1
|
Barros C, Alberro A, Fernandes A. Microglia and Immune cells interactions in multiple sclerosis cognitive impairment: a postmortem study. J Neuroinflammation 2024; 21:332. [PMID: 39741293 DOI: 10.1186/s12974-024-03326-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025] Open
Abstract
Multiple Sclerosis (MS), a neuroinflammatory disease of the central nervous system, is one of the commonest causes of non-traumatic disability among young adults. Impaired cognition arises as an impactful symptom affecting more than 50% of the patients and with substantial impact on social, economic, and individual wellbeing. Despite the lack of therapeutic strategies, many efforts have been made to understand the mechanisms behind cognitive impairment in MS patients. Here, we aimed to investigate whether microglia-derived synaptic elimination and immune interactions are exacerbated in MS patients with impaired cognition when compared to non-demented controls (NDC) and cognitively preserved MS patients, that may clarify the role of immune cell interplay in MS cognitive deficits. Postmortem hippocampal samples were obtained from NDCs and MS patients. Sixteen MS patients were categorized based on their cognitive status: preserved cognition (MSCP) and impaired cognition (MSCI). Immunohistochemistry studies were conducted to explore the density of microglia, their role in synaptic engulfment, and their interaction with CD8+ immune cells in the context of cognitive impairment in MS. In high synaptic density hippocampal regions, MSCI patients exhibited a massive presence of microglia cells actively engulfing both excitatory and inhibitory synapses, accompanied by morphological alterations. Additionally, there was an increased expression of the complement protein C1q particularly localized at inhibitory synapses within microglia cells, suggesting a preferential engulfment of complement-tagged inhibitory synapses in MSCI patients. Furthermore, in hippocampal lesions of MSCI patients, we detected a significant infiltration of microglia and CD8 T cells that may be contributing to the smouldering MS and cognitive deterioration. These findings demonstrate that cognitive deficits occurring in MS are associated with microglia engulfment of C1q-tagged inhibitory synapses, which may be driven by direct or indirect stimulation from CD8+ T cells.
Collapse
Affiliation(s)
- Catarina Barros
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal
| | - Ainhoa Alberro
- IIS Biogipuzkoa Health Research Institute, San Sebastian, Spain
| | - Adelaide Fernandes
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal.
- Department of Pharmaceutical Sciences and Medicines, Faculdade de Farmácia, Universidade de Lisboa, Avenida Professor Gama Pinto, Lisboa, 1600-083, Portugal.
| |
Collapse
|
2
|
Kalaitzidis G, Ezzedin O, Bacchetti A, Moussa H, Murphy OC, Filippatou AG, Ehrhardt H, Vasileiou E, Pellegrini N, Davis S, Douglas M, Fitzgerald KC, DuVal A, Douglas Newsome S, Sotirchos ES, Nourbakhsh B, Dewey BE, Prince J, Saidha S, Calabresi PA. Homonymous hemi-macular atrophy in multiple sclerosis. Mult Scler 2024; 30:1802-1814. [PMID: 39579046 PMCID: PMC11617262 DOI: 10.1177/13524585241297816] [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: 11/25/2024]
Abstract
BACKGROUND Retrograde trans-synaptic degeneration (TSD) following retro-chiasmal pathology, typically retro-geniculate in multiple sclerosis (MS), may manifest as homonymous hemi-macular atrophy (HHMA) of the ganglion cell/inner plexiform layer (GCIPL). OBJECTIVE To determine the frequency, association with clinical outcomes, and retinal and radiological features of HHMA in people with MS (PwMS). METHODS In this cross-sectional study, healthy controls (HC) and PwMS underwent retinal optical coherence tomography scanning. For quantitative identification of HHMA, a normalized asymmetry ratio was used, and its normative cutoffs were established from the HC. HHMA PwMS were propensity score matched 1:2 to non-HHMA PwMS. Mixed-effects linear regression models were used in analyses. RESULTS Based on normative data from 238 HC (466 eyes), 79 out of 942 PwMS exhibited HHMA (8.4%; 143 eyes). Compared to non-HHMA eyes from matched PwMS (158 PwMS; 308 eyes), HHMA eyes had lower average GCIPL (diff: -5.7 μm (95% CI -7.6 to -3.8); p < 0.001) but also inner nuclear layer (diff: -0.9 μm (95% CI -1.6 to -0.1); p = 0.02), and outer nuclear layer (diff: -1.9 μm (95% CI -3.4 to -0.4); p = 0.02) thicknesses; in further analyses, these differences were exclusive to the homonymous side of HHMA. HHMA participants also exhibited higher expanded disability status scale scores (diff: 0.5 (95% CI 0.1 to 0.9); p = 0.02), worse 100% and 2.5% visual acuity scores (diff: -3.2 (95% CI -4.1 to -1.0); p = 0.002, -5.4 (95% CI -7.5 to -3.5); p < 0.001), and higher frequency of microcystoid macular changes (10.1% vs. 3.2%; p = 0.03) compared to non-HHMA participants. CONCLUSIONS HHMA, possibly as a marker of TSD, may signify higher disability in MS.
Collapse
Affiliation(s)
- Grigorios Kalaitzidis
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Omar Ezzedin
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Anna Bacchetti
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Hussein Moussa
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Olwen C Murphy
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Angeliki G Filippatou
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Henrik Ehrhardt
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Eleni Vasileiou
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Nicole Pellegrini
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Simidele Davis
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Morgan Douglas
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Kathryn C Fitzgerald
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Anna DuVal
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Scott Douglas Newsome
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Elias S Sotirchos
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Bardia Nourbakhsh
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Blake E Dewey
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Jerry Prince
- Department of Electrical and Computer Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Shiv Saidha
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| | - Peter A Calabresi
- Division of Neuroimmunology and Neurological Infections, Department of Neurology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
3
|
Vander Wall R, Basavarajappa D, Palanivel V, Sharma S, Gupta V, Klistoner A, Graham S, You Y. VEP Latency Delay Reflects Demyelination Beyond the Optic Nerve in the Cuprizone Model. Invest Ophthalmol Vis Sci 2024; 65:50. [PMID: 39576623 PMCID: PMC11587907 DOI: 10.1167/iovs.65.13.50] [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: 06/20/2024] [Accepted: 09/24/2024] [Indexed: 11/24/2024] Open
Abstract
Purpose Remyelination therapies are advancing for multiple sclerosis, focusing on visual pathways and using visual evoked potentials (VEPs) for de/remyelination processes. While the cuprizone (CZ) model and VEPs are core tools in preclinical trials, many overlook the posterior visual pathway. This study aimed to assess functional and structural changes across the murine visual pathway during de/remyelination. Methods One group of C57BL/6 mice underwent a CZ diet for 6 weeks to simulate demyelination, with a subset returning to a regular diet to induce remyelination. An additional group was fed a protracted CZ diet for 12 weeks to maintain chronic demyelination. Visual function was evaluated using electrophysiological recordings, including scotopic threshold responses (STRs) and electroretinograms (ERGs), with VEPs serving as a key biomarker for overall pathway health. Tissues from eyes, brains, and optic nerves (ONs) were collected at different time points for structural analysis. Results Our results demonstrated significant effects on VEPs, including increased N1 latencies and reduced amplitudes in the CZ mouse model. However, retinal function remained unaffected, as evidenced by unchanged STRs, ERGs, and retinal ganglion cell counts. Analysis of ONs revealed morphological changes, characterized by a significantly decreased axon diameter in the core region compared to the subpial region. Additionally, there was a significant increase in the g-ratio of the core region at 12 weeks CZ compared to controls. Immunofluorescence further demonstrated a decrease in myelin basic protein levels at 6 and 12 weeks in CZ animals. Interestingly, the dorsal lateral geniculate nucleus and primary visual cortex (V1) exhibited similar myelin changes, correlating with VEP latency alterations. Conclusions These data reveal that interpreting VEP latency solely as a marker for ON demyelination is incomplete. Previous preclinical studies have overlooked the posterior visual pathways, necessitating a broader interpretation of VEP latency to cover the entire visual pathway.
Collapse
Affiliation(s)
- Roshana Vander Wall
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Devaraj Basavarajappa
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Viswanthram Palanivel
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Samridhi Sharma
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Vivek Gupta
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Alexander Klistoner
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Stuart Graham
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, Australia
- Save Sight Institute, Sydney University, Sydney, NSW, Australia
| | - Yuyi You
- Faculty of Medicine Health and Human Sciences, Macquarie University, North Ryde, NSW, Australia
- Save Sight Institute, Sydney University, Sydney, NSW, Australia
| |
Collapse
|
4
|
Kim NH, Choo HI, Lee YA. Effect of nanoplastic intake on the dopamine system during the development of male mice. Neuroscience 2024; 555:11-22. [PMID: 39033990 DOI: 10.1016/j.neuroscience.2024.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/23/2024]
Abstract
Exposure to environmental microplastics has been demonstrated to impact health. However, its effect on development remains unclear. This study investigated whether consumption of nanoplastics (NPx) during development affects social and cognitive functions in rodents. In this study, we utilized male Institute of Cancer Research mice; they were divided into five subgroups based on the duration of NPx administration. NPx (100 nm) was orally administered via gavage for 6 days from gestational day (GTD) 7, representing the mid-gestation period, and for 5-6 days from GTD13 to birth, representing the late-gestation period; the male offspring were used for experiments. NPx was orally administered for 15 days starting at postnatal day (PND) 21 as the juvenile, PND38 as the adolescent, and PND56 as adulthood. On PND77, offspring were assessed for locomotion, social behavior, and nest-building tests. We observed that NPx administration altered dopamine system responses in GTD13 and PND56 groups. Social behavior was similarly affected by NPx treatment, with GTD13 and PND56 groups displaying decreased familiarity. Additionally, NPx treatment enhanced local field potentials in the prefrontal cortex, nucleus accumbens, and amygdala of GTD7 group and in the striatum of GTD13 group, while amphetamine treatment induced changes of local field potentials compared to saline treatment in the prefrontal cortex and the ventral tegmental area of CTR, GTD7, PND21, and PND56 groups. Taken together, these results showed that NPx treatment induced changes in social behavior partly depending on developmental stage, and these changes are associated with neural circuits innervated by the dopamine system.
Collapse
Affiliation(s)
- Na-Hyun Kim
- Department of Food Science and Nutrition, Daegu Catholic University, 13-13 Hayang-Ro, Hayang-Eup, Gyeongsan, Gyeongbuk 38430, South Korea
| | - Hye-In Choo
- Department of Food Science and Nutrition, Daegu Catholic University, 13-13 Hayang-Ro, Hayang-Eup, Gyeongsan, Gyeongbuk 38430, South Korea
| | - Young-A Lee
- Department of Food Science and Nutrition, Daegu Catholic University, 13-13 Hayang-Ro, Hayang-Eup, Gyeongsan, Gyeongbuk 38430, South Korea.
| |
Collapse
|
5
|
Shen FS, Liu C, Sun HZ, Chen XY, Xue Y, Chen L. Emerging evidence of context-dependent synapse elimination by phagocytes in the CNS. J Leukoc Biol 2024; 116:511-522. [PMID: 38700080 DOI: 10.1093/jleuko/qiae098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/09/2024] [Accepted: 04/09/2024] [Indexed: 05/05/2024] Open
Abstract
Precise synapse elimination is essential for the establishment of a fully developed neural circuit during brain development and higher function in adult brain. Beyond immune and nutrition support, recent groundbreaking studies have revealed that phagocytic microglia and astrocytes can actively and selectively eliminate synapses in normal and diseased brains, thereby mediating synapse loss and maintaining circuit homeostasis. Multiple lines of evidence indicate that the mechanisms of synapse elimination by phagocytic glia are not universal but rather depend on specific contexts and detailed neuron-glia interactions. The mechanism of synapse elimination by phagocytic glia is dependent on neuron-intrinsic factors and many innate immune and local apoptosis-related molecules. During development, microglial synapse engulfment in the visual thalamus is primarily influenced by the classic complement pathway, whereas in the barrel cortex, the fractalkine pathway is dominant. In Alzheimer's disease, microglia employ complement-dependent mechanisms for synapse engulfment in tauopathy and early β-amyloid pathology, but microglia are not involved in synapse loss at late β-amyloid stages. Phagocytic microglia also engulf synapses in a complement-dependent way in schizophrenia, anxiety, and stress. In addition, phagocytic astrocytes engulf synapses in a MEGF10-dependent way during visual development, memory, and stroke. Furthermore, the mechanism of a phenomenon that phagocytes selectively eliminate excitatory and inhibitory synapses is also emphasized in this review. We hypothesize that elucidating context-dependent synapse elimination by phagocytic microglia and astrocytes may reveal the molecular basis of synapse loss in neural disorders and provide a rationale for developing novel candidate therapies that target synapse loss and circuit homeostasis.
Collapse
Affiliation(s)
- Fang-Shuai Shen
- Department of Physiology and Pathophysiology, School of Basic Medicine, No. 308 Ningxia Road, Shinan District, Qingdao University 266071, Qingdao, China
| | - Cui Liu
- Department of Physiology and Pathophysiology, School of Basic Medicine, No. 308 Ningxia Road, Shinan District, Qingdao University 266071, Qingdao, China
| | - Hui-Zhe Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, No. 308 Ningxia Road, Shinan District, Qingdao University 266071, Qingdao, China
| | - Xin-Yi Chen
- Department of International Medicine, No. 16 Jiangsu Road, Shinan District, Affiliated Hospital of Qingdao University 266000, Qingdao, China
| | - Yan Xue
- Department of Physiology and Pathophysiology, School of Basic Medicine, No. 308 Ningxia Road, Shinan District, Qingdao University 266071, Qingdao, China
| | - Lei Chen
- Department of Physiology and Pathophysiology, School of Basic Medicine, No. 308 Ningxia Road, Shinan District, Qingdao University 266071, Qingdao, China
| |
Collapse
|
6
|
Negro-Demontel L, Maleki AF, Reich DS, Kemper C. The complement system in neurodegenerative and inflammatory diseases of the central nervous system. Front Neurol 2024; 15:1396520. [PMID: 39022733 PMCID: PMC11252048 DOI: 10.3389/fneur.2024.1396520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Neurodegenerative and neuroinflammatory diseases, including Alzheimer's disease, Parkinson's disease, and multiple sclerosis, affect millions of people globally. As aging is a major risk factor for neurodegenerative diseases, the continuous increase in the elderly population across Western societies is also associated with a rising prevalence of these debilitating conditions. The complement system, a crucial component of the innate immune response, has gained increasing attention for its multifaceted involvement in the normal development of the central nervous system (CNS) and the brain but also as a pathogenic driver in several neuroinflammatory disease states. Although complement is generally understood as a liver-derived and blood or interstitial fluid operative system protecting against bloodborne pathogens or threats, recent research, particularly on the role of complement in the healthy and diseased CNS, has demonstrated the importance of locally produced and activated complement components. Here, we provide a succinct overview over the known beneficial and pathological roles of complement in the CNS with focus on local sources of complement, including a discussion on the potential importance of the recently discovered intracellularly active complement system for CNS biology and on infection-triggered neurodegeneration.
Collapse
Affiliation(s)
- Luciana Negro-Demontel
- National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Complement and Inflammation Research Section (CIRS), Bethesda, MD, United States
- Department of Histology and Embryology, Faculty of Medicine, UDELAR, Montevideo, Uruguay
- Neuroinflammation and Gene Therapy Laboratory, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Adam F. Maleki
- National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Complement and Inflammation Research Section (CIRS), Bethesda, MD, United States
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD, United States
| | - Daniel S. Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke (NINDS), NIH, Bethesda, MD, United States
| | - Claudia Kemper
- National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Complement and Inflammation Research Section (CIRS), Bethesda, MD, United States
| |
Collapse
|
7
|
Calabrese M, Preziosa P, Scalfari A, Colato E, Marastoni D, Absinta M, Battaglini M, De Stefano N, Di Filippo M, Hametner S, Howell OW, Inglese M, Lassmann H, Martin R, Nicholas R, Reynolds R, Rocca MA, Tamanti A, Vercellino M, Villar LM, Filippi M, Magliozzi R. Determinants and Biomarkers of Progression Independent of Relapses in Multiple Sclerosis. Ann Neurol 2024; 96:1-20. [PMID: 38568026 DOI: 10.1002/ana.26913] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/04/2024] [Accepted: 02/15/2024] [Indexed: 06/20/2024]
Abstract
Clinical, pathological, and imaging evidence in multiple sclerosis (MS) suggests that a smoldering inflammatory activity is present from the earliest stages of the disease and underlies the progression of disability, which proceeds relentlessly and independently of clinical and radiological relapses (PIRA). The complex system of pathological events driving "chronic" worsening is likely linked with the early accumulation of compartmentalized inflammation within the central nervous system as well as insufficient repair phenomena and mitochondrial failure. These mechanisms are partially lesion-independent and differ from those causing clinical relapses and the formation of new focal demyelinating lesions; they lead to neuroaxonal dysfunction and death, myelin loss, glia alterations, and finally, a neuronal network dysfunction outweighing central nervous system (CNS) compensatory mechanisms. This review aims to provide an overview of the state of the art of neuropathological, immunological, and imaging knowledge about the mechanisms underlying the smoldering disease activity, focusing on possible early biomarkers and their translation into clinical practice. ANN NEUROL 2024;96:1-20.
Collapse
Affiliation(s)
- Massimiliano Calabrese
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Paolo Preziosa
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Antonio Scalfari
- Centre of Neuroscience, Department of Medicine, Imperial College, London, UK
| | - Elisa Colato
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Damiano Marastoni
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Martina Absinta
- Translational Neuropathology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Battaglini
- Siena Imaging S.r.l., Siena, Italy
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Massimiliano Di Filippo
- Section of Neurology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Simon Hametner
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Owain W Howell
- Institute of Life Sciences, Swansea University Medical School, Swansea, UK
| | - Matilde Inglese
- Dipartimento di neuroscienze, riabilitazione, oftalmologia, genetica e scienze materno-infantili - DINOGMI, University of Genova, Genoa, Italy
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Roland Martin
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
- Therapeutic Design Unit, Center for Molecular Medicine, Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden
- Cellerys AG, Schlieren, Switzerland
| | - Richard Nicholas
- Department of Brain Sciences, Faculty of Medicine, Burlington Danes, Imperial College London, London, UK
| | - Richard Reynolds
- Division of Neuroscience, Department of Brain Sciences, Imperial College London, London, UK
| | - Maria A Rocca
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Agnese Tamanti
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| | - Marco Vercellino
- Multiple Sclerosis Center & Neurologia I U, Department of Neuroscience, University Hospital AOU Città della Salute e della Scienza di Torino, Turin, Italy
| | - Luisa Maria Villar
- Department of Immunology, Ramon y Cajal University Hospital. IRYCIS. REI, Madrid, Spain
| | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Roberta Magliozzi
- Department of Neurosciences and Biomedicine and Movement, The Multiple Sclerosis Center of University Hospital of Verona, Verona, Italy
| |
Collapse
|
8
|
Zhang L, Yao Q, Hu J, Qiu B, Xiao Y, Zhang Q, Zeng Y, Zheng S, Zhang Y, Wan Y, Zheng X, Zeng Q. Hotspots and trends of microglia in Alzheimer's disease: a bibliometric analysis during 2000-2022. Eur J Med Res 2024; 29:75. [PMID: 38268044 PMCID: PMC10807212 DOI: 10.1186/s40001-023-01602-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 12/17/2023] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Alzheimer's disease is one common type of dementia. Numerous studies have suggested a correlation between Alzheimer's disease and inflammation. Microglia mainly participate in the inflammatory response in the brain. Currently, ample evidence has shown that microglia are closely related to the occurrence and development of Alzheimer's disease. OBJECTIVE We opted for bibliometric analysis to comprehensively summarize the advancements in the study of microglia in Alzheimer's disease, aiming to provide researchers with current trends and future research directions. METHODS All articles and reviews pertaining to microglia in Alzheimer's disease from 2000 to 2022 were downloaded through Web of Science Core Collection. The results were subjected to bibliometric analysis using VOSviewer 1.6.18 and CiteSpace 6.1 R2. RESULTS Overall, 7449 publications were included. The number of publications was increasing yearly. The United States has published the most publications. Harvard Medical School has published the most papers of all institutions. Journal of Alzheimer's Disease and Journal of Neuroscience were the journals with the most studies and the most commonly cited, respectively. Mt Heneka is the author with the highest productivity and co-citation. After analysis, the most common keywords are neuroinflammation, amyloid-beta, inflammation, neurodegeneration. Gut microbiota, extracellular vesicle, dysfunction and meta-analysis are the hotspots of research at the present stage and are likely to continue. CONCLUSION NLRP3 inflammasome, TREM2, gut microbiota, mitochondrial dysfunction, exosomes are research hotspots. The relationship between microglia-mediated neuroinflammation and Alzheimer's disease have been the focus of current research and the development trend of future research.
Collapse
Affiliation(s)
- Lijie Zhang
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Qiuru Yao
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Nursing, Southern Medical University, Guangzhou, China
| | - Jinjing Hu
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Baizhi Qiu
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Nursing, Southern Medical University, Guangzhou, China
| | - Yupeng Xiao
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Qi Zhang
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Yuting Zeng
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shuqi Zheng
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China
| | - Youao Zhang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yantong Wan
- College of Anesthesiology, Southern Medical University, Guangzhou, China.
| | - Xiaoyan Zheng
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China.
| | - Qing Zeng
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
- School of Rehabilitation Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
9
|
Gillani RL, Kironde EN, Whiteman S, Zwang TJ, Bacskai BJ. Instability of excitatory synapses in experimental autoimmune encephalomyelitis and the outcome for excitatory circuit inputs to individual cortical neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.23.576662. [PMID: 38328177 PMCID: PMC10849614 DOI: 10.1101/2024.01.23.576662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Synapses are lost on a massive scale in the brain and spinal cord of people living with multiple sclerosis (PwMS), and this synaptic loss extends far beyond demyelinating lesions. Post-mortem studies show the long-term consequences of multiple sclerosis (MS) on synapses but do not inform on the early impacts of neuroinflammation on synapses that subsequently lead to synapse loss. How excitatory circuit inputs are altered across the dendritic tree of individual neurons under neuroinflammatory stress is not well understood. Here, we directly assessed the structural dynamics of labeled excitatory synapses in experimental autoimmune encephalomyelitis (EAE) as a model of immune-mediated cortical neuronal damage. We used in vivo two-photon imaging and a synthetic tissue-hydrogel super-resolution imaging technique to reveal the dynamics of excitatory synapses, map their location across the dendritic tree of individual neurons, and examine neurons at super-resolution for synaptic loss. We found that excitatory synapses are destabilized but not lost from dendritic spines in EAE, starting with the earliest imaging session before symptom onset. This led to dramatic changes in excitatory circuit inputs to individual cells. In EAE, stable synapses are replaced by synapses that appear or disappear across the imaging sessions or repeatedly change at the same location. These unstable excitatory inputs occur closer to one another in EAE than in healthy controls and are distributed across the dendritic tree. When imaged at super-resolution, we found that a small proportion of dendritic protrusions lost their presynapse and/or postsynapse. Our finding of diffuse destabilizing effects of neuroinflammation on excitatory synapses across cortical neurons may have significant functional consequences since normal dendritic spine dynamics and clustering are essential for learning and memory.
Collapse
|
10
|
Scheepstra KWF, Mizee MR, van Scheppingen J, Adelia A, Wever DD, Mason MRJ, Dubbelaar ML, Hsiao CC, Eggen BJL, Hamann J, Huitinga I. Microglia Transcriptional Profiling in Major Depressive Disorder Shows Inhibition of Cortical Gray Matter Microglia. Biol Psychiatry 2023; 94:619-629. [PMID: 37121366 DOI: 10.1016/j.biopsych.2023.04.020] [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: 02/15/2022] [Revised: 02/01/2023] [Accepted: 04/17/2023] [Indexed: 05/02/2023]
Abstract
BACKGROUND Microglia have been implicated in the pathophysiology of major depressive disorder (MDD), but information on biological mechanisms is limited. Therefore, we investigated the gene expression profile of microglial cells in relation to neuronal regulators of microglia activity in well-characterized MDD and control autopsy brains. METHODS Pure, intact microglia were isolated at brain autopsy from occipital cortex gray matter (GM) and corpus callosum white matter of 13 donors with MDD and 10 age-matched control donors for RNA sequencing. Top differentially expressed genes were validated using immunohistochemistry staining. Because gene expression changes were only detected in GM microglia, neuronal regulators of microglia were investigated in cortical tissue and synaptosomes from the cortex by reverse transcriptase-quantitative polymerase chain reaction and Western blot. RESULTS Transcriptome analysis revealed 92 genes differentially expressed in microglia isolated from GM, but none in microglia from white matter in donors with MDD, compared with control donors. Of these, 81 genes were less abundantly expressed in GM in MDD, including CD163, MKI67, SPP1, CD14, FCGR1A/C, and C1QA/B/C. Accordingly, pathways related to effector mechanisms, such as the complement system and phagocytosis, were differentially regulated in GM microglia in MDD. Immunohistochemistry staining revealed significantly lower expression of CD163 protein in MDD. Whole tissue analysis showed an increase in CD200 (p = .0009) and CD47 (p = .068) messenger RNA, and CD47 protein was significantly elevated (p = .0396) in synaptic fractions of MDD cases. CONCLUSIONS Transcriptional profiling indicates an immune-suppressed microglial phenotype in MDD that is possibly caused by neuronal regulation.
Collapse
Affiliation(s)
- Karel W F Scheepstra
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands; Psychiatric Program of the Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands; Department of Psychiatry, Amsterdam University Medical Centers, Amsterdam Neuroscience, Amsterdam, the Netherlands.
| | - Mark R Mizee
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands; Psychiatric Program of the Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Jackelien van Scheppingen
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands; Psychiatric Program of the Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Adelia Adelia
- Psychiatric Program of the Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Dennis D Wever
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands; Psychiatric Program of the Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Matthew R J Mason
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Marissa L Dubbelaar
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, Groningen, the Netherlands
| | - Cheng-Chih Hsiao
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Bart J L Eggen
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, Groningen, the Netherlands
| | - Jörg Hamann
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Inge Huitinga
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands; Psychiatric Program of the Netherlands Brain Bank, Netherlands Institute for Neuroscience, Amsterdam, the Netherlands; Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
11
|
Fan J, Dong X, Tang Y, Wang X, Lin D, Gong L, Chen C, Jiang J, Shen W, Xu A, Zhang X, Xie Y, Huang X, Zeng L. Preferential pruning of inhibitory synapses by microglia contributes to alteration of the balance between excitatory and inhibitory synapses in the hippocampus in temporal lobe epilepsy. CNS Neurosci Ther 2023; 29:2884-2900. [PMID: 37072932 PMCID: PMC10493672 DOI: 10.1111/cns.14224] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 03/31/2023] [Accepted: 04/05/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND A consensus has formed that neural circuits in the brain underlie the pathogenesis of temporal lobe epilepsy (TLE). In particular, the synaptic excitation/inhibition balance (E/I balance) has been implicated in shifting towards elevated excitation during the development of TLE. METHODS Sprague Dawley (SD) rats were intraperitoneally subjected to kainic acid (KA) to generate a model of TLE. Next, electroencephalography (EEG) recording was applied to verify the stability and detectability of spontaneous recurrent seizures (SRS) in rats. Moreover, hippocampal slices from rats and patients with mesial temporal lobe epilepsy (mTLE) were assessed using immunofluorescence to determine the alterations of excitatory and inhibitory synapses and microglial phagocytosis. RESULTS We found that KA induced stable SRSs 14 days after status epilepticus (SE) onset. Furthermore, we discovered a continuous increase in excitatory synapses during epileptogenesis, where the total area of vesicular glutamate transporter 1 (vGluT1) rose considerably in the stratum radiatum (SR) of cornu ammonis 1 (CA1), the stratum lucidum (SL) of CA3, and the polymorphic layer (PML) of the dentate gyrus (DG). In contrast, inhibitory synapses decreased significantly, with the total area of glutamate decarboxylase 65 (GAD65) in the SL and PML diminishing enormously. Moreover, microglia conducted active synaptic phagocytosis after the formation of SRSs, especially in the SL and PML. Finally, microglia preferentially pruned inhibitory synapses during recurrent seizures in both rat and human hippocampal slices, which contributed to the synaptic alteration in hippocampal subregions. CONCLUSIONS Our findings elaborately characterize the alteration of neural circuits and demonstrate the selectivity of synaptic phagocytosis mediated by microglia in TLE, which could strengthen the comprehension of the pathogenesis of TLE and inspire potential therapeutic targets for epilepsy treatment.
Collapse
Affiliation(s)
- Jianchen Fan
- College of Pharmaceutical Sciences, Institute of Pharmacology and Toxicology, Key Laboratory of Medical Neurobiology of the Ministry of Health of ChinaZhejiang UniversityHangzhouChina
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of MedicineHangzhou City UniversityHangzhouChina
| | - Xinyan Dong
- Department of NeurologyThe Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child HealthHangzhouChina
| | - Yejiao Tang
- College of Pharmaceutical Sciences, Institute of Pharmacology and Toxicology, Key Laboratory of Medical Neurobiology of the Ministry of Health of ChinaZhejiang UniversityHangzhouChina
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of MedicineHangzhou City UniversityHangzhouChina
| | - Xuehui Wang
- Department of NeurologyThe Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child HealthHangzhouChina
| | - Donghui Lin
- Department of NeurologyThe Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child HealthHangzhouChina
| | - Lifen Gong
- Department of NeurologyThe Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child HealthHangzhouChina
| | - Chen Chen
- Department of NeurologyThe Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child HealthHangzhouChina
| | - Jie Jiang
- Department of NeurologyThe Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child HealthHangzhouChina
| | - Weida Shen
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of MedicineHangzhou City UniversityHangzhouChina
| | - Anyu Xu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of MedicineHangzhou City UniversityHangzhouChina
| | - Xiangnan Zhang
- College of Pharmaceutical Sciences, Institute of Pharmacology and Toxicology, Key Laboratory of Medical Neurobiology of the Ministry of Health of ChinaZhejiang UniversityHangzhouChina
| | - Yicheng Xie
- Department of NeurologyThe Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center For Child HealthHangzhouChina
| | - Xin Huang
- Department of NeurosurgeryThe First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Linghui Zeng
- College of Pharmaceutical Sciences, Institute of Pharmacology and Toxicology, Key Laboratory of Medical Neurobiology of the Ministry of Health of ChinaZhejiang UniversityHangzhouChina
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of MedicineHangzhou City UniversityHangzhouChina
| |
Collapse
|
12
|
Michailidou I, Fluiter K, Boziki M, Grigoriadis N, Baas F. Editorial: Complement in nervous system disease. Front Cell Neurosci 2023; 17:1268023. [PMID: 37614913 PMCID: PMC10442514 DOI: 10.3389/fncel.2023.1268023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/25/2023] Open
Affiliation(s)
- Iliana Michailidou
- Laboratory of Experimental Neurology and Neuroimmunology and the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Kees Fluiter
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology and the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology and the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Frank Baas
- Department of Clinical Genetics, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
13
|
Magliozzi R, Howell OW, Calabrese M, Reynolds R. Meningeal inflammation as a driver of cortical grey matter pathology and clinical progression in multiple sclerosis. Nat Rev Neurol 2023:10.1038/s41582-023-00838-7. [PMID: 37400550 DOI: 10.1038/s41582-023-00838-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2023] [Indexed: 07/05/2023]
Abstract
Growing evidence from cerebrospinal fluid samples and post-mortem brain tissue from individuals with multiple sclerosis (MS) and rodent models indicates that the meninges have a key role in the inflammatory and neurodegenerative mechanisms underlying progressive MS pathology. The subarachnoid space and associated perivascular spaces between the membranes of the meninges are the access points for entry of lymphocytes, monocytes and macrophages into the brain parenchyma, and the main route for diffusion of inflammatory and cytotoxic molecules from the cerebrospinal fluid into the brain tissue. In addition, the meningeal spaces act as an exit route for CNS-derived antigens, immune cells and metabolites. A number of studies have demonstrated an association between chronic meningeal inflammation and a more severe clinical course of MS, suggesting that the build-up of immune cell aggregates in the meninges represents a rational target for therapeutic intervention. Therefore, understanding the precise cell and molecular mechanisms, timing and anatomical features involved in the compartmentalization of inflammation within the meningeal spaces in MS is vital. Here, we present a detailed review and discussion of the cellular, molecular and radiological evidence for a role of meningeal inflammation in MS, alongside the clinical and therapeutic implications.
Collapse
Affiliation(s)
- Roberta Magliozzi
- Neurology Section of Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy.
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK.
| | - Owain W Howell
- Neurology Section of Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
- Institute of Life Sciences, Swansea University, Swansea, UK
| | - Massimiliano Calabrese
- Neurology Section of Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Richard Reynolds
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
- Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
14
|
Distéfano-Gagné F, Bitarafan S, Lacroix S, Gosselin D. Roles and regulation of microglia activity in multiple sclerosis: insights from animal models. Nat Rev Neurosci 2023:10.1038/s41583-023-00709-6. [PMID: 37268822 DOI: 10.1038/s41583-023-00709-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2023] [Indexed: 06/04/2023]
Abstract
As resident macrophages of the CNS, microglia are critical immune effectors of inflammatory lesions and associated neural dysfunctions. In multiple sclerosis (MS) and its animal models, chronic microglial inflammatory activity damages myelin and disrupts axonal and synaptic activity. In contrast to these detrimental effects, the potent phagocytic and tissue-remodelling capabilities of microglia support critical endogenous repair mechanisms. Although these opposing capabilities have long been appreciated, a precise understanding of their underlying molecular effectors is only beginning to emerge. Here, we review recent advances in our understanding of the roles of microglia in animal models of MS and demyelinating lesions and the mechanisms that underlie their damaging and repairing activities. We also discuss how the structured organization and regulation of the genome enables complex transcriptional heterogeneity within the microglial cell population at demyelinating lesions.
Collapse
Affiliation(s)
- Félix Distéfano-Gagné
- Axe Neuroscience, Centre de Recherche du CHU de Québec - Université Laval, Québec, Québec, Canada
- Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Québec, Québec, Canada
| | - Sara Bitarafan
- Axe Neuroscience, Centre de Recherche du CHU de Québec - Université Laval, Québec, Québec, Canada
- Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Québec, Québec, Canada
| | - Steve Lacroix
- Axe Neuroscience, Centre de Recherche du CHU de Québec - Université Laval, Québec, Québec, Canada
- Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Québec, Québec, Canada
| | - David Gosselin
- Axe Neuroscience, Centre de Recherche du CHU de Québec - Université Laval, Québec, Québec, Canada.
- Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Québec, Québec, Canada.
| |
Collapse
|
15
|
Wen L, Yang X, Wu Z, Fu S, Zhan Y, Chen Z, Bi D, Shen Y. The complement inhibitor CD59 is required for GABAergic synaptic transmission in the dentate gyrus. Cell Rep 2023; 42:112349. [PMID: 37027303 DOI: 10.1016/j.celrep.2023.112349] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 01/31/2023] [Accepted: 03/21/2023] [Indexed: 04/08/2023] Open
Abstract
Complement-dependent microglia pruning of excitatory synapses has been widely reported in physiological and pathological conditions, with few reports concerning pruning of inhibitory synapses or direct regulation of synaptic transmission by complement components. Here, we report that loss of CD59, an important endogenous inhibitor of the complement system, leads to compromised spatial memory performance. Furthermore, CD59 deficiency impairs GABAergic synaptic transmission in the hippocampal dentate gyrus (DG). This depends on regulation of GABA release triggered by Ca2+ influx through voltage-gated calcium channels (VGCCs) rather than inhibitory synaptic pruning by microglia. Notably, CD59 colocalizes with inhibitory pre-synaptic terminals and regulates SNARE complex assembly. Together, these results demonstrate that the complement regulator CD59 plays an important role in normal hippocampal function.
Collapse
Affiliation(s)
- Lang Wen
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disease Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Xiaoli Yang
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disease Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Zujun Wu
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disease Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Shumei Fu
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disease Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Yaxi Zhan
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disease Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
| | - Zuolong Chen
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disease Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215000, China
| | - Danlei Bi
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disease Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei 230026, China; Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei 230026, China; CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230026, China; Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Yong Shen
- Department of Neurology and Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Neurodegenerative Disease Research Center, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China; Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230026, China; Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
16
|
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
|
17
|
Huiskamp M, Yaqub M, van Lingen MR, Pouwels PJW, de Ruiter LRJ, Killestein J, Schwarte LA, Golla SSV, van Berckel BNM, Boellaard R, Geurts JJG, Hulst HE. Cognitive performance in multiple sclerosis: what is the role of the gamma-aminobutyric acid system? Brain Commun 2023; 5:fcad140. [PMID: 37180993 PMCID: PMC10174207 DOI: 10.1093/braincomms/fcad140] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 01/26/2023] [Accepted: 04/28/2023] [Indexed: 05/16/2023] Open
Abstract
Cognitive impairment occurs in 40-65% of persons with multiple sclerosis and may be related to alterations in glutamatergic and GABAergic neurotransmission. Therefore, the aim of this study was to determine how glutamatergic and GABAergic changes relate to cognitive functioning in multiple sclerosis in vivo. Sixty persons with multiple sclerosis (mean age 45.5 ± 9.6 years, 48 females, 51 relapsing-remitting multiple sclerosis) and 22 age-matched healthy controls (45.6 ± 22.0 years, 17 females) underwent neuropsychological testing and MRI. Persons with multiple sclerosis were classified as cognitively impaired when scoring at least 1.5 standard deviations below normative scores on ≥30% of tests. Glutamate and GABA concentrations were determined in the right hippocampus and bilateral thalamus using magnetic resonance spectroscopy. GABA-receptor density was assessed using quantitative [11C]flumazenil positron emission tomography in a subset of participants. Positron emission tomography outcome measures were the influx rate constant (a measure predominantly reflecting perfusion) and volume of distribution, which is a measure of GABA-receptor density. Twenty persons with multiple sclerosis (33%) fulfilled the criteria for cognitive impairment. No differences were observed in glutamate or GABA concentrations between persons with multiple sclerosis and healthy controls, or between cognitively preserved, impaired and healthy control groups. Twenty-two persons with multiple sclerosis (12 cognitively preserved and 10 impaired) and 10 healthy controls successfully underwent [11C]flumazenil positron emission tomography. Persons with multiple sclerosis showed a lower influx rate constant in the thalamus, indicating lower perfusion. For the volume of distribution, persons with multiple sclerosis showed higher values than controls in deep grey matter, reflecting increased GABA-receptor density. When comparing cognitively impaired and preserved patients to controls, the preserved group showed a significantly higher volume of distribution in cortical and deep grey matter and hippocampus. Positive correlations were observed between both positron emission tomography measures and information processing speed in the multiple sclerosis group only. Whereas concentrations of glutamate and GABA did not differ between multiple sclerosis and control nor between cognitively impaired, preserved and control groups, increased GABA-receptor density was observed in preserved persons with multiple sclerosis that was not seen in cognitively impaired patients. In addition, GABA-receptor density correlated to cognition, in particular with information processing speed. This could indicate that GABA-receptor density is upregulated in the cognitively preserved phase of multiple sclerosis as a means to regulate neurotransmission and potentially preserve cognitive functioning.
Collapse
Affiliation(s)
- Marijn Huiskamp
- Correspondence to: M. Huiskamp Department of Anatomy & Neurosciences Amsterdam UMC, Location Vrije Universiteit PO Box 7057, 1007 MB Amsterdam, The Netherlands E-mail:
| | - Maqsood Yaqub
- Department of Radiology and nuclear medicine, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Marike R van Lingen
- MS Center Amsterdam, Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Petra J W Pouwels
- Department of Radiology and nuclear medicine, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Lodewijk R J de Ruiter
- MS Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Joep Killestein
- MS Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Lothar A Schwarte
- Department of Anesthesiology, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Sandeep S V Golla
- Department of Radiology and nuclear medicine, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Bart N M van Berckel
- Department of Radiology and nuclear medicine, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Ronald Boellaard
- Department of Radiology and nuclear medicine, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Jeroen J G Geurts
- MS Center Amsterdam, Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
| | - Hanneke E Hulst
- MS Center Amsterdam, Anatomy and Neurosciences, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, 1081 HZ, The Netherlands
- Health, Medical and Neuropsychology Unit, Institute of Psychology, Leiden University, Leiden, 2333 AK, The Netherlands
| |
Collapse
|
18
|
Evans R, Watkins LM, Hawkins K, Santiago G, Demetriou C, Naughton M, Dittmer M, Rees MI, Fitzgerald D, Morgan BP, Neal JW, Howell OW. Complement activation and increased anaphylatoxin receptor expression are associated with cortical grey matter lesions and the compartmentalised inflammatory response of multiple sclerosis. Front Cell Neurosci 2023; 17:1094106. [PMID: 37032838 PMCID: PMC10073739 DOI: 10.3389/fncel.2023.1094106] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 02/07/2023] [Indexed: 04/11/2023] Open
Abstract
Background The extent of cortical pathology is an important determinant of multiple sclerosis (MS) severity. Cortical demyelination and neurodegeneration are related to inflammation of the overlying leptomeninges, a more inflammatory CSF milieu and with parenchymal microglia and astroglia activation. These are all components of the compartmentalised inflammatory response. Compartmentalised inflammation is a feature of progressive MS, which is not targeted by disease modifying therapies. Complement is differentially expressed in the MS CSF and complement, and complement receptors, are associated with demyelination and neurodegeneration. Methods To better understand if complement activation in the leptomeninges is associated with underlying cortical demyelination, inflammation, and microglial activation, we performed a neuropathological study of progressive MS (n = 22, 14 females), neuroinflammatory (n = 8), and non-neurological disease controls (n = 10). We then quantified the relative extent of demyelination, connective tissue inflammation, complement, and complement receptor positive microglia/macrophages. Results Complement was elevated at the leptomeninges, subpial, and within and around vessels of the cortical grey matter. The extent of complement C1q immunoreactivity correlated with connective tissue infiltrates, whilst activation products C4d, Bb, and C3b associated with grey matter demyelination, and C3a receptor 1+ and C5a receptor 1+ microglia/macrophages closely apposed C3b labelled cells. The density of C3a receptor 1+ and C5a receptor 1+ cells was increased at the expanding edge of subpial and leukocortical lesions. C5a receptor 1+ cells expressed TNFα, iNOS and contained puncta immunoreactive for proteolipid protein, neurofilament and synaptophysin, suggesting their involvement in grey matter lesion expansion. Interpretation The presence of products of complement activation at the brain surfaces, their association with the extent of underlying pathology and increased complement anaphylatoxin receptor positive microglia/macrophages at expanding cortical grey matter lesions, could represent a target to modify compartmentalised inflammation and cortical demyelination.
Collapse
Affiliation(s)
- Rhian Evans
- Faculty of Medicine, Health and Life Sciences, Swansea University Medical School, Swansea, United Kingdom
| | - Lewis M. Watkins
- Faculty of Medicine, Health and Life Sciences, Swansea University Medical School, Swansea, United Kingdom
| | - Kristen Hawkins
- Faculty of Medicine, Health and Life Sciences, Swansea University Medical School, Swansea, United Kingdom
| | - Gabriella Santiago
- Faculty of Medicine, Health and Life Sciences, Swansea University Medical School, Swansea, United Kingdom
| | - Constantinos Demetriou
- Faculty of Medicine, Health and Life Sciences, Swansea University Medical School, Swansea, United Kingdom
| | - Michelle Naughton
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - Marie Dittmer
- Centre for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - Mark I. Rees
- Faculty of Medicine and Health, The University of Sydney, Darlington, NSW, Australia
| | - Denise Fitzgerald
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, Belfast, United Kingdom
| | - B. Paul Morgan
- School of Medicine, UK Dementia Research Institute Cardiff and Systems Immunity Research Institute, Cardiff University, Cardiff, United Kingdom
| | - James W. Neal
- Faculty of Medicine, Health and Life Sciences, Swansea University Medical School, Swansea, United Kingdom
| | - Owain W. Howell
- Faculty of Medicine, Health and Life Sciences, Swansea University Medical School, Swansea, United Kingdom
- *Correspondence: Owain W. Howell,
| |
Collapse
|
19
|
Zhang W, Chen Y, Pei H. C1q and central nervous system disorders. Front Immunol 2023; 14:1145649. [PMID: 37033981 PMCID: PMC10076750 DOI: 10.3389/fimmu.2023.1145649] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/07/2023] [Indexed: 04/11/2023] Open
Abstract
C1q is a crucial component of the complement system, which is activated through the classical pathway to perform non-specific immune functions, serving as the first line of defense against pathogens. C1q can also bind to specific receptors to carry out immune and other functions, playing a vital role in maintaining immune homeostasis and normal physiological functions. In the developing central nervous system (CNS), C1q functions in synapse formation and pruning, serving as a key player in the development and homeostasis of neuronal networks in the CNS. C1q has a close relationship with microglia and astrocytes, and under their influence, C1q may contribute to the development of CNS disorders. Furthermore, C1q can also have independent effects on neurological disorders, producing either beneficial or detrimental outcomes. Most of the evidence for these functions comes from animal models, with some also from human specimen studies. C1q is now emerging as a promising target for the treatment of a variety of diseases, and clinical trials are already underway for CNS disorders. This article highlights the role of C1q in CNS diseases, offering new directions for the diagnosis and treatment of these conditions.
Collapse
Affiliation(s)
- Wenjie Zhang
- Department of Emergency Intensive Care Unit, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of General Practice, Xingyang Sishui Central Health Center, Zhengzhou, China
| | - Yuan Chen
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hui Pei
- Department of Emergency Intensive Care Unit, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Hui Pei,
| |
Collapse
|
20
|
Piskorowski RA, Chevaleyre V. Hippocampal area CA2: interneuron disfunction during pathological states. Front Neural Circuits 2023; 17:1181032. [PMID: 37180763 PMCID: PMC10174260 DOI: 10.3389/fncir.2023.1181032] [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: 03/06/2023] [Accepted: 04/07/2023] [Indexed: 05/16/2023] Open
Abstract
Hippocampal area CA2 plays a critical role in social recognition memory and has unique cellular and molecular properties that distinguish it from areas CA1 and CA3. In addition to having a particularly high density of interneurons, the inhibitory transmission in this region displays two distinct forms of long-term synaptic plasticity. Early studies on human hippocampal tissue have reported unique alteration in area CA2 with several pathologies and psychiatric disorders. In this review, we present recent studies revealing changes in inhibitory transmission and plasticity of area CA2 in mouse models of multiple sclerosis, autism spectrum disorder, Alzheimer's disease, schizophrenia and the 22q11.2 deletion syndrome and propose how these changes could underly deficits in social cognition observed during these pathologies.
Collapse
Affiliation(s)
- Rebecca A. Piskorowski
- Université Paris Cité, INSERM UMRS 1266, Institute of Psychiatry and Neuroscience of Paris, GHU Paris Psychiatrie et Neurosciences, Paris, France
- Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR 8246, INSERM U1130, Sorbonne Université, Paris, France
- *Correspondence: Rebecca A. Piskorowski,
| | - Vivien Chevaleyre
- Université Paris Cité, INSERM UMRS 1266, Institute of Psychiatry and Neuroscience of Paris, GHU Paris Psychiatrie et Neurosciences, Paris, France
- Institute of Biology Paris Seine, Neuroscience Paris Seine, CNRS UMR 8246, INSERM U1130, Sorbonne Université, Paris, France
| |
Collapse
|
21
|
Kole K, Voesenek BJB, Brinia ME, Petersen N, Kole MHP. Parvalbumin basket cell myelination accumulates axonal mitochondria to internodes. Nat Commun 2022; 13:7598. [PMID: 36494349 PMCID: PMC9734141 DOI: 10.1038/s41467-022-35350-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
Parvalbumin-expressing (PV+) basket cells are fast-spiking inhibitory interneurons that exert critical control over local circuit activity and oscillations. PV+ axons are often myelinated, but the electrical and metabolic roles of interneuron myelination remain poorly understood. Here, we developed viral constructs allowing cell type-specific investigation of mitochondria with genetically encoded fluorescent probes. Single-cell reconstructions revealed that mitochondria selectively cluster to myelinated segments of PV+ basket cells, confirmed by analyses of a high-resolution electron microscopy dataset. In contrast to the increased mitochondrial densities in excitatory axons cuprizone-induced demyelination abolished mitochondrial clustering in PV+ axons. Furthermore, with genetic deletion of myelin basic protein the mitochondrial clustering was still observed at internodes wrapped by noncompacted myelin, indicating that compaction is dispensable. Finally, two-photon imaging of action potential-evoked calcium (Ca2+) responses showed that interneuron myelination attenuates both the cytosolic and mitochondrial Ca2+ transients. These findings suggest that oligodendrocyte ensheathment of PV+ axons assembles mitochondria to branch selectively fine-tune metabolic demands.
Collapse
Affiliation(s)
- Koen Kole
- grid.418101.d0000 0001 2153 6865Axonal Signaling Group, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
| | - Bas J. B. Voesenek
- grid.418101.d0000 0001 2153 6865Axonal Signaling Group, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
| | - Maria E. Brinia
- grid.418101.d0000 0001 2153 6865Axonal Signaling Group, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands ,grid.5216.00000 0001 2155 0800Medical School, National Kapodistrian University of Athens, Athens, 11527 Greece
| | - Naomi Petersen
- grid.418101.d0000 0001 2153 6865Axonal Signaling Group, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands
| | - Maarten H. P. Kole
- grid.418101.d0000 0001 2153 6865Axonal Signaling Group, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, 1105 BA Amsterdam, the Netherlands ,grid.5477.10000000120346234Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Padualaan 8, 3584 CH Utrecht, the Netherlands
| |
Collapse
|
22
|
Oligodendroglia are emerging players in several forms of learning and memory. Commun Biol 2022; 5:1148. [PMID: 36309567 PMCID: PMC9617857 DOI: 10.1038/s42003-022-04116-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 10/14/2022] [Indexed: 11/15/2022] Open
Abstract
Synaptic plasticity is the fundamental cellular mechanism of learning and memory, but recent research reveals that myelin-forming glia, oligodendrocytes (OL), are also involved. They contribute in ways that synaptic plasticity cannot, and the findings have not been integrated into the established conceptual framework used in the field of learning and memory. OLs and their progenitors are involved in long-term memory, memory consolidation, working memory, and recall in associative learning. They also contribute to short-term memory and non-associative learning by affecting synaptic transmission, intrinsic excitability of axons, and neural oscillations. Oligodendroglial involvement expands the field beyond synaptic plasticity to system-wide network function, where precise spike time arrival and neural oscillations are critical in information processing, storage, and retrieval. A Perspective highlights current evidence that supports oligodendrocytes and their progenitors’ involvement in cognition and proposes that our understanding of learning and memory can be expanded beyond the classic view of synaptic plasticity to a system-wide network function.
Collapse
|
23
|
Saez-Calveras N, Brewster AL, Stuve O. The validity of animal models to explore the pathogenic role of the complement system in multiple sclerosis: A review. Front Mol Neurosci 2022; 15:1017484. [PMID: 36311030 PMCID: PMC9606595 DOI: 10.3389/fnmol.2022.1017484] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/26/2022] [Indexed: 11/26/2022] Open
Abstract
Animal models of multiple sclerosis (MS) have been extensively used to characterize the disease mechanisms in MS, as well as to identify potential pharmacologic targets for this condition. In recent years, the immune complement system has gained increased attention as an important effector in the pathogenesis of MS. Evidence from histological, serum, and CSF studies of patients supports an involvement of complement in both relapsing-remitting and progressive MS. In this review, we discuss the history and advances made on the use of MS animal models to profile the effects of the complement system in this condition. The first studies that explored the complement system in the context of MS used cobra venom factor (CVF) as a complement depleting agent in experimental autoimmune encephalomyelitis (EAE) Lewis rats. Since then, multiple mice and rat models of MS have revealed a role of C3 and the alternative complement cascade in the opsonization and phagocytosis of myelin by microglia and myeloid cells. Studies using viral vectors, genetic knockouts and pharmacologic complement inhibitors have also shown an effect of complement in synaptic loss. Antibody-mediated EAE models have revealed an involvement of the C1 complex and the classical complement as an effector of the humoral response in this disease. C1q itself may also be involved in modulating microglia activation and oligodendrocyte differentiation in these animals. In addition, animal and in vitro models have revealed that multiple complement factors may act as modulators of both the innate and adaptive immune responses. Finally, evidence gathered from mice models suggests that the membrane attack complex (MAC) may even exert protective roles in the chronic stages of EAE. Overall, this review summarizes the importance of MS animal models to better characterize the role of the complement system and guide future therapeutic approaches in this condition.
Collapse
Affiliation(s)
- Nil Saez-Calveras
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Neurology Section, Parkland Hospital, Dallas, TX, United States
| | - Amy L. Brewster
- Department of Biological Sciences, Southern Methodist University, Dallas, TX, United States
| | - Olaf Stuve
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX, United States
- Neurology Section, VA North Texas Health Care System, Dallas, TX, United States
- Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States
- *Correspondence: Olaf Stuve,
| |
Collapse
|
24
|
Huiskamp M, Kiljan S, Kulik S, Witte ME, Jonkman LE, Gjm Bol J, Schenk GJ, Hulst HE, Tewarie P, Schoonheim MM, Geurts JJ. Inhibitory synaptic loss drives network changes in multiple sclerosis: An ex vivo to in silico translational study. Mult Scler 2022; 28:2010-2019. [PMID: 36189828 PMCID: PMC9574900 DOI: 10.1177/13524585221125381] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Background: Synaptic and neuronal loss contribute to network dysfunction and disability
in multiple sclerosis (MS). However, it is unknown whether excitatory or
inhibitory synapses and neurons are more vulnerable and how their losses
impact network functioning. Objective: To quantify excitatory and inhibitory synapses and neurons and to investigate
how synaptic loss affects network functioning through computational
modeling. Methods: Using immunofluorescent staining and confocal microscopy, densities of
glutamatergic and GABAergic synapses and neurons were compared between
post-mortem MS and non-neurological control cases. Then, a corticothalamic
biophysical model was employed to study how MS-induced excitatory and
inhibitory synaptic loss affect network functioning. Results: In layer VI of normal-appearing MS cortex, excitatory and inhibitory synaptic
densities were significantly lower than controls (reductions up to 14.9%),
but demyelinated cortex showed larger losses of inhibitory synapses (29%).
In our computational model, reducing inhibitory synapses impacted the
network most, leading to a disinhibitory increase in neuronal activity and
connectivity. Conclusion: In MS, excitatory and inhibitory synaptic losses were observed, predominantly
for inhibitory synapses in demyelinated cortex. Inhibitory synaptic loss
affected network functioning most, leading to increased neuronal activity
and connectivity. As network disinhibition relates to cognitive impairment,
inhibitory synaptic loss seems particularly relevant in MS.
Collapse
Affiliation(s)
- Marijn Huiskamp
- Anatomy and Neurosciences, MS Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Svenja Kiljan
- Anatomy and Neurosciences, MS Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Shanna Kulik
- Anatomy and Neurosciences, MS Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Maarteen E Witte
- Molecular Cell Biology and Inflammation, MS Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Laura E Jonkman
- Anatomy and Neurosciences, MS Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - John Gjm Bol
- Anatomy and Neurosciences, MS Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Geert J Schenk
- Anatomy and Neurosciences, MS Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Hanneke E Hulst
- Anatomy and Neurosciences, MS Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands/Health, Medical and Neuropsychology Unit, Institute of Psychology, Leiden University, Leiden, The Netherlands
| | - Prejaas Tewarie
- Neurology, MS center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands/Clinical Neurophysiology and MEG Center, MS Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Menno M Schoonheim
- Anatomy and Neurosciences, MS Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Jeroen Jg Geurts
- Anatomy and Neurosciences, MS Center Amsterdam, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Zuo M, Fettig NM, Bernier LP, Pössnecker E, Spring S, Pu A, Ma XI, Lee DS, Ward LA, Sharma A, Kuhle J, Sled JG, Pröbstel AK, MacVicar BA, Osborne LC, Gommerman JL, Ramaglia V. Age-dependent gray matter demyelination is associated with leptomeningeal neutrophil accumulation. JCI Insight 2022; 7:e158144. [PMID: 35536649 PMCID: PMC9309059 DOI: 10.1172/jci.insight.158144] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/04/2022] [Indexed: 11/21/2022] Open
Abstract
People living with multiple sclerosis (MS) experience episodic CNS white matter lesions instigated by autoreactive T cells. With age, patients with MS show evidence of gray matter demyelination and experience devastating nonremitting symptomology. What drives progression is unclear and studying this has been hampered by the lack of suitable animal models. Here, we show that passive experimental autoimmune encephalomyelitis (EAE) induced by an adoptive transfer of young Th17 cells induced a nonremitting clinical phenotype that was associated with persistent leptomeningeal inflammation and cortical pathology in old, but not young, SJL/J mice. Although the quantity and quality of T cells did not differ in the brains of old versus young EAE mice, an increase in neutrophils and a decrease in B cells were observed in the brains of old mice. Neutrophils were also found in the leptomeninges of a subset of progressive MS patient brains that showed evidence of leptomeningeal inflammation and subpial cortical demyelination. Taken together, our data show that while Th17 cells initiate CNS inflammation, subsequent clinical symptoms and gray matter pathology are dictated by age and associated with other immune cells, such as neutrophils.
Collapse
Affiliation(s)
- Michelle Zuo
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Naomi M. Fettig
- Department of Microbiology and Immunology and Life Sciences Institute, and
| | - Louis-Philippe Bernier
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Elisabeth Pössnecker
- Multiple Sclerosis Center & Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), Departments of Neurology, Biomedicine, and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
| | - Shoshana Spring
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Annie Pu
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Xianjie I. Ma
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Dennis S.W. Lee
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Lesley A. Ward
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Anshu Sharma
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Jens Kuhle
- Multiple Sclerosis Center & Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), Departments of Neurology, Biomedicine, and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
| | - John G. Sled
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Anne-Katrin Pröbstel
- Multiple Sclerosis Center & Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), Departments of Neurology, Biomedicine, and Clinical Research, University Hospital and University of Basel, Basel, Switzerland
| | - Brian A. MacVicar
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Lisa C. Osborne
- Department of Microbiology and Immunology and Life Sciences Institute, and
| | | | - Valeria Ramaglia
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
26
|
Scaroni F, Visconte C, Serpente M, Golia MT, Gabrielli M, Huiskamp M, Hulst HE, Carandini T, De Riz M, Pietroboni A, Rotondo E, Scarpini E, Galimberti D, Teunissen CE, van Dam M, de Jong BA, Fenoglio C, Verderio C. miR-150-5p and let-7b-5p in Blood Myeloid Extracellular Vesicles Track Cognitive Symptoms in Patients with Multiple Sclerosis. Cells 2022; 11:cells11091551. [PMID: 35563859 PMCID: PMC9104242 DOI: 10.3390/cells11091551] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 04/30/2022] [Accepted: 05/02/2022] [Indexed: 02/04/2023] Open
Abstract
Cognitive deficits strongly affect the quality of life of patients with multiple sclerosis (MS). However, no cognitive MS biomarkers are currently available. Extracellular vesicles (EVs) contain markers of parental cells and are able to pass from the brain into blood, representing a source of disease biomarkers. The aim of this study was to investigate whether small non-coding microRNAs (miRNAs) targeting synaptic genes and packaged in plasma EVs may reflect cognitive deficits in MS patients. Total EVs were precipitated by Exoquick from the plasma of twenty-six cognitively preserved (CP) and twenty-three cognitively impaired (CI) MS patients belonging to two independent cohorts. Myeloid EVs were extracted by affinity capture from total EVs using Isolectin B4 (IB4). Fourteen miRNAs targeting synaptic genes were selected and measured by RT-PCR in both total and myeloid EVs. Myeloid EVs from CI patients expressed higher levels of miR-150-5p and lower levels of let-7b-5p compared to CP patients. Stratification for progressive MS (PMS) and relapsing-remitting MS (RRMS) and correlation with clinical parameters suggested that these alterations might be attributable to cognitive deficits rather than disease progression. This study identifies miR-150-5p and let-7b-5p packaged in blood myeloid EVs as possible biomarkers for cognitive deficits in MS.
Collapse
Affiliation(s)
- Federica Scaroni
- Institute of Neuroscience, CNR, Via Follereau 3, 20854 Vedano al Lambro, Italy; (F.S.); (M.T.G.); (M.G.)
| | - Caterina Visconte
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via F. Sforza 35, 20122 Milan, Italy; (C.V.); (E.S.); (D.G.)
- Centro Dino Ferrari, University of Milan, 20122 Milan, Italy; (M.S.); (T.C.); (M.D.R.); (A.P.); (E.R.)
| | - Maria Serpente
- Centro Dino Ferrari, University of Milan, 20122 Milan, Italy; (M.S.); (T.C.); (M.D.R.); (A.P.); (E.R.)
- Fondazione IRCCS Ca’ Granda, Ospedale Policlinico, 20122 Milan, Italy
| | - Maria Teresa Golia
- Institute of Neuroscience, CNR, Via Follereau 3, 20854 Vedano al Lambro, Italy; (F.S.); (M.T.G.); (M.G.)
| | - Martina Gabrielli
- Institute of Neuroscience, CNR, Via Follereau 3, 20854 Vedano al Lambro, Italy; (F.S.); (M.T.G.); (M.G.)
| | - Marijn Huiskamp
- MS Center Amsterdam, Amsterdam Neuroscience, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam UMC, De Boelelaan 1117, 1081 Amsterdam, The Netherlands; (M.H.); (M.v.D.)
| | - Hanneke E. Hulst
- Health-, Medical- and Neuropsychology Unit, Institute of Psychology, Leiden University, 2300 Leiden, The Netherlands;
| | - Tiziana Carandini
- Centro Dino Ferrari, University of Milan, 20122 Milan, Italy; (M.S.); (T.C.); (M.D.R.); (A.P.); (E.R.)
- Fondazione IRCCS Ca’ Granda, Ospedale Policlinico, 20122 Milan, Italy
| | - Milena De Riz
- Centro Dino Ferrari, University of Milan, 20122 Milan, Italy; (M.S.); (T.C.); (M.D.R.); (A.P.); (E.R.)
- Fondazione IRCCS Ca’ Granda, Ospedale Policlinico, 20122 Milan, Italy
| | - Anna Pietroboni
- Centro Dino Ferrari, University of Milan, 20122 Milan, Italy; (M.S.); (T.C.); (M.D.R.); (A.P.); (E.R.)
- Fondazione IRCCS Ca’ Granda, Ospedale Policlinico, 20122 Milan, Italy
| | - Emanuela Rotondo
- Centro Dino Ferrari, University of Milan, 20122 Milan, Italy; (M.S.); (T.C.); (M.D.R.); (A.P.); (E.R.)
- Fondazione IRCCS Ca’ Granda, Ospedale Policlinico, 20122 Milan, Italy
| | - Elio Scarpini
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via F. Sforza 35, 20122 Milan, Italy; (C.V.); (E.S.); (D.G.)
- Centro Dino Ferrari, University of Milan, 20122 Milan, Italy; (M.S.); (T.C.); (M.D.R.); (A.P.); (E.R.)
- Fondazione IRCCS Ca’ Granda, Ospedale Policlinico, 20122 Milan, Italy
| | - Daniela Galimberti
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via F. Sforza 35, 20122 Milan, Italy; (C.V.); (E.S.); (D.G.)
- Centro Dino Ferrari, University of Milan, 20122 Milan, Italy; (M.S.); (T.C.); (M.D.R.); (A.P.); (E.R.)
- Fondazione IRCCS Ca’ Granda, Ospedale Policlinico, 20122 Milan, Italy
| | - Charlotte E. Teunissen
- MS Center Amsterdam, Amsterdam Neuroscience, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 Amsterdam, The Netherlands; (C.E.T.); (B.A.d.J.)
| | - Maureen van Dam
- MS Center Amsterdam, Amsterdam Neuroscience, Department of Anatomy and Neurosciences, Amsterdam UMC, Vrije Universiteit Amsterdam UMC, De Boelelaan 1117, 1081 Amsterdam, The Netherlands; (M.H.); (M.v.D.)
| | - Brigit A. de Jong
- MS Center Amsterdam, Amsterdam Neuroscience, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 Amsterdam, The Netherlands; (C.E.T.); (B.A.d.J.)
| | - Chiara Fenoglio
- MS Center Amsterdam, Amsterdam Neuroscience, Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 Amsterdam, The Netherlands; (C.E.T.); (B.A.d.J.)
- Department of Neuropathology and Transplantation, University of Milan, Via F. Sforza 35, 20122 Milan, Italy
- Correspondence: (C.F.); (C.V.); Tel.: +39-0264488386 (C.V.)
| | - Claudia Verderio
- Institute of Neuroscience, CNR, Via Follereau 3, 20854 Vedano al Lambro, Italy; (F.S.); (M.T.G.); (M.G.)
- Correspondence: (C.F.); (C.V.); Tel.: +39-0264488386 (C.V.)
| |
Collapse
|
27
|
Cooze BJ, Dickerson M, Loganathan R, Watkins LM, Grounds E, Pearson BR, Bevan RJ, Morgan BP, Magliozzi R, Reynolds R, Neal JW, Howell OW. The association between neurodegeneration and local complement activation in the thalamus to progressive multiple sclerosis outcome. Brain Pathol 2022; 32:e13054. [PMID: 35132719 PMCID: PMC9425007 DOI: 10.1111/bpa.13054] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/17/2021] [Accepted: 01/17/2022] [Indexed: 01/22/2023] Open
Abstract
The extent of grey matter demyelination and neurodegeneration in the progressive multiple sclerosis (PMS) brains at post‐mortem associates with more severe disease. Regional tissue atrophy, especially affecting the cortical and deep grey matter, including the thalamus, is prognostic for poor outcomes. Microglial and complement activation are important in the pathogenesis and contribute to damaging processes that underlie tissue atrophy in PMS. We investigated the extent of pathology and innate immune activation in the thalamus in comparison to cortical grey and white matter in blocks from 21 cases of PMS and 10 matched controls. Using a digital pathology workflow, we show that the thalamus is invariably affected by demyelination and had a far higher proportion of active inflammatory lesions than forebrain cortical tissue blocks from the same cases. Lesions were larger and more frequent in the medial nuclei near the ventricular margin, whilst neuronal loss was greatest in the lateral thalamic nuclei. The extent of thalamic neuron loss was not associated with thalamic demyelination but correlated with the burden of white matter pathology in other forebrain areas (Spearman r = 0.79, p < 0.0001). Only thalamic neuronal loss, and not that seen in other forebrain cortical areas, correlated with disease duration (Spearman r = −0.58, p = 0.009) and age of death (Spearman r = −0.47, p = 0.045). Immunoreactivity for the complement pattern recognition molecule C1q, and products of complement activation (C4d, Bb and C3b) were elevated in thalamic lesions with an active inflammatory pathology. Complement regulatory protein, C1 inhibitor, was unchanged in expression. We conclude that active inflammatory demyelination, neuronal loss and local complement synthesis and activation in the thalamus, are important to the pathological and clinical disease outcomes of PMS.
Collapse
Affiliation(s)
- Benjamin J Cooze
- Faculty of Medical, Health and Life Sciences, Swansea University, Swansea, UK
| | - Matthew Dickerson
- Faculty of Medical, Health and Life Sciences, Swansea University, Swansea, UK
| | | | - Lewis M Watkins
- Faculty of Medical, Health and Life Sciences, Swansea University, Swansea, UK
| | - Ethan Grounds
- Faculty of Medical, Health and Life Sciences, Swansea University, Swansea, UK
| | - Ben R Pearson
- Faculty of Medical, Health and Life Sciences, Swansea University, Swansea, UK
| | - Ryan Jack Bevan
- UK Dementia Research Institute at Cardiff University, Cardiff, UK
| | - B Paul Morgan
- UK Dementia Research Institute at Cardiff University, Cardiff, UK
| | - Roberta Magliozzi
- Department of Neurological and Movement Sciences, University of Verona, Italy
| | | | - James W Neal
- Faculty of Medical, Health and Life Sciences, Swansea University, Swansea, UK
| | - Owain W Howell
- Faculty of Medical, Health and Life Sciences, Swansea University, Swansea, UK
| |
Collapse
|
28
|
Dubey M, Pascual-Garcia M, Helmes K, Wever DD, Hamada MS, Kushner SA, Kole MHP. Myelination synchronizes cortical oscillations by consolidating parvalbumin-mediated phasic inhibition. eLife 2022; 11:73827. [PMID: 35001871 PMCID: PMC8887893 DOI: 10.7554/elife.73827] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/28/2021] [Indexed: 12/18/2022] Open
Abstract
Parvalbumin-positive (PV+) γ-aminobutyric acid (GABA) interneurons are critically involved in producing rapid network oscillations and cortical microcircuit computations, but the significance of PV+ axon myelination to the temporal features of inhibition remains elusive. Here, using toxic and genetic mouse models of demyelination and dysmyelination, respectively, we find that loss of compact myelin reduces PV+ interneuron presynaptic terminals and increases failures, and the weak phasic inhibition of pyramidal neurons abolishes optogenetically driven gamma oscillations in vivo. Strikingly, during behaviors of quiet wakefulness selectively theta rhythms are amplified and accompanied by highly synchronized interictal epileptic discharges. In support of a causal role of impaired PV-mediated inhibition, optogenetic activation of myelin-deficient PV+ interneurons attenuated the power of slow theta rhythms and limited interictal spike occurrence. Thus, myelination of PV axons is required to consolidate fast inhibition of pyramidal neurons and enable behavioral state-dependent modulation of local circuit synchronization.
Collapse
Affiliation(s)
- Mohit Dubey
- Department of Axonal Signaling, Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, Netherlands
| | | | - Koke Helmes
- Department of Axonal Signaling, Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, Netherlands
| | - Dennis D Wever
- Department of Axonal Signaling, Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, Netherlands
| | - Mustafa S Hamada
- Department of Axonal Signaling, Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, Netherlands.,Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Steven A Kushner
- Department of Psychiatry, Erasmus Medical Centre, Rotterdam, Netherlands
| | - Maarten H P Kole
- Department of Axonal Signaling, Netherlands Institute for Neuroscience (NIN), Royal Netherlands Academy of Arts and Sciences (KNAW), Amsterdam, Netherlands.,Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
29
|
Age-related changes in multiple sclerosis and experimental autoimmune encephalomyelitis. Semin Immunol 2022; 59:101631. [PMID: 35752572 DOI: 10.1016/j.smim.2022.101631] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 06/03/2022] [Accepted: 06/13/2022] [Indexed: 01/15/2023]
Abstract
A better understanding of the pathological mechanisms that drive neurodegeneration in people living with multiple sclerosis (MS) is needed to design effective therapies to treat and/or prevent disease progression. We propose that CNS-intrinsic inflammation and re-modelling of the sub-arachnoid space of the leptomeninges sets the stage for neurodegeneration from the earliest stages of MS. While neurodegenerative processes are clinically silent early in disease, ageing results in neurodegenerative changes that become clinically manifest as progressive disability. Here we review pathological correlates of MS disease progression, highlight emerging mouse models that mimic key progressive changes in MS, and provide new perspectives on therapeutic approaches to protect against MS-associated neurodegeneration.
Collapse
|
30
|
Yates RL, Pansieri J, Li Q, Bell JS, Yee SA, Palace J, Esiri MM, DeLuca GC. The influence of HLA-DRB1*15 on the relationship between microglia and neurons in multiple sclerosis normal appearing cortical grey matter. Brain Pathol 2021; 32:e13041. [PMID: 34904300 PMCID: PMC9245937 DOI: 10.1111/bpa.13041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 11/06/2022] Open
Abstract
Cortical tissue injury is common in multiple sclerosis (MS) and associates with disability progression. We have previously shown that HLA‐DRB1*15 genotype status associates with the extent of cortical inflammatory pathology. In the current study, we sought to examine the influence of HLA‐DRB1*15 on relationships between inflammation and neurodegeneration in MS. Human post‐mortem MS cases (n = 47) and controls (n = 10) were used. Adjacent sections of motor cortex were stained for microglia (Iba1+, CD68+, TMEM119+), lymphocytes (CD3+, CD8+), GFAP+ astrocytes, and neurons (NeuN+). A subset of MS cases (n = 20) and controls (n = 7) were double‐labeled for neurofilament and glutamic acid decarboxylase 65/67 (GAD+) to assess the extent of the inhibitory synaptic loss. In MS cases, microglial protein expression positively correlated with neuron density (Iba1+: r = 0.548, p < 0.001, CD68+: r = 0.498, p = 0.001, TMEM119+ r = 0.437, p = 0.003). This finding was restricted to MS cases not carrying HLA‐DRB1*15. Evidence of a 14% reduction in inhibitory synapses in MS was detected (MS: 0.299 ± 0.006 synapses/μm2 neuronal membrane versus control: 0.348 ± 0.009 synapses/μm2 neuronal membrane, p = 0.005). Neurons expressing inhibitory synapses were 24% smaller in MS cases compared to the control (MS: 403 ± 15 μm2 versus control: 531 ± 29 μm2, p = 0.001), a finding driven by HLA‐DRB1*15+ cases (15+: 376 ± 21 μm2 vs. 15−: 432 ± 22 μm2, p = 0.018). Taken together, our results demonstrate that HLA‐DRB1*15 modulates the relationship between microglial inflammation, inhibitory synapses, and neuronal density in the MS cortex.
Collapse
Affiliation(s)
- Richard L Yates
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Jonathan Pansieri
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Qizhu Li
- Department of Engineering Science, University of Oxford, Oxford, UK
| | - Jack S Bell
- Salford Royal NHS Foundation Trust, Salford, UK
| | - Sydney A Yee
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Jacqueline Palace
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Margaret M Esiri
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Gabriele C DeLuca
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
31
|
Complement component 3 from astrocytes mediates retinal ganglion cell loss during neuroinflammation. Acta Neuropathol 2021; 142:899-915. [PMID: 34487221 DOI: 10.1007/s00401-021-02366-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 08/13/2021] [Accepted: 08/27/2021] [Indexed: 12/14/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) characterized by varying degrees of secondary neurodegeneration. Retinal ganglion cells (RGC) are lost in MS in association with optic neuritis but the mechanisms of neuronal injury remain unclear. Complement component C3 has been implicated in retinal and cerebral synaptic pathology that may precede neurodegeneration. Herein, we examined post-mortem MS retinas, and then used a mouse model, experimental autoimmune encephalomyelitis (EAE), to examine the role of C3 in the pathogenesis of RGC loss associated with optic neuritis. First, we show extensive C3 expression in astrocytes (C3+/GFAP+ cells) and significant RGC loss (RBPMS+ cells) in post-mortem retinas from people with MS compared to retinas from non-MS individuals. A patient with progressive MS with a remote history of optic neuritis showed marked reactive astrogliosis with C3 expression in the inner retina extending into deeper layers in the affected eye more than the unaffected eye. To study whether C3 mediates retinal degeneration, we utilized global C3-/- EAE mice and found that they had less RGC loss and partially preserved neurites in the retina compared with C3+/+ EAE mice. C3-/- EAE mice had fewer axonal swellings in the optic nerve, reflecting reduced axonal injury, but had no changes in demyelination or T cell infiltration into the CNS. Using a C3-tdTomato reporter mouse line, we show definitive evidence of C3 expression in astrocytes in the retina and optic nerves of EAE mice. Conditional deletion of C3 in astrocytes showed RGC protection replicating the effects seen in the global knockouts. These data implicate astrocyte C3 expression as a critical mediator of retinal neuronal pathology in EAE and MS, and are consistent with recent studies showing C3 gene variants are associated with faster rates of retinal neurodegeneration in human disease.
Collapse
|
32
|
Gomez-Arboledas A, Acharya MM, Tenner AJ. The Role of Complement in Synaptic Pruning and Neurodegeneration. Immunotargets Ther 2021; 10:373-386. [PMID: 34595138 PMCID: PMC8478425 DOI: 10.2147/itt.s305420] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 09/04/2021] [Indexed: 12/14/2022] Open
Abstract
The complement system, an essential part of the innate immune system, is composed of a group of secreted and membrane proteins that collectively participate in maintaining the function of the healthy and diseased brain. However, an inappropriate activation of the complement system has been related to an inflammatory response in multiple diseases, such as stroke, traumatic brain injury, multiple sclerosis, and Alzheimer's disease, as well as Zika infection and radiotherapy. In addition, C1q and C3 (initial activation components of the complement cascade) have been shown to play a key beneficial role in the refinement of synaptic circuits during developmental stages and adult plasticity. Nevertheless, excessive synaptic pruning in the adult brain can be detrimental and has been associated with synaptic loss in several pathological conditions. In this brief review, we will discuss the role of the complement system in synaptic pruning as well as its contribution to neurodegeneration and cognitive deficits. We also mention potential therapeutic approaches to target the complement system to treat several neuroinflammatory diseases and unintended consequences of radiotherapy.
Collapse
Affiliation(s)
- Angela Gomez-Arboledas
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
| | - Munjal M Acharya
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, USA
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, USA
| | - Andrea J Tenner
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, University of California, Irvine, School of Medicine, Irvine, CA, USA
| |
Collapse
|