1
|
Tripathi A, Rai NK, Perles A, Courtney H, Jones C, Sapra A, Plemel J, Dutta R. Dicer deficiency affects microglial function during demyelination and impairs remyelination. Neurobiol Dis 2025; 208:106879. [PMID: 40120829 DOI: 10.1016/j.nbd.2025.106879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/10/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025] Open
Abstract
Microglia are essential regulators of central nervous system (CNS) homeostasis, playing key roles in demyelination and remyelination. Dysregulated microglial activity contributes to pathological inflammation and impaired repair processes in demyelinating diseases. Here, we investigate the role of Dicer1, a critical enzyme in microRNA biogenesis, in affecting microglial function, demyelination, and remyelination. Loss of Dicer1 in microglia resulted in amplified inflammatory responses, defective myelin debris clearance, and disruption of metabolic homeostasis, leading to exacerbated demyelination and delayed remyelination. Transcriptomic analysis revealed significant upregulation of inflammatory pathways, including interferon signaling and JAK/STAT activation, alongside a loss of homeostatic microglial gene expression. Protein-level validation confirmed sustained secretion of pro-inflammatory cytokines such as IFN-γ, IL-16, and CXCL12, creating a chronic inflammatory environment that impaired remyelination. Furthermore, Dicer1-deficient microglia failed to support oligodendrocyte progenitor cells (OPCs) differentiation/maturation, with increased apoptosis of mature oligodendrocytes (OLs), contributing to remyelination failure. These findings identify Dicer1 as a critical regulator of microglial homeostasis and inflammation resolution, highlighting its potential as a therapeutic target to mitigate inflammation and promote repair in demyelinating diseases.
Collapse
Affiliation(s)
- Ajai Tripathi
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, USA
| | | | - Aaron Perles
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, USA
| | - Haley Courtney
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, USA
| | - Claire Jones
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, USA
| | - Adya Sapra
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, USA
| | - Jason Plemel
- Neuroscience and Mental Health Institute, Department of Medicine, Division of Neurology, Department of Medical Microbiology and Immunology, University of Alberta, Canada
| | - Ranjan Dutta
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH, USA; Cleveland Clinic Lerner College of Medicine, Cleveland, OH, USA.
| |
Collapse
|
2
|
Aisenberg WH, O'Brien CA, Sangster M, Yaqoob F, Zhang Y, Temsamrit B, Thom S, Gosse L, Chaluvadi S, Elfayomi B, Lee G, Polam V, Levitt EM, Liu G, Lombroso SI, Nemec KM, Clowry G, Nieves C, Rawat P, Church E, Martinez D, Shoffler C, Kancheva D, Petucci C, Taylor D, Kofler J, Erskine D, Movahedi K, Bennett ML, Bennett FC. Direct microglia replacement reveals pathologic and therapeutic contributions of brain macrophages to a monogenic neurological disease. Immunity 2025:S1074-7613(25)00138-4. [PMID: 40311614 DOI: 10.1016/j.immuni.2025.03.019] [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: 07/03/2024] [Revised: 12/20/2024] [Accepted: 03/26/2025] [Indexed: 05/03/2025]
Abstract
Krabbe disease, also named globoid cell (GC) leukodystrophy (GLD) for its distinct lipid-laden macrophages, is a severe leukodystrophy caused by galactosylceramidase (GALC) mutations. Hematopoietic stem cell transplant (HSCT) ameliorates disease and is associated with central nervous system (CNS) engraftment of GALC+ donor macrophages. Yet, the role of macrophages in GLD pathophysiology and HSCT remains unclear. Using single-cell sequencing, we revealed early interferon response signatures that preceded progressively severe macrophage dyshomeostasis and identified a molecular signature of GCs, which we validated in human brain specimens. Genetic depletion and direct microglia replacement by CNS monocyte injection rapidly replaced >80% of endogenous microglia with healthy macrophages in the twitcher (GalcW355∗) mouse model of GLD. Perinatal microglia replacement completely normalized transcriptional signatures, rescued histopathology, and doubled average survival. Overall, we uncovered distinct forms of microglial dysfunction and evidence that direct, CNS-limited microglia replacement improves a monogenic neurodegenerative disease, identifying a promising therapeutic target.
Collapse
Affiliation(s)
- William H Aisenberg
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carleigh A O'Brien
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Madison Sangster
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Fazeela Yaqoob
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuanchao Zhang
- Department of Biomedical Informatics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Brian Temsamrit
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Searlait Thom
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Luca Gosse
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Sai Chaluvadi
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bilal Elfayomi
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gavin Lee
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Vidhur Polam
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eli M Levitt
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gary Liu
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sonia I Lombroso
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kelsey M Nemec
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gavin Clowry
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Cassaundra Nieves
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Priyanka Rawat
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Emily Church
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Martinez
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Clarissa Shoffler
- Penn Metabolomics Core, Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daliya Kancheva
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Christopher Petucci
- Penn Metabolomics Core, Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Deanne Taylor
- Department of Biomedical Informatics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Julia Kofler
- Division of Neuropathology, Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Daniel Erskine
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Kiavash Movahedi
- Brain and Systems Immunology Laboratory, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mariko L Bennett
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - F Chris Bennett
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Hill BM, Holloway RK, Forbes LH, Davies CL, Monteiro JK, Brown CM, Rose J, Fudge N, Plant PJ, Mahmood A, Brand-Arzamendi K, Kent SA, Molina-Gonzalez I, Gyoneva S, Ransohoff RM, Wipke B, Priller J, Schneider R, Moore CS, Miron VE. Monocyte-secreted Wnt reduces the efficiency of central nervous system remyelination. PLoS Biol 2025; 23:e3003073. [PMID: 40233100 PMCID: PMC12052099 DOI: 10.1371/journal.pbio.3003073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/05/2025] [Accepted: 02/18/2025] [Indexed: 04/17/2025] Open
Abstract
The regeneration of myelin in the central nervous system (CNS) reinstates nerve health and function, yet its decreased efficiency with aging and progression of neurodegenerative disease contributes to axonal loss and/or degeneration. Although CNS myeloid cells have been implicated in regulating the efficiency of remyelination, the distinct contribution of blood monocytes versus that of resident microglia is unclear. Here, we reveal that monocytes have non-redundant functions compared to microglia in regulating remyelination. Using a transgenic mouse in which classical monocytes have reduced egress from bone marrow (Ccr2-/-), we demonstrate that monocytes drive the timely onset of oligodendrocyte differentiation and myelin protein expression, yet impede myelin production. Ribonucleic acid sequencing revealed a Wnt signature in wild-type mouse lesion monocytes, which was confirmed in monocytes from multiple sclerosis white matter lesions and blood. Genetic or pharmacological inhibition of Wnt release by monocytes increased remyelination. Our findings reveal monocytes to be critical regulators of remyelination and identify monocytic Wnt signaling as a promising therapeutic target to inhibit for increased efficiency of CNS regeneration.
Collapse
Affiliation(s)
- Bianca M. Hill
- Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, Ontario, Canada
- BARLO Multiple Sclerosis Centre, St. Michael’s Hospital, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Rebecca K. Holloway
- Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, Ontario, Canada
- BARLO Multiple Sclerosis Centre, St. Michael’s Hospital, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Lindsey H. Forbes
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Claire L. Davies
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Jonathan K. Monteiro
- Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, Ontario, Canada
- BARLO Multiple Sclerosis Centre, St. Michael’s Hospital, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Christina M. Brown
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Jamie Rose
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Neva Fudge
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Newfoundland, Canada
| | - Pamela J. Plant
- Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Ayisha Mahmood
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Koroboshka Brand-Arzamendi
- Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, Ontario, Canada
- BARLO Multiple Sclerosis Centre, St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Sarah A. Kent
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Irene Molina-Gonzalez
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Stefka Gyoneva
- Previously at Biogen Ltd, Cambridge, Massachusetts, United States of America
| | - Richard M. Ransohoff
- Previously at Biogen Ltd, Cambridge, Massachusetts, United States of America
- Third Rock Ventures, Boston, Massachusetts, United States of America
| | - Brian Wipke
- Previously at Biogen Ltd, Cambridge, Massachusetts, United States of America
- Manifold Bio, Boston, Massachusetts, United States of America
| | - Josef Priller
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Clinical Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin and DZNE, Berlin, Germany
| | - Raphael Schneider
- Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, Ontario, Canada
- BARLO Multiple Sclerosis Centre, St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Craig S. Moore
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Newfoundland, Canada
| | - Veronique E. Miron
- Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, Ontario, Canada
- BARLO Multiple Sclerosis Centre, St. Michael’s Hospital, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| |
Collapse
|
4
|
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
|
5
|
Gross PS, Durán-Laforet V, Ho LT, Melchor GS, Zia S, Manavi Z, Barclay WE, Lee SH, Shults N, Selva S, Alvarez E, Plemel JR, Fu MM, Schafer DP, Huang JK. Senescent-like microglia limit remyelination through the senescence associated secretory phenotype. Nat Commun 2025; 16:2283. [PMID: 40055369 PMCID: PMC11889183 DOI: 10.1038/s41467-025-57632-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
The capacity to regenerate myelin in the central nervous system diminishes with age. This decline is particularly evident in multiple sclerosis (MS), a chronic demyelinating disease. Whether cellular senescence, a hallmark of aging, contributes to remyelination impairment remains unknown. Here, we show that senescent cells accumulate within demyelinated lesions after injury, and treatments with senolytics enhances remyelination in young and middle-aged mice but not aged mice. In young mice, we observe the upregulation of senescence-associated transcripts, primarily in microglia and macrophages, after demyelination, followed by a reduction during remyelination. However, in aged mice, senescence-associated factors persist within lesions, correlating with inefficient remyelination. Proteomic analysis of the senescence-associated secretory phenotype (SASP) reveals elevated levels of CCL11/Eotaxin-1 in lesions of aged mice, which is found to inhibit oligodendrocyte maturation. These results suggest therapeutic targeting of SASP components, such as CCL11, may improve remyelination in aging and MS.
Collapse
Affiliation(s)
- Phillip S Gross
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, USA
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Violeta Durán-Laforet
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Lana T Ho
- Department of Molecular and Cell Biology (MCB), University of California Berkeley, Berkeley, CA, USA
| | - George S Melchor
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, USA
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Sameera Zia
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Zeeba Manavi
- Department of Biology, Georgetown University, Washington, DC, USA
| | - William E Barclay
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Sung Hyun Lee
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Nataliia Shults
- Department of Biology, Georgetown University, Washington, DC, USA
| | - Sean Selva
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Enrique Alvarez
- Department of Neurology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jason R Plemel
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Meng-Meng Fu
- Department of Molecular and Cell Biology (MCB), University of California Berkeley, Berkeley, CA, USA
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jeffrey K Huang
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, USA.
- Department of Biology, Georgetown University, Washington, DC, USA.
| |
Collapse
|
6
|
Zhou H, Zhou M, Liao X, Zhang L, Wei H, Lu Y, Zhang Y, Huang H, Hu Y, Chen T, Lv Z. The Innate Immune Sensor Zbp1 Mediates Central Nervous System Inflammation Induced by Angiostrongylus Cantonensis by Promoting Macrophage Inflammatory Phenotypes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413675. [PMID: 39853924 PMCID: PMC11923990 DOI: 10.1002/advs.202413675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/14/2025] [Indexed: 01/26/2025]
Abstract
Angiostrongylus cantonensis (AC) is the leading cause of eosinophilic meningoencephalitis worldwide. The neuroimmune interactions between peripheral and central immune systems in angiostrongyliasis remain unclear. In this study, significant infiltration of eosinophils, myeloid cells, macrophages, neutrophils, and Ly6C monocytes is observed in the brains of AC-infected mice, with macrophages being the most abundant. RNA-seq and SMART-seq analysis of pattern recognition receptor (PRR) and DNA sensor gene sets revealed a marked increase in Z-DNA binding protein 1 (Zbp1) expression in infected mice. Confocal microscopy, RT-qPCR, western blotting, and immunohistochemistry further confirmed that Zbp1 is specifically upregulated in macrophages and microglia. Using Zbp1-knockout mice and flow cytometry, it is found that knockout of Zbp1 enhanced lymphocyte infiltration and natural killer cell cytotoxicity, modulating the immune microenvironment in the central nervous system (CNS) during AC infection. Mechanistically, it is revealed that in macrophage Zbp1 directly binds to receptor-interacting protein 3 (RIP3) to promote its phosphorylation, subsequently facilitating the phosphorylation of mixed lineage kinase domain-like protein (Mlkl). The activated Zbp1-pRIP3-pMlkl axis leads to necroptosis and upregulates pro-inflammatory cytokines including TNF-α, IL-1α, CXCL9, CXCL10 in macrophages, which recruits and activates immune cells. These findings offer new insights into the pathogenic mechanisms of angiostrongyliasis and suggest potential therapeutic strategies.
Collapse
Affiliation(s)
- Hongli Zhou
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Clinical Medical Research CenterThe Second Affiliated HospitalArmy Medical UniversityChongqing400037China
| | - Minyu Zhou
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
| | - XiPing Liao
- Clinical Medical Research CenterThe Second Affiliated HospitalArmy Medical UniversityChongqing400037China
| | - Liangyu Zhang
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
| | - Hang Wei
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
| | - Yuting Lu
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
| | - Yiqing Zhang
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
| | - Hui Huang
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
| | - Yue Hu
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
| | - Tao Chen
- Department of NeurologyHainan General Hospital,Hainan Affiliated Hospital of Hainan Medical UniversityHaikouHainan570311China
| | - Zhiyue Lv
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
- Department of NeurologyHainan General Hospital,Hainan Affiliated Hospital of Hainan Medical UniversityHaikouHainan570311China
- Key Laboratory of Tropical Translational Medicine of Ministry of EducationHainan Medical UniversityHaikouHainan570216China
| |
Collapse
|
7
|
Ju H, Kim ID, Pavlova I, Mu S, Park KW, Minkler J, Madkoor A, Wang W, Wang X, Wu Z, Yang J, Febbraio M, Cave JW, Cho S. Ischemic Conditioning Promotes Transneuronal Survival and Stroke Recovery via CD36-Mediated Efferocytosis. Circ Res 2025; 136:e34-e51. [PMID: 39886760 PMCID: PMC11867857 DOI: 10.1161/circresaha.124.325428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Remote ischemic conditioning (RIC) has been implicated in cross-organ protection in cerebrovascular disease, including stroke. However, the lack of a consensus protocol and controversy over the clinical therapeutic outcomes of RIC suggest an inadequate mechanistic understanding of RIC. The current study identifies RIC-induced molecular and cellular events in the blood, which enhance long-term functional recovery in experimental cerebral ischemia. METHODS Naive mice or mice subjected to transient ischemic stroke were randomly selected to receive sham conditioning or RIC in the hindlimb at 2 hours post-stroke. At 3 days post-stroke, monocyte composition in the blood was analyzed, and brain tissue was examined for monocyte-derived macrophage (Mφ), levels of efferocytosis, and CD36 expression. Mouse with a specific deletion of CD36 in monocytes/Mφs was used to establish the role of CD36 in RIC-mediated modulation of efferocytosis, transneuronal degeneration, and recovery following stroke. RESULTS RIC applied 2 hours after stroke increased the entry of monocytes into the injured brain. In the postischemic brain, Mφ had increased levels of CD36 expression and efferocytosis. These changes in brain Mφ were derived from RIC-induced changes in circulating monocytes. In the blood, RIC increased CD36 expression in circulating monocytes and shifted monocytes to a proinflammatory Lymphocyte antigen 6 complex (LY6C)High state. Conditional deletion of CD36 in Mφ abrogated the RIC-induced monocyte shift in the blood and efferocytosis in the brain. During the recovery phase of stroke, RIC rescued the loss of the volume and of tyrosine hydroxylase+ neurons in substantia nigra and behavioral deficits in wild-type mice but not in mice with a specific deletion of CD36 in monocytes/Mφs. CONCLUSIONS RIC induces a shift in monocytes to a proinflammatory state with elevated CD36 levels, and this is associated with CD36-dependent efferocytosis in Mφs that rescues delayed transneuronal degeneration in the postischemic brain and promotes stroke recovery. Together, these findings provide novel insight into our mechanistic understanding of how RIC improves poststroke recovery.
Collapse
Affiliation(s)
- Hyunwoo Ju
- Burke Neurological Institute, 785 Mamaroneck Ave, White Plains, NY 10605, USA
- Feil Brain Mind Research Institute, Weill Cornell Medicine, 1600 York Avenue, New York, NY, USA
| | - Il-doo Kim
- Department of Anatomy, Inha University School of Medicine, 1018, 60 Anniversary Hall, 100 Inharo, Incheon, South Korea
| | - Ina Pavlova
- Burke Neurological Institute, 785 Mamaroneck Ave, White Plains, NY 10605, USA
| | - Shang Mu
- Helen & Robert Appel Alzheimer’s Disease Research Institute, Weill Cornell Medicine, 413 E 69th St, New York, NY 10021, USA
- Feil Brain Mind Research Institute, Weill Cornell Medicine, 1600 York Avenue, New York, NY, USA
| | - Keun Woo Park
- Burke Neurological Institute, 785 Mamaroneck Ave, White Plains, NY 10605, USA
- Feil Brain Mind Research Institute, Weill Cornell Medicine, 1600 York Avenue, New York, NY, USA
| | - Joseph Minkler
- Burke Neurological Institute, 785 Mamaroneck Ave, White Plains, NY 10605, USA
| | - Ahmed Madkoor
- Burke Neurological Institute, 785 Mamaroneck Ave, White Plains, NY 10605, USA
| | - Wei Wang
- Helen & Robert Appel Alzheimer’s Disease Research Institute, Weill Cornell Medicine, 413 E 69th St, New York, NY 10021, USA
- Feil Brain Mind Research Institute, Weill Cornell Medicine, 1600 York Avenue, New York, NY, USA
| | - Xiaoman Wang
- Helen & Robert Appel Alzheimer’s Disease Research Institute, Weill Cornell Medicine, 413 E 69th St, New York, NY 10021, USA
- Feil Brain Mind Research Institute, Weill Cornell Medicine, 1600 York Avenue, New York, NY, USA
| | - Zhuhao Wu
- Helen & Robert Appel Alzheimer’s Disease Research Institute, Weill Cornell Medicine, 413 E 69th St, New York, NY 10021, USA
- Feil Brain Mind Research Institute, Weill Cornell Medicine, 1600 York Avenue, New York, NY, USA
| | - Jiwon Yang
- Innovation & Product Development, The Jackson Laboratory, Sacramento, CA, USA
| | - Maria Febbraio
- Department of Dentistry and Dental Hygiene, University of Alberta, Edmonton, Alberta, Canada
| | | | - Sunghee Cho
- Burke Neurological Institute, 785 Mamaroneck Ave, White Plains, NY 10605, USA
- Feil Brain Mind Research Institute, Weill Cornell Medicine, 1600 York Avenue, New York, NY, USA
| |
Collapse
|
8
|
John RK, Vogel SP, Zia S, Lee KV, Nguyen AT, Torres-Espin A, Fenrich KK, Ng C, Schmidt EKA, Vavrek R, Raposo PJF, Smith K, Fouad K, Plemel JR. Reawakening inflammation in the chronically injured spinal cord using lipopolysaccharide induces diverse microglial states. J Neuroinflammation 2025; 22:56. [PMID: 40022205 PMCID: PMC11871772 DOI: 10.1186/s12974-025-03379-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/14/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Rehabilitative training is an effective method to promote recovery following spinal cord injury (SCI), with lower training efficacy observed in the chronic stage. The increased training efficacy during the subacute period is associated with a shift towards a more adaptive or proreparative state induced by the SCI. A potential link is SCI-induced inflammation, which is elevated in the subacute period, and, as injection of lipopolysaccharide (LPS) alongside training improves recovery in chronic SCI, suggesting LPS could reopen a window of plasticity late after injury. Microglia may play a role in LPS-mediated plasticity as they react to LPS and are implicated in facilitating recovery following SCI. However, it is unknown how microglia change in response to LPS following SCI to promote neuroplasticity. MAIN BODY Here we used single-cell RNA sequencing to examine microglial responses in subacute and chronic SCI with and without an LPS injection. We show that subacute SCI is characterized by a disease-associated microglial (DAM) signature, while chronic SCI is highly heterogeneous, with both injury-induced and homeostatic states. DAM states exhibit predicted metabolic pathway activity and neuronal interactions that are associated with potential mediators of plasticity. With LPS injection, microglia shifted away from the homeostatic signature to a primed, translation-associated state and increased DAM in degenerated tracts caudal to the injury. CONCLUSION Microglial states following an inflammatory stimulus in chronic injury incompletely recapitulate the subacute injury environment, showing both overlapping and distinct microglial signatures across time and with LPS injection. Our results contribute to an understanding of how microglia and LPS-induced neuroinflammation contribute to plasticity following SCI.
Collapse
Affiliation(s)
- Rebecca K John
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Sadie P Vogel
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Sameera Zia
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Kelly V Lee
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, Canada
| | - Antoinette T Nguyen
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Abel Torres-Espin
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
- School of Public Health Sciences, Faculty of Health, University of Waterloo, Waterloo, Canada
| | - Keith K Fenrich
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Carmen Ng
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Emma K A Schmidt
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Romana Vavrek
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Pamela J F Raposo
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Keira Smith
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Karim Fouad
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada.
- Department of Physical Therapy, Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada.
| | - Jason R Plemel
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada.
- Department of Medicine, Division of Neurology, University of Alberta, Edmonton, Canada.
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Canada.
- Li Ka Shing Institute of Virology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada.
| |
Collapse
|
9
|
Liu Y, Li T, Xiong J. Alzheimer's disease and diabetes-associated cognitive dysfunction: the microglia link? Metab Brain Dis 2025; 40:85. [PMID: 39754611 DOI: 10.1007/s11011-024-01516-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 12/18/2024] [Indexed: 01/06/2025]
Abstract
Alzheimer's disease (AD) is characterized by the accumulation of amyloid-β (Aβ) plaques and the aggregation of tau protein, resulting in intense memory loss and dementia. Diabetes-associated cognitive dysfunction (DACD) is a complication of diabetes mellitus, which is associated with decreased cognitive function and impaired memory. A growing body of literature emphasize the involvement of microglia in AD and DACD. Although AD and DACD share some common features related to symptomatology and pathophysiology, the characteristics and heterogeneity of microglia remain largely unknown in these two diseases. In this study, multiple bioinformatics analyses were performed to analyze the frequency, altered genes, cell-cell communication, and subtypes of microglia in AD and DACD mouse models based on two publicly single-nucleus RNA sequencing (snRNA-Seq) datasets. The results revealed that the frequency of microglia was increased in both AD and DACD mouse models when compared with control mice. After analyzing the differentially expressed genes of microglia from the two mouse models, only six common upregulated genes were found. The CellChat analysis revealed the complex cell-cell communication network (microglia clusters with other cell types) in 5XFAD vs. control mice and db/db vs. control mice. The microglia subtypes and their transcription factor activity profile in 5XFAD mice were different from that in db/db mice. In summary, this study provided some insights into the alterations of microglia in 5XFAD and db/db mice, which might open up potential avenues for the microglial-targeted therapy in AD and DACD.
Collapse
Affiliation(s)
- Yaqiong Liu
- Regenerative Medicine Institute (REMEDI), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Tao Li
- Regenerative Medicine Institute (REMEDI), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Juliang Xiong
- Department of Pharmacy, the Second Affiliated Hospital of Shaoyang University, Shaoyang, Hunan, China.
| |
Collapse
|
10
|
Sharma T, Mehan S, Tiwari A, Khan Z, Gupta GD, Narula AS. Targeting Oligodendrocyte Dynamics and Remyelination: Emerging Therapies and Personalized Approaches in Multiple Sclerosis Management. Curr Neurovasc Res 2025; 21:359-417. [PMID: 39219420 DOI: 10.2174/0115672026336440240822063430] [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: 06/27/2024] [Revised: 01/01/1970] [Accepted: 07/12/2024] [Indexed: 09/04/2024]
Abstract
Multiple sclerosis (MS) is a progressive autoimmune condition that primarily affects young people and is characterized by demyelination and neurodegeneration of the central nervous system (CNS). This in-depth review explores the complex involvement of oligodendrocytes, the primary myelin- producing cells in the CNS, in the pathophysiology of MS. It discusses the biochemical processes and signalling pathways required for oligodendrocytes to function and remain alive, as well as how they might fail and cause demyelination to occur. We investigate developing therapeutic options that target remyelination, a fundamental component of MS treatment. Remyelination approaches promote the survival and differentiation of oligodendrocyte precursor cells (OPCs), restoring myelin sheaths. This improves nerve fibre function and may prevent MS from worsening. We examine crucial parameters influencing remyelination success, such as OPC density, ageing, and signalling pathway regulation (e.g., Retinoid X receptor, LINGO-1, Notch). The review also examines existing neuroprotective and antiinflammatory medications being studied to see if they can assist oligodendrocytes in surviving and reducing the severity of MS symptoms. The review focuses on medicines that target the myelin metabolism in oligodendrocytes. Altering oligodendrocyte metabolism has been linked to reversing demyelination and improving MS patient outcomes through various mechanisms. We also explore potential breakthroughs, including innovative antisense technologies, deep brain stimulation, and the impact of gut health and exercise on MS development. The article discusses the possibility of personalized medicine in MS therapy, emphasizing the importance of specific medicines based on individual molecular profiles. The study emphasizes the need for reliable biomarkers and improved imaging tools for monitoring disease progression and therapy response. Finally, this review focuses on the importance of oligodendrocytes in MS and the potential for remyelination therapy. It also underlines the importance of continued research to develop more effective treatment regimens, taking into account the complexities of MS pathology and the different factors that influence disease progression and treatment.
Collapse
Affiliation(s)
- Tarun Sharma
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Aarti Tiwari
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | | | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| |
Collapse
|
11
|
Stoller F, Hinds E, Ionescu T, Khatamsaz E, Marston HM, Hengerer B. Forceps minor control of social behaviour. Sci Rep 2024; 14:30492. [PMID: 39681620 DOI: 10.1038/s41598-024-81930-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 11/30/2024] [Indexed: 12/18/2024] Open
Abstract
The PRISM project, funded by the EU's Innovative Medicines Initiative, has identified a transdiagnostic, pathophysiological relationship between the integrity of the default mode network (DMN) and social dysfunction. To explore the causal link between DMN integrity and social behaviour, we employed a preclinical back-translation approach, using focal demyelination of the forceps minor to disrupt DMN connectivity in mice. By applying advanced techniques such as functional ultrasound imaging and automated analysis of social behaviour, we demonstrated that reduced DMN connectivity leads to impaired social interactions and increased anxiety in mice. Notably, these effects were reversible, indicating that the forceps minor, a critical fibre tract connecting key DMN regions in the prefrontal cortex, plays a crucial role in social function. This study provides direct evidence for a causal relationship between DMN integrity and social dysfunction, with potential implications for developing targeted treatments in precision psychiatry.
Collapse
Affiliation(s)
| | - Eleanor Hinds
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Tudor Ionescu
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | - Hugh M Marston
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | |
Collapse
|
12
|
Langlois J, Lange S, Ebeling M, Macnair W, Schmucki R, Li C, DeGeer J, Sudharshan TJJ, Yong VW, Shen YA, Harp C, Collin L, Keaney J. Fenebrutinib, a Bruton's tyrosine kinase inhibitor, blocks distinct human microglial signaling pathways. J Neuroinflammation 2024; 21:276. [PMID: 39465429 PMCID: PMC11514909 DOI: 10.1186/s12974-024-03267-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Bruton's tyrosine kinase (BTK) is an intracellular signaling enzyme that regulates B-lymphocyte and myeloid cell functions. Due to its involvement in both innate and adaptive immune compartments, BTK inhibitors have emerged as a therapeutic option in autoimmune disorders such as multiple sclerosis (MS). Brain-penetrant, small-molecule BTK inhibitors may also address compartmentalized neuroinflammation, which is proposed to underlie MS disease progression. BTK is expressed by microglia, which are the resident innate immune cells of the brain; however, the precise roles of microglial BTK and impact of BTK inhibitors on microglial functions are still being elucidated. Research on the effects of BTK inhibitors has been limited to rodent disease models. This is the first study reporting effects in human microglia. METHODS Here we characterize the pharmacological and functional properties of fenebrutinib, a potent, highly selective, noncovalent, reversible, brain-penetrant BTK inhibitor, in human microglia and complex human brain cell systems, including brain organoids. RESULTS We find that fenebrutinib blocks the deleterious effects of microglial Fc gamma receptor (FcγR) activation, including cytokine and chemokine release, microglial clustering and neurite damage in diverse human brain cell systems. Gene expression analyses identified pathways linked to inflammation, matrix metalloproteinase production and cholesterol metabolism that were modulated by fenebrutinib treatment. In contrast, fenebrutinib had no significant impact on human microglial pathways linked to Toll-like receptor 4 (TLR4) and NACHT, LRR and PYD domains-containing protein 3 (NLRP3) signaling or myelin phagocytosis. CONCLUSIONS Our study enhances the understanding of BTK functions in human microglial signaling that are relevant to MS pathogenesis and suggests that fenebrutinib could attenuate detrimental microglial activity associated with FcγR activation in people with MS.
Collapse
Affiliation(s)
- Julie Langlois
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Simona Lange
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Martin Ebeling
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Will Macnair
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Roland Schmucki
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Cenxiao Li
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, Canada
| | - Jonathan DeGeer
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Tania J J Sudharshan
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - V Wee Yong
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, Canada
| | - Yun-An Shen
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA
| | | | - Ludovic Collin
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - James Keaney
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland.
| |
Collapse
|
13
|
Hoffmann A, Miron VE. CNS macrophage contributions to myelin health. Immunol Rev 2024; 327:53-70. [PMID: 39484853 DOI: 10.1111/imr.13416] [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] [Indexed: 11/03/2024]
Abstract
Myelin is the membrane surrounding neuronal axons in the central nervous system (CNS), produced by oligodendrocytes to provide insulation for electrical impulse conduction and trophic/metabolic support. CNS dysfunction occurs following poor development of myelin in infancy, myelin damage in neurological diseases, and impaired regeneration of myelin with disease progression in aging. The lack of approved therapies aimed at supporting myelin health highlights the critical need to identify the cellular and molecular influences on oligodendrocytes. CNS macrophages have been shown to influence the development, maintenance, damage and regeneration of myelin, revealing critical interactions with oligodendrocyte lineage cells. CNS macrophages are comprised of distinct populations, including CNS-resident microglia and cells associated with CNS border regions (the meninges, vasculature, and choroid plexus), in addition to macrophages derived from monocytes infiltrating from the blood. Importantly, the distinct contribution of these macrophage populations to oligodendrocyte lineage responses and myelin health are only just beginning to be uncovered, with the advent of new tools to specifically identify, track, and target macrophage subsets. Here, we summarize the current state of knowledge on the roles of CNS macrophages in myelin health, and recent developments in distinguishing the roles of macrophage populations in development, homeostasis, and disease.
Collapse
Affiliation(s)
- Alana Hoffmann
- BARLO Multiple Sclerosis Centre and Keenan Research Centre for Biomedical Science at St. Michael's Hospital, Toronto, Ontario, Canada
- Department of Immunology, The University of Toronto, Toronto, Ontario, Canada
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Veronique E Miron
- BARLO Multiple Sclerosis Centre and Keenan Research Centre for Biomedical Science at St. Michael's Hospital, Toronto, Ontario, Canada
- Department of Immunology, The University of Toronto, Toronto, Ontario, Canada
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
14
|
Groh AMR, Caporicci-Dinucci N, Afanasiev E, Bigotte M, Lu B, Gertsvolf J, Smith MD, Garton T, Callahan-Martin L, Allot A, Hatrock DJ, Mamane V, Drake S, Tai H, Ding J, Fournier AE, Larochelle C, Calabresi PA, Stratton JA. Ependymal cells undergo astrocyte-like reactivity in response to neuroinflammation. J Neurochem 2024; 168:3449-3466. [PMID: 38702968 DOI: 10.1111/jnc.16120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 04/08/2024] [Accepted: 04/15/2024] [Indexed: 05/06/2024]
Abstract
Ependymal cells form a specialized brain-cerebrospinal fluid (CSF) interface and regulate local CSF microcirculation. It is becoming increasingly recognized that ependymal cells assume a reactive state in response to aging and disease, including conditions involving hypoxia, hydrocephalus, neurodegeneration, and neuroinflammation. Yet what transcriptional signatures govern these reactive states and whether this reactivity shares any similarities with classical descriptions of glial reactivity (i.e., in astrocytes) remain largely unexplored. Using single-cell transcriptomics, we interrogated this phenomenon by directly comparing the reactive ependymal cell transcriptome to the reactive astrocyte transcriptome using a well-established model of autoimmune-mediated neuroinflammation (MOG35-55 EAE). In doing so, we unveiled core glial reactivity-associated genes that defined the reactive ependymal cell and astrocyte response to MOG35-55 EAE. Interestingly, known reactive astrocyte genes from other CNS injury/disease contexts were also up-regulated by MOG35-55 EAE ependymal cells, suggesting that this state may be conserved in response to a variety of pathologies. We were also able to recapitulate features of the reactive ependymal cell state acutely using a classic neuroinflammatory cocktail (IFNγ/LPS) both in vitro and in vivo. Taken together, by comparing reactive ependymal cells and astrocytes, we identified a conserved signature underlying glial reactivity that was present in several neuroinflammatory contexts. Future work will explore the mechanisms driving ependymal reactivity and assess downstream functional consequences.
Collapse
Affiliation(s)
- Adam M R Groh
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, Quebec, Canada
| | - Nina Caporicci-Dinucci
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, Quebec, Canada
| | - Elia Afanasiev
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, Quebec, Canada
| | - Maxime Bigotte
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, Quebec, Canada
| | - Brianna Lu
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, Quebec, Canada
| | - Joshua Gertsvolf
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, Quebec, Canada
| | - Matthew D Smith
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Thomas Garton
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Liam Callahan-Martin
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, Quebec, Canada
| | - Alexis Allot
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, Quebec, Canada
| | - Dale J Hatrock
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, Quebec, Canada
| | - Victoria Mamane
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montréal, Quebec, Canada
| | - Sienna Drake
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, Quebec, Canada
| | - Huilin Tai
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montréal, Quebec, Canada
- Department of Medicine, McGill University Health Centre, Montréal, Quebec, Canada
| | - Jun Ding
- Meakins-Christie Laboratories, Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montréal, Quebec, Canada
- Department of Medicine, McGill University Health Centre, Montréal, Quebec, Canada
| | - Alyson E Fournier
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, Quebec, Canada
| | - Catherine Larochelle
- Department of Neuroscience, Faculty of Medicine, Université de Montréal, Montréal, Quebec, Canada
| | - Peter A Calabresi
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins Hospital, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jo Anne Stratton
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, Quebec, Canada
| |
Collapse
|
15
|
Wuerch EC, Mirzaei R, Yong VW. Niacin produces an inconsistent treatment response in the EAE model of multiple sclerosis. J Neuroimmunol 2024; 394:578421. [PMID: 39088907 DOI: 10.1016/j.jneuroim.2024.578421] [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: 04/30/2024] [Revised: 06/21/2024] [Accepted: 07/25/2024] [Indexed: 08/03/2024]
Abstract
Niacin was found in the lysolecithin model of multiple sclerosis (MS) to promote the phagocytic clearance of debris and enhance remyelination. Lysolecithin lesions have prominent microglia/macrophages but lack lymphocytes that populate plaques of MS or its experimental autoimmune encephalomyelitis (EAE) model. Thus, the current study assessed the efficacy of niacin in EAE. We found that niacin inconsistently affects EAE clinical score, and largely does not ameliorate neuropathology. In culture, niacin enhances phagocytosis by macrophages, but does not reduce T cell proliferation. We suggest that studies of niacin for potential remyelination in MS should include a therapeutic that targets adaptive immunity.
Collapse
Affiliation(s)
- Emily C Wuerch
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Reza Mirzaei
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - V Wee Yong
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
16
|
Tiwari V, Prajapati B, Asare Y, Damkou A, Ji H, Liu L, Naser N, Gouna G, Leszczyńska KB, Mieczkowski J, Dichgans M, Wang Q, Kawaguchi R, Shi Z, Swarup V, Geschwind DH, Prinz M, Gokce O, Simons M. Innate immune training restores pro-reparative myeloid functions to promote remyelination in the aged central nervous system. Immunity 2024; 57:2173-2190.e8. [PMID: 39053462 DOI: 10.1016/j.immuni.2024.07.001] [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: 04/26/2023] [Revised: 11/21/2023] [Accepted: 07/01/2024] [Indexed: 07/27/2024]
Abstract
The reduced ability of the central nervous system to regenerate with increasing age limits functional recovery following demyelinating injury. Previous work has shown that myelin debris can overwhelm the metabolic capacity of microglia, thereby impeding tissue regeneration in aging, but the underlying mechanisms are unknown. In a model of demyelination, we found that a substantial number of genes that were not effectively activated in aged myeloid cells displayed epigenetic modifications associated with restricted chromatin accessibility. Ablation of two class I histone deacetylases in microglia was sufficient to restore the capacity of aged mice to remyelinate lesioned tissue. We used Bacillus Calmette-Guerin (BCG), a live-attenuated vaccine, to train the innate immune system and detected epigenetic reprogramming of brain-resident myeloid cells and functional restoration of myelin debris clearance and lesion recovery. Our results provide insight into aging-associated decline in myeloid function and how this decay can be prevented by innate immune reprogramming.
Collapse
Affiliation(s)
- Vini Tiwari
- Institute of Neuronal Cell Biology, Technical University Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Bharat Prajapati
- Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 41390 Gothenburg, Sweden
| | - Yaw Asare
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Alkmini Damkou
- Institute of Neuronal Cell Biology, Technical University Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Hao Ji
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Lu Liu
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Nawraa Naser
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Garyfallia Gouna
- Institute of Neuronal Cell Biology, Technical University Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Katarzyna B Leszczyńska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 02093 Warsaw, Poland
| | - Jakub Mieczkowski
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 02093 Warsaw, Poland; 3P-Medicine Laboratory, Medical University of Gdańsk, 80211 Gdańsk, Poland
| | - Martin Dichgans
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), 81377 Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Qing Wang
- Departments of Neurology and Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Riki Kawaguchi
- Departments of Neurology and Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Psychiatry, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zechuan Shi
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Daniel H Geschwind
- Departments of Neurology and Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79085 Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Ozgun Gokce
- Munich Cluster of Systems Neurology (SyNergy), 81377 Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany; Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, 53127 Bonn, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), 81377 Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany.
| |
Collapse
|
17
|
Lozinski BM, Ta K, Dong Y. Emerging role of galectin 3 in neuroinflammation and neurodegeneration. Neural Regen Res 2024; 19:2004-2009. [PMID: 38227529 DOI: 10.4103/1673-5374.391181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 11/15/2023] [Indexed: 01/17/2024] Open
Abstract
Neuroinflammation and neurodegeneration are key processes that mediate the development and progression of neurological diseases. However, the mechanisms modulating these processes in different diseases remain incompletely understood. Advances in single cell based multi-omic analyses have helped to identify distinct molecular signatures such as Lgals3 that is associated with neuroinflammation and neurodegeneration in the central nervous system (CNS). Lgals3 encodes galectin-3 (Gal3), a β-galactoside and glycan binding glycoprotein that is frequently upregulated by reactive microglia/macrophages in the CNS during various neurological diseases. While Gal3 has previously been associated with non-CNS inflammatory and fibrotic diseases, recent studies highlight Gal3 as a prominent regulator of inflammation and neuroaxonal damage in the CNS during diseases such as multiple sclerosis, Alzheimer's disease, and Parkinson's disease. In this review, we summarize the pleiotropic functions of Gal3 and discuss evidence that demonstrates its detrimental role in neuroinflammation and neurodegeneration during different neurological diseases. We also consider the challenges of translating preclinical observations into targeting Gal3 in the human CNS.
Collapse
Affiliation(s)
- Brian M Lozinski
- Department of Clinical Neuroscience, University of Calgary, Calgary, AB, Canada
| | - Khanh Ta
- Deparment of Biochemistry, Microbiology & Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Yifei Dong
- Deparment of Biochemistry, Microbiology & Immunology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
18
|
Olveda GE, Barasa MN, Hill RA. Microglial phagocytosis of single dying oligodendrocytes is mediated by CX3CR1 but not MERTK. Cell Rep 2024; 43:114385. [PMID: 38935500 PMCID: PMC11304498 DOI: 10.1016/j.celrep.2024.114385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/10/2024] [Accepted: 06/05/2024] [Indexed: 06/29/2024] Open
Abstract
Oligodendrocyte death is common in aging and neurodegenerative disease. In these conditions, dying oligodendrocytes must be efficiently removed to allow remyelination and to prevent a feedforward degenerative cascade. Removal of this cellular debris is thought to primarily be carried out by resident microglia. To investigate the cellular dynamics underlying how microglia do this, we use a single-cell cortical demyelination model combined with longitudinal intravital imaging of dual-labeled transgenic mice. Following phagocytosis, single microglia clear the targeted oligodendrocyte and its myelin sheaths in one day via a precise, rapid, and stereotyped sequence. Deletion of the fractalkine receptor, CX3CR1, delays the microglial phagocytosis of the cell soma but has no effect on clearance of myelin sheaths. Unexpectedly, deletion of the phosphatidylserine receptor, MERTK, has no effect on oligodendrocyte or myelin sheath clearance. Thus, separate molecular signals are used to detect, engage, and clear distinct sub-compartments of dying oligodendrocytes to maintain tissue homeostasis.
Collapse
Affiliation(s)
- Genaro E Olveda
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - Maryanne N Barasa
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - Robert A Hill
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA.
| |
Collapse
|
19
|
Shui X, Chen J, Fu Z, Zhu H, Tao H, Li Z. Microglia in Ischemic Stroke: Pathogenesis Insights and Therapeutic Challenges. J Inflamm Res 2024; 17:3335-3352. [PMID: 38800598 PMCID: PMC11128258 DOI: 10.2147/jir.s461795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/14/2024] [Indexed: 05/29/2024] Open
Abstract
Ischemic stroke is the most common type of stroke, which is the main cause of death and disability on a global scale. As the primary immune cells in the brain that are crucial for preserving homeostasis of the central nervous system microenvironment, microglia have been found to exhibit dual or even multiple effects at different stages of ischemic stroke. The anti-inflammatory polarization of microglia and release of neurotrophic factors may provide benefits by promoting neurological recovery at the lesion in the early phase after ischemic stroke. However, the pro-inflammatory polarization of microglia and secretion of inflammatory factors in the later phase of injury may exacerbate the ischemic lesion, suggesting the therapeutic potential of modulating the balance of microglial polarization to predispose them to anti-inflammatory transformation in ischemic stroke. Microglia-mediated signaling crosstalk with other cells may also be key to improving functional outcomes following ischemic stroke. Thus, this review provides an overview of microglial functions and responses under physiological and ischemic stroke conditions, including microglial activation, polarization, and interactions with other cells. We focus on approaches that promote anti-inflammatory polarization of microglia, inhibit microglial activation, and enhance beneficial cell-to-cell interactions. These targets may hold promise for the creation of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Xinyao Shui
- Clinical Medical College, Southwest Medical University, Luzhou, People’s Republic of China
| | - Jingsong Chen
- Department of Laboratory Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, People’s Republic of China
- Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| | - Ziyue Fu
- Clinical Medical College, Southwest Medical University, Luzhou, People’s Republic of China
| | - Haoyue Zhu
- Clinical Medical College, Southwest Medical University, Luzhou, People’s Republic of China
| | - Hualin Tao
- Department of Laboratory Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, People’s Republic of China
- Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| | - Zhaoyinqian Li
- Department of Laboratory Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, People’s Republic of China
- Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| |
Collapse
|
20
|
You Y, Chen Z, Hu WW. The role of microglia heterogeneity in synaptic plasticity and brain disorders: Will sequencing shed light on the discovery of new therapeutic targets? Pharmacol Ther 2024; 255:108606. [PMID: 38346477 DOI: 10.1016/j.pharmthera.2024.108606] [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: 10/31/2023] [Revised: 01/05/2024] [Accepted: 02/02/2024] [Indexed: 02/18/2024]
Abstract
Microglia play a crucial role in interacting with neuronal synapses and modulating synaptic plasticity. This function is particularly significant during postnatal development, as microglia are responsible for removing excessive synapses to prevent neurodevelopmental deficits. Dysregulation of microglial synaptic function has been well-documented in various pathological conditions, notably Alzheimer's disease and multiple sclerosis. The recent application of RNA sequencing has provided a powerful and unbiased means to decipher spatial and temporal microglial heterogeneity. By identifying microglia with varying gene expression profiles, researchers have defined multiple subgroups of microglia associated with specific pathological states, including disease-associated microglia, interferon-responsive microglia, proliferating microglia, and inflamed microglia in multiple sclerosis, among others. However, the functional roles of these distinct subgroups remain inadequately characterized. This review aims to refine our current understanding of the potential roles of heterogeneous microglia in regulating synaptic plasticity and their implications for various brain disorders, drawing from recent sequencing research and functional studies. This knowledge may aid in the identification of pathogenetic biomarkers and potential factors contributing to pathogenesis, shedding new light on the discovery of novel drug targets. The field of sequencing-based data mining is evolving toward a multi-omics approach. With advances in viral tools for precise microglial regulation and the development of brain organoid models, we are poised to elucidate the functional roles of microglial subgroups detected through sequencing analysis, ultimately identifying valuable therapeutic targets.
Collapse
Affiliation(s)
- Yi You
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhong Chen
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Wei-Wei Hu
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
21
|
Lombardi M, Scaroni F, Gabrielli M, Raffaele S, Bonfanti E, Filipello F, Giussani P, Picciolini S, de Rosbo NK, Uccelli A, Golia MT, D’Arrigo G, Rubino T, Hooshmand K, Legido-Quigley C, Fenoglio C, Gualerzi A, Fumagalli M, Verderio C. Extracellular vesicles released by microglia and macrophages carry endocannabinoids which foster oligodendrocyte differentiation. Front Immunol 2024; 15:1331210. [PMID: 38464529 PMCID: PMC10921360 DOI: 10.3389/fimmu.2024.1331210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/01/2024] [Indexed: 03/12/2024] Open
Abstract
Introduction Microglia and macrophages can influence the evolution of myelin lesions through the production of extracellular vesicles (EVs). While microglial EVs promote in vitro differentiation of oligodendrocyte precursor cells (OPCs), whether EVs derived from macrophages aid or limit OPC maturation is unknown. Methods Immunofluorescence analysis for the myelin protein MBP was employed to evaluate the impact of EVs from primary rat macrophages on cultured OPC differentiation. Raman spectroscopy and liquid chromatography-mass spectrometry was used to define the promyelinating lipid components of myelin EVs obtained in vitro and isolated from human plasma. Results and discussion Here we show that macrophage-derived EVs do not promote OPC differentiation, and those released from macrophages polarized towards an inflammatory state inhibit OPC maturation. However, their lipid cargo promotes OPC maturation in a similar manner to microglial EVs. We identify the promyelinating endocannabinoids anandamide and 2-arachidonoylglycerol in EVs released by both macrophages and microglia in vitro and circulating in human plasma. Analysis of OPC differentiation in the presence of the endocannabinoid receptor antagonists SR141716A and AM630 reveals a key role of vesicular endocannabinoids in OPC maturation. From this study, EV-associated endocannabinoids emerge as important mediators in microglia/macrophage-oligodendrocyte crosstalk, which may be exploited to enhance myelin repair.
Collapse
Affiliation(s)
- Marta Lombardi
- Department of Biomedical Sciences, National Research Council (CNR) Institute of Neuroscience, Vedano al Lambro, Italy
- NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| | - Federica Scaroni
- Department of Biomedical Sciences, National Research Council (CNR) Institute of Neuroscience, Vedano al Lambro, Italy
| | - Martina Gabrielli
- Department of Biomedical Sciences, National Research Council (CNR) Institute of Neuroscience, Vedano al Lambro, Italy
- NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| | - Stefano Raffaele
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Milan, Italy
| | - Elisabetta Bonfanti
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Milan, Italy
| | - Fabia Filipello
- Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Humanitas Research Hospital, Rozzano, Italy
| | - Paola Giussani
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Segrate, Italy
| | - Silvia Picciolini
- Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Fondazione Don Carlo Gnocchi Onlus, Milan, Italy
| | - Nicole Kerlero de Rosbo
- Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Ospedale Policlinico San Martino, Genoa, Italy
- TomaLab, Institute of Nanotechnology, CNR, Rome, Italy
| | - Antonio Uccelli
- Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Ospedale Policlinico San Martino, Genoa, Italy
- Department of Neurology, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Maria Teresa Golia
- Department of Biomedical Sciences, National Research Council (CNR) Institute of Neuroscience, Vedano al Lambro, Italy
- NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| | - Giulia D’Arrigo
- Department of Biomedical Sciences, National Research Council (CNR) Institute of Neuroscience, Vedano al Lambro, Italy
- NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| | - Tiziana Rubino
- Department of Biotechnology and Life Sciences (DBSV) and Neuroscience Center, University of Insubria, Busto Arsizio, Italy
| | - Kourosh Hooshmand
- System Medicine, Steno Diabetes Center Copenhagen, Copenhagen, Denmark
| | - Cristina Legido-Quigley
- System Medicine, Steno Diabetes Center Copenhagen, Copenhagen, Denmark
- Institute of Pharmaceutical Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| | - Chiara Fenoglio
- Department of Biomedical, Surgical and Dental Sciences, Università degli Studi di Milano, Milan, Italy
- Fondazione Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Ca’ Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Alice Gualerzi
- Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Fondazione Don Carlo Gnocchi Onlus, Milan, Italy
| | - Marta Fumagalli
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”, Università degli Studi di Milano, Milan, Italy
| | - Claudia Verderio
- Department of Biomedical Sciences, National Research Council (CNR) Institute of Neuroscience, Vedano al Lambro, Italy
- NeuroMI Milan Center for Neuroscience, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
22
|
Lan Y, Zhang X, Liu S, Guo C, Jin Y, Li H, Wang L, Zhao J, Hao Y, Li Z, Liu Z, Ginhoux F, Xie Q, Xu H, Jia JM, He D. Fate mapping of Spp1 expression reveals age-dependent plasticity of disease-associated microglia-like cells after brain injury. Immunity 2024; 57:349-363.e9. [PMID: 38309272 DOI: 10.1016/j.immuni.2024.01.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 10/22/2023] [Accepted: 01/09/2024] [Indexed: 02/05/2024]
Abstract
Microglial reactivity to injury and disease is emerging as a heterogeneous, dynamic, and crucial determinant in neurological disorders. However, the plasticity and fate of disease-associated microglia (DAM) remain largely unknown. We established a lineage tracing system, leveraging the expression dynamics of secreted phosphoprotein 1(Spp1) to label and track DAM-like microglia during brain injury and recovery. Fate mapping of Spp1+ microglia during stroke in juvenile mice revealed an irreversible state of DAM-like microglia that were ultimately eliminated from the injured brain. By contrast, DAM-like microglia in the neonatal stroke models exhibited high plasticity, regaining a homeostatic signature and integrating into the microglial network after recovery. Furthermore, neonatal injury had a lasting impact on microglia, rendering them intrinsically sensitized to subsequent immune challenges. Therefore, our findings highlight the plasticity and innate immune memory of neonatal microglia, shedding light on the fate of DAM-like microglia in various neuropathological conditions.
Collapse
Affiliation(s)
- Yangning Lan
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Laboratory of Neuroimmunology, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Xiaoxuan Zhang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Laboratory of Neurovascular Biology, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Shaorui Liu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Laboratory of Neuroimmunology, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Laboratory of Systems Immunology, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Chen Guo
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Cancer Stem Cell and Tumor Microenvironment lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Yuxiao Jin
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Laboratory of Neurovascular Biology, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Hui Li
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Laboratory of Neuroimmunology, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Laboratory of Systems Immunology, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Linyixiao Wang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Laboratory of Neuroimmunology, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Jinghong Zhao
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Laboratory of Neuroimmunology, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Yilin Hao
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Laboratory of Neuroimmunology, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Zhicheng Li
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Laboratory of Systems Immunology, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Zhaoyuan Liu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Florent Ginhoux
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Singapore Immunology Network, Agency for Science, Technology and Research, Singapore 138648, Singapore; Gustave Roussy Cancer Campus, Villejuif 94800, France; Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale contre le Cancer, Villejuif, France; Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Qi Xie
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Cancer Stem Cell and Tumor Microenvironment lab, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Heping Xu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Laboratory of Systems Immunology, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Jie-Min Jia
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Laboratory of Neurovascular Biology, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
| | - Danyang He
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Laboratory of Neuroimmunology, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
| |
Collapse
|
23
|
Xu L, Min H, Saha A, Gunaratne A, Schwartzman J, Parrott R, Kurtzberg J, Filiano AJ. Mesenchymal stromal cells suppress microglial activation and tumor necrosis factor production. Cytotherapy 2024; 26:185-193. [PMID: 38054911 DOI: 10.1016/j.jcyt.2023.11.006] [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: 07/14/2023] [Revised: 11/03/2023] [Accepted: 11/16/2023] [Indexed: 12/07/2023]
Abstract
BACKGROUND AIMS White matter diseases are commonly associated with microglial activation and neuroinflammation. Mesenchymal stromal cells (MSCs) have immunomodulatory properties and thus have the potential to be developed as cell therapy for white matter disease. MSCs interact with resident macrophages to alter the trajectory of inflammation; however, the impact MSCs have on central nervous system macrophages and the effect this has on the progression of white matter disease are unclear. METHODS In this study, we utilized numerous assays of varying complexity to model different aspects of white matter disease. These assays ranged from an in vivo spinal cord acute demyelination model to a simple microglial cell line activation assay. Our goal was to investigate the influence of human umbilical cord tissue MSCs on the activation of microglia. RESULTS MSCs reduced the production of tumor necrosis factor (TNF) by microglia and decreased demyelinated lesions in the spinal cord after acute focal injury. To determine if MSCs could directly suppress the activation of microglia and to develop an efficient potency assay, we utilized isolated primary microglia from mouse brains and the Immortalized MicroGlial Cell Line (IMG). MSCs suppressed the activation of microglia and the release of TNF after stimulation with lipopolysaccharide, a toll-like receptor agonist. CONCLUSIONS In this study, we demonstrated that MSCs altered the immune response after acute injury in the spinal cord. In numerous assays, MSCs suppressed activation of microglia and release of the pro-inflammatory cytokine TNF. Of these assays, IMG could be standardized and used as an effective potency assay to determine the efficacy of MSCs for treating white matter disease or other neuroinflammatory conditions associated with microglial activation.
Collapse
Affiliation(s)
- Li Xu
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA
| | - Hyunjung Min
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA
| | - Arjun Saha
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA
| | - Aruni Gunaratne
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA; Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | | | - Roberta Parrott
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA
| | - Joanne Kurtzberg
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA; Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | - Anthony J Filiano
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA; Department of Neurosurgery, Duke University, Durham, North Carolina, USA; Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA; Department of Pathology, Duke University, Durham, North Carolina, USA.
| |
Collapse
|
24
|
孙 晓, 史 航, 张 磊, 刘 中, 李 克, 钱 玲, 朱 星, 杨 康, 付 强, 丁 华. [Exosomes from ectoderm mesenchymal stem cells inhibits lipopolysaccharide-induced microglial M1 polarization and promotes survival of H 2O 2-exposed PC12 cells by suppressing inflammatory response and oxidative stress]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:119-128. [PMID: 38293983 PMCID: PMC10878899 DOI: 10.12122/j.issn.1673-4254.2024.01.14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Indexed: 02/01/2024]
Abstract
OBJECTIVE To investigate the potential value of exosomes derived from rat ectoderm mesenchymal stem cells (EMSCs-exo) for repairing secondary spinal cord injury. METHODS EMSCs-exo were obtained using ultracentrifugation from EMSCs isolated from rat nasal mucosa, identified by transmission electron microscope, nanoparticle tracking analysis (NTA), and Western blotting, and quantified using the BCA method. Neonatal rat microglia purified by differential attachment were induced with 100 μg/L lipopolysaccharide (LPS) and treated with 37.5 or 75 mg/L EMSCs-exo. PC12 cells were exposed to 400 μmol/L H2O2 and treated with EMSCs-exo at 37.5 or 75 mg/L. The protein and mRNA expressions of Arg1 and iNOS in the treated cells were determined with Western blotting and qRT- PCR, and the concentrations of IL- 6, IL-10, and IGF-1 in the supernatants were measured with ELISA. The viability and apoptosis of PC12 cells were detected using CCK-8 assay and flow cytometry. RESULTS The isolated rat EMSCs showed high expressions of nestin, CD44, CD105, and vimentin. The obtained EMSCs-exo had a typical cup-shaped structure under transmission electron microscope with an average particle size of 142 nm and positivity for CD63, CD81, and TSG101 but not vimentin. In LPS-treated microglia, EMSCs-exo treatment at 75 mg/L significantly increased Arg1 protein level and lowered iNOS protein expression (P < 0.05). EMSCs-exo treatment at 75 mg/L, as compared with the lower concentration at 37.5 mg/L, more strongly increased Arg1 mRNA expression and IGF-1 and IL-10 production and decreased iNOS mRNA expression and IL-6 production in LPS-induced microglia, and more effectively promoted cell survival and decreased apoptosis rate of H2O2-induced PC12 cells (P < 0.05). CONCLUSION EMSCs-exo at 75 mg/L can effectively reduce the proportion of M1 microglia and alleviate neuronal apoptosis under oxidative stress to promote neuronal survival, suggesting its potential in controlling secondary spinal cord injury.
Collapse
Affiliation(s)
- 晓鹏 孙
- 江苏大学附属人民医院骨科,江苏 镇江 212000Department of Orthopedics, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212000, China
| | - 航 史
- 江苏大学附属人民医院骨科,江苏 镇江 212000Department of Orthopedics, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212000, China
| | - 磊 张
- 江苏大学附属人民医院骨科,江苏 镇江 212000Department of Orthopedics, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212000, China
| | - 中 刘
- 上海交通大学医学院附属第一人民医院骨科,上海 200080Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - 克威 李
- 上海交通大学医学院附属第一人民医院骨科,上海 200080Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - 玲玲 钱
- 江苏大学医学院,江苏 镇江 212013School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - 星宇 朱
- 江苏大学附属人民医院骨科,江苏 镇江 212000Department of Orthopedics, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212000, China
| | - 康佳 杨
- 江苏大学附属人民医院骨科,江苏 镇江 212000Department of Orthopedics, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212000, China
| | - 强 付
- 上海交通大学医学院附属第一人民医院骨科,上海 200080Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - 华 丁
- 江苏大学附属人民医院骨科,江苏 镇江 212000Department of Orthopedics, Affiliated People's Hospital of Jiangsu University, Zhenjiang 212000, China
| |
Collapse
|
25
|
Hu J, Melchor GS, Ladakis D, Reger J, Kim HW, Chamberlain KA, Shults NV, Oft HC, Smith VN, Rosko LM, Li E, Baydyuk M, Fu MM, Bhargava P, Huang JK. Myeloid cell-associated aromatic amino acid metabolism facilitates CNS myelin regeneration. NPJ Regen Med 2024; 9:1. [PMID: 38167866 PMCID: PMC10762216 DOI: 10.1038/s41536-023-00345-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024] Open
Abstract
Regulation of myeloid cell activity is critical for successful myelin regeneration (remyelination) in demyelinating diseases, such as multiple sclerosis (MS). Here, we show aromatic alpha-keto acids (AKAs) generated from the amino acid oxidase, interleukin-4 induced 1 (IL4I1), promote efficient remyelination in mouse models of MS. During remyelination, myeloid cells upregulated the expression of IL4I1. Conditionally knocking out IL4I1 in myeloid cells impaired remyelination efficiency. Mice lacking IL4I1 expression exhibited a reduction in the AKAs, phenylpyruvate, indole-3-pyruvate, and 4-hydroxyphenylpyruvate, in remyelinating lesions. Decreased AKA levels were also observed in people with MS, particularly in the progressive phase when remyelination is impaired. Oral administration of AKAs modulated myeloid cell-associated inflammation, promoted oligodendrocyte maturation, and enhanced remyelination in mice with focal demyelinated lesions. Transcriptomic analysis revealed AKA treatment induced a shift in metabolic pathways in myeloid cells and upregulated aryl hydrocarbon receptor activity in lesions. Our results suggest myeloid cell-associated aromatic amino acid metabolism via IL4I1 produces AKAs in demyelinated lesions to enable efficient remyelination. Increasing AKA levels or targeting related pathways may serve as a strategy to facilitate the regeneration of myelin in inflammatory demyelinating conditions.
Collapse
Affiliation(s)
- Jingwen Hu
- Department of Biology, Georgetown University, Washington, DC, 20007, USA
| | - George S Melchor
- Department of Biology, Georgetown University, Washington, DC, 20007, USA
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, 20007, USA
| | - Dimitrios Ladakis
- Division of Neuroimmunology and Neurological Infections, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Joan Reger
- Department of Biology, Georgetown University, Washington, DC, 20007, USA
- National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Hee Won Kim
- Department of Biology, Georgetown University, Washington, DC, 20007, USA
| | - Kelly A Chamberlain
- Department of Biology, Georgetown University, Washington, DC, 20007, USA
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, 20007, USA
| | - Nataliia V Shults
- Department of Biology, Georgetown University, Washington, DC, 20007, USA
| | - Helena C Oft
- Department of Biology, Georgetown University, Washington, DC, 20007, USA
| | - Victoria N Smith
- Department of Biology, Georgetown University, Washington, DC, 20007, USA
| | - Lauren M Rosko
- Department of Biology, Georgetown University, Washington, DC, 20007, USA
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, 20007, USA
| | - Erqiu Li
- Department of Biology, Georgetown University, Washington, DC, 20007, USA
| | - Maryna Baydyuk
- Department of Biology, Georgetown University, Washington, DC, 20007, USA
| | - Meng-Meng Fu
- National Institute of Neurological Disorders and Stroke (NINDS), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Pavan Bhargava
- Division of Neuroimmunology and Neurological Infections, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Jeffrey K Huang
- Department of Biology, Georgetown University, Washington, DC, 20007, USA.
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, 20007, USA.
| |
Collapse
|
26
|
Kent SA, Miron VE. Microglia regulation of central nervous system myelin health and regeneration. Nat Rev Immunol 2024; 24:49-63. [PMID: 37452201 DOI: 10.1038/s41577-023-00907-4] [Citation(s) in RCA: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2023] [Indexed: 07/18/2023]
Abstract
Microglia are resident macrophages of the central nervous system that have key functions in its development, homeostasis and response to damage and infection. Although microglia have been increasingly implicated in contributing to the pathology that underpins neurological dysfunction and disease, they also have crucial roles in neurological homeostasis and regeneration. This includes regulation of the maintenance and regeneration of myelin, the membrane that surrounds neuronal axons, which is required for axonal health and function. Myelin is damaged with normal ageing and in several neurodegenerative diseases, such as multiple sclerosis and Alzheimer disease. Given the lack of approved therapies targeting myelin maintenance or regeneration, it is imperative to understand the mechanisms by which microglia support and restore myelin health to identify potential therapeutic approaches. However, the mechanisms by which microglia regulate myelin loss or integrity are still being uncovered. In this Review, we discuss recent work that reveals the changes in white matter with ageing and neurodegenerative disease, how this relates to microglia dynamics during myelin damage and regeneration, and factors that influence the regenerative functions of microglia.
Collapse
Affiliation(s)
- Sarah A Kent
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Veronique E Miron
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK.
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK.
- Barlo Multiple Sclerosis Centre, St Michael's Hospital, Toronto, Ontario, Canada.
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, Toronto, Ontario, Canada.
- Department of Immunology, The University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
27
|
Hammond BP, Panda SP, Kaushik DK, Plemel JR. Microglia and Multiple Sclerosis. ADVANCES IN NEUROBIOLOGY 2024; 37:445-456. [PMID: 39207707 DOI: 10.1007/978-3-031-55529-9_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Multiple sclerosis (MS) is a devastating autoimmune disease that leads to profound disability. This disability arises from the stochastic, regional loss of myelin-the insulating sheath surrounding neurons-in the central nervous system (CNS). The demyelinated regions are dominated by the brain's resident macrophages: microglia. Microglia perform a variety of functions in MS and are thought to initiate and perpetuate demyelination through their interactions with peripheral immune cells that traffic into the brain. However, microglia are also likely essential for recruiting and promoting the differentiation of cells that can restore lost myelin in a process known as remyelination. Given these seemingly opposing functions, an overarching beneficial or detrimental role is yet to be ascribed to these immune cells. In this chapter, we will discuss microglia dynamics throughout the MS disease course and probe the apparent dichotomy of microglia as the drivers of both demyelination and remyelination.
Collapse
Affiliation(s)
- Brady P Hammond
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Sharmistha P Panda
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Deepak K Kaushik
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Jason R Plemel
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada.
- Division of Neurology, Department of Medicine, University of Alberta, Edmonton, AB, Canada.
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada.
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
28
|
Liu F, Cheng X, Zhao C, Zhang X, Liu C, Zhong S, Liu Z, Lin X, Qiu W, Zhang X. Single-Cell Mapping of Brain Myeloid Cell Subsets Reveals Key Transcriptomic Changes Favoring Neuroplasticity after Ischemic Stroke. Neurosci Bull 2024; 40:65-78. [PMID: 37755676 PMCID: PMC10774469 DOI: 10.1007/s12264-023-01109-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 05/27/2023] [Indexed: 09/28/2023] Open
Abstract
Interactions between brain-resident and peripheral infiltrated immune cells are thought to contribute to neuroplasticity after cerebral ischemia. However, conventional bulk sequencing makes it challenging to depict this complex immune network. Using single-cell RNA sequencing, we mapped compositional and transcriptional features of peri-infarct immune cells. Microglia were the predominant cell type in the peri-infarct region, displaying a more diverse activation pattern than the typical pro- and anti-inflammatory state, with axon tract-associated microglia (ATMs) being associated with neuronal regeneration. Trajectory inference suggested that infiltrated monocyte-derived macrophages (MDMs) exhibited a gradual fate trajectory transition to activated MDMs. Inter-cellular crosstalk between MDMs and microglia orchestrated anti-inflammatory and repair-promoting microglia phenotypes and promoted post-stroke neurogenesis, with SOX2 and related Akt/CREB signaling as the underlying mechanisms. This description of the brain's immune landscape and its relationship with neurogenesis provides new insight into promoting neural repair by regulating neuroinflammatory responses.
Collapse
Affiliation(s)
- Fangxi Liu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Xi Cheng
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Chuansheng Zhao
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
- Stroke Center, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Xiaoqian Zhang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Chang Liu
- Stroke Center, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Shanshan Zhong
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Zhouyang Liu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Xinyu Lin
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| | - Xiuchun Zhang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China.
| |
Collapse
|
29
|
Li L, Li T, Qu X, Sun G, Fu Q, Han G. Stress/cell death pathways, neuroinflammation, and neuropathic pain. Immunol Rev 2024; 321:33-51. [PMID: 37688390 DOI: 10.1111/imr.13275] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/14/2023] [Accepted: 08/27/2023] [Indexed: 09/10/2023]
Abstract
Neuropathic pain is a common and debilitating modality of chronic pain induced by a lesion or disease of the somatosensory nervous system. Albeit the elucidation of numerous pathophysiological mechanisms and the development of potential treatment compounds, safe and reliable therapies of neuropathic pain remain poor. Multiple stress/cell death pathways have been shown to be implicated in neuroinflammation during neuropathic pain. Here, we summarize the current knowledge of stress/cell death pathways and present an overview of the roles and molecular mechanisms of stress/cell death pathways in neuroinflammation during neuropathic pain, covering intrinsic and extrinsic apoptosis, autophagy, mitophagy, ferroptosis, pyroptosis, necroptosis, and phagoptosis. Small molecule compounds that modulate stress/cell death pathways in alleviating neuropathic pain are discussed mainly based on preclinical neuropathic pain models. These findings will contribute to in-depth understanding of the pathological processes during neuropathic pain as well as bridge the gap between basic and translational research to uncover new neuroprotective interventions.
Collapse
Affiliation(s)
- Lu Li
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xinyu Qu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Guangwei Sun
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qi Fu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Guang Han
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
30
|
Baaklini CS, Ho MFS, Lange T, Hammond BP, Panda SP, Zirngibl M, Zia S, Himmelsbach K, Rana H, Phillips B, Antoszko D, Ibanga J, Lopez M, Lee KV, Keough MB, Caprariello AV, Kerr BJ, Plemel JR. Microglia promote remyelination independent of their role in clearing myelin debris. Cell Rep 2023; 42:113574. [PMID: 38100356 DOI: 10.1016/j.celrep.2023.113574] [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: 05/18/2023] [Revised: 10/17/2023] [Accepted: 11/27/2023] [Indexed: 12/17/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory disease characterized by myelin loss. While therapies exist to slow MS progression, no treatment currently exists for remyelination. Remyelination, linked to reduced disability in MS, relies on microglia and monocyte-derived macrophages (MDMs). This study aims to understand the role of microglia during remyelination by lineage tracing and depleting them. Microglial lineage tracing reveals that both microglia and MDMs initially accumulate, but microglia later dominate the lesion. Microglia and MDMs engulf equal amounts of inhibitory myelin debris, but after microglial depletion, MDMs compensate by engulfing more myelin debris. Microglial depletion does, however, reduce the recruitment and proliferation of oligodendrocyte progenitor cells (OPCs) and impairs their subsequent differentiation and remyelination. These findings underscore the essential role of microglia during remyelination and offer insights for enhancing this process by understanding microglial regulation of remyelination.
Collapse
Affiliation(s)
- Charbel S Baaklini
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Madelene F S Ho
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Tristan Lange
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Brady P Hammond
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Sharmistha P Panda
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Martin Zirngibl
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Sameera Zia
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Kassandre Himmelsbach
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Heli Rana
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Braxton Phillips
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Daria Antoszko
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Jeremies Ibanga
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Mizuki Lopez
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Kelly V Lee
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Michael B Keough
- Division of Neurosurgery, Department of Surgery, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Andrew V Caprariello
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Cumming School of Medicine, Calgary, AB T2N 1N4, Canada
| | - Bradley J Kerr
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Jason R Plemel
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2R3, Canada; Department of Medicine, Division of Neurology, University of Alberta, Edmonton, AB T6G 2R3, Canada.
| |
Collapse
|
31
|
Kaffe D, Kaplanis SI, Karagogeos D. The Roles of Caloric Restriction Mimetics in Central Nervous System Demyelination and Remyelination. Curr Issues Mol Biol 2023; 45:9526-9548. [PMID: 38132442 PMCID: PMC10742427 DOI: 10.3390/cimb45120596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
The dysfunction of myelinating glial cells, the oligodendrocytes, within the central nervous system (CNS) can result in the disruption of myelin, the lipid-rich multi-layered membrane structure that surrounds most vertebrate axons. This leads to axonal degeneration and motor/cognitive impairments. In response to demyelination in the CNS, the formation of new myelin sheaths occurs through the homeostatic process of remyelination, facilitated by the differentiation of newly formed oligodendrocytes. Apart from oligodendrocytes, the two other main glial cell types of the CNS, microglia and astrocytes, play a pivotal role in remyelination. Following a demyelination insult, microglia can phagocytose myelin debris, thus permitting remyelination, while the developing neuroinflammation in the demyelinated region triggers the activation of astrocytes. Modulating the profile of glial cells can enhance the likelihood of successful remyelination. In this context, recent studies have implicated autophagy as a pivotal pathway in glial cells, playing a significant role in both their maturation and the maintenance of myelin. In this Review, we examine the role of substances capable of modulating the autophagic machinery within the myelinating glial cells of the CNS. Such substances, called caloric restriction mimetics, have been shown to decelerate the aging process by mitigating age-related ailments, with their mechanisms of action intricately linked to the induction of autophagic processes.
Collapse
Affiliation(s)
- Despoina Kaffe
- Department of Biology, University of Crete, Vassilika Vouton, 70013 Heraklion, Greece;
| | - Stefanos Ioannis Kaplanis
- Department of Basic Science, School of Medicine, University of Crete, Vassilika Vouton, 70013 Heraklion, Greece;
- Institute of Molecular Biology & Biotechnology (IMBB), Foundation for Research and Technology-Hellas (FORTH), Vassilika Vouton, 70013 Heraklion, Greece
| | - Domna Karagogeos
- Department of Basic Science, School of Medicine, University of Crete, Vassilika Vouton, 70013 Heraklion, Greece;
- Institute of Molecular Biology & Biotechnology (IMBB), Foundation for Research and Technology-Hellas (FORTH), Vassilika Vouton, 70013 Heraklion, Greece
| |
Collapse
|
32
|
Zou P, Wu C, Liu TCY, Duan R, Yang L. Oligodendrocyte progenitor cells in Alzheimer's disease: from physiology to pathology. Transl Neurodegener 2023; 12:52. [PMID: 37964328 PMCID: PMC10644503 DOI: 10.1186/s40035-023-00385-7] [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: 08/10/2023] [Accepted: 11/01/2023] [Indexed: 11/16/2023] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) play pivotal roles in myelin formation and phagocytosis, communicating with neighboring cells and contributing to the integrity of the blood-brain barrier (BBB). However, under the pathological circumstances of Alzheimer's disease (AD), the brain's microenvironment undergoes detrimental changes that significantly impact OPCs and their functions. Starting with OPC functions, we delve into the transformation of OPCs to myelin-producing oligodendrocytes, the intricate signaling interactions with other cells in the central nervous system (CNS), and the fascinating process of phagocytosis, which influences the function of OPCs and affects CNS homeostasis. Moreover, we discuss the essential role of OPCs in BBB formation and highlight the critical contribution of OPCs in forming CNS-protective barriers. In the context of AD, the deterioration of the local microenvironment in the brain is discussed, mainly focusing on neuroinflammation, oxidative stress, and the accumulation of toxic proteins. The detrimental changes disturb the delicate balance in the brain, impacting the regenerative capacity of OPCs and compromising myelin integrity. Under pathological conditions, OPCs experience significant alterations in migration and proliferation, leading to impaired differentiation and a reduced ability to produce mature oligodendrocytes. Moreover, myelin degeneration and formation become increasingly active in AD, contributing to progressive neurodegeneration. Finally, we summarize the current therapeutic approaches targeting OPCs in AD. Strategies to revitalize OPC senescence, modulate signaling pathways to enhance OPC differentiation, and explore other potential therapeutic avenues are promising in alleviating the impact of AD on OPCs and CNS function. In conclusion, this review highlights the indispensable role of OPCs in CNS function and their involvement in the pathogenesis of AD. The intricate interplay between OPCs and the AD brain microenvironment underscores the complexity of neurodegenerative diseases. Insights from studying OPCs under pathological conditions provide a foundation for innovative therapeutic strategies targeting OPCs and fostering neurodegeneration. Future research will advance our understanding and management of neurodegenerative diseases, ultimately offering hope for effective treatments and improved quality of life for those affected by AD and related disorders.
Collapse
Affiliation(s)
- Peibin Zou
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Chongyun Wu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Timon Cheng-Yi Liu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Rui Duan
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou, 510006, China.
| |
Collapse
|
33
|
Li H, Ghorbani S, Zhang R, Ebacher V, Stephenson EL, Keough MB, Yong VW, Xue M. Prominent elevation of extracellular matrix molecules in intracerebral hemorrhage. Front Mol Neurosci 2023; 16:1251432. [PMID: 38025264 PMCID: PMC10658787 DOI: 10.3389/fnmol.2023.1251432] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Background Intracerebral hemorrhage (ICH) is the predominant type of hemorrhagic stroke with high mortality and disability. In other neurological conditions, the deposition of extracellular matrix (ECM) molecules is a prominent obstacle for regenerative processes and an enhancer of neuroinflammation. Whether ECM molecules alter in composition after ICH, and which ECM members may inhibit repair, remain largely unknown in hemorrhagic stroke. Methods The collagenase-induced ICH mouse model and an autopsied human ICH specimen were investigated for expression of ECM members by immunofluorescence microscopy. Confocal image z-stacks were analyzed with Imaris 3D to assess the association of immune cells and ECM molecules. Sections from a mouse model of multiple sclerosis were used as disease and staining controls. Tissue culture was employed to examine the roles of ECM members on oligodendrocyte precursor cells (OPCs). Results Among the lectican chondroitin sulfate proteoglycan (CSPG) members, neurocan but not aggrecan, versican-V1 and versican-V2 was prominently expressed in perihematomal tissue and lesion core compared to the contralateral area in murine ICH. Fibrinogen, fibronectin and heparan sulfate proteoglycan (HSPG) were also elevated after murine ICH while thrombospondin and tenascin-C was not. Confocal microscopy with Imaris 3D rendering co-localized neurocan, fibrinogen, fibronectin and HSPG molecules to Iba1+ microglia/macrophages or GFAP+ astrocytes. Marked differentiation from the multiple sclerosis model was observed, the latter with high versican-V1 and negligible neurocan. In culture, purified neurocan inhibited adhesion and process outgrowth of OPCs, which are early steps in myelination in vivo. The prominent expression of neurocan in murine ICH was corroborated in human ICH sections. Conclusion ICH caused distinct alterations in ECM molecules. Among CSPG members, neurocan was selectively upregulated in both murine and human ICH. In tissue culture, neurocan impeded the properties of oligodendrocyte lineage cells. Alterations to the ECM in ICH may adversely affect reparative outcomes after stroke.
Collapse
Affiliation(s)
- Hongmin Li
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Samira Ghorbani
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Ruiyi Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Vincent Ebacher
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
| | - Erin L. Stephenson
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - Michael B. Keough
- Division of Neurosurgery, University of Alberta, Edmonton, AB, Canada
| | - V. Wee Yong
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
34
|
Zhang X, Yuan J, Zhang S, Li W, Xu Y, Li H, Zhang L, Chen X, Ding W, Zhu J, Song J, Wang X, Zhu C. Germinal matrix hemorrhage induces immune responses, brain injury, and motor impairment in neonatal rats. J Cereb Blood Flow Metab 2023; 43:49-65. [PMID: 36545808 PMCID: PMC10638988 DOI: 10.1177/0271678x221147091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 11/17/2022] [Accepted: 11/24/2022] [Indexed: 12/24/2022]
Abstract
Germinal matrix hemorrhage (GMH) is a major complication of prematurity that causes secondary brain injury and is associated with long-term neurological disabilities. This study used a postnatal day 5 rat model of GMH to explore immune response, brain injury, and neurobehavioral changes after hemorrhagic injury. The results showed that CD45high/CD11b+ immune cells increased in the brain after GMH and were accompanied by increased macrophage-related chemokine/cytokines and inflammatory mediators. Hematoma formed as early as 2 h after injection of collagenase VII and white matter injury appeared not only in the external capsule and hippocampus, but also in the thalamus. In addition, GMH caused abnormal motor function as revealed by gait analysis, and locomotor hyperactivity in the elevated plus maze, though no other obvious anxiety or recognition/memory function changes were noted when examined by the open field test and novel object recognition test. The animal model used here partially reproduces the GMH-induced brain injury and motor dysfunction seen in human neonates and therefore can be used as a valid tool in experimental studies for the development of effective therapeutic strategies for GMH-induced brain injury.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou, China
| | - Jing Yuan
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou, China
| | - Shan Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou, China
| | - Wendong Li
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou, China
| | - Yiran Xu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Birth Defects Prevention, Henan Key Laboratory of Population Defects Prevention, Zhengzhou, China
| | - Hongwei Li
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou, China
| | - Lingling Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou, China
| | - Xi Chen
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou, China
| | - Wenjun Ding
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou, China
| | - Jinjin Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou, China
| | - Juan Song
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou, China
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou, China
- Center for Perinatal Medicine and Health, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Third Affiliated Hospital and Institute of Neuroscience of Zhengzhou University, Zhengzhou, China
- Center for Bran Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
35
|
Mokhtarzadeh Khanghahi A, Rayatpour A, Baharvand H, Javan M. Neuroglial components of brain lesions may provide new therapeutic strategies for multiple sclerosis. Neurol Sci 2023; 44:3795-3807. [PMID: 37410268 DOI: 10.1007/s10072-023-06915-5] [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: 12/30/2022] [Accepted: 06/14/2023] [Indexed: 07/07/2023]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune and demyelinating disease of the central nervous system (CNS) which leads to focal demyelinated lesions in the brain and spinal cord. Failure of remyelination contributes to chronic disability in young adults. Characterization of events occurring during the demyelination and remyelination processes and those of which subsequently limit remyelination or contribute to demyelination can provide the possibility of new therapies development for MS. Most of the currently available therapies and investigations modulate immune responses and mediators. Since most therapeutic strategies have unsatisfied outcomes, developing new therapies that enhance brain lesion repair is a priority. A close look at cellular and chemical components of MS lesions will pave the way to a better understanding of lesions pathology and will provide possible opportunities for repair strategies and targeted pharmacotherapy. This review summarizes the lesion components and features, particularly the detrimental elements, and discusses the possibility of suggesting new potential targets as therapies for demyelinating diseases like MS.
Collapse
Affiliation(s)
- Akram Mokhtarzadeh Khanghahi
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Atefeh Rayatpour
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Institute for Brain and Cognition, Tarbiat Modares University, Tehran, Iran
| | - Hossein Baharvand
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mohammad Javan
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
- Institute for Brain and Cognition, Tarbiat Modares University, Tehran, Iran.
- International Collaboration on Repair Discoveries (ICORD), the University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
36
|
Tripathi A, Rai N, Perles A, Jones C, Dutta R. Dicer deficiency in microglia leads to accelerated demyelination and failed remyelination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.17.562812. [PMID: 37905110 PMCID: PMC10614879 DOI: 10.1101/2023.10.17.562812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Microglia are the resident immune cells of the central nervous system (CNS) and are important regulators of normal brain functions. In CNS demyelinating diseases like multiple sclerosis (MS), the functions of these cells are of particular interest. Here we probed the impact of microRNA (miRNA)-mediated post-transcriptional gene regulation using a mouse model lacking microglia/macrophage-specific Dicer expression during demyelination and remyelination. Conditional Dicer ablation and loss of miRNAs in adult microglia led to extensive demyelination and impaired myelin processing. Interestingly, demyelination was accompanied by increased apoptosis of mature oligodendrocytes (OLs) and arresting OL progenitor cells (OPCs) in the precursor stage. At the transcriptional level, Dicer -deficient microglia led to downregulation of microglial homeostatic genes, increased cell proliferation, and a shift towards a disease-associated phenotype. Loss of remyelination efficiency in these mice was accompanied by stalling of OPCs in the precursor stage. Collectively, these results highlight a new role of microglial miRNAs in promoting a pro-regenerative phenotype in addition to promoting OPC maturation and differentiation during demyelination and remyelination.
Collapse
|
37
|
Yonemoto K, Fujii F, Taira R, Ohgidani M, Eguchi K, Okuzono S, Ichimiya Y, Sonoda Y, Chong PF, Goto H, Kanemasa H, Motomura Y, Ishimura M, Koga Y, Tsujimura K, Hashiguchi T, Torisu H, Kira R, Kato TA, Sakai Y, Ohga S. Heterogeneity and mitochondrial vulnerability configurate the divergent immunoreactivity of human induced microglia-like cells. Clin Immunol 2023; 255:109756. [PMID: 37678717 DOI: 10.1016/j.clim.2023.109756] [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: 06/01/2023] [Accepted: 09/02/2023] [Indexed: 09/09/2023]
Abstract
Microglia play versatile roles in progression of and protection against neuroinflammatory diseases. Little is known, however, about the mechanisms underlying the diverse reactivity of microglia to inflammatory conditions. We investigated how human induced microglia-like (iMG) cells respond to innate immune ligands. Quantitative PCR showed that poly-I:C and lipopolysaccharide (LPS) activated the expression of IL1B and TNF. Immunoreactivity of iMG did not differ between controls (n = 11) and patients with neuroinflammatory diseases (n = 24). Flow cytometry revealed that CD14high cells expressed interleukin (IL) -1β after LPS treatment. Immunoblotting showed that poly-I:C and LPS differentially activated inflammatory pathways but commonly induced mitochondrial instability and the expression of pyruvate kinase isoform M2 (PKM2). Furthermore, a potent stimulator of PKM2 (DASA-58) alleviated IL-1β production after LPS treatment. These data indicate that heterogeneous cell populations and mitochondrial stability underlie the divergent immunoreactivity of human iMG in environments.
Collapse
Affiliation(s)
- Kousuke Yonemoto
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Fumihiko Fujii
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryoji Taira
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masahiro Ohgidani
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Functional Anatomy and Neuroscience, Asahikawa Medical University, Hokkaido, Japan
| | - Katsuhide Eguchi
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Sayaka Okuzono
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Section of Pediatrics, Department of Medicine, Fukuoka Dental College, Fukuoka, Japan
| | - Yuko Ichimiya
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuri Sonoda
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Pin Fee Chong
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hironori Goto
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hikaru Kanemasa
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshitomo Motomura
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masataka Ishimura
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuhki Koga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Keita Tsujimura
- Group of Brain Function and Development, Neuroscience Institute of the Graduate School of Science, Nagoya University, Aichi, Japan; Research Unit for Developmental Disorders, Institute for Advanced Research, Nagoya University, Aichi, Japan
| | - Takao Hashiguchi
- Laboratory of Medical Virology, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Hiroyuki Torisu
- Section of Pediatrics, Department of Medicine, Fukuoka Dental College, Fukuoka, Japan
| | - Ryutaro Kira
- Department of Pediatric Neurology, Fukuoka Children's Hospital, Fukuoka, Japan
| | - Takahiro A Kato
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yasunari Sakai
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
38
|
Touil H, Li R, Zuroff L, Moore CS, Healy L, Cignarella F, Piccio L, Ludwin S, Prat A, Gommerman J, Bennett FC, Jacobs D, Benjamins JA, Lisak RP, Antel JP, Bar-Or A. Cross-talk between B cells, microglia and macrophages, and implications to central nervous system compartmentalized inflammation and progressive multiple sclerosis. EBioMedicine 2023; 96:104789. [PMID: 37703640 PMCID: PMC10505984 DOI: 10.1016/j.ebiom.2023.104789] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 08/07/2023] [Accepted: 08/22/2023] [Indexed: 09/15/2023] Open
Abstract
BACKGROUND B cells can be enriched within meningeal immune-cell aggregates of multiple sclerosis (MS) patients, adjacent to subpial cortical demyelinating lesions now recognized as important contributors to progressive disease. This subpial demyelination is notable for a 'surface-in' gradient of neuronal loss and microglial activation, potentially reflecting the effects of soluble factors secreted into the CSF. We previously demonstrated that MS B-cell secreted products are toxic to oligodendrocytes and neurons. The potential for B-cell-myeloid cell interactions to propagate progressive MS is of considerable interest. METHODS Secreted products of MS-implicated pro-inflammatory effector B cells or IL-10-expressing B cells with regulatory potential were applied to human brain-derived microglia or monocyte-derived macrophages, with subsequent assessment of myeloid phenotype and function through measurement of their expression of pro-inflammatory, anti-inflammatory and homeostatic/quiescent molecules, and phagocytosis (using flow cytometry, ELISA and fluorescently-labeled myelin). Effects of secreted products of differentially activated microglia on B-cell survival and activation were further studied. FINDINGS Secreted products of MS-implicated pro-inflammatory B cells (but not IL-10 expressing B cells) substantially induce pro-inflammatory cytokine (IL-12, IL-6, TNFα) expression by both human microglia and macrophage (in a GM-CSF dependent manner), while down-regulating their expression of IL-10 and of quiescence-associated molecules, and suppressing their myelin phagocytosis. In contrast, secreted products of IL-10 expressing B cells upregulate both human microglia and macrophage expression of quiescence-associated molecules and enhance their myelin phagocytosis. Secreted factors from pro-inflammatory microglia enhance B-cell activation. INTERPRETATION Potential cross-talk between disease-relevant human B-cell subsets and both resident CNS microglia and infiltrating macrophages may propagate CNS-compartmentalized inflammation and injury associated with MS disease progression. These interaction represents an attractive therapeutic target for agents such as Bruton's tyrosine kinase inhibitors (BTKi) that modulate responses of both B cells and myeloid cells. FUNDING Stated in Acknowledgments section of manuscript.
Collapse
Affiliation(s)
- Hanane Touil
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rui Li
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Leah Zuroff
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Craig S Moore
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Luke Healy
- Neuroimmunology Unit, Montréal Neurological Institute, McGill University, Canada
| | - Francesca Cignarella
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, St Louis, MO, USA
| | - Laura Piccio
- Charles Perkins Centre and School of Medical Sciences, The University of Sydney, Camperdown, NSW, Australia
| | - Samuel Ludwin
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Alexandre Prat
- Université de Montréal Centre de Recherche du CHUM (CRCHUM) and Department of Neuroscience, Université de Montréal, 900 Saint Denis Street, Montréal, QC, H2X 0A9, Canada
| | - Jennifer Gommerman
- Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Frederick C Bennett
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dina Jacobs
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joyce A Benjamins
- Departments of Neurology and Biochemistry, Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Robert P Lisak
- Departments of Neurology and Biochemistry, Immunology and Microbiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jack P Antel
- Neuroimmunology Unit, Montréal Neurological Institute, McGill University, Canada
| | - Amit Bar-Or
- Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
39
|
Wang J, Brown K, Danehy C, Mérigeon E, Goralski S, Rice S, Torgbe K, Thomas F, Block D, Olsen H, Strome SE, Fitzpatrick EA. Fc multimers effectively treat murine models of multiple sclerosis. Front Immunol 2023; 14:1199747. [PMID: 37638040 PMCID: PMC10451071 DOI: 10.3389/fimmu.2023.1199747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/25/2023] [Indexed: 08/29/2023] Open
Abstract
Multiple Sclerosis (MS) is a chronic neurodegenerative disease with limited therapeutic options. Recombinant Fc multimers (rFc), designed to mirror many of the anti-inflammatory activities of Intravenous Immunoglobulin (IVIG), have been shown to effectively treat numerous immune-mediated diseases in rodents. In this study we used the experimental autoimmune encephalomyelitis (EAE) murine model of MS to test the efficacy of a rFc, M019, that consists of multimers of the Fc portion of IgG2, in inhibiting disease severity. We show that M019 effectively reduced clinical symptoms when given either pre- or post-symptom onset compared to vehicle treated EAE induced mice. M019 was effective in reducing symptoms in both SJL model of relapsing remitting MS as well as the B6 model of chronic disease. M019 binds to FcγR bearing-monocytes both in vivo and in vitro and prevented immune cell infiltration into the CNS of treated mice. The lack of T cell infiltration into the spinal cord was not due to a decrease in T cell priming; there was an equivalent frequency of Th17 cells in the spleens of M019 and vehicle treated EAE induced mice. Surprisingly, there was an increase in chemokines in the sera but not in the CNS of M019 treated mice compared to vehicle treated animals. We postulate that M019 interacts with a FcγR rich monocyte intermediary to prevent T cell migration into the CNS and demyelination.
Collapse
Affiliation(s)
- Jin Wang
- Dept. of Microbiology Immunology and Biochemistry, UTHSC, Memphis, TN, United States
| | - Kellie Brown
- Dept. of Microbiology Immunology and Biochemistry, UTHSC, Memphis, TN, United States
| | - Caroline Danehy
- College of Graduate Health Sciences, UTHSC, Memphis, TN, United States
| | | | | | - Samuel Rice
- College of Medicine, UTHSC, Memphis, TN, United States
| | - Kwame Torgbe
- Dept. of Pathology, UTHSC, Memphis, TN, United States
| | - Fridtjof Thomas
- Div. of Biostatistics, Dept. of Preventive Medicine, UTHSC, Memphis, TN, United States
| | | | | | - Scott E. Strome
- Dept. of Microbiology Immunology and Biochemistry, UTHSC, Memphis, TN, United States
| | | |
Collapse
|
40
|
Salvador AFM, Dykstra T, Rustenhoven J, Gao W, Blackburn SM, Bhasiin K, Dong MQ, Guimarães RM, Gonuguntla S, Smirnov I, Kipnis J, Herz J. Age-dependent immune and lymphatic responses after spinal cord injury. Neuron 2023; 111:2155-2169.e9. [PMID: 37148871 PMCID: PMC10523880 DOI: 10.1016/j.neuron.2023.04.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 02/13/2023] [Accepted: 04/12/2023] [Indexed: 05/08/2023]
Abstract
Spinal cord injury (SCI) causes lifelong debilitating conditions. Previous works demonstrated the essential role of the immune system in recovery after SCI. Here, we explored the temporal changes of the response after SCI in young and aged mice in order to characterize multiple immune populations within the mammalian spinal cord. We revealed substantial infiltration of myeloid cells to the spinal cord in young animals, accompanied by changes in the activation state of microglia. In contrast, both processes were blunted in aged mice. Interestingly, we discovered the formation of meningeal lymphatic structures above the lesion site, and their role has not been examined after contusive injury. Our transcriptomic data predicted lymphangiogenic signaling between myeloid cells in the spinal cord and lymphatic endothelial cells (LECs) in the meninges after SCI. Together, our findings delineate how aging affects the immune response following SCI and highlight the participation of the spinal cord meninges in supporting vascular repair.
Collapse
Affiliation(s)
- Andrea Francesca M Salvador
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA; Neuroscience Graduate Program, University of Virginia, Charlottesville, VA 22903, USA
| | - Taitea Dykstra
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Justin Rustenhoven
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Auckland 1023, New Zealand
| | - Wenqing Gao
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Susan M Blackburn
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Kesshni Bhasiin
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Michael Q Dong
- Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Rafaela Mano Guimarães
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA; Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo 14049-900, Brazil
| | - Sriharsha Gonuguntla
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Igor Smirnov
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Jonathan Kipnis
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA.
| | - Jasmin Herz
- Brain Immunology and Glia (BIG) Center, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Immunobiology, Washington University in St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
41
|
Boghozian R, Sharma S, Narayana K, Cheema M, Brown CE. Sex and interferon gamma signaling regulate microglia migration in the adult mouse cortex in vivo. Proc Natl Acad Sci U S A 2023; 120:e2302892120. [PMID: 37428916 PMCID: PMC10629543 DOI: 10.1073/pnas.2302892120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 06/02/2023] [Indexed: 07/12/2023] Open
Abstract
Although microglia possess the unique ability to migrate, whether mobility is evident in all microglia, is sex dependent, and what molecular mechanisms drive this, is not well understood in the adult brain. Using longitudinal in vivo two-photon imaging of sparsely labeled microglia, we find a relatively small population of microglia (~5%) are mobile under normal conditions. Following injury (microbleed), the fraction of mobile microglia increased in a sex-dependent manner, with male microglia migrating significantly greater distances toward the microbleed relative to their female counterparts. To understand the signaling pathways involved, we interrogated the role of interferon gamma (IFNγ). Our data show that in male mice, stimulating microglia with IFNγ promotes migration whereas inhibiting IFNγ receptor 1 signaling inhibits them. By contrast, female microglia were generally unaffected by these manipulations. These findings highlight the diversity of microglia migratory responses to injury, its dependence on sex and the signaling mechanisms that modulate this behavior.
Collapse
Affiliation(s)
- Roobina Boghozian
- Division of Medical Sciences, University of Victoria, Victoria, BCV8P 5C2, Canada
| | - Sorabh Sharma
- Division of Medical Sciences, University of Victoria, Victoria, BCV8P 5C2, Canada
| | - Kamal Narayana
- Division of Medical Sciences, University of Victoria, Victoria, BCV8P 5C2, Canada
| | - Manjinder Cheema
- Division of Medical Sciences, University of Victoria, Victoria, BCV8P 5C2, Canada
| | - Craig E. Brown
- Division of Medical Sciences, University of Victoria, Victoria, BCV8P 5C2, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, BCV6T 2A1, Canada
| |
Collapse
|
42
|
Androvic P, Schifferer M, Perez Anderson K, Cantuti-Castelvetri L, Jiang H, Ji H, Liu L, Gouna G, Berghoff SA, Besson-Girard S, Knoferle J, Simons M, Gokce O. Spatial Transcriptomics-correlated Electron Microscopy maps transcriptional and ultrastructural responses to brain injury. Nat Commun 2023; 14:4115. [PMID: 37433806 DOI: 10.1038/s41467-023-39447-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 06/14/2023] [Indexed: 07/13/2023] Open
Abstract
Understanding the complexity of cellular function within a tissue necessitates the combination of multiple phenotypic readouts. Here, we developed a method that links spatially-resolved gene expression of single cells with their ultrastructural morphology by integrating multiplexed error-robust fluorescence in situ hybridization (MERFISH) and large area volume electron microscopy (EM) on adjacent tissue sections. Using this method, we characterized in situ ultrastructural and transcriptional responses of glial cells and infiltrating T-cells after demyelinating brain injury in male mice. We identified a population of lipid-loaded "foamy" microglia located in the center of remyelinating lesion, as well as rare interferon-responsive microglia, oligodendrocytes, and astrocytes that co-localized with T-cells. We validated our findings using immunocytochemistry and lipid staining-coupled single-cell RNA sequencing. Finally, by integrating these datasets, we detected correlations between full-transcriptome gene expression and ultrastructural features of microglia. Our results offer an integrative view of the spatial, ultrastructural, and transcriptional reorganization of single cells after demyelinating brain injury.
Collapse
Affiliation(s)
- Peter Androvic
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Martina Schifferer
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Katrin Perez Anderson
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Ludovico Cantuti-Castelvetri
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Hanyi Jiang
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Hao Ji
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Lu Liu
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Garyfallia Gouna
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Stefan A Berghoff
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Simon Besson-Girard
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Johanna Knoferle
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
| | - Mikael Simons
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Ozgun Gokce
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany.
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany.
| |
Collapse
|
43
|
Xue S, Lozinski BM, Ghorbani S, Ta K, D'Mello C, Yong VW, Dong Y. Elevated Galectin-3 Is Associated with Aging, Multiple Sclerosis, and Oxidized Phosphatidylcholine-Induced Neurodegeneration. J Neurosci 2023; 43:4725-4737. [PMID: 37208177 PMCID: PMC10286945 DOI: 10.1523/jneurosci.2312-22.2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 04/25/2023] [Accepted: 05/05/2023] [Indexed: 05/21/2023] Open
Abstract
Aging is a significant risk factor associated with the progression of CNS neurodegenerative diseases including multiple sclerosis (MS). Microglia, the resident macrophages of the CNS parenchyma, are a major population of immune cells that accumulate in MS lesions. While they normally regulate tissue homeostasis and facilitate the clearance of neurotoxic molecules including oxidized phosphatidylcholines (OxPCs), their transcriptome and neuroprotective functions are reprogrammed by aging. Thus, determining the factors that instigate aging associated microglia dysfunction can lead to new insights for promoting CNS repair and for halting MS disease progression. Through single-cell RNA sequencing (scRNAseq), we identified Lgals3, which encodes for galectin-3 (Gal3), as an age upregulated gene by microglia responding to OxPC. Consistently, excess Gal3 accumulated in OxPC and lysolecithin-induced focal spinal cord white matter (SCWM) lesions of middle-aged mice compared with young mice. Gal3 was also elevated in mouse experimental autoimmune encephalomyelitis (EAE) lesions and more importantly in MS brain lesions from two male and one female individuals. While Gal3 delivery alone into the mouse spinal cord did not induce damage, its co-delivery with OxPC increased cleaved caspase 3 and IL-1β within white matter lesions and exacerbated OxPC-induced injury. Conversely, OxPC-mediated neurodegeneration was reduced in Gal3-/- mice compared with Gal3+/+ mice. Thus, Gal3 is associated with increased neuroinflammation and neurodegeneration and its overexpression by microglia/macrophages may be detrimental for lesions within the aging CNS.SIGNIFICANCE STATEMENT Aging accelerates the progression of neurodegenerative diseases such as multiple sclerosis (MS). Understanding the molecular mechanisms of aging that increases the susceptibility of the CNS to damage could lead to new strategies to manage MS progression. Here, we highlight that microglia/macrophage-associated galectin-3 (Gal3) was upregulated with age exacerbated neurodegeneration in the mouse spinal cord white matter (SCWM) and in MS lesions. More importantly, co-injection of Gal3 with oxidized phosphatidylcholines (OxPCs), which are neurotoxic lipids found in MS lesions, caused greater neurodegeneration compared with injection of OxPC alone, whereas genetic loss of Gal3 reduced OxPC damage. These results demonstrate that Gal3 overexpression is detrimental to CNS lesions and suggest its deposition in MS lesions may contribute to neurodegeneration.
Collapse
Affiliation(s)
- Sara Xue
- Hotchkiss Brain Institute and the Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Brian M Lozinski
- Hotchkiss Brain Institute and the Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Samira Ghorbani
- Hotchkiss Brain Institute and the Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Khanh Ta
- Department of Biochemistry, Microbiology, and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Charlotte D'Mello
- Hotchkiss Brain Institute and the Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and the Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Yifei Dong
- Department of Biochemistry, Microbiology, and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| |
Collapse
|
44
|
Wishart CL, Spiteri AG, Locatelli G, King NJC. Integrating transcriptomic datasets across neurological disease identifies unique myeloid subpopulations driving disease-specific signatures. Glia 2023; 71:904-925. [PMID: 36527260 PMCID: PMC10952672 DOI: 10.1002/glia.24314] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 11/06/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022]
Abstract
Microglia and bone marrow-derived monocytes are key elements of central nervous system (CNS) inflammation, both capable of enhancing and dampening immune-mediated pathology. However, the study-specific focus on individual cell types, disease models or experimental approaches has limited our ability to infer common and disease-specific responses. This meta-analysis integrates bulk and single-cell transcriptomic datasets of microglia and monocytes from disease models of autoimmunity, neurodegeneration, sterile injury, and infection to build a comprehensive resource connecting myeloid responses across CNS disease. We demonstrate that the bulk microglial and monocyte program is highly contingent on the disease environment, challenging the notion of a universal microglial disease signature. Integration of six single-cell RNA-sequencing datasets revealed that these disease-specific signatures are likely driven by differing proportions of unique myeloid subpopulations that were individually expanded in different disease settings. These subsets were functionally-defined as neurodegeneration-associated, inflammatory, interferon-responsive, phagocytic, antigen-presenting, and lipopolysaccharide-responsive cellular states, revealing a core set of myeloid responses at the single-cell level that are conserved across CNS pathology. Showcasing the predictive and practical value of this resource, we performed differential expression analysis on microglia and monocytes across disease and identified Cd81 as a new neuroinflammatory-stable gene that accurately identified microglia and distinguished them from monocyte-derived cells across all experimental models at both the bulk and single-cell level. Together, this resource dissects the influence of disease environment on shared immune response programmes to build a unified perspective of myeloid behavior across CNS pathology.
Collapse
Affiliation(s)
- Claire L. Wishart
- Infection, Immunity, Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
- Sydney Cytometry FacilityThe University of Sydney and Centenary InstituteSydneyNew South WalesAustralia
- Ramaciotti Facility for Human Systems BiologyThe University of Sydney and Centenary InstituteSydneyNew South WalesAustralia
- Charles Perkins CentreThe University of SydneySydneyNew South WalesAustralia
| | - Alanna G. Spiteri
- Infection, Immunity, Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
- Sydney Cytometry FacilityThe University of Sydney and Centenary InstituteSydneyNew South WalesAustralia
- Ramaciotti Facility for Human Systems BiologyThe University of Sydney and Centenary InstituteSydneyNew South WalesAustralia
- Charles Perkins CentreThe University of SydneySydneyNew South WalesAustralia
| | - Giuseppe Locatelli
- Theodor Kocher InstituteUniversity of BernBernSwitzerland
- Novartis Institutes for BioMedical ResearchNovartisBaselSwitzerland
| | - Nicholas J. C. King
- Infection, Immunity, Inflammation Research Theme, School of Medical Sciences, Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
- Sydney Cytometry FacilityThe University of Sydney and Centenary InstituteSydneyNew South WalesAustralia
- Ramaciotti Facility for Human Systems BiologyThe University of Sydney and Centenary InstituteSydneyNew South WalesAustralia
- Charles Perkins CentreThe University of SydneySydneyNew South WalesAustralia
- Sydney Institute for Infectious Diseases, Faculty of Medicine and HealthThe University of SydneySydneyNew South WalesAustralia
- The University of Sydney Nano Institute, Faculty of ScienceThe University of SydneySydneyNew South WalesAustralia
| |
Collapse
|
45
|
Pilipović I, Stojić-Vukanić Z, Prijić I, Jasnić N, Djordjević J, Leposavić G. β-Adrenoceptor Blockade Moderates Neuroinflammation in Male and Female EAE Rats and Abrogates Sexual Dimorphisms in the Major Neuroinflammatory Pathways by Being More Efficient in Males. Cell Mol Neurobiol 2023; 43:1237-1265. [PMID: 35798933 PMCID: PMC11414456 DOI: 10.1007/s10571-022-01246-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/18/2022] [Indexed: 11/03/2022]
Abstract
Our previous studies showed more severe experimental autoimmune encephalomyelitis (EAE) in male compared with female adult rats, and moderating effect of propranolol-induced β-adrenoceptor blockade on EAE in females, the effect associated with transcriptional stimulation of Nrf2/HO-1 axis in spinal cord microglia. This study examined putative sexual dimorphism in propranolol action on EAE severity. Propranolol treatment beginning from the onset of clinical EAE mitigated EAE severity in rats of both sexes, but to a greater extent in males exhibiting higher noradrenaline levels and myeloid cell β2-adrenoceptor expression in spinal cord. This correlated with more prominent stimulatory effects of propranolol not only on CX3CL1/CX3CR1/Nrf2/HO-1 cascade, but also on Stat3/Socs3 signaling axis in spinal cord microglia/myeloid cells (mirrored in the decreased Stat3 and the increased Socs3 expression) from male rats compared with their female counterparts. Propranolol diminished the frequency of activated cells among microglia, increased their phagocyting/endocyting capacity, and shifted cytokine secretory profile of microglia/blood-borne myeloid cells towards an anti-inflammatory/neuroprotective phenotype. Additionally, it downregulated the expression of chemokines (CCL2, CCL19/21) driving T-cell/monocyte trafficking into spinal cord. Consequently, in propranolol-treated rats fewer activated CD4+ T cells and IL-17+ T cells, including CD4+IL17+ cells coexpressing IFN-γ/GM-CSF, were recovered from spinal cord of propranolol-treated rats compared with sex-matched saline-injected controls. All the effects of propranolol were more prominent in males. The study as a whole disclosed that sexual dimorphism in multiple molecular mechanisms implicated in EAE development may be responsible for greater severity of EAE in male rats and sexually dimorphic action of substances affecting them. Propranolol moderated EAE severity more effectively in male rats, exhibiting greater spinal cord noradrenaline (NA) levels and myeloid cell β2-adrenoceptor (β2-AR) expression than females. Propranolol affected CX3CR1/Nrf2/HO-1 and Stat3/Socs3 signaling axes in myeloid cells, favored their anti-inflammatory/neuroprotective phenotype and, consequently, reduced Th cell reactivation and differentiation into highly pathogenic IL-17/IFN-γ/GM-CSF-producing cells.
Collapse
Affiliation(s)
- Ivan Pilipović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Ivana Prijić
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", Belgrade, Serbia
| | - Nebojša Jasnić
- Institute for Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Jelena Djordjević
- Institute for Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Gordana Leposavić
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, 450 Vojvode Stepe, 11221, Belgrade, Serbia.
| |
Collapse
|
46
|
Vainchtein ID, Alsema AM, Dubbelaar ML, Grit C, Vinet J, van Weering HRJ, Al‐Izki S, Biagini G, Brouwer N, Amor S, Baker D, Eggen BJL, Boddeke EWGM, Kooistra SM. Characterizing microglial gene expression in a model of secondary progressive multiple sclerosis. Glia 2023; 71:588-601. [PMID: 36377669 PMCID: PMC10100411 DOI: 10.1002/glia.24297] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 09/30/2022] [Accepted: 10/26/2022] [Indexed: 11/16/2022]
Abstract
Multiple sclerosis (MS) is the most common inflammatory, demyelinating and neurodegenerative disease of the central nervous system in young adults. Chronic-relapsing experimental autoimmune encephalomyelitis (crEAE) in Biozzi ABH mice is an experimental model of MS. This crEAE model is characterized by an acute phase with severe neurological disability, followed by remission of disease, relapse of neurological disease and remission that eventually results in a chronic progressive phase that mimics the secondary progressive phase (SPEAE) of MS. In both MS and SPEAE, the role of microglia is poorly defined. We used a crEAE model to characterize microglia in the different phases of crEAE phases using morphometric and RNA sequencing analyses. At the initial, acute inflammation phase, microglia acquired a pro-inflammatory phenotype. At the remission phase, expression of standard immune activation genes was decreased while expression of genes associated with lipid metabolism and tissue remodeling were increased. Chronic phase microglia partially regain inflammatory gene sets and increase expression of genes associated with proliferation. Together, the data presented here indicate that microglia obtain different features at different stages of crEAE and a particularly mixed phenotype in the chronic stage. Understanding the properties of microglia that are present at the chronic phase of EAE will help to understand the role of microglia in secondary progressive MS, to better aid the development of therapies for this phase of the disease.
Collapse
Affiliation(s)
- Ilia D. Vainchtein
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Astrid M. Alsema
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Marissa L. Dubbelaar
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Corien Grit
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Jonathan Vinet
- Department of Biomedical, Metabolic and Neural SciencesUniversity of Modena and Reggio EmiliaModenaItaly
| | - Hilmar R. J. van Weering
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Sarah Al‐Izki
- Department of NeuroimmunologyBlizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of LondonLondonUK
| | - Giuseppe Biagini
- Department of Biomedical, Metabolic and Neural SciencesUniversity of Modena and Reggio EmiliaModenaItaly
| | - Nieske Brouwer
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Sandra Amor
- Department of NeuroimmunologyBlizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of LondonLondonUK
- Department of PathologyVUMCAmsterdamThe Netherlands
| | - David Baker
- Department of NeuroimmunologyBlizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of LondonLondonUK
| | - Bart J. L. Eggen
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| | - Erik W. G. M. Boddeke
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
- Department of Cellular and Molecular MedicineCenter for Healthy Ageing, University of CopenhagenCopenhagenDenmark
| | - Susanne M. Kooistra
- Department of Biomedical Sciences of Cells & Systems, Section Molecular NeurobiologyUniversity of Groningen, University Medical Center GroningenGroningenThe Netherlands
| |
Collapse
|
47
|
Hu X, Li S, Shi Z, Lin WJ, Yang Y, Li Y, Li H, Xu Y, Zhou M, Tang Y. Partial Ablation of Astrocytes Exacerbates Cerebral Infiltration of Monocytes and Neuronal Loss After Brain Stab Injury in Mice. Cell Mol Neurobiol 2023; 43:893-905. [PMID: 35437650 PMCID: PMC11415208 DOI: 10.1007/s10571-022-01224-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/31/2022] [Indexed: 11/03/2022]
Abstract
In traumatic brain injury (TBI), mechanical injury results in instantaneous tissue damages accompanied by subsequent pro-inflammatory cascades composed of microgliosis and astrogliosis. However, the interactive roles between microglia and astrocytes during the pathogenesis of TBI remain unclear and sometimes debatable. In this study, we used a forebrain stab injury mouse model to investigate the pathological role of reactive astrocytes in cellular and molecular changes of inflammatory response following TBI. In the ipsilateral hemisphere of stab-injured brain, monocyte infiltration and neuronal loss, as well as increased elevated astrogliosis, microglia activation and inflammatory cytokines were observed. To verify the role of reactive astrocytes in TBI, local and partial ablation of astrocytes was achieved by stereotactic injection of diphtheria toxin in the forebrain of Aldh1l1-CreERT2::Ai9::iDTR transgenic mice which expressed diphtheria toxin receptor (DTR) in astrocytes after tamoxifen induction. This strategy achieved about 20% of astrocytes reduction at the stab site as validated by immunofluorescence co-staining of GFAP with tdTomato-positive astrocytes. Interestingly, reduction of astrocytes showed increased microglia activation and monocyte infiltration, accompanied with increased severity in stab injury-induced neuronal loss when compared with DTR-/- mice, together with elevation of inflammatory chemokines such as CCL2, CCL5 and CXCL10 in astrogliosis-reduced mice. Collectively, our data verified the interactive role of astrocytes as an immune modulator in suppressing inflammatory responses in the injured brain. Schematic diagram shows monocyte infiltration and neuronal loss, as well as increased elevated astrogliosis, microglia activation and chemokines were observed in the injured site after stab injury. Local and partial ablation of astrocytes led to increased microglia activation and monocyte infiltration, accompanied with increased severity in neuronal loss together with elevation of inflammatory chemokines as compared with control mice subjected stab injury.
Collapse
Affiliation(s)
- Xia Hu
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Shaojian Li
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat‑Sen University, Guangzhou, 510120, China
| | - Zhongshan Shi
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat‑Sen University, Guangzhou, 510120, China
| | - Wei-Jye Lin
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Yuhua Yang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat‑Sen University, Guangzhou, 510120, China
| | - Yi Li
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat‑Sen University, Guangzhou, 510120, China
| | - Honghong Li
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat‑Sen University, Guangzhou, 510120, China
| | - Yongteng Xu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat‑Sen University, Guangzhou, 510120, China
| | - Meijuan Zhou
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China.
| | - Yamei Tang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat‑Sen University, Guangzhou, 510120, China.
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-Sen University, Guangzhou, 510275, China.
| |
Collapse
|
48
|
Drake SS, Zaman A, Simas T, Fournier AE. Comparing RNA-sequencing datasets from astrocytes, oligodendrocytes, and microglia in multiple sclerosis identifies novel dysregulated genes relevant to inflammation and myelination. WIREs Mech Dis 2023; 15:e1594. [PMID: 36600404 DOI: 10.1002/wsbm.1594] [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: 06/20/2022] [Revised: 09/25/2022] [Accepted: 12/14/2022] [Indexed: 01/06/2023]
Abstract
Central nervous system (CNS) inflammation is a key factor in multiple sclerosis (MS). Invasion of peripheral immune cells into the CNS resulting from an unknown signal or combination of signals results in activation of resident immune cells and the hallmark feature of the disease: demyelinating lesions. These lesion sites are an amalgam of reactive peripheral and central immune cells, astrocytes, damaged and dying oligodendrocytes, and injured neurons and axons. Sustained inflammation affects cells directly located within the lesion site and further abnormalities are apparent diffusely throughout normal-appearing white matter and grey matter. It is only relatively recently, using animal models, new tissue sampling techniques, and next-generation sequencing, that molecular changes occurring in CNS resident cells have been broadly captured. Advances in cell isolation through Fluorescence Activated Cell Sorting (FACS) and laser-capture microdissection together with the emergence of single-cell sequencing have enabled researchers to investigate changes in gene expression in astrocytes, microglia, and oligodendrocytes derived from animal models of MS as well as from primary patient tissue. The contribution of some dysregulated pathways has been followed up in individual studies; however, corroborating results often go unreported between sequencing studies. To this end, we have consolidated results from numerous RNA-sequencing studies to identify and review novel patterns of differentially regulated genes and pathways occurring within CNS glial cells in MS. This article is categorized under: Neurological Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Sienna S Drake
- McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Aliyah Zaman
- McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Tristan Simas
- McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada
| | - Alyson E Fournier
- McGill University, Montreal Neurological Institute, Montreal, Quebec, Canada
| |
Collapse
|
49
|
Muñoz-Castro C, Mejias-Ortega M, Sanchez-Mejias E, Navarro V, Trujillo-Estrada L, Jimenez S, Garcia-Leon JA, Fernandez-Valenzuela JJ, Sanchez-Mico MV, Romero-Molina C, Moreno-Gonzalez I, Baglietto-Vargas D, Vizuete M, Gutierrez A, Vitorica J. Monocyte-derived cells invade brain parenchyma and amyloid plaques in human Alzheimer's disease hippocampus. Acta Neuropathol Commun 2023; 11:31. [PMID: 36855152 PMCID: PMC9976401 DOI: 10.1186/s40478-023-01530-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 03/02/2023] Open
Abstract
Microglia are brain-resident myeloid cells and play a major role in the innate immune responses of the CNS and the pathogenesis of Alzheimer's disease (AD). However, the contribution of nonparenchymal or brain-infiltrated myeloid cells to disease progression remains to be demonstrated. Here, we show that monocyte-derived cells (MDC) invade brain parenchyma in advanced stages of AD continuum using transcriptional analysis and immunohistochemical characterization in post-mortem human hippocampus. Our findings demonstrated that a high proportion (60%) of demented Braak V-VI individuals was associated with up-regulation of genes rarely expressed by microglial cells and abundant in monocytes, among which stands the membrane-bound scavenger receptor for haptoglobin/hemoglobin complexes or Cd163. These Cd163-positive MDC invaded the hippocampal parenchyma, acquired a microglial-like morphology, and were located in close proximity to blood vessels. Moreover, and most interesting, these invading monocytes infiltrated the nearby amyloid plaques contributing to plaque-associated myeloid cell heterogeneity. However, in aged-matched control individuals with hippocampal amyloid pathology, no signs of MDC brain infiltration or plaque invasion were found. The previously reported microglial degeneration/dysfunction in AD hippocampus could be a key pathological factor inducing MDC recruitment. Our data suggest a clear association between MDC infiltration and endothelial activation which in turn may contribute to damage of the blood brain barrier integrity. The recruitment of monocytes could be a consequence rather than the cause of the severity of the disease. Whether monocyte infiltration is beneficial or detrimental to AD pathology remains to be fully elucidated. These findings open the opportunity to design targeted therapies, not only for microglia but also for the peripheral immune cell population to modulate amyloid pathology and provide a better understanding of the immunological mechanisms underlying the progression of AD.
Collapse
Affiliation(s)
- Clara Muñoz-Castro
- Dpto. Bioquimica Y Biologia Molecular, Facultad de Farmacia, Universidad de Sevilla, C/ Prof. Garcia Gonzalez 2, 41012, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013, Seville, Spain
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Marina Mejias-Ortega
- Dpto. Biologia Celular, Genetica y Fisiologia, Instituto de Investigación Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Campus de Teatinos S/N, 29071, Malaga, Spain
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Elisabeth Sanchez-Mejias
- Dpto. Biologia Celular, Genetica y Fisiologia, Instituto de Investigación Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Campus de Teatinos S/N, 29071, Malaga, Spain
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Victoria Navarro
- Dpto. Bioquimica Y Biologia Molecular, Facultad de Farmacia, Universidad de Sevilla, C/ Prof. Garcia Gonzalez 2, 41012, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013, Seville, Spain
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Laura Trujillo-Estrada
- Dpto. Biologia Celular, Genetica y Fisiologia, Instituto de Investigación Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Campus de Teatinos S/N, 29071, Malaga, Spain
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Sebastian Jimenez
- Dpto. Bioquimica Y Biologia Molecular, Facultad de Farmacia, Universidad de Sevilla, C/ Prof. Garcia Gonzalez 2, 41012, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013, Seville, Spain
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Juan Antonio Garcia-Leon
- Dpto. Biologia Celular, Genetica y Fisiologia, Instituto de Investigación Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Campus de Teatinos S/N, 29071, Malaga, Spain
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Juan Jose Fernandez-Valenzuela
- Dpto. Biologia Celular, Genetica y Fisiologia, Instituto de Investigación Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Campus de Teatinos S/N, 29071, Malaga, Spain
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Maria Virtudes Sanchez-Mico
- Dpto. Bioquimica Y Biologia Molecular, Facultad de Farmacia, Universidad de Sevilla, C/ Prof. Garcia Gonzalez 2, 41012, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013, Seville, Spain
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Carmen Romero-Molina
- Dpto. Bioquimica Y Biologia Molecular, Facultad de Farmacia, Universidad de Sevilla, C/ Prof. Garcia Gonzalez 2, 41012, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013, Seville, Spain
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Ines Moreno-Gonzalez
- Dpto. Biologia Celular, Genetica y Fisiologia, Instituto de Investigación Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Campus de Teatinos S/N, 29071, Malaga, Spain
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - David Baglietto-Vargas
- Dpto. Biologia Celular, Genetica y Fisiologia, Instituto de Investigación Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Campus de Teatinos S/N, 29071, Malaga, Spain
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Marisa Vizuete
- Dpto. Bioquimica Y Biologia Molecular, Facultad de Farmacia, Universidad de Sevilla, C/ Prof. Garcia Gonzalez 2, 41012, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013, Seville, Spain
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain
| | - Antonia Gutierrez
- Dpto. Biologia Celular, Genetica y Fisiologia, Instituto de Investigación Biomedica de Malaga-IBIMA, Facultad de Ciencias, Universidad de Malaga, Campus de Teatinos S/N, 29071, Malaga, Spain.
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.
| | - Javier Vitorica
- Dpto. Bioquimica Y Biologia Molecular, Facultad de Farmacia, Universidad de Sevilla, C/ Prof. Garcia Gonzalez 2, 41012, Seville, Spain.
- Instituto de Biomedicina de Sevilla (IBiS)-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013, Seville, Spain.
- Centro de Investigacion Biomedica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031, Madrid, Spain.
| |
Collapse
|
50
|
Li JL, Fu GQ, Wang YY, Bian MM, Xu YM, Zhang L, Chen YQ, Zhang N, Ding SQ, Wang R, Fang R, Tang J, Hu JG, Lü HZ. The polarization of microglia and infiltrated macrophages in the injured mice spinal cords: a dynamic analysis. PeerJ 2023; 11:e14929. [PMID: 36846458 PMCID: PMC9951800 DOI: 10.7717/peerj.14929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 01/30/2023] [Indexed: 02/23/2023] Open
Abstract
Background Following spinal cord injury (SCI), a large number of peripheral monocytes infiltrate into the lesion area and differentiate into macrophages (Mø). These monocyte-derived Mø are very difficult to distinguish from the local activated microglia (MG). Therefore, the term Mø/MG are often used to define the infiltrated Mø and/or activated MG. It has been recognized that pro-inflammatory M1-type Mø/MG play "bad" roles in the SCI pathology. Our recent research showed that local M1 cells are mainly CD45-/lowCD68+CD11b+ in the subacute stage of SCI. Thus, we speculated that the M1 cells in injured spinal cords mainly derived from MG rather than infiltrating Mø. So far, their dynamics following SCI are not yet entirely clear. Methods Female C57BL/6 mice were used to establish SCI model, using an Infinite Horizon impactor with a 1.3 mm diameter rod and a 50 Kdynes force. Sham-operated (sham) mice only underwent laminectomy without contusion. Flow cytometry and immunohistofluorescence were combined to analyze the dynamic changes of polarized Mø and MG in the acute (1 day), subacute (3, 7 and 14 days) and chronic (21 and 28 days) phases of SCI. Results The total Mø/MG gradually increased and peaked at 7 days post-injury (dpi), and maintained at high levels 14, 21 and 28 dpi. Most of the Mø/MG were activated, and the Mø increased significantly at 1 and 3 dpi. However, with the pathological process, activated MG increased nearly to 90% at 7, 14, 21 and 28 dpi. Both M1 and M2 Mø were increased significantly at 1 and 3 dpi. However, they decreased to very low levels from 7 to 28 dpi. On the contrary, the M2-type MG decreased significantly following SCI and maintained at a low level during the pathological process.
Collapse
Affiliation(s)
- Jing-Lu Li
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China,Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Gui-Qiang Fu
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China,Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yang-Yang Wang
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China,Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Ming-Ming Bian
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China,Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yao-Mei Xu
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China,Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Lin Zhang
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China,Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Yu-Qing Chen
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China,Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Nan Zhang
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China,Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Shu-Qin Ding
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China,Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Rui Wang
- Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Rui Fang
- Department of Clinical Medical, Bengbu Medical College, Bengbu, China
| | - Jie Tang
- Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Jian-Guo Hu
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China,Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - He-Zuo Lü
- Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China,Anhui Key Laboratory of Tissue Transplantation, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| |
Collapse
|