid cells facilitate axon regeneration and nerve repair. Recent advance in single-cell RNA-sequencing and identification of microglia-specific markers could improve our understanding on microglial biology and to facilitate the development of novel therapeutic strategies aiming to switch resident retinal microglia’s phenotype to foster neuroprotection.
Collapse
Affiliation(s)
- Ngan Pan Bennett Au
- Department of Neuroscience, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Chi Him Eddie Ma
- Department of Neuroscience, City University of Hong Kong, Hong Kong, Hong Kong SAR, China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- *Correspondence: Chi Him Eddie Ma,
| |
Collapse
|
27
|
Sen MK, Mahns DA, Coorssen JR, Shortland PJ. The roles of microglia and astrocytes in phagocytosis and myelination: Insights from the cuprizone model of multiple sclerosis. Glia 2022; 70:1215-1250. [PMID: 35107839 PMCID: PMC9302634 DOI: 10.1002/glia.24148] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/12/2022]
Abstract
In human demyelinating diseases such as multiple sclerosis (MS), an imbalance between demyelination and remyelination can trigger progressive degenerative processes. The clearance of myelin debris (phagocytosis) from the site of demyelination by microglia is critically important to achieve adequate remyelination and to slow the progression of the disease. However, how microglia phagocytose the myelin debris, and why clearance is impaired in MS, is not fully known; likewise, the role of the microglia in remyelination remains unclear. Recent studies using cuprizone (CPZ) as an animal model of central nervous system demyelination revealed that the up‐regulation of signaling proteins in microglia facilitates effective phagocytosis of myelin debris. Moreover, during demyelination, protective mediators are released from activated microglia, resulting in the acceleration of remyelination in the CPZ model. In contrast, inadequate microglial activation or recruitment to the site of demyelination, and the production of toxic mediators, impairs remyelination resulting in progressive demyelination. In addition to the microglia‐mediated phagocytosis, astrocytes play an important role in the phagocytic process by recruiting microglia to the site of demyelination and producing regenerative mediators. The current review is an update of these emerging findings from the CPZ animal model, discussing the roles of microglia and astrocytes in phagocytosis and myelination.
Collapse
Affiliation(s)
- Monokesh K Sen
- School of Medicine, Western Sydney University, Penrith, Australia
| | - David A Mahns
- School of Medicine, Western Sydney University, Penrith, Australia
| | - Jens R Coorssen
- Faculty of Applied Health Sciences and Faculty of Mathematics & Science, Brock University, St. Cathari, Canada
| | | |
Collapse
|
28
|
Pajarskienė J, Kašėta V, Vaikšnoraitė K, Tunaitis V, Pivoriūnas A. MicroRNA-124 acts as a positive regulator of IFN-β signaling in the lipopolysaccharide-stimulated human microglial cells. Int Immunopharmacol 2021; 101:108262. [PMID: 34688135 DOI: 10.1016/j.intimp.2021.108262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/20/2021] [Accepted: 10/11/2021] [Indexed: 10/20/2022]
Abstract
miR-124 is ubiquitously expressed in the nervous tissue and acts as a negative regulator of neuroinflammation. In the present study, we analyzed the possible role of miR-124 in response to LPS in the human microglial cell line. Our data revealed that the miR-124 anti-inflammatory effect is based not only on the suppression of MyD88 - NFκB pathway and downregulation of pro-inflammatory cytokines IL-1β and IL-6 but also on the enhancement of TRAM-TRIF signaling and increased IFN-β expression. Furthermore, the NFκB reporter assay demonstrated that specific miR-124 - induced NFκB activity changes could be detected only using NFκB reporter promoters lacking ATF/CREB binding site.
Collapse
Affiliation(s)
- Justina Pajarskienė
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102, Vilnius, Lithuania
| | - Vytautas Kašėta
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102, Vilnius, Lithuania
| | - Kristina Vaikšnoraitė
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102, Vilnius, Lithuania
| | - Virginijus Tunaitis
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102, Vilnius, Lithuania
| | - Augustas Pivoriūnas
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102, Vilnius, Lithuania.
| |
Collapse
|
29
|
Muzio L, Viotti A, Martino G. Microglia in Neuroinflammation and Neurodegeneration: From Understanding to Therapy. Front Neurosci 2021; 15:742065. [PMID: 34630027 PMCID: PMC8497816 DOI: 10.3389/fnins.2021.742065] [Citation(s) in RCA: 261] [Impact Index Per Article: 65.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/25/2021] [Indexed: 12/19/2022] Open
Abstract
Microglia are the resident macrophages of the central nervous system (CNS) acting as the first line of defense in the brain by phagocytosing harmful pathogens and cellular debris. Microglia emerge from early erythromyeloid progenitors of the yolk sac and enter the developing brain before the establishment of a fully mature blood-brain barrier. In physiological conditions, during brain development, microglia contribute to CNS homeostasis by supporting cell proliferation of neural precursors. In post-natal life, such cells contribute to preserving the integrity of neuronal circuits by sculpting synapses. After a CNS injury, microglia change their morphology and down-regulate those genes supporting homeostatic functions. However, it is still unclear whether such changes are accompanied by molecular and functional modifications that might contribute to the pathological process. While comprehensive transcriptome analyses at the single-cell level have identified specific gene perturbations occurring in the "pathological" microglia, still the precise protective/detrimental role of microglia in neurological disorders is far from being fully elucidated. In this review, the results so far obtained regarding the role of microglia in neurodegenerative disorders will be discussed. There is solid and sound evidence suggesting that regulating microglia functions during disease pathology might represent a strategy to develop future therapies aimed at counteracting brain degeneration in multiple sclerosis, Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Luca Muzio
- Neuroimmunology Unit, Division of Neuroscience, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | | | | |
Collapse
|
30
|
Radandish M, Khalilian P, Esmaeil N. The Role of Distinct Subsets of Macrophages in the Pathogenesis of MS and the Impact of Different Therapeutic Agents on These Populations. Front Immunol 2021; 12:667705. [PMID: 34489926 PMCID: PMC8417824 DOI: 10.3389/fimmu.2021.667705] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 07/31/2021] [Indexed: 01/03/2023] Open
Abstract
Multiple sclerosis (MS) is a demyelinating inflammatory disorder of the central nervous system (CNS). Besides the vital role of T cells, other immune cells, including B cells, innate immune cells, and macrophages (MФs), also play a critical role in MS pathogenesis. Tissue-resident MФs in the brain’s parenchyma, known as microglia and monocyte-derived MФs, enter into the CNS following alterations in CNS homeostasis that induce inflammatory responses in MS. Although the neuroprotective and anti-inflammatory actions of monocyte-derived MФs and resident MФs are required to maintain CNS tolerance, they can release inflammatory cytokines and reactivate primed T cells during neuroinflammation. In the CNS of MS patients, elevated myeloid cells and activated MФs have been found and associated with demyelination and axonal loss. Thus, according to the role of MФs in neuroinflammation, they have attracted attention as a therapeutic target. Also, due to their different origin, location, and turnover, other strategies may require to target the various myeloid cell populations. Here we review the role of distinct subsets of MФs in the pathogenesis of MS and different therapeutic agents that target these cells.
Collapse
Affiliation(s)
- Maedeh Radandish
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parvin Khalilian
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nafiseh Esmaeil
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Environment Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
31
|
Papadopoulos VE, Skarlis C, Evangelopoulos ME, Mavragani CP. Type I interferon detection in autoimmune diseases: challenges and clinical applications. Expert Rev Clin Immunol 2021; 17:883-903. [PMID: 34096436 DOI: 10.1080/1744666x.2021.1939686] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Accumulating data highlights that the dysregulation of type I interferon (IFN) pathways plays a central role in the pathogenesis of several systemic and organ-specific autoimmune diseases. Advances in understanding the role of type I IFNs in these disorders can lead to targeted drug development as well as establishing potential disease biomarkers. AREAS COVERED Here, we summarize current knowledge regarding the role of type I IFNs in the major systemic, as well as organ-specific, autoimmune disorders, including prominent inflammatory CNS disorders like multiple sclerosis. EXPERT OPINION Type I IFN involvement and its clinical associations in a wide spectrum of autoimmune diseases represents a promising area for research aiming to unveil common pathogenetic pathways in systemic and organ-specific autoimmunity.
Collapse
Affiliation(s)
- Vassilis E Papadopoulos
- Demyelinating Diseases Unit, First Department of Neurology, Eginition Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Charalampos Skarlis
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria-Eleftheria Evangelopoulos
- Demyelinating Diseases Unit, First Department of Neurology, Eginition Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Clio P Mavragani
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Joint Academic Rheumatology Program, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
32
|
Regnier-Golanov AS, Dündar F, Zumbo P, Betel D, Hernandez MS, Peterson LE, Lo EH, Golanov EV, Britz GW. Hippocampal Transcriptome Changes After Subarachnoid Hemorrhage in Mice. Front Neurol 2021; 12:691631. [PMID: 34354664 PMCID: PMC8329593 DOI: 10.3389/fneur.2021.691631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/11/2021] [Indexed: 11/13/2022] Open
Abstract
After subarachnoid hemorrhage (SAH), up to 95% of surviving patients suffer from post-SAH syndrome, which includes cognitive deficits with impaired memory, executive functions, and emotional disturbances. Although these long-term cognitive deficits are thought to result from damage to temporomesial-hippocampal areas, the underlying mechanisms remain unknown. To fill this gap in knowledge, we performed a systematic RNA sequencing screen of the hippocampus in a mouse model of SAH. SAH was induced by perforation of the circle of Willis in mice. Four days later, hippocampal RNA was obtained from SAH and control (sham perforation) mice. Next-generation RNA sequencing was used to determine differentially expressed genes in the whole bilateral hippocampi remote from the SAH bleeding site. Functional analyses and clustering tools were used to define molecular pathways. Differential gene expression analysis detected 642 upregulated and 398 downregulated genes (false discovery rate <0.10) in SAH compared to Control group. Functional analyses using IPA suite, Gene Ontology terms, REACTOME pathways, and MsigDB Hallmark gene set collections revealed suppression of oligodendrocytes/myelin related genes, and overexpression of genes related to complement system along with genes associated with innate and adaptive immunity, and extracellular matrix reorganization. Interferon regulatory factors, TGF-β1, and BMP were identified as major orchestrating elements in the hippocampal tissue response. The MEME-Suite identified binding motifs of Krüppel-like factors, zinc finger transcription factors, and interferon regulatory factors as overrepresented DNA promoter motifs. This study provides the first systematic gene and pathway database of the hippocampal response after SAH. Our findings suggest that damage of the entorhinal cortex by subarachnoid blood may remotely trigger specific hippocampal responses, which include suppression of oligodendrocyte function. Identification of these novel pathways may allow for development of new therapeutic approaches for post-SAH cognitive deficits.
Collapse
Affiliation(s)
- Angelique S. Regnier-Golanov
- Laboratory of Cerebrovascular Research, Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| | - Friederike Dündar
- Applied Bioinformatics Core, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States
| | - Paul Zumbo
- Applied Bioinformatics Core, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States
| | - Doron Betel
- Applied Bioinformatics Core, Weill Cornell Medicine, New York, NY, United States
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, United States
| | - Magda S. Hernandez
- Laboratory of Cerebrovascular Research, Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| | - Leif E. Peterson
- Center for Biostatistics, Houston Methodist Research Institute, Houston, TX, United States
| | - Eng H. Lo
- Laboratory of Neuroprotection Research, Harvard Medical School, Massachusetts General Hospital, Charlestown, MA, United States
| | - Eugene V. Golanov
- Laboratory of Cerebrovascular Research, Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| | - Gavin W. Britz
- Laboratory of Cerebrovascular Research, Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
33
|
Simankova A, Bizen N, Saitoh S, Shibata S, Ohno N, Abe M, Sakimura K, Takebayashi H. Ddx20, DEAD box helicase 20, is essential for the differentiation of oligodendrocyte and maintenance of myelin gene expression. Glia 2021; 69:2559-2574. [PMID: 34231259 DOI: 10.1002/glia.24058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 12/17/2022]
Abstract
Oligodendrocytes form myelin sheaths that surround axons, contributing to saltatory conduction and proper central nervous system (CNS) function. Oligodendrocyte progenitor cells (OPCs) are generated during the embryonic stage and differentiate into myelinating oligodendrocytes postnatally. Ddx20 is a multifunctional, DEAD-box helicase involved in multiple cellular processes, including transcription, splicing, microRNA biogenesis, and translation. Although defects in each of these processes result in abnormal oligodendrocyte differentiation and myelination, the involvement of Ddx20 in oligodendrocyte terminal differentiation remains unknown. To address this question, we used Mbp-Cre mice to generate Ddx20 conditional knockout (cKO) mice to allow for the deletion of Ddx20 from mature oligodendrocytes. Mbp-Cre;Ddx20 cKO mice demonstrated small body sizes, behavioral abnormalities, muscle weakness, and short lifespans, with mortality by the age of 2 months old. Histological analyses demonstrated significant reductions in the number of mature oligodendrocytes and drastic reductions in the expression levels of myelin-associated mRNAs, such as Mbp and Plp at postnatal day 42. The number of OPCs did not change. A thin myelin layer was observed for large-diameter axons in Ddx20 cKO mice, based on electron microscopic analysis. A bromodeoxyuridine (BrdU) labeling experiment demonstrated that terminal differentiation was perturbed from ages 2 weeks to 7 weeks in the CNS of Mbp-Cre;Ddx20 cKO mice. The activation of mitogen-activated protein (MAP) kinase, which promotes myelination, was downregulated in the Ddx20 cKO mice based on immunohistochemical detection. These results indicate that Ddx20 is an essential factor for terminal differentiation of oligodendrocytes and maintenance of myelin gene expression.
Collapse
Affiliation(s)
- Anna Simankova
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Norihisa Bizen
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Sei Saitoh
- Section of Electron Microscopy, Supportive Center for Brain Research, National Institute for Physiological Sciences, Okazaki, Japan.,Department of Biomedical Molecular Sciences (Anatomy II), Fujita Health University School of Medicine, Toyoake, Japan
| | - Shinsuke Shibata
- Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Nobuhiko Ohno
- Department of Anatomy, Division of Histology and Cell Biology, School of Medicine, Jichi Medical University, Shimotsuke, Japan.,Division of Ultrastructural Research, National Institute for Physiological Sciences, Okazaki, Japan
| | - Manabu Abe
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, Japan
| | - Kenji Sakimura
- Department of Animal Model Development, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.,Center for Coordination of Research Facilities, Niigata University, Niigata, Japan
| |
Collapse
|
34
|
Sefiani A, Geoffroy CG. The Potential Role of Inflammation in Modulating Endogenous Hippocampal Neurogenesis After Spinal Cord Injury. Front Neurosci 2021; 15:682259. [PMID: 34220440 PMCID: PMC8249862 DOI: 10.3389/fnins.2021.682259] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/17/2021] [Indexed: 12/24/2022] Open
Abstract
Currently there are approximately 291,000 people suffering from a spinal cord injury (SCI) in the United States. SCI is associated with traumatic changes in mobility and neuralgia, as well as many other long-term chronic health complications, including metabolic disorders, diabetes mellitus, non-alcoholic steatohepatitis, osteoporosis, and elevated inflammatory markers. Due to medical advances, patients with SCI survive much longer than previously. This increase in life expectancy exposes them to novel neurological complications such as memory loss, cognitive decline, depression, and Alzheimer's disease. In fact, these usually age-associated disorders are more prevalent in people living with SCI. A common factor of these disorders is the reduction in hippocampal neurogenesis. Inflammation, which is elevated after SCI, plays a major role in modulating hippocampal neurogenesis. While there is no clear consensus on the mechanism of the decline in hippocampal neurogenesis and cognition after SCI, we will examine in this review how SCI-induced inflammation could modulate hippocampal neurogenesis and provoke age-associated neurological disorders. Thereafter, we will discuss possible therapeutic options which may mitigate the influence of SCI associated complications on hippocampal neurogenesis.
Collapse
|
35
|
Rasouli J, Casella G, Ishikawa LLW, Thome R, Boehm A, Ertel A, Melo-Silva CR, Mari ER, Porazzi P, Zhang W, Xiao D, Sigal LJ, Fortina P, Zhang GX, Rostami A, Ciric B. IFN-β Acts on Monocytes to Ameliorate CNS Autoimmunity by Inhibiting Proinflammatory Cross-Talk Between Monocytes and Th Cells. Front Immunol 2021; 12:679498. [PMID: 34149716 PMCID: PMC8213026 DOI: 10.3389/fimmu.2021.679498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/12/2021] [Indexed: 01/18/2023] Open
Abstract
IFN-β has been the treatment for multiple sclerosis (MS) for almost three decades, but understanding the mechanisms underlying its beneficial effects remains incomplete. We have shown that MS patients have increased numbers of GM-CSF+ Th cells in circulation, and that IFN-β therapy reduces their numbers. GM-CSF expression by myelin-specific Th cells is essential for the development of experimental autoimmune encephalomyelitis (EAE), an animal model of MS. These findings suggested that IFN-β therapy may function via suppression of GM-CSF production by Th cells. In the current study, we elucidated a feedback loop between monocytes and Th cells that amplifies autoimmune neuroinflammation, and found that IFN-β therapy ameliorates central nervous system (CNS) autoimmunity by inhibiting this proinflammatory loop. IFN-β suppressed GM-CSF production in Th cells indirectly by acting on monocytes, and IFN-β signaling in monocytes was required for EAE suppression. IFN-β increased IL-10 expression by monocytes, and IL-10 was required for the suppressive effects of IFN-β. IFN-β treatment suppressed IL-1β expression by monocytes in the CNS of mice with EAE. GM-CSF from Th cells induced IL-1β production by monocytes, and, in a positive feedback loop, IL-1β augmented GM-CSF production by Th cells. In addition to GM-CSF, TNF and FASL expression by Th cells was also necessary for IL-1β production by monocyte. IFN-β inhibited GM-CSF, TNF, and FASL expression by Th cells to suppress IL-1β secretion by monocytes. Overall, our study describes a positive feedback loop involving several Th cell- and monocyte-derived molecules, and IFN-β actions on monocytes disrupting this proinflammatory loop.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- Autoimmunity/drug effects
- Cell Communication/genetics
- Cell Communication/immunology
- Cytokines/metabolism
- Disease Models, Animal
- Disease Susceptibility/immunology
- Encephalomyelitis, Autoimmune, Experimental/etiology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis
- Interferon-beta/metabolism
- Interferon-beta/pharmacology
- Mice
- Mice, Knockout
- Monocytes/drug effects
- Monocytes/immunology
- Monocytes/metabolism
- T-Lymphocytes, Helper-Inducer/drug effects
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
Collapse
Affiliation(s)
- Javad Rasouli
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Giacomo Casella
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | | | - Rodolfo Thome
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Alexandra Boehm
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Adam Ertel
- Sidney Kimmel Cancer Center, Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Carolina R. Melo-Silva
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Elisabeth R. Mari
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Patrizia Porazzi
- Sidney Kimmel Cancer Center, Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Weifeng Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Dan Xiao
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Luis J. Sigal
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Paolo Fortina
- Sidney Kimmel Cancer Center, Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
- Department of Translation and Precision Medicine, Sapienza University, Rome, Italy
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Abdolmohamad Rostami
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Bogoljub Ciric
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
36
|
Xu Y, Yu L, Liu Y, Tang X, Wang X. Lipopolysaccharide-Induced Microglial Neuroinflammation: Attenuation by FK866. Neurochem Res 2021; 46:1291-1304. [PMID: 33713324 DOI: 10.1007/s11064-021-03267-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/21/2021] [Accepted: 02/03/2021] [Indexed: 12/12/2022]
Abstract
Alleviating microglia-mediated neuroinflammation bears great promise to reduce neurodegeneration. Nicotinamide phosphoribosyltransferase (NAMPT) may exert cytokine-like effect in the brain. However, it remains unclear about role of NAMPT in microglial inflammation. Also, it remains unknown about effect of NAMPT inhibition on microglial inflammation. In the present study, we observed that FK866 (a specific noncompetitive NAMPT inhibitor) dose-dependently inhibited lipopolysaccharide (LPS)-induced proinflammatory mediator (interleukin (IL)-6, IL-1β, inducible nitric oxide synthase, nitric oxide and reactive species) level increase in BV2 microglia cultures. FK866 also significantly inhibited LPS-induced polarization change in microglia. Furthermore, LPS significantly increased NAMPT expression and nuclear factor kappa B (NF-κB) phosphorylation in microglia. FK866 significantly decreased NAMPT expression and NF-κB phosphorylation in LPS-treated microglia. Finally, conditioned medium from microglia cultures co-treated with FK866 and LPS significantly increased SH-SY5Y and PC12 cell viability compared with conditioned medium from microglia cultures treated with LPS alone. Our study strongly indicates that NAMPT may be a promising target for microglia modulation and NAMPT inhibition may attenuate microglial inflammation.
Collapse
Affiliation(s)
- Yaling Xu
- Department of Neurology, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, People's Republic of China
| | - Lijia Yu
- Department of Neurology, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, People's Republic of China
| | - Ying Liu
- Department of Neurology, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, People's Republic of China
| | - Xiaohui Tang
- Department of Neurology, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, People's Republic of China
| | - Xijin Wang
- Department of Neurology, Xinhua Hospital Affiliated To Shanghai Jiao Tong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, People's Republic of China.
| |
Collapse
|
37
|
Mimouna S, Rollins DA, Shibu G, Tharmalingam B, Deochand DK, Chen X, Oliver D, Chinenov Y, Rogatsky I. Transcription cofactor GRIP1 differentially affects myeloid cell-driven neuroinflammation and response to IFN-β therapy. J Exp Med 2021; 218:e20192386. [PMID: 33045064 PMCID: PMC7555412 DOI: 10.1084/jem.20192386] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 07/29/2020] [Accepted: 09/04/2020] [Indexed: 11/18/2022] Open
Abstract
Macrophages (MФ) and microglia (MG) are critical in the pathogenesis of multiple sclerosis (MS) and its mouse model, experimental autoimmune encephalomyelitis (EAE). Glucocorticoids (GCs) and interferon β (IFN-β) are frontline treatments for MS, and disrupting each pathway in mice aggravates EAE. Glucocorticoid receptor-interacting protein 1 (GRIP1) facilitates both GR and type I IFN transcriptional actions; hence, we evaluated the role of GRIP1 in neuroinflammation. Surprisingly, myeloid cell-specific loss of GRIP1 dramatically reduced EAE severity, immune cell infiltration of the CNS, and MG activation and demyelination specifically during the neuroinflammatory phase of the disease, yet also blunted therapeutic properties of IFN-β. MФ/MG transcriptome analyses at the bulk and single-cell levels revealed that GRIP1 deletion attenuated nuclear receptor, inflammatory and, interestingly, type I IFN pathways and promoted the persistence of a homeostatic MG signature. Together, these results uncover the multifaceted function of type I IFN in MS/EAE pathogenesis and therapy, and an unexpectedly permissive role of myeloid cell GRIP1 in neuroinflammation.
Collapse
Affiliation(s)
- Sanda Mimouna
- The David Z. Rosensweig Genomics Center, Hospital for Special Surgery Research Institute, New York, NY
| | - David A. Rollins
- The David Z. Rosensweig Genomics Center, Hospital for Special Surgery Research Institute, New York, NY
- Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY
| | - Gayathri Shibu
- The David Z. Rosensweig Genomics Center, Hospital for Special Surgery Research Institute, New York, NY
- Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY
| | - Bowranigan Tharmalingam
- The David Z. Rosensweig Genomics Center, Hospital for Special Surgery Research Institute, New York, NY
| | - Dinesh K. Deochand
- The David Z. Rosensweig Genomics Center, Hospital for Special Surgery Research Institute, New York, NY
| | - Xi Chen
- The David Z. Rosensweig Genomics Center, Hospital for Special Surgery Research Institute, New York, NY
- Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY
| | - David Oliver
- The David Z. Rosensweig Genomics Center, Hospital for Special Surgery Research Institute, New York, NY
| | - Yurii Chinenov
- The David Z. Rosensweig Genomics Center, Hospital for Special Surgery Research Institute, New York, NY
| | - Inez Rogatsky
- The David Z. Rosensweig Genomics Center, Hospital for Special Surgery Research Institute, New York, NY
- Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY
| |
Collapse
|
38
|
Fox LE, Locke MC, Lenschow DJ. Context Is Key: Delineating the Unique Functions of IFNα and IFNβ in Disease. Front Immunol 2020; 11:606874. [PMID: 33408718 PMCID: PMC7779635 DOI: 10.3389/fimmu.2020.606874] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/11/2020] [Indexed: 12/15/2022] Open
Abstract
Type I interferons (IFNs) are critical effector cytokines of the immune system and were originally known for their important role in protecting against viral infections; however, they have more recently been shown to play protective or detrimental roles in many disease states. Type I IFNs consist of IFNα, IFNβ, IFNϵ, IFNκ, IFNω, and a few others, and they all signal through a shared receptor to exert a wide range of biological activities, including antiviral, antiproliferative, proapoptotic, and immunomodulatory effects. Though the individual type I IFN subtypes possess overlapping functions, there is growing appreciation that they also have unique properties. In this review, we summarize some of the mechanisms underlying differential expression of and signaling by type I IFNs, and we discuss examples of differential functions of IFNα and IFNβ in models of infectious disease, cancer, and autoimmunity.
Collapse
Affiliation(s)
- Lindsey E. Fox
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, United States
| | - Marissa C. Locke
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, United States
| | - Deborah J. Lenschow
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, United States
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States
| |
Collapse
|
39
|
Wlodarczyk A, Khorooshi R, Marczynska J, Holtman IR, Burton M, Jensen KN, Blaabjerg M, Meyer M, Thomassen M, Eggen BJL, Asgari N, Owens T. Type I interferon-activated microglia are critical for neuromyelitis optica pathology. Glia 2020; 69:943-953. [PMID: 33241604 DOI: 10.1002/glia.23938] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 10/13/2020] [Accepted: 11/11/2020] [Indexed: 01/26/2023]
Abstract
Neuromyelitis optica (NMO) is an inflammatory disease of the central nervous system (CNS) most frequently mediated by serum autoantibodies against the water channel aquaporin 4, expressed on CNS astrocytes, resulting in primary astrocytopathy. There is no cure for NMO, and treatment with Type I interferon (IFNI)-IFNβ is ineffective or even detrimental. We have previously shown that both NMO lesions and associated microglial activation were reduced in mice lacking the receptor for IFNβ. However, the role of microglia in NMO is not well understood. In this study, we clarify the pathomechanism for IFNI dependence of and the role of microglia in experimental NMO. Transcriptome analysis showed a strong IFNI footprint in affected CNS tissue as well as in microglial subpopulations. Treatment with IFNβ led to exacerbated pathology and further microglial activation as evidenced by expansion of a CD11c+ subset of microglia. Importantly, depletion of microglia led to suppression of pathology and decrease of IFNI signature genes. Our data show a pro-pathologic role for IFNI-activated microglia in NMO and open new perspectives for microglia-targeted therapies.
Collapse
Affiliation(s)
- Agnieszka Wlodarczyk
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Brain Research InterDisciplinary Guided Excellence (BRIDGE), University of Southern Denmark, Denmark
| | - Reza Khorooshi
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Brain Research InterDisciplinary Guided Excellence (BRIDGE), University of Southern Denmark, Denmark
| | - Joanna Marczynska
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Brain Research InterDisciplinary Guided Excellence (BRIDGE), University of Southern Denmark, Denmark
| | - Inge R Holtman
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Mark Burton
- Department of Genetics, Odense University Hospital, Odense, Denmark
| | - Kirstine Nolling Jensen
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Brain Research InterDisciplinary Guided Excellence (BRIDGE), University of Southern Denmark, Denmark
| | - Morten Blaabjerg
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Brain Research InterDisciplinary Guided Excellence (BRIDGE), University of Southern Denmark, Denmark.,Department of Neurology, Odense University Hospital and Neurology Research Unit, Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark
| | - Morten Meyer
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Brain Research InterDisciplinary Guided Excellence (BRIDGE), University of Southern Denmark, Denmark
| | - Mads Thomassen
- Department of Genetics, Odense University Hospital, Odense, Denmark
| | - Bart J L Eggen
- Brain Research InterDisciplinary Guided Excellence (BRIDGE), University of Southern Denmark, Denmark
| | - Nasrin Asgari
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Brain Research InterDisciplinary Guided Excellence (BRIDGE), University of Southern Denmark, Denmark.,Department of Neurology, Slagelse Hospital, Slagelse, Denmark
| | - Trevor Owens
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Brain Research InterDisciplinary Guided Excellence (BRIDGE), University of Southern Denmark, Denmark
| |
Collapse
|
40
|
González LF, Acuña E, Arellano G, Morales P, Sotomayor P, Oyarzun-Ampuero F, Naves R. Intranasal delivery of interferon-β-loaded nanoparticles induces control of neuroinflammation in a preclinical model of multiple sclerosis: A promising simple, effective, non-invasive, and low-cost therapy. J Control Release 2020; 331:443-459. [PMID: 33220325 DOI: 10.1016/j.jconrel.2020.11.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 12/21/2022]
Abstract
Multiple sclerosis (MS) is an autoimmune disease affecting the central nervous system (CNS). Interferon (IFN)-β constitutes one of the first-line therapies to treat MS, but has limited efficacy due to the injectable systemic administration, short half-life, and limited CNS access. To address these limitations, we developed IFN-β-loaded chitosan/sulfobutylether-β-cyclodextrin nanoparticles (IFN-β-NPs) for delivery of IFN-β into the CNS via the intranasal (i.n.) route. The nanoparticles (NPs) (≈200 nm, polydispersity ≈0.1, and zeta potential ≈20 mV) were prepared by mixing two aqueous solutions and associated human or murine IFN-β with high efficiency (90%). Functional in vitro assays showed that IFN-β-NPs were safe and that IFN-β was steadily released while retaining biological activity. Biodistribution analysis showed an early and high fluorescence in the brain after nasal administration of fluorescent probe-loaded NPs. Remarkably, mice developing experimental autoimmune encephalomyelitis (EAE), an experimental model of MS, exhibited a significant improvement of clinical symptoms in response to intranasal IFN-β-NPs (inIFN-β-NPs), whereas a similar dose of intranasal or systemic free IFN-β had no effect. Importantly, inIFN-β-NPs treatment was equally effective despite a reduction of 78% in the total amount of weekly administered IFN-β. Spinal cords obtained from inIFN-β-NPs-treated EAE mice showed fewer inflammatory foci and demyelination, lower expression of antigen-presenting and costimulatory proteins on CD11b+ cells, and lower astrocyte and microglia activation than control mice. Therefore, IFN-β treatment at tested doses was effective in promoting clinical recovery and control of neuroinflammation in EAE only when associated with NPs. Overall, inIFN-β-NPs represent a potential, effective, non-invasive, and low-cost therapy for MS.
Collapse
Affiliation(s)
- Luis F González
- Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Eric Acuña
- Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Gabriel Arellano
- Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Paola Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences and Department of Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Paula Sotomayor
- Center for Integrative Medicine and Innovative Science, Universidad Andrés Bello, Santiago, Chile
| | - Felipe Oyarzun-Ampuero
- Departamento de Ciencias y Tecnología Farmacéuticas, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile; Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile.
| | - Rodrigo Naves
- Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
41
|
Vezzani B, Carinci M, Patergnani S, Pasquin MP, Guarino A, Aziz N, Pinton P, Simonato M, Giorgi C. The Dichotomous Role of Inflammation in the CNS: A Mitochondrial Point of View. Biomolecules 2020; 10:E1437. [PMID: 33066071 PMCID: PMC7600410 DOI: 10.3390/biom10101437] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/07/2020] [Accepted: 10/10/2020] [Indexed: 12/14/2022] Open
Abstract
Innate immune response is one of our primary defenses against pathogens infection, although, if dysregulated, it represents the leading cause of chronic tissue inflammation. This dualism is even more present in the central nervous system, where neuroinflammation is both important for the activation of reparatory mechanisms and, at the same time, leads to the release of detrimental factors that induce neurons loss. Key players in modulating the neuroinflammatory response are mitochondria. Indeed, they are responsible for a variety of cell mechanisms that control tissue homeostasis, such as autophagy, apoptosis, energy production, and also inflammation. Accordingly, it is widely recognized that mitochondria exert a pivotal role in the development of neurodegenerative diseases, such as multiple sclerosis, Parkinson's and Alzheimer's diseases, as well as in acute brain damage, such in ischemic stroke and epileptic seizures. In this review, we will describe the role of mitochondria molecular signaling in regulating neuroinflammation in central nervous system (CNS) diseases, by focusing on pattern recognition receptors (PRRs) signaling, reactive oxygen species (ROS) production, and mitophagy, giving a hint on the possible therapeutic approaches targeting mitochondrial pathways involved in inflammation.
Collapse
Affiliation(s)
- Bianca Vezzani
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (B.V.); (M.C.); (S.P.); (M.P.P.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
| | - Marianna Carinci
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (B.V.); (M.C.); (S.P.); (M.P.P.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
| | - Simone Patergnani
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (B.V.); (M.C.); (S.P.); (M.P.P.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
| | - Matteo P. Pasquin
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (B.V.); (M.C.); (S.P.); (M.P.P.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
| | - Annunziata Guarino
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
- Department of BioMedical and Specialist Surgical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Nimra Aziz
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
- Department of BioMedical and Specialist Surgical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Paolo Pinton
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (B.V.); (M.C.); (S.P.); (M.P.P.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola (RA), Italy
| | - Michele Simonato
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
- Department of BioMedical and Specialist Surgical Sciences, University of Ferrara, 44121 Ferrara, Italy
- School of Medicine, University Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Carlotta Giorgi
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (B.V.); (M.C.); (S.P.); (M.P.P.); (P.P.)
- Laboratory of Technologies for Advanced Therapy (LTTA), Technopole of Ferrara, 44121 Ferrara, Italy; (A.G.); (N.A.); (M.S.)
| |
Collapse
|
42
|
Owens T, Benmamar-Badel A, Wlodarczyk A, Marczynska J, Mørch MT, Dubik M, Arengoth DS, Asgari N, Webster G, Khorooshi R. Protective roles for myeloid cells in neuroinflammation. Scand J Immunol 2020; 92:e12963. [PMID: 32851668 DOI: 10.1111/sji.12963] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/14/2020] [Accepted: 08/19/2020] [Indexed: 12/20/2022]
Abstract
Myeloid cells represent the major cellular component of innate immune responses. Myeloid cells include monocytes and macrophages, granulocytes (neutrophils, basophils and eosinophils) and dendritic cells (DC). The role of myeloid cells has been broadly described both in physiological and in pathological conditions. All tissues or organs are equipped with resident myeloid cells, such as parenchymal microglia in the brain, which contribute to maintaining homeostasis. Moreover, in case of infection or tissue damage, other myeloid cells such as monocytes or granulocytes (especially neutrophils) can be recruited from the circulation, at first to promote inflammation and later to participate in repair and regeneration. This review aims to address the regulatory roles of myeloid cells in inflammatory diseases of the central nervous system (CNS), with a particular focus on recent work showing induction of suppressive function via stimulation of innate signalling in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE).
Collapse
Affiliation(s)
- Trevor Owens
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark
| | - Anouk Benmamar-Badel
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark.,Department of Neurology, Slagelse Hospital, Slagelse, Denmark
| | - Agnieszka Wlodarczyk
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark
| | - Joanna Marczynska
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark
| | - Marlene T Mørch
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark
| | - Magdalena Dubik
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark
| | - Dina S Arengoth
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark
| | - Nasrin Asgari
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark.,Department of Neurology, Slagelse Hospital, Slagelse, Denmark
| | - Gill Webster
- Innate Immunotherapeutics, Auckland, New Zealand
| | - Reza Khorooshi
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark
| |
Collapse
|
43
|
Plastini MJ, Desu HL, Brambilla R. Dynamic Responses of Microglia in Animal Models of Multiple Sclerosis. Front Cell Neurosci 2020; 14:269. [PMID: 32973458 PMCID: PMC7468479 DOI: 10.3389/fncel.2020.00269] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 07/31/2020] [Indexed: 12/20/2022] Open
Abstract
Microglia play an essential role in maintaining central nervous system (CNS) homeostasis, as well as responding to injury and disease. Most neurological disorders feature microglial activation, a process whereby microglia undergo profound morphological and transcriptional changes aimed at containing CNS damage and promoting repair, but often resulting in overt inflammation that sustains and propagates the neurodegenerative process. This is especially evident in multiple sclerosis (MS), were microglial activation and microglia-driven neuroinflammation are considered key events in the onset, progression, and resolution of the disease. Our understanding of microglial functions in MS has widened exponentially in the last decade by way of new tools and markers to discriminate microglia from other myeloid populations. Consequently, the complex functional and phenotypical diversity of microglia can now be appreciated. This, in combination with a variety of animal models that mimic specific features and processes of MS, has contributed to filling the gap of knowledge in the cascade of events underlying MS pathophysiology. The purpose of this review is to present the most up to date knowledge of the dynamic responses of microglia in the commonly used animal models of MS, specifically the immune-mediated experimental autoimmune encephalomyelitis (EAE) model, and the chemically-induced cuprizone and lysolecithin models. Elucidating the spectrum of microglial functions in these models, from detrimental to protective, is essential to identify emerging targets for therapy and guide drug discovery efforts.
Collapse
Affiliation(s)
- Melanie J Plastini
- The Miami Project To Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States.,The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Haritha L Desu
- The Miami Project To Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States.,The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Roberta Brambilla
- The Miami Project To Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States.,The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL, United States.,Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,BRIDGE-Brain Research Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
44
|
Castellani G, Schwartz M. Immunological Features of Non-neuronal Brain Cells: Implications for Alzheimer's Disease Immunotherapy. Trends Immunol 2020; 41:794-804. [PMID: 32800704 DOI: 10.1016/j.it.2020.07.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/08/2020] [Accepted: 07/11/2020] [Indexed: 12/23/2022]
Abstract
An interaction network exists among cells within the brain, maintaining brain homeostasis and ensuring its functional plasticity. In addition to neurons, participating cells include astrocytes, oligodendrocytes, and microglia. Peripheral immune cells, such as monocytes and lymphocytes, have also been found to play an important role in supporting the brain in health and assisting in its repair. Here, we describe the multiple immune-specific modes of cellular dialogue among cells within the mammalian brain and their crosstalk with the periphery in both health and disease. We further suggest that interventions directed at boosting the peripheral immune response can restore the balance between the brain and the immune system and can rewire their communication to modify chronic neurodegenerative diseases.
Collapse
Affiliation(s)
- Giulia Castellani
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Schwartz
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
45
|
Hayden L, Semenoff T, Schultz V, Merz SF, Chapple KJ, Rodriguez M, Warrington AE, Shi X, McKimmie CS, Edgar JM, Thümmler K, Linington C, Pingen M. Lipid-specific IgMs induce antiviral responses in the CNS: implications for progressive multifocal leukoencephalopathy in multiple sclerosis. Acta Neuropathol Commun 2020; 8:135. [PMID: 32792006 PMCID: PMC7427287 DOI: 10.1186/s40478-020-01011-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/01/2020] [Indexed: 01/07/2023] Open
Abstract
Progressive multi-focal leukoencephalopathy (PML) is a potentially fatal encephalitis caused by JC polyomavirus (JCV). PML principally affects people with a compromised immune system, such as patients with multiple sclerosis (MS) receiving treatment with natalizumab. However, intrathecal synthesis of lipid-reactive IgM in MS patients is associated with a markedly lower incidence of natalizumab-associated PML compared to those without this antibody repertoire. Here we demonstrate that a subset of lipid-reactive human and murine IgMs induce a functional anti-viral response that inhibits replication of encephalitic Alpha and Orthobunyaviruses in multi-cellular central nervous system cultures. These lipid-specific IgMs trigger microglia to produce IFN-β in a cGAS-STING-dependent manner, which induces an IFN-α/β-receptor 1-dependent antiviral response in glia and neurons. These data identify lipid-reactive IgM as a mediator of anti-viral activity in the nervous system and provide a rational explanation why intrathecal synthesis of lipid-reactive IgM correlates with a reduced incidence of iatrogenic PML in MS.
Collapse
|
46
|
Béland LC, Markovinovic A, Jakovac H, De Marchi F, Bilic E, Mazzini L, Kriz J, Munitic I. Immunity in amyotrophic lateral sclerosis: blurred lines between excessive inflammation and inefficient immune responses. Brain Commun 2020; 2:fcaa124. [PMID: 33134918 PMCID: PMC7585698 DOI: 10.1093/braincomms/fcaa124] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/07/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022] Open
Abstract
Despite wide genetic, environmental and clinical heterogeneity in amyotrophic lateral sclerosis, a rapidly fatal neurodegenerative disease targeting motoneurons, neuroinflammation is a common finding. It is marked by local glial activation, T cell infiltration and systemic immune system activation. The immune system has a prominent role in the pathogenesis of various chronic diseases, hence some of them, including some types of cancer, are successfully targeted by immunotherapeutic approaches. However, various anti-inflammatory or immunosuppressive therapies in amyotrophic lateral sclerosis have failed. This prompted increased scrutiny over the immune-mediated processes underlying amyotrophic lateral sclerosis. Perhaps the biggest conundrum is that amyotrophic lateral sclerosis pathogenesis exhibits features of three otherwise distinct immune dysfunctions-excessive inflammation, autoimmunity and inefficient immune responses. Epidemiological and genome-wide association studies show only minimal overlap between amyotrophic lateral sclerosis and autoimmune diseases, so excessive inflammation is usually thought to be secondary to protein aggregation, mitochondrial damage or other stresses. In contrast, several recently characterized amyotrophic lateral sclerosis-linked mutations, including those in TBK1, OPTN, CYLD and C9orf72, could lead to inefficient immune responses and/or damage pile-up, suggesting that an innate immunodeficiency may also be a trigger and/or modifier of this disease. In such cases, non-selective immunosuppression would further restrict neuroprotective immune responses. Here we discuss multiple layers of immune-mediated neuroprotection and neurotoxicity in amyotrophic lateral sclerosis. Particular focus is placed on individual patient mutations that directly or indirectly affect the immune system, and the mechanisms by which these mutations influence disease progression. The topic of immunity in amyotrophic lateral sclerosis is timely and relevant, because it is one of the few common and potentially malleable denominators in this heterogenous disease. Importantly, amyotrophic lateral sclerosis progression has recently been intricately linked to patient T cell and monocyte profiles, as well as polymorphisms in cytokine and chemokine receptors. For this reason, precise patient stratification based on immunophenotyping will be crucial for efficient therapies.
Collapse
Affiliation(s)
| | - Andrea Markovinovic
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
- ENCALS Center Zagreb, 10000 Zagreb, Croatia
| | - Hrvoje Jakovac
- Department of Physiology and Immunology, Medical Faculty, University of Rijeka, 51000 Rijeka, Croatia
| | - Fabiola De Marchi
- Department of Neurology, ALS Centre, University of Piemonte Orientale, “Maggiore della Carità” Hospital, 28100 Novara, Italy
| | - Ervina Bilic
- Department of Neurology, Clinical Hospital Centre Zagreb, 10000 Zagreb, Croatia
- ENCALS Center Zagreb, 10000 Zagreb, Croatia
| | - Letizia Mazzini
- Department of Neurology, ALS Centre, University of Piemonte Orientale, “Maggiore della Carità” Hospital, 28100 Novara, Italy
| | - Jasna Kriz
- CERVO Research Centre, Laval University, Quebec City, Quebec G1J 2G3, Canada
| | - Ivana Munitic
- Laboratory for Molecular Immunology, Department of Biotechnology, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
47
|
Wang SS, Bi HZ, Chu SF, Dong YX, He WB, Tian YJ, Zang YD, Zhang DM, Zhang Z, Chen NH. CZ-7, a new derivative of Claulansine F, promotes remyelination induced by cuprizone by enhancing myelin debris clearance. Brain Res Bull 2020; 159:67-78. [PMID: 32289743 DOI: 10.1016/j.brainresbull.2020.03.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 03/18/2020] [Accepted: 03/23/2020] [Indexed: 12/11/2022]
Abstract
The mechanism of demyelinating diseases is controversial, while demyelination and remyeliantion disorder is the acknowledged etiology and therapeutic target. Untill now, there is no efficient therapy for these diseases. CZ-7, a new derivative of Claulansine F, which has been reported before, were investigated its pro-remyelination effect and its associated mechanism in cuprizone (CPZ)-induced demyelination model. In this study, male C57BL/6 mice were subjected to CPZ (300 mg/kg) through intragastric gavage and were orally administered CZ-7 (20 mg/kg) meanwhile. The results of weight monitoring and behavioral testing showed that CZ-7 can significantly improve behavior dysfunction in the demyelinating mice. Luxol-fast blue (LFB) staining, myelin basic protein (MBP) immunostaining, transmission electron microscopy (TEM) and QPCR results indicated the therapeutic effect of CZ-7 on CPZ mice model. Furthermore, degraded myelin basic protein (dMBP) immunofluorescent staining and oil red O staining showed that CZ-7 contributed to the clearance of degraded myelin debris. More microglia displayed phagocytic shape assembled in corpus callosum (CC) and there was an active process of phagocytosis in microglia after CZ-7 treatment. Immunofluorescent staining and QPCR analysis revealed the M2-polarized phenotype switch of microglia in the process of myelin debris removel, which demostrated the microenvironment improvement of CZ-7. Moreover, immunofluorescent staining of NG2 and O4 demonstated that more oligodendrocyte precursor cells (OPCs) existed in CC after CZ-7 treatment. In conclusion, our results demonstrated CZ-7 has a potential therapeutic effect for MS and other demyelinating diseases through enhancing myelin debris clearance to improve the microenvironment.
Collapse
Affiliation(s)
- Sha-Sha Wang
- Institute of Pharmaceutical & Food Engineering, Shanxi University of Traditional Chinese Medicine, Taiyuan, 030619, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Hao-Zhi Bi
- Institute of Pharmaceutical & Food Engineering, Shanxi University of Traditional Chinese Medicine, Taiyuan, 030619, China
| | - Shi-Feng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yi-Xiao Dong
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Wen-Bin He
- Institute of Pharmaceutical & Food Engineering, Shanxi University of Traditional Chinese Medicine, Taiyuan, 030619, China
| | - Ya-Juan Tian
- Institute of Pharmaceutical & Food Engineering, Shanxi University of Traditional Chinese Medicine, Taiyuan, 030619, China
| | - Ying-Da Zang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Dong-Ming Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Nai-Hong Chen
- Institute of Pharmaceutical & Food Engineering, Shanxi University of Traditional Chinese Medicine, Taiyuan, 030619, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| |
Collapse
|
48
|
Moore Z, Mobilio F, Walker FR, Taylor JM, Crack PJ. Abrogation of type-I interferon signalling alters the microglial response to Aβ 1-42. Sci Rep 2020; 10:3153. [PMID: 32081950 PMCID: PMC7035268 DOI: 10.1038/s41598-020-59917-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/31/2020] [Indexed: 11/08/2022] Open
Abstract
Neuroinflammation and accompanying microglial dysfunction are now appreciated to be involved in Alzheimer's disease (AD) pathogenesis. Critical to the process of neuroinflammation are the type-I interferon (IFN) family of cytokines. Efforts to phenotypically characterize microglia within AD identify distinct populations associated with type-I IFN signalling, yet how this affects underlying microglial function is yet to be fully elucidated. Here we demonstrate that Aβ1-42 exposure increases bioactive levels of type-I IFN produced by primary microglia alongside increased expression of type-I IFN related genes. Primary microglia isolated from brains of APPswePS1ΔE9 mice with ablated type-I IFN signalling show an increased phagocytic ability to uptake FITC-Aβ1-42. Correlative assessment of plaque sizes in aged APPswePS1ΔE9 mice with abrogated type-I IFN signalling show unchanged deposition levels. Microglia from these mice did however show alterations in morphology. This data further highlights the role of type-I IFN signalling within microglia and identifies a role in phagocytosis. As such, targeting both microglial and global type-I IFN signalling presents as a novel therapeutic strategy for AD management.
Collapse
Affiliation(s)
- Zachery Moore
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Australia
| | - Frank Mobilio
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Australia
| | - Frederick R Walker
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, Australia
| | - Juliet M Taylor
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Australia.
| | - Peter J Crack
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Australia.
| |
Collapse
|
49
|
Glial Factors Regulating White Matter Development and Pathologies of the Cerebellum. Neurochem Res 2020; 45:643-655. [PMID: 31974933 PMCID: PMC7058568 DOI: 10.1007/s11064-020-02961-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/07/2020] [Accepted: 01/09/2020] [Indexed: 12/31/2022]
Abstract
The cerebellum is a brain region that undergoes extremely dynamic growth during perinatal and postnatal development which is regulated by the proper interaction between glial cells and neurons with a complex concert of growth factors, chemokines, cytokines, neurotransmitters and transcriptions factors. The relevance of cerebellar functions for not only motor performance but also for cognition, emotion, memory and attention is increasingly being recognized and acknowledged. Since perturbed circuitry of cerebro-cerebellar trajectories can play a role in many central nervous system pathologies and thereby contribute to neurological symptoms in distinct neurodevelopmental and neurodegenerative diseases, is it the aim with this mini-review to highlight the pathways of glia–glia interplay being involved. The designs of future treatment strategies may hence be targeted to molecular pathways also playing a role in development and disease of the cerebellum.
Collapse
|
50
|
Khorooshi R, Marczynska J, Dieu RS, Wais V, Hansen CR, Kavan S, Thomassen M, Burton M, Kruse T, Webster GA, Owens T. Innate signaling within the central nervous system recruits protective neutrophils. Acta Neuropathol Commun 2020; 8:2. [PMID: 31915070 PMCID: PMC6950927 DOI: 10.1186/s40478-019-0876-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 12/20/2019] [Indexed: 01/27/2023] Open
Abstract
There is great interest in understanding how the central nervous system (CNS) communicates with the immune system for recruitment of protective responses. Infiltrating phagocytic monocytes and granulocytes are implicated in neuroinflammation in multiple sclerosis and its animal model experimental autoimmune encephalomyelitis (EAE). To investigate how CNS endogenous signals can be harnessed to promote anti-inflammatory programs, we have used a particulate Toll-like receptor 9 and nucleotide-oligomerization domain 2 bispecific innate ligand (MIS416), to address whether its phagocytosis within the CNS recruits protective myeloid cells. We find that MIS416 injected intrathecally into the cerebrospinal fluid via the cisterna magna induced a local chemokine response that recruited blood-derived monocytes and neutrophils to the CNS. These cells phagocytosed MIS416. The increase in EAE severity normally seen from time of onset did not occur in mice receiving MIS416. This suppression of disease symptoms was dependent on expression of the type I interferon receptor (IFNAR). Transfer of intrathecal MIS416-induced neutrophils suppressed EAE in recipient mice, while monocytes did not transfer protection. MIS416-induced neutrophils showed increased IL-10 expression that was IFNAR1-driven. In contrast to intrathecal administration, intravenous administration of MIS416 led to monocyte but not neutrophil infiltration to the CNS. We thus identify a CNS-intrinsic and -specific phagocytosis-induced recruitment of anti-inflammatory neutrophils that contribute to CNS homeostasis and may have therapeutic potential.
Collapse
|