1
|
Pan M, Li X, Tian X, Yang LZ, Fang W. Eugenol Alleviates Cerebral Ischemia-Reperfusion Injury in Mice by Promoting the Phagocytosis of Microglia via Up-Regulating Tripartite Motif Protein 59. Basic Clin Pharmacol Toxicol 2025; 137:e70058. [PMID: 40432346 DOI: 10.1111/bcpt.70058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/29/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025]
Abstract
Ischemic stroke (IS) is one of the most sinister diseases and the second leading cause of death in the world. Eugenol (EUG) is a natural and biologically active component that can be extracted from various plants. Studies have found that EUG can alleviate middle cerebral artery occlusion and reperfusion (MCAO/R) injury in mice, but the specific mechanism remains vague. Tripartite motif protein 59 (TRIM59) is a member of TRIM protein family, a group of E3 ubiquitin ligases. In this article, we conducted both in vivo and in vitro experiments to determine the effect of EUG on ischemia-reperfusion injury and to explore the underlying mechanisms by manipulating the expression of TRIM59. Results showed that EUG alleviates acute injury and promotes functional repair of mouse IS by enhancing the phagocytosis of microglia through up-regulating the TRIM59, activating the STAT3 pathway and promoting the expression of CD11b.
Collapse
Affiliation(s)
- Mengtian Pan
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Xiang Li
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Xinjuan Tian
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Lele Zixin Yang
- State College, The Pennsylvania State University, Eberly College of Science, Pennsylvania, USA
| | - Weirong Fang
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
2
|
Chen M, Chen J, Liu Y, Wang X, Yao M, Chen J, Zhang J, Huang Q. Senescent Macrophages Promote Age-Related Revascularization Impairment by Increasing Antiangiogenic VEGF-A165B Expression. Aging Cell 2025:e70059. [PMID: 40243169 DOI: 10.1111/acel.70059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 03/05/2025] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
Peripheral arterial disease is a common vascular disease in the elderly. Therapeutic revascularization, including angiogenic and arteriogenic therapy, is a promising treatment approach for peripheral arterial disease. However, the progress of clinical trials is not ideal, possibly due to insufficiency of preclinical models, such as not taking into account the effect of aging on vascular regeneration. Macrophages are crucial in angiogenesis and arteriogenesis. The aging microenvironment typically makes recruited monocytes and macrophages more susceptible to senescence. However, the feature of macrophages in ischemic hindlimb muscle of old individuals and their underlying role remains unclear. In this study, we reveal that macrophages of ischemic skeletal muscle in old mice are more senescent and proinflammatory. By transplanting macrophages into mice following hindlimb ischemia, we find senescent macrophages inhibit revascularization. Mechanistically, these senescent macrophages induce endothelial dysfunction via increasing vascular endothelial growth factor A-165B (VEGF-A165B) expression and secretion, and eventually impair revascularization. Notably, plasma VEGF-A165B levels are elevated in old patients with PAD and positively associated with a lower ankle brachial index (ABI). Our study suggests that targeting the senescent macrophages presents an avenue to improve age-related revascularization damage.
Collapse
Affiliation(s)
- Minghong Chen
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Junyu Chen
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yu Liu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of General and Vascular Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xuerui Wang
- Department of Cardiology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Meilian Yao
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jing Chen
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jian Zhang
- Center of Coronary Circulation, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qun Huang
- Department of Child Health Care, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
| |
Collapse
|
3
|
Vanherle S, Loix M, Miron VE, Hendriks JJA, Bogie JFJ. Lipid metabolism, remodelling and intercellular transfer in the CNS. Nat Rev Neurosci 2025; 26:214-231. [PMID: 39972160 DOI: 10.1038/s41583-025-00908-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2025] [Indexed: 02/21/2025]
Abstract
Lipid metabolism encompasses the catabolism and anabolism of lipids, and is fundamental for the maintenance of cellular homeostasis, particularly within the lipid-rich CNS. Increasing evidence further underscores the importance of lipid remodelling and transfer within and between glial cells and neurons as key orchestrators of CNS lipid homeostasis. In this Review, we summarize and discuss the complex landscape of processes involved in lipid metabolism, remodelling and intercellular transfer in the CNS. Highlighted are key pathways, including those mediating lipid (and lipid droplet) biogenesis and breakdown, lipid oxidation and phospholipid metabolism, as well as cell-cell lipid transfer mediated via lipoproteins, extracellular vesicles and tunnelling nanotubes. We further explore how the dysregulation of these pathways contributes to the onset and progression of neurodegenerative diseases, and examine the homeostatic and pathogenic impacts of environment, diet and lifestyle on CNS lipid metabolism.
Collapse
Affiliation(s)
- Sam Vanherle
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Centre, Hasselt University, Hasselt, Belgium
| | - Melanie Loix
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Centre, Hasselt University, Hasselt, Belgium
| | - Veronique E Miron
- Keenan Research Centre for Biomedical Science and Barlo Multiple Sclerosis Centre, St Michael's Hospital, Toronto, Ontario, Canada
- Department of Immunology, The University of Toronto, Toronto, Ontario, Canada
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
| | - Jerome J A Hendriks
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- University MS Centre, Hasselt University, Hasselt, Belgium
| | - Jeroen F J Bogie
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Hasselt, Belgium.
- University MS Centre, Hasselt University, Hasselt, Belgium.
| |
Collapse
|
4
|
Basavarajappa D, Chitranshi N, Oddin Mirshahvaladi SS, Gupta VB, Palanivel V, Parrilla GE, Salkar A, Mirzaei M, Komáromy AM, Krezel W, Graham SL, Gupta V. Retinoid X receptor agonist 9CDHRA mitigates retinal ganglion cell apoptosis and neuroinflammation in a mouse model of glaucoma. FASEB J 2025; 39:e70465. [PMID: 40079201 PMCID: PMC11904862 DOI: 10.1096/fj.202402642r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/26/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025]
Abstract
Glaucoma, a leading cause of irreversible blindness, is characterized by the progressive loss of retinal ganglion cells (RGCs) and optic nerve damage, often associated with elevated intraocular pressure (IOP). Retinoid X receptors (RXRs) are ligand-activated transcription factors crucial for neuroprotection, as they regulate gene expression to promote neuronal survival via several biochemical networks and reduce neuroinflammation. This study investigated the therapeutic potential of 9-cis-13,14-dihydroretinoic acid (9CDHRA), an endogenous retinoid RXR agonist, in mitigating RGC degeneration in a high-IOP-induced experimental model of glaucoma. We administered 9CDHRA to glaucomatous mice eyes via intravitreal injections and assessed its effects on endoplasmic reticulum (ER) stress markers, glial cell activation, and RGC survival. Our findings demonstrated that 9CDHRA treatment significantly protected inner retinal function and retinal laminar structure in high-IOP glaucoma. The treatment reduced ER stress markers, increased protein lysine acetylation, and diminished glial cell activation, leading to a significant decrease in apoptotic cells under glaucomatous conditions. These results suggest that 9CDHRA exerts neuroprotective effects by modulating key pathogenic pathways in glaucoma, highlighting its potential as a novel therapeutic strategy for preserving vision in glaucoma.
Collapse
Affiliation(s)
- Devaraj Basavarajappa
- Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Nitin Chitranshi
- Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | | | - Veer B. Gupta
- School of MedicineDeakin UniversityGeelongVictoriaAustralia
| | - Viswanthram Palanivel
- Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Gabriella E. Parrilla
- Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Akanksha Salkar
- Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Mehdi Mirzaei
- Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - András M. Komáromy
- Department of Small Animal Clinical Sciences, College of Veterinary MedicineMichigan State UniversityEast LansingMichiganUSA
| | - Wojciech Krezel
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Institut de la Santé et de la Recherche Médicale (U1258), Centre National de la Recherche Scientifique (UMR7104)Université de Strasbourg, Fédération de Médecine Translationnelle de StrasbourgIllkirchFrance
| | - Stuart L. Graham
- Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Vivek Gupta
- Macquarie Medical School, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| |
Collapse
|
5
|
Jana R, Das Sarma J. The crosstalk between CNS resident glial cells and peripheral immune cells is critical for age-dependent demyelination and subsequent remyelination. Biogerontology 2025; 26:74. [PMID: 40085264 DOI: 10.1007/s10522-025-10213-2] [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: 12/30/2024] [Accepted: 03/01/2025] [Indexed: 03/16/2025]
Abstract
White-matter diseases like multiple sclerosis begin in young adulthood. Aging, being a risk factor, contributes to the progression of these diseases and makes neurological disabilities worsen. Aging causes white matter alteration due to myelin loss, axonal degeneration, and hyperintensities, resulting in cognitive impairment and neurological disorders. Aging also negatively affects central nervous system resident glial cells and peripheral immune cells, contributing to myelin degeneration and diminished myelin renewal process. Restoration of myelin failure with aging accelerates the progression of cognitive decline. This review will mainly focus on how age-related altered functions of glial and peripheral cells will affect myelin sheath alteration and myelin restoration. This understanding can give us insights into the underlying mechanisms of demyelination and failure of remyelination with aging concerning altered glial and peripheral immune cell function and their crosstalk. Also, we will explain the therapeutic strategies to enhance the remyelination process of an aging brain to improve the cognitive health of an aging person.
Collapse
Affiliation(s)
- Rishika Jana
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India.
- Departments of Ophthalmology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
6
|
Kornberg MD, Calabresi PA. Multiple Sclerosis and Other Acquired Demyelinating Diseases of the Central Nervous System. Cold Spring Harb Perspect Biol 2025; 17:a041374. [PMID: 38806240 PMCID: PMC11875095 DOI: 10.1101/cshperspect.a041374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Acquired demyelinating diseases of the central nervous system (CNS) comprise inflammatory conditions, including multiple sclerosis (MS) and related diseases, as well as noninflammatory conditions caused by toxic, metabolic, infectious, traumatic, and neurodegenerative insults. Here, we review the spectrum of diseases producing acquired CNS demyelination before focusing on the prototypical example of MS, exploring the pathologic mechanisms leading to myelin injury in relapsing and progressive MS and summarizing the mechanisms and modulators of remyelination. We highlight the complex interplay between the immune system, oligodendrocytes and oligodendrocyte progenitor cells (OPCs), and other CNS glia cells such as microglia and astrocytes in the pathogenesis and clinical course of MS. Finally, we review emerging therapeutic strategies that exploit our growing understanding of disease mechanisms to limit progression and promote remyelination.
Collapse
Affiliation(s)
- Michael D Kornberg
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21287, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21287, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland 21205, USA
| |
Collapse
|
7
|
Zhang G, Yao Q, Long C, Yi P, Song J, Wu L, Wan W, Rao X, Lin Y, Wei G, Ying J, Hua F. Infiltration by monocytes of the central nervous system and its role in multiple sclerosis: reflections on therapeutic strategies. Neural Regen Res 2025; 20:779-793. [PMID: 38886942 PMCID: PMC11433895 DOI: 10.4103/nrr.nrr-d-23-01508] [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: 09/06/2023] [Revised: 12/12/2023] [Accepted: 02/18/2024] [Indexed: 06/20/2024] Open
Abstract
Mononuclear macrophage infiltration in the central nervous system is a prominent feature of neuroinflammation. Recent studies on the pathogenesis and progression of multiple sclerosis have highlighted the multiple roles of mononuclear macrophages in the neuroinflammatory process. Monocytes play a significant role in neuroinflammation, and managing neuroinflammation by manipulating peripheral monocytes stands out as an effective strategy for the treatment of multiple sclerosis, leading to improved patient outcomes. This review outlines the steps involved in the entry of myeloid monocytes into the central nervous system that are targets for effective intervention: the activation of bone marrow hematopoiesis, migration of monocytes in the blood, and penetration of the blood-brain barrier by monocytes. Finally, we summarize the different monocyte subpopulations and their effects on the central nervous system based on phenotypic differences. As activated microglia resemble monocyte-derived macrophages, it is important to accurately identify the role of monocyte-derived macrophages in disease. Depending on the roles played by monocyte-derived macrophages at different stages of the disease, several of these processes can be interrupted to limit neuroinflammation and improve patient prognosis. Here, we discuss possible strategies to target monocytes in neurological diseases, focusing on three key aspects of monocyte infiltration into the central nervous system, to provide new ideas for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Guangyong Zhang
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Qing Yao
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Chubing Long
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Pengcheng Yi
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Jiali Song
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Luojia Wu
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Wei Wan
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Xiuqin Rao
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Yue Lin
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Gen Wei
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Jun Ying
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Key Laboratory of Anesthesiology of Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Fuzhou Hua
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
8
|
Sharma T, Mehan S, Tiwari A, Khan Z, Gupta GD, Narula AS. Targeting Oligodendrocyte Dynamics and Remyelination: Emerging Therapies and Personalized Approaches in Multiple Sclerosis Management. Curr Neurovasc Res 2025; 21:359-417. [PMID: 39219420 DOI: 10.2174/0115672026336440240822063430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 01/01/1970] [Accepted: 07/12/2024] [Indexed: 09/04/2024]
Abstract
Multiple sclerosis (MS) is a progressive autoimmune condition that primarily affects young people and is characterized by demyelination and neurodegeneration of the central nervous system (CNS). This in-depth review explores the complex involvement of oligodendrocytes, the primary myelin- producing cells in the CNS, in the pathophysiology of MS. It discusses the biochemical processes and signalling pathways required for oligodendrocytes to function and remain alive, as well as how they might fail and cause demyelination to occur. We investigate developing therapeutic options that target remyelination, a fundamental component of MS treatment. Remyelination approaches promote the survival and differentiation of oligodendrocyte precursor cells (OPCs), restoring myelin sheaths. This improves nerve fibre function and may prevent MS from worsening. We examine crucial parameters influencing remyelination success, such as OPC density, ageing, and signalling pathway regulation (e.g., Retinoid X receptor, LINGO-1, Notch). The review also examines existing neuroprotective and antiinflammatory medications being studied to see if they can assist oligodendrocytes in surviving and reducing the severity of MS symptoms. The review focuses on medicines that target the myelin metabolism in oligodendrocytes. Altering oligodendrocyte metabolism has been linked to reversing demyelination and improving MS patient outcomes through various mechanisms. We also explore potential breakthroughs, including innovative antisense technologies, deep brain stimulation, and the impact of gut health and exercise on MS development. The article discusses the possibility of personalized medicine in MS therapy, emphasizing the importance of specific medicines based on individual molecular profiles. The study emphasizes the need for reliable biomarkers and improved imaging tools for monitoring disease progression and therapy response. Finally, this review focuses on the importance of oligodendrocytes in MS and the potential for remyelination therapy. It also underlines the importance of continued research to develop more effective treatment regimens, taking into account the complexities of MS pathology and the different factors that influence disease progression and treatment.
Collapse
Affiliation(s)
- Tarun Sharma
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Aarti Tiwari
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | | | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| |
Collapse
|
9
|
Kasheke GDS, Hendy BAM, Dorighello GG, Uccelli NA, Gothié JDM, Novorolsky RJ, Oulton MJ, Asainayagam J, Makarov AI, Fraser KS, Vuligonda V, Sanders ME, Kennedy TE, Robertson GS. Selective retinoid X receptor agonism promotes functional recovery and myelin repair in experimental autoimmune encephalomyelitis. Acta Neuropathol Commun 2024; 12:197. [PMID: 39707547 DOI: 10.1186/s40478-024-01904-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: 10/11/2024] [Accepted: 11/29/2024] [Indexed: 12/23/2024] Open
Abstract
Evidence that myelin repair is crucial for functional recovery in multiple sclerosis (MS) led to the identification of bexarotene (BXT). This clinically promising remyelinating agent activates multiple nuclear hormone receptor subtypes implicated in myelin repair. However, BXT produces unacceptable hyperlipidemia. In contrast, IRX4204 selectively activates the retinoid X receptor (RXR). Given compelling links between RXR activation and increased myelin repair, we employed IRX4204 to investigate the impact of RXR agonism alone on functional recovery in mice subjected to experimental autoimmune encephalomyelitis (EAE). Since gait deficits are common in MS, we used machine learning to obtain highly sensitive and reliable measurements of sagittal hindleg joint movements for mice walking on a treadmill. IRX4204 not only blocked the progressive loss of knee and ankle movements but also reversed joint movement impairments in EAE mice. Our biochemical, transcriptional and histological measurements in spinal cord suggest these gait improvements reflect increased axon survival and remyelination and reduced inflammation. Using microglia, astrocytes and oligodendrocyte progenitor cells, we present additional data suggesting that IRX4204 may act on multiple glial subtypes to orchestrate myelin repair. These results inform the discovery of restorative neural therapeutics for MS by demonstrating that selective RXR agonism is sufficient for effective myelin repair. Moreover, our findings support the therapeutic potential of IRX4204 to promote functional recovery in MS.
Collapse
Affiliation(s)
- Gracious D S Kasheke
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada
- Brain Repair Centre, Life Sciences Research Institute, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Basmah A M Hendy
- Brain Repair Centre, Life Sciences Research Institute, Dalhousie University, Halifax, NS, B3H 4R2, Canada
- Department of Psychology and Neuroscience, Faculty of Science, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Gabriel G Dorighello
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Nonthué A Uccelli
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Jean-David M Gothié
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, H3A 2B4, Canada
| | - Robyn J Novorolsky
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada
- Brain Repair Centre, Life Sciences Research Institute, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Madison J Oulton
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada
- Brain Repair Centre, Life Sciences Research Institute, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Jude Asainayagam
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada
- Brain Repair Centre, Life Sciences Research Institute, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Adam I Makarov
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada
- Brain Repair Centre, Life Sciences Research Institute, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | - Kaitlyn S Fraser
- Brain Repair Centre, Life Sciences Research Institute, Dalhousie University, Halifax, NS, B3H 4R2, Canada
- Department of Psychology and Neuroscience, Faculty of Science, Dalhousie University, Halifax, NS, B3H 4R2, Canada
| | | | | | - Timothy E Kennedy
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, H3A 2B4, Canada
| | - George S Robertson
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2, Canada.
- Brain Repair Centre, Life Sciences Research Institute, Dalhousie University, Halifax, NS, B3H 4R2, Canada.
- Department of Psychiatry, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 2E2, Canada.
- , 1348 Summer St, Halifax, NS, B3H 0A8, Canada.
| |
Collapse
|
10
|
Zhou Y, Xu T, Zhou Y, Han W, Wu Z, Yang C, Chen X. A review focuses on a neglected and controversial component of SCI: myelin debris. Front Immunol 2024; 15:1436031. [PMID: 39650659 PMCID: PMC11621000 DOI: 10.3389/fimmu.2024.1436031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 10/22/2024] [Indexed: 12/11/2024] Open
Abstract
Myelin sheath, as the multilayer dense structure enclosing axons in humans and other higher organisms, may rupture due to various injury factors after spinal cord injury, thus producing myelin debris. The myelin debris contains a variety of myelin-associated inhibitors (MAIs) and lipid, all inhibiting the repair after spinal cord injury. Through summary and analysis, the present authors found that the inhibition of myelin debris can be mainly divided into two categories: firstly, the direct inhibition mediated by MAIs; secondly, the indirect inhibition mediated by lipid such as cholesterol. It is worth noting that phagocytes are required in the latter indirect inhibition, such as professional phagocytes (macrophages et al.) and non-professional phagocytes (astrocytes et al.). Moreover, complement and the immune system also participate in the phagocytosis of myelin debris, working together with phagocytes to aggravate spinal cord injury. In conclusion, this paper focuses on the direct and indirect effects of myelin debris on spinal cord injury, aiming to provide new inspiration and reflection for the basic research of spinal cord injury and the conception of related treatment.
Collapse
Affiliation(s)
- Yuchen Zhou
- Department of Spine Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Tao Xu
- Medical School of Nantong University, Nantong, China
- Department of Orthopedics, Yancheng Dafeng People's Hospital, Yancheng, China
| | - Yiyan Zhou
- Department of Spine Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Wei Han
- Institute of Regenerative Biology and Medicine, Helmholtz Zentrum München, Munich, Germany
| | - Zhengchao Wu
- Department of Spine Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Changwei Yang
- Department of Spine Surgery, Affiliated Hospital of Nantong University, Nantong, China
- Medical School of Nantong University, Nantong, China
| | - Xiaoqing Chen
- Department of Spine Surgery, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
11
|
Jaing TH, Wang YL, Chiu CC. Time to Rethink Bronchiolitis Obliterans Syndrome Following Lung or Hematopoietic Cell Transplantation in Pediatric Patients. Cancers (Basel) 2024; 16:3715. [PMID: 39518153 PMCID: PMC11545638 DOI: 10.3390/cancers16213715] [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: 10/08/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Background: Similar in histological characteristics and clinical manifestations, bronchiolitis obliterans syndrome (BOS) can develop following lung transplantation (LTx) or hematopoietic cell transplantation (HCT). In contrast to lung transplantation, where BOS is restricted to the lung allograft, HCT-related systemic graft-versus-host disease (GVHD) is the root cause of BOS. Because lung function declines following HCT, diagnosis becomes more difficult. Given the lack of proven effective medicines, treatment is based on empirical evidence. Methods: Cross-disciplinary learning is crucial, and novel therapies are under investigation to improve survival and avoid LTx. Recent advances have focused on updating the understanding of the etiology, clinical features, and pathobiology of BOS. It emphasizes the significance of learning from experts in other transplant modalities, promoting cross-disciplinary knowledge. Results: Our treatment algorithms are derived from extensive research and expert clinical input. It is important to ensure that immunosuppression is optimized and that any other conditions or contributing factors are addressed, if possible. Clear treatment algorithms are provided for each condition, drawing from the published literature and consensus clinical opinion. There are several novel therapies currently being investigated, such as aerosolized liposomal cyclosporine, Janus kinase inhibitors, antifibrotic therapies, and B-cell-directed therapies. Conclusions: We urgently need innovative treatments that can greatly increase survival rates and eliminate the need for LTx or re-transplantation.
Collapse
Affiliation(s)
- Tang-Her Jaing
- Division of Hematology and Oncology, Department of Pediatrics, Chang Gung Memorial Hospital, 5 Fu-Shin Street, Kwei-Shan, Taoyuan 33315, Taiwan;
| | - Yi-Lun Wang
- Division of Hematology and Oncology, Department of Pediatrics, Chang Gung Memorial Hospital, 5 Fu-Shin Street, Kwei-Shan, Taoyuan 33315, Taiwan;
| | - Chia-Chi Chiu
- Division of Nursing, Chang Gung Memorial Hospital, 5 Fu-Shin Street, Kwei-Shan, Taoyuan 33315, Taiwan;
| |
Collapse
|
12
|
Holland C, Dravecz N, Owens L, Benedetto A, Dias I, Gow A, Broughton S. Understanding exogenous factors and biological mechanisms for cognitive frailty: A multidisciplinary scoping review. Ageing Res Rev 2024; 101:102461. [PMID: 39278273 DOI: 10.1016/j.arr.2024.102461] [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: 12/21/2023] [Revised: 07/15/2024] [Accepted: 08/15/2024] [Indexed: 09/18/2024]
Abstract
Cognitive frailty (CF) is the conjunction of cognitive impairment without dementia and physical frailty. While predictors of each element are well-researched, mechanisms of their co-occurrence have not been integrated, particularly in terms of relationships between social, psychological, and biological factors. This interdisciplinary scoping review set out to categorise a heterogenous multidisciplinary literature to identify potential pathways and mechanisms of CF, and research gaps. Studies were included if they used the definition of CF OR focused on conjunction of cognitive impairment and frailty (by any measure), AND excluded studies on specific disease populations, interventions, epidemiology or prediction of mortality. Searches used Web of Science, PubMed and Science Direct. Search terms included "cognitive frailty" OR (("cognitive decline" OR "cognitive impairment") AND (frail*)), with terms to elicit mechanisms, predictors, causes, pathways and risk factors. To ensure inclusion of animal and cell models, keywords such as "behavioural" or "cognitive decline" or "senescence", were added. 206 papers were included. Descriptive analysis provided high-level categorisation of determinants from social and environmental through psychological to biological. Patterns distinguishing CF from Alzheimer's disease were identified and social and psychological moderators and mediators of underlying biological and physiological changes and of trajectories of CF development were suggested as foci for further research.
Collapse
Affiliation(s)
- Carol Holland
- Division of Health Research, Health Innovation One, Sir John Fisher Drive, Lancaster University, Lancaster LA1 4YW, UK.
| | - Nikolett Dravecz
- Division of Health Research, Health Innovation One, Sir John Fisher Drive, Lancaster University, Lancaster LA1 4YW, UK.
| | - Lauren Owens
- Division of Biomedical and Life Sciences, Furness College, Lancaster University, LA1 4YG, UK.
| | - Alexandre Benedetto
- Division of Biomedical and Life Sciences, Furness College, Lancaster University, LA1 4YG, UK.
| | - Irundika Dias
- Aston University Medical School, Aston University, Birmingham B4 7ET, UK.
| | - Alan Gow
- Centre for Applied Behavioural Sciences, Department of Psychology, School of Social Sciences, Heriot-Watt University, Edinburgh EH14 4AS, UK.
| | - Susan Broughton
- Division of Biomedical and Life Sciences, Furness College, Lancaster University, LA1 4YG, UK.
| |
Collapse
|
13
|
Fransson J, Bachelin C, Ichou F, Guillot-Noël L, Ponnaiah M, Gloaguen A, Maillart E, Stankoff B, Tenenhaus A, Fontaine B, Mochel F, Louapre C, Zujovic V. Multiple Sclerosis Patient Macrophages Impaired Metabolism Leads to an Altered Response to Activation Stimuli. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200312. [PMID: 39467238 PMCID: PMC11521098 DOI: 10.1212/nxi.0000000000200312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 08/05/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND AND OBJECTIVES In multiple sclerosis (MS), immune cells invade the CNS and destroy myelin. Macrophages contribute to demyelination and myelin repair, and their role in each process depends on their ability to acquire specific phenotypes in response to external signals. In this article, we assess whether defects in MS patient macrophage responses may lead to increased inflammation or lack of neuroregenerative effects. METHODS CD14+CD16- monocytes from patients with MS and healthy controls (HCs) were activated in vitro to obtain homeostatic-like, proinflammatory, and proregenerative macrophages. Macrophage activation profiles were assessed through RNA sequencing and metabolomics. Surface molecule expression of CD14, CD16, and HLA-DR and myelin phagocytic capacity were evaluated with flow cytometry. Macrophage supernatant capacity to influence oligodendrocyte precursor cell differentiation toward an astrocytic or oligodendroglia fate was also tested. RESULTS We observed that MS patient monocytes ex vivo recapitulate their preferential activation toward the CD16+ phenotype, a subset of proinflammatory cells overrepresented in MS lesions. Functionally, MS patient macrophages display a decreased capacity to phagocytose human myelin and a deficit of processing myelin after ingestion. In addition, MS patient macrophage supernatant favors astrocytes over oligodendrocyte differentiation when compared with HC macrophage supernatant. Furthermore, even when exposed to homeostatic or proregenerative stimuli, MS patient macrophages uphold a proinflammatory transcriptomic profile with higher levels of cytokine/chemokine. Of interest, MS patient macrophages exhibit a distinct metabolic signature with a mitochondrial energy metabolism blockage. Transcriptomic data are further substantiated by metabolomics studies that reveal perturbations in the corresponding metabolic pathways. DISCUSSION Our results show an intrinsic defect of MS patient macrophages, reminiscent of innate immune cell memory in MS, lifting macrophage importance in the disease and as potential therapeutic targets.
Collapse
Affiliation(s)
- Jennifer Fransson
- From the Sorbonne Université (J.F., C.B., L.G.-N., E.M., A.T., F.M., C.L., V.Z.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital Pitié Salpétrière Univ. Hosp., DMU Neuroscience 6; Inst. of Cardiometabolism and Nutrition (F.I., M.P.), Sorbonne-universités-Upmc 06, INSERM, CNRS; Laboratoire des Signaux et Systèmes (L2S) (A.G., A.T.), CNRS-CentraleSupélec, Université Paris-Saclay; Sorbonne Université (B.S.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital St. Antoine-HUEP; and INSERM (B.F.), SU, AP-HP, Centre de recherche en Myologie-UMR974 and Service of Neuro-Myology, Institute of Myology, University hospital Pitié-Salpêtriere
| | - Corinne Bachelin
- From the Sorbonne Université (J.F., C.B., L.G.-N., E.M., A.T., F.M., C.L., V.Z.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital Pitié Salpétrière Univ. Hosp., DMU Neuroscience 6; Inst. of Cardiometabolism and Nutrition (F.I., M.P.), Sorbonne-universités-Upmc 06, INSERM, CNRS; Laboratoire des Signaux et Systèmes (L2S) (A.G., A.T.), CNRS-CentraleSupélec, Université Paris-Saclay; Sorbonne Université (B.S.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital St. Antoine-HUEP; and INSERM (B.F.), SU, AP-HP, Centre de recherche en Myologie-UMR974 and Service of Neuro-Myology, Institute of Myology, University hospital Pitié-Salpêtriere
| | - Farid Ichou
- From the Sorbonne Université (J.F., C.B., L.G.-N., E.M., A.T., F.M., C.L., V.Z.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital Pitié Salpétrière Univ. Hosp., DMU Neuroscience 6; Inst. of Cardiometabolism and Nutrition (F.I., M.P.), Sorbonne-universités-Upmc 06, INSERM, CNRS; Laboratoire des Signaux et Systèmes (L2S) (A.G., A.T.), CNRS-CentraleSupélec, Université Paris-Saclay; Sorbonne Université (B.S.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital St. Antoine-HUEP; and INSERM (B.F.), SU, AP-HP, Centre de recherche en Myologie-UMR974 and Service of Neuro-Myology, Institute of Myology, University hospital Pitié-Salpêtriere
| | - Léna Guillot-Noël
- From the Sorbonne Université (J.F., C.B., L.G.-N., E.M., A.T., F.M., C.L., V.Z.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital Pitié Salpétrière Univ. Hosp., DMU Neuroscience 6; Inst. of Cardiometabolism and Nutrition (F.I., M.P.), Sorbonne-universités-Upmc 06, INSERM, CNRS; Laboratoire des Signaux et Systèmes (L2S) (A.G., A.T.), CNRS-CentraleSupélec, Université Paris-Saclay; Sorbonne Université (B.S.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital St. Antoine-HUEP; and INSERM (B.F.), SU, AP-HP, Centre de recherche en Myologie-UMR974 and Service of Neuro-Myology, Institute of Myology, University hospital Pitié-Salpêtriere
| | - Maharajah Ponnaiah
- From the Sorbonne Université (J.F., C.B., L.G.-N., E.M., A.T., F.M., C.L., V.Z.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital Pitié Salpétrière Univ. Hosp., DMU Neuroscience 6; Inst. of Cardiometabolism and Nutrition (F.I., M.P.), Sorbonne-universités-Upmc 06, INSERM, CNRS; Laboratoire des Signaux et Systèmes (L2S) (A.G., A.T.), CNRS-CentraleSupélec, Université Paris-Saclay; Sorbonne Université (B.S.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital St. Antoine-HUEP; and INSERM (B.F.), SU, AP-HP, Centre de recherche en Myologie-UMR974 and Service of Neuro-Myology, Institute of Myology, University hospital Pitié-Salpêtriere
| | - Arnaud Gloaguen
- From the Sorbonne Université (J.F., C.B., L.G.-N., E.M., A.T., F.M., C.L., V.Z.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital Pitié Salpétrière Univ. Hosp., DMU Neuroscience 6; Inst. of Cardiometabolism and Nutrition (F.I., M.P.), Sorbonne-universités-Upmc 06, INSERM, CNRS; Laboratoire des Signaux et Systèmes (L2S) (A.G., A.T.), CNRS-CentraleSupélec, Université Paris-Saclay; Sorbonne Université (B.S.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital St. Antoine-HUEP; and INSERM (B.F.), SU, AP-HP, Centre de recherche en Myologie-UMR974 and Service of Neuro-Myology, Institute of Myology, University hospital Pitié-Salpêtriere
| | - Elisabeth Maillart
- From the Sorbonne Université (J.F., C.B., L.G.-N., E.M., A.T., F.M., C.L., V.Z.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital Pitié Salpétrière Univ. Hosp., DMU Neuroscience 6; Inst. of Cardiometabolism and Nutrition (F.I., M.P.), Sorbonne-universités-Upmc 06, INSERM, CNRS; Laboratoire des Signaux et Systèmes (L2S) (A.G., A.T.), CNRS-CentraleSupélec, Université Paris-Saclay; Sorbonne Université (B.S.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital St. Antoine-HUEP; and INSERM (B.F.), SU, AP-HP, Centre de recherche en Myologie-UMR974 and Service of Neuro-Myology, Institute of Myology, University hospital Pitié-Salpêtriere
| | - Bruno Stankoff
- From the Sorbonne Université (J.F., C.B., L.G.-N., E.M., A.T., F.M., C.L., V.Z.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital Pitié Salpétrière Univ. Hosp., DMU Neuroscience 6; Inst. of Cardiometabolism and Nutrition (F.I., M.P.), Sorbonne-universités-Upmc 06, INSERM, CNRS; Laboratoire des Signaux et Systèmes (L2S) (A.G., A.T.), CNRS-CentraleSupélec, Université Paris-Saclay; Sorbonne Université (B.S.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital St. Antoine-HUEP; and INSERM (B.F.), SU, AP-HP, Centre de recherche en Myologie-UMR974 and Service of Neuro-Myology, Institute of Myology, University hospital Pitié-Salpêtriere
| | - Arthur Tenenhaus
- From the Sorbonne Université (J.F., C.B., L.G.-N., E.M., A.T., F.M., C.L., V.Z.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital Pitié Salpétrière Univ. Hosp., DMU Neuroscience 6; Inst. of Cardiometabolism and Nutrition (F.I., M.P.), Sorbonne-universités-Upmc 06, INSERM, CNRS; Laboratoire des Signaux et Systèmes (L2S) (A.G., A.T.), CNRS-CentraleSupélec, Université Paris-Saclay; Sorbonne Université (B.S.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital St. Antoine-HUEP; and INSERM (B.F.), SU, AP-HP, Centre de recherche en Myologie-UMR974 and Service of Neuro-Myology, Institute of Myology, University hospital Pitié-Salpêtriere
| | - Bertrand Fontaine
- From the Sorbonne Université (J.F., C.B., L.G.-N., E.M., A.T., F.M., C.L., V.Z.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital Pitié Salpétrière Univ. Hosp., DMU Neuroscience 6; Inst. of Cardiometabolism and Nutrition (F.I., M.P.), Sorbonne-universités-Upmc 06, INSERM, CNRS; Laboratoire des Signaux et Systèmes (L2S) (A.G., A.T.), CNRS-CentraleSupélec, Université Paris-Saclay; Sorbonne Université (B.S.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital St. Antoine-HUEP; and INSERM (B.F.), SU, AP-HP, Centre de recherche en Myologie-UMR974 and Service of Neuro-Myology, Institute of Myology, University hospital Pitié-Salpêtriere
| | - Fanny Mochel
- From the Sorbonne Université (J.F., C.B., L.G.-N., E.M., A.T., F.M., C.L., V.Z.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital Pitié Salpétrière Univ. Hosp., DMU Neuroscience 6; Inst. of Cardiometabolism and Nutrition (F.I., M.P.), Sorbonne-universités-Upmc 06, INSERM, CNRS; Laboratoire des Signaux et Systèmes (L2S) (A.G., A.T.), CNRS-CentraleSupélec, Université Paris-Saclay; Sorbonne Université (B.S.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital St. Antoine-HUEP; and INSERM (B.F.), SU, AP-HP, Centre de recherche en Myologie-UMR974 and Service of Neuro-Myology, Institute of Myology, University hospital Pitié-Salpêtriere
| | - Celine Louapre
- From the Sorbonne Université (J.F., C.B., L.G.-N., E.M., A.T., F.M., C.L., V.Z.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital Pitié Salpétrière Univ. Hosp., DMU Neuroscience 6; Inst. of Cardiometabolism and Nutrition (F.I., M.P.), Sorbonne-universités-Upmc 06, INSERM, CNRS; Laboratoire des Signaux et Systèmes (L2S) (A.G., A.T.), CNRS-CentraleSupélec, Université Paris-Saclay; Sorbonne Université (B.S.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital St. Antoine-HUEP; and INSERM (B.F.), SU, AP-HP, Centre de recherche en Myologie-UMR974 and Service of Neuro-Myology, Institute of Myology, University hospital Pitié-Salpêtriere
| | - Violetta Zujovic
- From the Sorbonne Université (J.F., C.B., L.G.-N., E.M., A.T., F.M., C.L., V.Z.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital Pitié Salpétrière Univ. Hosp., DMU Neuroscience 6; Inst. of Cardiometabolism and Nutrition (F.I., M.P.), Sorbonne-universités-Upmc 06, INSERM, CNRS; Laboratoire des Signaux et Systèmes (L2S) (A.G., A.T.), CNRS-CentraleSupélec, Université Paris-Saclay; Sorbonne Université (B.S.), Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital St. Antoine-HUEP; and INSERM (B.F.), SU, AP-HP, Centre de recherche en Myologie-UMR974 and Service of Neuro-Myology, Institute of Myology, University hospital Pitié-Salpêtriere
| |
Collapse
|
14
|
Li L, Zhang T, Xiao M, Lu Y, Gao L. Brain macrophage senescence in glioma. Semin Cancer Biol 2024; 104-105:46-60. [PMID: 39098625 DOI: 10.1016/j.semcancer.2024.07.005] [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: 06/07/2024] [Revised: 07/20/2024] [Accepted: 07/29/2024] [Indexed: 08/06/2024]
Abstract
Gliomas are a diverse group of primary central nervous system neoplasms with no curative therapies available. Brain macrophages comprise microglia in the brain parenchyma, border-associated macrophages in the meningeal-choroid plexus-perivascular space and monocyte-derived macrophages infiltrating the brain. With the great improvement of our recognition of brain macrophages, diverse macrophage populations have been found in the context of glioma, which exhibit functional and phenotypic heterogeneity. We have long thought that brain macrophage senescence is detrimental, manifested by specialized forms of persistent cell cycle arrest and chronic low-grade inflammation. Persistent senescence of macrophages may result in immune dysfunction, potentially contributing to glioma initiation and development. Given the crucial roles played by brain macrophages in glioma, we unravel how brain macrophages undergo reprogramming and their contribution to glioma. We outline general molecular alterations and specific biomarkers in senescent brain macrophages, as well as functional changes (such as metabolism, autophagy, phagocytosis, antigen presentation, and infiltration and recruitment). In addition, recent advances in genetic regulation and mechanisms linked to senescent brain macrophages are discussed. In particular, this review emphasizes the contribution of senescent brain macrophages to glioma, which may drive translational efforts to utilize brain macrophages as a prognostic marker or/and treatment target in glioma. An in-depth comprehending of how brain macrophage senescence functionally influences the tumor microenvironment will be key to our development of innovative therapeutics for glioma.
Collapse
Affiliation(s)
- Lu Li
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Tianhe Zhang
- Department of Neurosurgery, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, Liaoning 110016, China
| | - Meiling Xiao
- Department of Rehabilitation, The Central Hospital of Shenyang Medical College, Shenyang, Liaoning 110024, China
| | - Yu Lu
- Rehabilitation Medicine Department, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, Liaoning 110016, China.
| | - Lin Gao
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| |
Collapse
|
15
|
Chen S, Tan Y, Tian L. Immunophenotypes in psychosis: is it a premature inflamm-aging disorder? Mol Psychiatry 2024; 29:2834-2848. [PMID: 38532012 PMCID: PMC11420084 DOI: 10.1038/s41380-024-02539-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 03/28/2024]
Abstract
Immunopsychiatric field has rapidly accumulated evidence demonstrating the involvement of both innate and adaptive immune components in psychotic disorders such as schizophrenia. Nevertheless, researchers are facing dilemmas of discrepant findings of immunophenotypes both outside and inside the brains of psychotic patients, as discovered by recent meta-analyses. These discrepancies make interpretations and interrogations on their roles in psychosis remain vague and even controversial, regarding whether certain immune cells are more activated or less so, and whether they are causal or consequential, or beneficial or harmful for psychosis. Addressing these issues for psychosis is not at all trivial, as immune cells either outside or inside the brain are an enormously heterogeneous and plastic cell population, falling into a vast range of lineages and subgroups, and functioning differently and malleably in context-dependent manners. This review aims to overview the currently known immunophenotypes of patients with psychosis, and provocatively suggest the premature immune "burnout" or inflamm-aging initiated since organ development as a potential primary mechanism behind these immunophenotypes and the pathogenesis of psychotic disorders.
Collapse
Affiliation(s)
- Song Chen
- Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, PR China
| | - Yunlong Tan
- Peking University HuiLongGuan Clinical Medical School, Beijing Huilongguan Hospital, Beijing, PR China
| | - Li Tian
- Department of Psychology and Logopedics, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
16
|
Wang H, Guo Y, Zhang X, Zhou X. Bexarotene Induce Differentiation of Myeloid-Derived Suppressor Cells through Arg-1 Signalling Pathway. Transplant Proc 2024; 56:1469-1477. [PMID: 38981763 DOI: 10.1016/j.transproceed.2024.03.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 02/21/2024] [Accepted: 03/18/2024] [Indexed: 07/11/2024]
Abstract
BACKGROUND Cellular therapy has emerged as a promising strategy to minimize the use of conventional immunosuppressive drugs and ultimately induce long-term graft survival. Myeloid-derived suppressor cells (MDSCs) can be used for immunosuppressive treatment of solid organ transplants. METHODS Granular macrophage colony-stimulating factor (GM-CSF) and bexarotene, an X receptor-selective retinoid, were used for in vitro MDSC induction. Cell phenotypes were detected using flow cytometry, while mRNA was detected via real-time PCR. A mouse skin transplantation model was used to verify the inhibitory effects of this treatment. RESULTS The combination of GM-CSF and bexarotene-induced MDSC differentiation. MDSCs induce immune tolerance by inhibiting T-cell proliferation, influencing cytokine secretion, and inducing T-cell transformation into Treg cells. Combination treatment significantly up-regulated Arg-1 expression in MDSCs. The Arg-1 inhibitor nor-NOHA neutralized the immunosuppressive activity of MDSCs, suggesting the involvement of Arg-1 in MDSC-mediated immunosuppression. GM-CSF and bexarotene-induced MDSCs prolong graft survival in mouse skin transplants, exhibiting in vivo immunosuppressive effects. CONCLUSIONS A new method for inducing MDSCs is presented. The combination of GM-CSF and bexarotene induces MDSCs with remarkable regulatory functions. Adoptive transfer of the induced MDSCs extended allograft survival. These results suggest that MDSCs can potentially be used in future clinical transplants to inhibit rejection, reduce adverse events, and induce operative tolerance.
Collapse
Affiliation(s)
- Haozhou Wang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yinan Guo
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xiaodong Zhang
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Xiaoguang Zhou
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
17
|
Di Pietro AA, Pasquini LA. A novel in vitro model for investigating oligodendroglial maturation and myelin deposition under demyelinating and remyelinating conditions: Impact of microglial depletion and repopulation. Mol Cell Neurosci 2024; 129:103937. [PMID: 38796120 DOI: 10.1016/j.mcn.2024.103937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/14/2024] [Accepted: 05/17/2024] [Indexed: 05/28/2024] Open
Abstract
Experimental models of multiple sclerosis (MS) have significantly contributed to our understanding of pathophysiology and the development of therapeutic interventions. Various in vivo animal models have successfully replicated key features of MS and associated pathophysiological processes, shedding light on the sequence of events leading to disease initiation, progression, and resolution. Nevertheless, these models often entail substantial costs and prolonged treatment periods. In contrast, in vitro models offer distinct advantages, including cost-effectiveness and precise control over experimental conditions, thereby facilitating more reproducible results. We have developed a novel in vitro model tailored to the study of oligodendroglial maturation and myelin deposition under demyelinating and remyelinating conditions, which encompasses all the cell types present in the central nervous system (CNS). Of note, our model enables the evaluation of microglial cell commitment through a protocol involving their depletion and subsequent repopulation. Given that the development and survival of microglia are critically reliant on colony-stimulating factor-1 receptor (CSF-1R) signaling, we have employed CSF-1R inhibition to effectively deplete microglia. This versatile model holds promise for the assessment of potential therapies aimed at promoting oligodendroglial differentiation to safeguard and repair myelin, hence mitigate neurodegenerative processes.
Collapse
Affiliation(s)
- Anabella Ayelen Di Pietro
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Cátedra de Química Biológica Patológica, Buenos Aires, Argentina; Universidad de Buenos Airess, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini, Facultad de Farmacia y Bioquímica, Buenos Aire, Argentina.
| | - Laura Andrea Pasquini
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Cátedra de Química Biológica Patológica, Buenos Aires, Argentina; Universidad de Buenos Airess, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas Prof. Dr. Alejandro C. Paladini, Facultad de Farmacia y Bioquímica, Buenos Aire, Argentina.
| |
Collapse
|
18
|
Bánáti D, Hellman-Regen J, Mack I, Young HA, Benton D, Eggersdorfer M, Rohn S, Dulińska-Litewka J, Krężel W, Rühl R. Defining a vitamin A5/X specific deficiency - vitamin A5/X as a critical dietary factor for mental health. INT J VITAM NUTR RES 2024; 94:443-475. [PMID: 38904956 DOI: 10.1024/0300-9831/a000808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
A healthy and balanced diet is an important factor to assure a good functioning of the central and peripheral nervous system. Retinoid X receptor (RXR)-mediated signaling was identified as an important mechanism of transmitting major diet-dependent physiological and nutritional signaling such as the control of myelination and dopamine signalling. Recently, vitamin A5/X, mainly present in vegetables as provitamin A5/X, was identified as a new concept of a vitamin which functions as the nutritional precursor for enabling RXR-mediated signaling. The active form of vitamin A5/X, 9-cis-13,14-dehydroretinoic acid (9CDHRA), induces RXR-activation, thereby acting as the central switch for enabling various heterodimer-RXR-signaling cascades involving various partner heterodimers like the fatty acid and eicosanoid receptors/peroxisome proliferator-activated receptors (PPARs), the cholesterol receptors/liver X receptors (LXRs), the vitamin D receptor (VDR), and the vitamin A(1) receptors/retinoic acid receptors (RARs). Thus, nutritional supply of vitamin A5/X might be a general nutritional-dependent switch for enabling this large cascade of hormonal signaling pathways and thus appears important to guarantee an overall organism homeostasis. RXR-mediated signaling was shown to be dependent on vitamin A5/X with direct effects for beneficial physiological and neuro-protective functions mediated systemically or directly in the brain. In summary, through control of dopamine signaling, amyloid β-clearance, neuro-protection and neuro-inflammation, the vitamin A5/X - RXR - RAR - vitamin A(1)-signaling might be "one of" or even "the" critical factor(s) necessary for good mental health, healthy brain aging, as well as for preventing drug addiction and prevention of a large array of nervous system diseases. Likewise, vitamin A5/X - RXR - non-RAR-dependent signaling relevant for myelination/re-myelination and phagocytosis/brain cleanup will contribute to such regulations too. In this review we discuss the basic scientific background, logical connections and nutritional/pharmacological expert recommendations for the nervous system especially considering the ageing brain.
Collapse
Affiliation(s)
- Diána Bánáti
- Department of Food Engineering, Faculty of Engineering, University of Szeged, Hungary
| | - Julian Hellman-Regen
- Department of Psychiatry, Charité-Campus Benjamin Franklin, Section Neurobiology, University Medicine Berlin, Germany
| | - Isabelle Mack
- Department of Psychosomatic Medicine and Psychotherapy, University Hospital Tübingen, Germany
| | - Hayley A Young
- Faculty of Medicine, Health and Life Sciences, Swansea University, UK
| | - David Benton
- Faculty of Medicine, Health and Life Sciences, Swansea University, UK
| | - Manfred Eggersdorfer
- Department of Healthy Ageing, University Medical Center Groningen (UMCG), The Netherlands
| | - Sascha Rohn
- Department of Food Chemistry and Analysis, Institute of Food Technology and Food Chemistry, Technische Universität Berlin, Germany
| | | | - Wojciech Krężel
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Inserm U1258, CNRS UMR 7104, Université de Strasbourg, Illkirch, France
| | | |
Collapse
|
19
|
Moss CE, Johnston SA, Kimble JV, Clements M, Codd V, Hamby S, Goodall AH, Deshmukh S, Sudbery I, Coca D, Wilson HL, Kiss-Toth E. Aging-related defects in macrophage function are driven by MYC and USF1 transcriptional programs. Cell Rep 2024; 43:114073. [PMID: 38578825 DOI: 10.1016/j.celrep.2024.114073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/15/2024] [Accepted: 03/21/2024] [Indexed: 04/07/2024] Open
Abstract
Macrophages are central innate immune cells whose function declines with age. The molecular mechanisms underlying age-related changes remain poorly understood, particularly in human macrophages. We report a substantial reduction in phagocytosis, migration, and chemotaxis in human monocyte-derived macrophages (MDMs) from older (>50 years old) compared with younger (18-30 years old) donors, alongside downregulation of transcription factors MYC and USF1. In MDMs from young donors, knockdown of MYC or USF1 decreases phagocytosis and chemotaxis and alters the expression of associated genes, alongside adhesion and extracellular matrix remodeling. A concordant dysregulation of MYC and USF1 target genes is also seen in MDMs from older donors. Furthermore, older age and loss of either MYC or USF1 in MDMs leads to an increased cell size, altered morphology, and reduced actin content. Together, these results define MYC and USF1 as key drivers of MDM age-related functional decline and identify downstream targets to improve macrophage function in aging.
Collapse
Affiliation(s)
- Charlotte E Moss
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK; Healthy Lifespan Institute, University of Sheffield, Sheffield, UK
| | - Simon A Johnston
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Joshua V Kimble
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK; Healthy Lifespan Institute, University of Sheffield, Sheffield, UK
| | - Martha Clements
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Veryan Codd
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK; National Institute for Healthcare Research, Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Stephen Hamby
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK; National Institute for Healthcare Research, Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Alison H Goodall
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK; National Institute for Healthcare Research, Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Sumeet Deshmukh
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Ian Sudbery
- School of Biosciences, University of Sheffield, Sheffield, UK
| | - Daniel Coca
- Healthy Lifespan Institute, University of Sheffield, Sheffield, UK; Department of Autonomic Control and Systems Engineering, University of Sheffield, Sheffield, UK
| | - Heather L Wilson
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK; Healthy Lifespan Institute, University of Sheffield, Sheffield, UK.
| | - Endre Kiss-Toth
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK; Healthy Lifespan Institute, University of Sheffield, Sheffield, UK; Biological Research Centre, Szeged, Hungary.
| |
Collapse
|
20
|
Goyne CE, Fair AE, Sumowski PE, Graves JS. The Impact of Aging on Multiple Sclerosis. Curr Neurol Neurosci Rep 2024; 24:83-93. [PMID: 38416310 DOI: 10.1007/s11910-024-01333-2] [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] [Accepted: 01/17/2024] [Indexed: 02/29/2024]
Abstract
PURPOSE OF REVIEW Multiple sclerosis (MS) is a chronic, immune-mediated demyelinating disorder of the central nervous system. Age is one of the most important factors in determining MS phenotype. This review provides an overview of how age influences MS clinical characteristics, pathology, and treatment. RECENT FINDINGS New methods for measuring aging have improved our understanding of the aging process in MS. New studies have characterized the molecular and cellular composition of chronic active or smoldering plaques in MS. These lesions are important contributors to disability progression in MS. These studies highlight the important role of immunosenescence and the innate immune system in sustaining chronic inflammation. Given these changes in immune function, several studies have assessed optimal treatment strategies in aging individuals with MS. MS phenotype is intimately linked with chronologic age and immunosenescence. While there are many unanswered questions, there has been much progress in understanding this relationship which may lead to more effective treatments for progressive disease.
Collapse
Affiliation(s)
- Christopher E Goyne
- Department of Neurosciences, University of California San Diego, 9452 Medical Center Drive, Ste 4W-222, La Jolla, San Diego, CA, 92037, USA
| | - Ashley E Fair
- Department of Neurosciences, University of California San Diego, 9452 Medical Center Drive, Ste 4W-222, La Jolla, San Diego, CA, 92037, USA
| | - Paige E Sumowski
- Department of Neurosciences, University of California San Diego, 9452 Medical Center Drive, Ste 4W-222, La Jolla, San Diego, CA, 92037, USA
| | - Jennifer S Graves
- Department of Neurosciences, University of California San Diego, 9452 Medical Center Drive, Ste 4W-222, La Jolla, San Diego, CA, 92037, USA.
| |
Collapse
|
21
|
Rokach M, Portioli C, Brahmachari S, Estevão BM, Decuzzi P, Barak B. Tackling myelin deficits in neurodevelopmental disorders using drug delivery systems. Adv Drug Deliv Rev 2024; 207:115218. [PMID: 38403255 DOI: 10.1016/j.addr.2024.115218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/27/2024] [Accepted: 02/20/2024] [Indexed: 02/27/2024]
Abstract
Interest in myelin and its roles in almost all brain functions has been greatly increasing in recent years, leading to countless new studies on myelination, as a dominant process in the development of cognitive functions. Here, we explore the unique role myelin plays in the central nervous system and specifically discuss the results of altered myelination in neurodevelopmental disorders. We present parallel developmental trajectories involving myelination that correlate with the onset of cognitive impairment in neurodevelopmental disorders and discuss the key challenges in the treatment of these chronic disorders. Recent developments in drug repurposing and nano/micro particle-based therapies are reviewed as a possible pathway to circumvent some of the main hurdles associated with early intervention, including patient's adherence and compliance, side effects, relapse, and faster route to possible treatment of these disorders. The strategy of drug encapsulation overcomes drug solubility and metabolism, with the possibility of drug targeting to a specific compartment, reducing side effects upon systemic administration.
Collapse
Affiliation(s)
- May Rokach
- Sagol School of Neuroscience, Tel-Aviv University, Israel
| | - Corinne Portioli
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Sayanti Brahmachari
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Bianca Martins Estevão
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genova, Italy
| | - Boaz Barak
- Sagol School of Neuroscience, Tel-Aviv University, Israel; Faculty of Social Sciences, The School of Psychological Sciences, Tel-Aviv University, Israel.
| |
Collapse
|
22
|
Dong X, Zhang Z, Shu X, Zhuang Z, Liu P, Liu R, Xia S, Bao X, Xu Y, Chen Y. MFG-E8 Alleviates Cognitive Impairments Induced by Chronic Cerebral Hypoperfusion by Phagocytosing Myelin Debris and Promoting Remyelination. Neurosci Bull 2024; 40:483-499. [PMID: 37979054 PMCID: PMC11003935 DOI: 10.1007/s12264-023-01147-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 06/22/2023] [Indexed: 11/19/2023] Open
Abstract
Chronic cerebral hypoperfusion is one of the pathophysiological mechanisms contributing to cognitive decline by causing white matter injury. Microglia phagocytosing myelin debris in a timely manner can promote remyelination and contribute to the repair of white matter. However, milk fat globule-epidermal growth factor-factor 8 (MFG-E8), a microglial phagocytosis-related protein, has not been well studied in hypoperfusion-related cognitive dysfunction. We found that the expression of MFG-E8 was significantly decreased in the brain of mice after bilateral carotid artery stenosis (BCAS). MFG-E8 knockout mice demonstrated more severe BCAS-induced cognitive impairments in the behavioral tests. In addition, we discovered that the deletion of MFG-E8 aggravated white matter damage and the destruction of myelin microstructure through fluorescent staining and electron microscopy. Meanwhile, MFG-E8 overexpression by AAV improved white matter injury and increased the number of mature oligodendrocytes after BCAS. Moreover, in vitro and in vivo experiments showed that MFG-E8 could enhance the phagocytic function of microglia via the αVβ3/αVβ5/Rac1 pathway and IGF-1 production to promote the differentiation of oligodendrocyte progenitor cells into mature oligodendrocytes. Interestingly, we found that MFG-E8 was mainly derived from astrocytes, not microglia. Our findings suggest that MFG-E8 is a potential therapeutic target for cognitive impairments following cerebral hypoperfusion.
Collapse
Affiliation(s)
- Xiaohong Dong
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
| | - Zhi Zhang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
| | - Xin Shu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China
| | - Zi Zhuang
- Department of Neurology, Drum Tower Hospital of Nanjing Medical University, Nanjing, 210008, China
| | - Pinyi Liu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, 210008, China
| | - Renyuan Liu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, 210008, China
| | - Shengnan Xia
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, 210008, China
| | - Xinyu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, 210008, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210008, China.
- Department of Neurology, Drum Tower Hospital of Nanjing Medical University, Nanjing, 210008, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China.
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, 210008, China.
| | - Yan Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, 210008, China.
- Jiangsu Provincial Key Discipline of Neurology, Nanjing, 210008, China.
| |
Collapse
|
23
|
Wang L, Hong W, Zhu H, He Q, Yang B, Wang J, Weng Q. Macrophage senescence in health and diseases. Acta Pharm Sin B 2024; 14:1508-1524. [PMID: 38572110 PMCID: PMC10985037 DOI: 10.1016/j.apsb.2024.01.008] [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: 08/19/2023] [Revised: 10/16/2023] [Accepted: 12/06/2023] [Indexed: 04/05/2024] Open
Abstract
Macrophage senescence, manifested by the special form of durable cell cycle arrest and chronic low-grade inflammation like senescence-associated secretory phenotype, has long been considered harmful. Persistent senescence of macrophages may lead to maladaptation, immune dysfunction, and finally the development of age-related diseases, infections, autoimmune diseases, and malignancies. However, it is a ubiquitous, multi-factorial, and dynamic complex phenomenon that also plays roles in remodeled processes, including wound repair and embryogenesis. In this review, we summarize some general molecular changes and several specific biomarkers during macrophage senescence, which may bring new sight to recognize senescent macrophages in different conditions. Also, we take an in-depth look at the functional changes in senescent macrophages, including metabolism, autophagy, polarization, phagocytosis, antigen presentation, and infiltration or recruitment. Furthermore, some degenerations and diseases associated with senescent macrophages as well as the mechanisms or relevant genetic regulations of senescent macrophages are integrated, not only emphasizing the possibility of regulating macrophage senescence to benefit age-associated diseases but also has an implication on the finding of potential targets or drugs clinically.
Collapse
Affiliation(s)
- Longling Wang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
| | - Wenxiang Hong
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hong Zhu
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
| | - Bo Yang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Wang
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
- Taizhou Institute of Zhejiang University, Taizhou 318000, China
| | - Qinjie Weng
- Center for Drug Safety Evaluation and Research, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
- Taizhou Institute of Zhejiang University, Taizhou 318000, China
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| |
Collapse
|
24
|
de la Fuente AG, Dittmer M, Heesbeen EJ, de la Vega Gallardo N, White JA, Young A, McColgan T, Dashwood A, Mayne K, Cabeza-Fernández S, Falconer J, Rodriguez-Baena FJ, McMurran CE, Inayatullah M, Rawji KS, Franklin RJM, Dooley J, Liston A, Ingram RJ, Tiwari VK, Penalva R, Dombrowski Y, Fitzgerald DC. Ageing impairs the regenerative capacity of regulatory T cells in mouse central nervous system remyelination. Nat Commun 2024; 15:1870. [PMID: 38467607 PMCID: PMC10928230 DOI: 10.1038/s41467-024-45742-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 01/31/2024] [Indexed: 03/13/2024] Open
Abstract
Myelin regeneration (remyelination) is essential to prevent neurodegeneration in demyelinating diseases such as Multiple Sclerosis, however, its efficiency declines with age. Regulatory T cells (Treg) recently emerged as critical players in tissue regeneration, including remyelination. However, the effect of ageing on Treg-mediated regenerative processes is poorly understood. Here, we show that expansion of aged Treg does not rescue age-associated remyelination impairment due to an intrinsically diminished capacity of aged Treg to promote oligodendrocyte differentiation and myelination in male and female mice. This decline in regenerative Treg functions can be rescued by a young environment. We identified Melanoma Cell Adhesion Molecule 1 (MCAM1) and Integrin alpha 2 (ITGA2) as candidates of Treg-mediated oligodendrocyte differentiation that decrease with age. Our findings demonstrate that ageing limits the neuroregenerative capacity of Treg, likely limiting their remyelinating therapeutic potential in aged patients, and describe two mechanisms implicated in Treg-driven remyelination that may be targetable to overcome this limitation.
Collapse
Affiliation(s)
- Alerie Guzman de la Fuente
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK.
- Institute for Health and Biomedical Sciences of Alicante (ISABIAL), Alicante, 03010, Spain.
- Instituto de Neurosciencias CSIC-UMH, San Juan de Alicante, Alicante, 03550, Spain.
| | - Marie Dittmer
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Elise J Heesbeen
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
- Division of Pharmacology, Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Nira de la Vega Gallardo
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Jessica A White
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Andrew Young
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Tiree McColgan
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Amy Dashwood
- Department of Pathology, University of Cambridge, CB2 1QP, Cambridge, UK
- Babraham Institute, CB22 3AT, Cambridge, UK
| | - Katie Mayne
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Sonia Cabeza-Fernández
- Institute for Health and Biomedical Sciences of Alicante (ISABIAL), Alicante, 03010, Spain
- Instituto de Neurosciencias CSIC-UMH, San Juan de Alicante, Alicante, 03550, Spain
| | - John Falconer
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
- CRUK Beatson Institute, G61 1BD, Glasgow, UK
| | | | - Christopher E McMurran
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Department of Medical Epidemiology and Biostatistics, Karolinska Institute, Stockholm, Sweden
| | - Mohammed Inayatullah
- Institute of Molecular Medicine, University of Southern Denmark, 5000, Odense, Denmark
- Danish Institute for Advanced Study (DIAS), 5230, Odense, Denmark
| | - Khalil S Rawji
- Altos Labs - Cambridge Institute of Science, Granta Park, Cambridge, CB21 6GP, UK
| | - Robin J M Franklin
- Altos Labs - Cambridge Institute of Science, Granta Park, Cambridge, CB21 6GP, UK
| | - James Dooley
- Department of Pathology, University of Cambridge, CB2 1QP, Cambridge, UK
| | - Adrian Liston
- Department of Pathology, University of Cambridge, CB2 1QP, Cambridge, UK
| | - Rebecca J Ingram
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Vijay K Tiwari
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
- Institute of Molecular Medicine, University of Southern Denmark, 5000, Odense, Denmark
- Danish Institute for Advanced Study (DIAS), 5230, Odense, Denmark
- Department of Clinical Genetics, Odense University Hospital, 5000, Odense, Denmark
| | - Rosana Penalva
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Yvonne Dombrowski
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK
| | - Denise C Fitzgerald
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast, BT9 7BL, Northern Ireland, UK.
| |
Collapse
|
25
|
Gingele S, Möllenkamp TM, Henkel F, Schröder L, Hümmert MW, Skripuletz T, Stangel M, Gudi V. Automated analysis of gray matter damage in aged mice reveals impaired remyelination in the cuprizone model. Brain Pathol 2024; 34:e13218. [PMID: 37927164 PMCID: PMC10901622 DOI: 10.1111/bpa.13218] [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: 06/22/2023] [Accepted: 10/14/2023] [Indexed: 11/07/2023] Open
Abstract
Multiple sclerosis is a chronic autoimmune disease of the central nervous system characterized by myelin loss, axonal damage, and glial scar formation. Still, the underlying processes remain unclear, as numerous pathways and factors have been found to be involved in the development and progression of the disease. Therefore, it is of great importance to find suitable animal models as well as reliable methods for their precise and reproducible analysis. Here, we describe the impact of demyelination on clinically relevant gray matter regions of the hippocampus and cerebral cortex, using the previously established cuprizone model for aged mice. We could show that bioinformatic image analysis methods are not only suitable for quantification of cell populations, but also for the assessment of de- and remyelination processes, as numerous objective parameters can be considered for reproducible measurements. After cuprizone-induced demyelination, subsequent remyelination proceeded slowly and remained incomplete in all gray matter areas studied. There were regional differences in the number of mature oligodendrocytes during remyelination suggesting region-specific differences in the factors accounting for remyelination failure, as, even in the presence of oligodendrocytes, remyelination in the cortex was found to be impaired. Upon cuprizone administration, synaptic density and dendritic volume in the gray matter of aged mice decreased. The intensity of synaptophysin staining gradually restored during the subsequent remyelination phase, however the expression of MAP2 did not fully recover. Microgliosis persisted in the gray matter of aged animals throughout the remyelination period, whereas extensive astrogliosis was of short duration as compared to white matter structures. In conclusion, we demonstrate that the application of the cuprizone model in aged mice mimics the impaired regeneration ability seen in human pathogenesis more accurately than commonly used protocols with young mice and therefore provides an urgently needed animal model for the investigation of remyelination failure and remyelination-enhancing therapies.
Collapse
Affiliation(s)
- Stefan Gingele
- Department of NeurologyHannover Medical SchoolHannoverGermany
| | | | - Florian Henkel
- Department of NeurologyHannover Medical SchoolHannoverGermany
| | | | | | | | - Martin Stangel
- Department of NeurologyHannover Medical SchoolHannoverGermany
- Department of Translational Medicine NeuroscienceNovartis Institute for BioMedical ResearchBaselSwitzerland
| | - Viktoria Gudi
- Department of NeurologyHannover Medical SchoolHannoverGermany
| |
Collapse
|
26
|
You Y, Chen Z, Hu WW. The role of microglia heterogeneity in synaptic plasticity and brain disorders: Will sequencing shed light on the discovery of new therapeutic targets? Pharmacol Ther 2024; 255:108606. [PMID: 38346477 DOI: 10.1016/j.pharmthera.2024.108606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/05/2024] [Accepted: 02/02/2024] [Indexed: 02/18/2024]
Abstract
Microglia play a crucial role in interacting with neuronal synapses and modulating synaptic plasticity. This function is particularly significant during postnatal development, as microglia are responsible for removing excessive synapses to prevent neurodevelopmental deficits. Dysregulation of microglial synaptic function has been well-documented in various pathological conditions, notably Alzheimer's disease and multiple sclerosis. The recent application of RNA sequencing has provided a powerful and unbiased means to decipher spatial and temporal microglial heterogeneity. By identifying microglia with varying gene expression profiles, researchers have defined multiple subgroups of microglia associated with specific pathological states, including disease-associated microglia, interferon-responsive microglia, proliferating microglia, and inflamed microglia in multiple sclerosis, among others. However, the functional roles of these distinct subgroups remain inadequately characterized. This review aims to refine our current understanding of the potential roles of heterogeneous microglia in regulating synaptic plasticity and their implications for various brain disorders, drawing from recent sequencing research and functional studies. This knowledge may aid in the identification of pathogenetic biomarkers and potential factors contributing to pathogenesis, shedding new light on the discovery of novel drug targets. The field of sequencing-based data mining is evolving toward a multi-omics approach. With advances in viral tools for precise microglial regulation and the development of brain organoid models, we are poised to elucidate the functional roles of microglial subgroups detected through sequencing analysis, ultimately identifying valuable therapeutic targets.
Collapse
Affiliation(s)
- Yi You
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zhong Chen
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China; Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Wei-Wei Hu
- Department of Pharmacology and Department of Pharmacy of the Second Affiliated Hospital, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
27
|
Franklin RJM, Bodini B, Goldman SA. Remyelination in the Central Nervous System. Cold Spring Harb Perspect Biol 2024; 16:a041371. [PMID: 38316552 PMCID: PMC10910446 DOI: 10.1101/cshperspect.a041371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
The inability of the mammalian central nervous system (CNS) to undergo spontaneous regeneration has long been regarded as a central tenet of neurobiology. However, while this is largely true of the neuronal elements of the adult mammalian CNS, save for discrete populations of granule neurons, the same is not true of its glial elements. In particular, the loss of oligodendrocytes, which results in demyelination, triggers a spontaneous and often highly efficient regenerative response, remyelination, in which new oligodendrocytes are generated and myelin sheaths are restored to denuded axons. Yet remyelination in humans is not without limitation, and a variety of demyelinating conditions are associated with sustained and disabling myelin loss. In this work, we will (1) review the biology of remyelination, including the cells and signals involved; (2) describe when remyelination occurs and when and why it fails, including the consequences of its failure; and (3) discuss approaches for therapeutically enhancing remyelination in demyelinating diseases of both children and adults, both by stimulating endogenous oligodendrocyte progenitor cells and by transplanting these cells into demyelinated brain.
Collapse
Affiliation(s)
- Robin J M Franklin
- Altos Labs Cambridge Institute of Science, Cambridge CB21 6GH, United Kingdom
| | - Benedetta Bodini
- Sorbonne Université, Paris Brain Institute, CNRS, INSERM, Paris 75013, France
- Saint-Antoine Hospital, APHP, Paris 75012, France
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York 14642, USA
- University of Copenhagen Faculty of Medicine, Copenhagen 2200, Denmark
| |
Collapse
|
28
|
Ananthamohan K, Stelzer JE, Sadayappan S. Hypertrophic cardiomyopathy in MYBPC3 carriers in aging. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:9. [PMID: 38406555 PMCID: PMC10883298 DOI: 10.20517/jca.2023.29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Hypertrophic cardiomyopathy (HCM) is characterized by abnormal thickening of the myocardium, leading to arrhythmias, heart failure, and elevated risk of sudden cardiac death, particularly among the young. This inherited disease is predominantly caused by mutations in sarcomeric genes, among which those in the cardiac myosin binding protein-C3 (MYBPC3) gene are major contributors. HCM associated with MYBPC3 mutations usually presents in the elderly and ranges from asymptomatic to symptomatic forms, affecting numerous cardiac functions and presenting significant health risks with a spectrum of clinical manifestations. Regulation of MYBPC3 expression involves various transcriptional and translational mechanisms, yet the destiny of mutant MYBPC3 mRNA and protein in late-onset HCM remains unclear. Pathogenesis related to MYBPC3 mutations includes nonsense-mediated decay, alternative splicing, and ubiquitin-proteasome system events, leading to allelic imbalance and haploinsufficiency. Aging further exacerbates the severity of HCM in carriers of MYBPC3 mutations. Advancements in high-throughput omics techniques have identified crucial molecular events and regulatory disruptions in cardiomyocytes expressing MYBPC3 variants. This review assesses the pathogenic mechanisms that promote late-onset HCM through the lens of transcriptional, post-transcriptional, and post-translational modulation of MYBPC3, underscoring its significance in HCM across carriers. The review also evaluates the influence of aging on these processes and MYBPC3 levels during HCM pathogenesis in the elderly. While pinpointing targets for novel medical interventions to conserve cardiac function remains challenging, the emergence of personalized omics offers promising avenues for future HCM treatments, particularly for late-onset cases.
Collapse
Affiliation(s)
- Kalyani Ananthamohan
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Julian E. Stelzer
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH 45267, USA
| | - Sakthivel Sadayappan
- Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
29
|
Jia J, Xu S, Hu J, Gan Y, Sun M, Xia S, Bao X, Zhang M, Xu Y. Growth arrest specific protein 6 alleviated white matter injury after experimental ischemic stroke. J Cereb Blood Flow Metab 2024; 44:77-93. [PMID: 37794790 PMCID: PMC10905636 DOI: 10.1177/0271678x231205078] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/09/2023] [Accepted: 09/11/2023] [Indexed: 10/06/2023]
Abstract
Ischemic white matter injury leads to long-term neurological deficits and lacks effective medication. Growth arrest specific protein 6 (Gas6) clears myelin debris, which is hypothesized to promote white matter integrity in experimental stroke models. By the middle cerebral artery occlusion (MCAO) stroke model, we observed that Gas6 reduced infarcted volume and behavior deficits 4 weeks after MCAO. Compared with control mice, Gas6-treatment mice represented higher FA values in the ipsilateral external capsules by MRI DTI scan. The SMI32/MBP ratio of the ipsilateral cortex and striatum was profoundly alleviated by Gas6 administration. Gas6-treatment group manifested thicker myelin sheaths than the control group by electron microscopy. We observed that Gas6 mainly promoted OPC maturation, which was closely related to microglia. Mechanically, Gas6 accelerated microglia-mediated myelin debris clearance and cholesterol transport protein expression (abca1, abcg1, apoc1, apoe) in vivo and in vitro, accordingly less myelin debris and lipid deposited in Gas6 treated stroke mice. HX531 (RXR inhibitor) administration mitigated the functions of Gas6 in speeding up debris clearance and cholesterol transport protein expression. Generally, we concluded that Gas6 cleared myelin debris and promoted cholesterol transportation protein expression through activating RXR, which could be one critical mechanism contributing to white matter repair after stroke.
Collapse
Affiliation(s)
- Junqiu Jia
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Siyi Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Zhenjiang, China
| | - Jinglong Hu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yonghui Gan
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Min Sun
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shengnan Xia
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinyu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Meijuan Zhang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Zhenjiang, China
- Department of Neurology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
- Jiangsu Provincial medical key discipline, Nanjing, China
| |
Collapse
|
30
|
Hammond BP, Panda SP, Kaushik DK, Plemel JR. Microglia and Multiple Sclerosis. ADVANCES IN NEUROBIOLOGY 2024; 37:445-456. [PMID: 39207707 DOI: 10.1007/978-3-031-55529-9_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Multiple sclerosis (MS) is a devastating autoimmune disease that leads to profound disability. This disability arises from the stochastic, regional loss of myelin-the insulating sheath surrounding neurons-in the central nervous system (CNS). The demyelinated regions are dominated by the brain's resident macrophages: microglia. Microglia perform a variety of functions in MS and are thought to initiate and perpetuate demyelination through their interactions with peripheral immune cells that traffic into the brain. However, microglia are also likely essential for recruiting and promoting the differentiation of cells that can restore lost myelin in a process known as remyelination. Given these seemingly opposing functions, an overarching beneficial or detrimental role is yet to be ascribed to these immune cells. In this chapter, we will discuss microglia dynamics throughout the MS disease course and probe the apparent dichotomy of microglia as the drivers of both demyelination and remyelination.
Collapse
Affiliation(s)
- Brady P Hammond
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Sharmistha P Panda
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Deepak K Kaushik
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Jason R Plemel
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada.
- Division of Neurology, Department of Medicine, University of Alberta, Edmonton, AB, Canada.
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada.
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
31
|
Stefik D, Vranic V, Ivkovic N, Bozic-Nedeljkovic B, Supic G. Genetic variants in the retinoid X receptor gene contribute to osteoarthritis susceptibility. ARCH BIOL SCI 2024; 76:151-160. [DOI: 10.2298/abs240306010s] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Osteoarthritis (OA) is a progressive disease of the joints that causes a gradual loss of function, resulting in limited mobility. Chronic inflammation is the main molecular process that triggers and propagates this disease. The retinoid X receptor (RXR), a member of the nuclear receptor family, is involved in modulating inflammatory pathways by influencing key procatabolic inflammatory cytokines, chemokines, and enzymes responsible for instigating and sustaining chronic joint inflammation. We evaluated the association between OA risk and genetic variants in the RXR? isoform. Compared to control individuals, a statistically significant difference in genotype distribution was detected for the rs7864987 polymorphism (P=0.008), while a positive inclination toward association was noted for rs3118523 (P=0.077). According to our findings based on the additive model, it appears that RXR? rs7864987 is linked to a higher risk of OA (adjusted odds ratio (OR)=1.846, P=0.012), whereas rs3118523 is associated with decreased risk of OA (adjusted OR=0.569, P=0.030). These results suggest that RXR? could be a significant inflammation-related gene involved in the complex network underlying the immunopathology of osteoarthritis. RXR? polymorphisms could potentially drive individualized retinoid therapy for OA based on genetic profile.
Collapse
Affiliation(s)
- Debora Stefik
- Institute for Medical Research, Military Medical Academy, Belgrade, Serbia
| | - Vladimir Vranic
- Clinic for Orthopedic Surgery and Traumatology, Military Medical Academy, Belgrade, Serbia
| | - Nemanja Ivkovic
- Institute for Medical Research, Military Medical Academy, Belgrade, Serbia
| | | | - Gordana Supic
- Institute for Medical Research, Military Medical Academy, Belgrade, Serbia + Medical Faculty of Military Medical Academy, University of Defense, Belgrade, Serbia
| |
Collapse
|
32
|
Baaklini CS, Ho MFS, Lange T, Hammond BP, Panda SP, Zirngibl M, Zia S, Himmelsbach K, Rana H, Phillips B, Antoszko D, Ibanga J, Lopez M, Lee KV, Keough MB, Caprariello AV, Kerr BJ, Plemel JR. Microglia promote remyelination independent of their role in clearing myelin debris. Cell Rep 2023; 42:113574. [PMID: 38100356 DOI: 10.1016/j.celrep.2023.113574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 10/17/2023] [Accepted: 11/27/2023] [Indexed: 12/17/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory disease characterized by myelin loss. While therapies exist to slow MS progression, no treatment currently exists for remyelination. Remyelination, linked to reduced disability in MS, relies on microglia and monocyte-derived macrophages (MDMs). This study aims to understand the role of microglia during remyelination by lineage tracing and depleting them. Microglial lineage tracing reveals that both microglia and MDMs initially accumulate, but microglia later dominate the lesion. Microglia and MDMs engulf equal amounts of inhibitory myelin debris, but after microglial depletion, MDMs compensate by engulfing more myelin debris. Microglial depletion does, however, reduce the recruitment and proliferation of oligodendrocyte progenitor cells (OPCs) and impairs their subsequent differentiation and remyelination. These findings underscore the essential role of microglia during remyelination and offer insights for enhancing this process by understanding microglial regulation of remyelination.
Collapse
Affiliation(s)
- Charbel S Baaklini
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Madelene F S Ho
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Tristan Lange
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Brady P Hammond
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Sharmistha P Panda
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Martin Zirngibl
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Sameera Zia
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Kassandre Himmelsbach
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Heli Rana
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Braxton Phillips
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Daria Antoszko
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Jeremies Ibanga
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Mizuki Lopez
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Kelly V Lee
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Michael B Keough
- Division of Neurosurgery, Department of Surgery, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Andrew V Caprariello
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Cumming School of Medicine, Calgary, AB T2N 1N4, Canada
| | - Bradley J Kerr
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Jason R Plemel
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada; Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2R3, Canada; Department of Medicine, Division of Neurology, University of Alberta, Edmonton, AB T6G 2R3, Canada.
| |
Collapse
|
33
|
Kaffe D, Kaplanis SI, Karagogeos D. The Roles of Caloric Restriction Mimetics in Central Nervous System Demyelination and Remyelination. Curr Issues Mol Biol 2023; 45:9526-9548. [PMID: 38132442 PMCID: PMC10742427 DOI: 10.3390/cimb45120596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
The dysfunction of myelinating glial cells, the oligodendrocytes, within the central nervous system (CNS) can result in the disruption of myelin, the lipid-rich multi-layered membrane structure that surrounds most vertebrate axons. This leads to axonal degeneration and motor/cognitive impairments. In response to demyelination in the CNS, the formation of new myelin sheaths occurs through the homeostatic process of remyelination, facilitated by the differentiation of newly formed oligodendrocytes. Apart from oligodendrocytes, the two other main glial cell types of the CNS, microglia and astrocytes, play a pivotal role in remyelination. Following a demyelination insult, microglia can phagocytose myelin debris, thus permitting remyelination, while the developing neuroinflammation in the demyelinated region triggers the activation of astrocytes. Modulating the profile of glial cells can enhance the likelihood of successful remyelination. In this context, recent studies have implicated autophagy as a pivotal pathway in glial cells, playing a significant role in both their maturation and the maintenance of myelin. In this Review, we examine the role of substances capable of modulating the autophagic machinery within the myelinating glial cells of the CNS. Such substances, called caloric restriction mimetics, have been shown to decelerate the aging process by mitigating age-related ailments, with their mechanisms of action intricately linked to the induction of autophagic processes.
Collapse
Affiliation(s)
- Despoina Kaffe
- Department of Biology, University of Crete, Vassilika Vouton, 70013 Heraklion, Greece;
| | - Stefanos Ioannis Kaplanis
- Department of Basic Science, School of Medicine, University of Crete, Vassilika Vouton, 70013 Heraklion, Greece;
- Institute of Molecular Biology & Biotechnology (IMBB), Foundation for Research and Technology-Hellas (FORTH), Vassilika Vouton, 70013 Heraklion, Greece
| | - Domna Karagogeos
- Department of Basic Science, School of Medicine, University of Crete, Vassilika Vouton, 70013 Heraklion, Greece;
- Institute of Molecular Biology & Biotechnology (IMBB), Foundation for Research and Technology-Hellas (FORTH), Vassilika Vouton, 70013 Heraklion, Greece
| |
Collapse
|
34
|
Thakolwiboon S, Mills EA, Yang J, Doty J, Belkin MI, Cho T, Schultz C, Mao-Draayer Y. Immunosenescence and multiple sclerosis: inflammaging for prognosis and therapeutic consideration. FRONTIERS IN AGING 2023; 4:1234572. [PMID: 37900152 PMCID: PMC10603254 DOI: 10.3389/fragi.2023.1234572] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 09/29/2023] [Indexed: 10/31/2023]
Abstract
Aging is associated with a progressive decline of innate and adaptive immune responses, called immunosenescence. This phenomenon links to different multiple sclerosis (MS) disease courses among different age groups. While clinical relapse and active demyelination are mainly related to the altered adaptive immunity, including invasion of T- and B-lymphocytes, impairment of innate immune cell (e.g., microglia, astrocyte) function is the main contributor to disability progression and neurodegeneration. Most patients with MS manifest the relapsing-remitting phenotype at a younger age, while progressive phenotypes are mainly seen in older patients. Current disease-modifying therapies (DMTs) primarily targeting adaptive immunity are less efficacious in older patients, suggesting that immunosenescence plays a role in treatment response. This review summarizes the recent immune mechanistic studies regarding immunosenescence in patients with MS and discusses the clinical implications of these findings.
Collapse
Affiliation(s)
| | - Elizabeth A. Mills
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Jennifer Yang
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Jonathan Doty
- Michigan Institute for Neurological Disorders, Farmington Hills, MI, United States
| | - Martin I. Belkin
- Michigan Institute for Neurological Disorders, Farmington Hills, MI, United States
| | - Thomas Cho
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Charles Schultz
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | - Yang Mao-Draayer
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- Michigan Institute for Neurological Disorders, Farmington Hills, MI, United States
- Autoimmune Center of Excellence, University of Michigan, Ann Arbor, MI, United States
- Graduate Program in Immunology, Program in Biomedical Sciences, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
35
|
Liu Y, Wang P, Jin G, Shi P, Zhao Y, Guo J, Yin Y, Shao Q, Li P, Yang P. The novel function of bexarotene for neurological diseases. Ageing Res Rev 2023; 90:102021. [PMID: 37495118 DOI: 10.1016/j.arr.2023.102021] [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: 05/28/2023] [Revised: 07/13/2023] [Accepted: 07/20/2023] [Indexed: 07/28/2023]
Abstract
Bexarotene, a retinoid X receptor (RXR) agonist, is approved by FDA to treat cutaneous T-cell lymphoma. However, it has also demonstrated promising therapeutic potential for neurological diseases such as stroke, traumatic brain injury, Parkinson's disease, and particularly Alzheimer's disease(AD). In AD, bexarotene inhibits the production and aggregation of amyloid β (Aβ), activates Liver X Receptor/RXR heterodimers to increase lipidated apolipoprotein E to remove Aβ, mitigates the negative impact of Aβ, regulates neuroinflammation, and ultimately improves cognitive function. For other neurological diseases, its mechanisms of action include inhibiting inflammatory responses, up-regulating microglial phagocytosis, and reducing misfolded protein aggregation, all of which aid in alleviating neurological damage. Here, we briefly discuss the characteristics, applications, and adverse effects of bexarotene, summarize its pharmacological mechanisms and therapeutic results in various neurological diseases, and elaborate on the problems encountered in preclinical research, with the aim of providing help for the further application of bexarotene in central nervous system diseases.
Collapse
Affiliation(s)
- Yangtao Liu
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China; College of Third Clinical, Xinxiang Medical University, Xinxiang, China
| | - Pengwei Wang
- Department of Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang Medical University, Weihui, China
| | - Guofang Jin
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
| | - Peijie Shi
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China; Xinxiang First People's Hospital, The Affiliated People's Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yonghui Zhao
- Xinxiang First People's Hospital, The Affiliated People's Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jiayi Guo
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China
| | - Yaling Yin
- School of Basic Medical Sciences, Department of Physiology and Pathophysiology, Sino-UK Joint Laboratory of Brain Function and Injury and Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China
| | - Qianhang Shao
- Department of Pharmacy, People's Hospital of Peking University, Beijing, China.
| | - Peng Li
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.
| | - Pengfei Yang
- College of Pharamacy, Xinxiang Medical University, Xinxiang, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, China.
| |
Collapse
|
36
|
Novorolsky RJ, Kasheke GDS, Hakim A, Foldvari M, Dorighello GG, Sekler I, Vuligonda V, Sanders ME, Renden RB, Wilson JJ, Robertson GS. Preserving and enhancing mitochondrial function after stroke to protect and repair the neurovascular unit: novel opportunities for nanoparticle-based drug delivery. Front Cell Neurosci 2023; 17:1226630. [PMID: 37484823 PMCID: PMC10360135 DOI: 10.3389/fncel.2023.1226630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 06/22/2023] [Indexed: 07/25/2023] Open
Abstract
The neurovascular unit (NVU) is composed of vascular cells, glia, and neurons that form the basic component of the blood brain barrier. This intricate structure rapidly adjusts cerebral blood flow to match the metabolic needs of brain activity. However, the NVU is exquisitely sensitive to damage and displays limited repair after a stroke. To effectively treat stroke, it is therefore considered crucial to both protect and repair the NVU. Mitochondrial calcium (Ca2+) uptake supports NVU function by buffering Ca2+ and stimulating energy production. However, excessive mitochondrial Ca2+ uptake causes toxic mitochondrial Ca2+ overloading that triggers numerous cell death pathways which destroy the NVU. Mitochondrial damage is one of the earliest pathological events in stroke. Drugs that preserve mitochondrial integrity and function should therefore confer profound NVU protection by blocking the initiation of numerous injury events. We have shown that mitochondrial Ca2+ uptake and efflux in the brain are mediated by the mitochondrial Ca2+ uniporter complex (MCUcx) and sodium/Ca2+/lithium exchanger (NCLX), respectively. Moreover, our recent pharmacological studies have demonstrated that MCUcx inhibition and NCLX activation suppress ischemic and excitotoxic neuronal cell death by blocking mitochondrial Ca2+ overloading. These findings suggest that combining MCUcx inhibition with NCLX activation should markedly protect the NVU. In terms of promoting NVU repair, nuclear hormone receptor activation is a promising approach. Retinoid X receptor (RXR) and thyroid hormone receptor (TR) agonists activate complementary transcriptional programs that stimulate mitochondrial biogenesis, suppress inflammation, and enhance the production of new vascular cells, glia, and neurons. RXR and TR agonism should thus further improve the clinical benefits of MCUcx inhibition and NCLX activation by increasing NVU repair. However, drugs that either inhibit the MCUcx, or stimulate the NCLX, or activate the RXR or TR, suffer from adverse effects caused by undesired actions on healthy tissues. To overcome this problem, we describe the use of nanoparticle drug formulations that preferentially target metabolically compromised and damaged NVUs after an ischemic or hemorrhagic stroke. These nanoparticle-based approaches have the potential to improve clinical safety and efficacy by maximizing drug delivery to diseased NVUs and minimizing drug exposure in healthy brain and peripheral tissues.
Collapse
Affiliation(s)
- Robyn J. Novorolsky
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Brain Repair Centre, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Gracious D. S. Kasheke
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Brain Repair Centre, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Antoine Hakim
- School of Pharmacy, Faculty of Science, University of Waterloo, Waterloo, ON, Canada
| | - Marianna Foldvari
- School of Pharmacy, Faculty of Science, University of Waterloo, Waterloo, ON, Canada
| | - Gabriel G. Dorighello
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Brain Repair Centre, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| | - Israel Sekler
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben Gurion University, Beersheva, Israel
| | | | | | - Robert B. Renden
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, NV, United States
| | - Justin J. Wilson
- Department of Chemistry and Chemical Biology, College of Arts and Sciences, Cornell University, Ithaca, NY, United States
| | - George S. Robertson
- Department of Pharmacology, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Brain Repair Centre, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
- Department of Psychiatry, Faculty of Medicine, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
37
|
Packer D, Fresenko EE, Harrington EP. Remyelination in animal models of multiple sclerosis: finding the elusive grail of regeneration. Front Mol Neurosci 2023; 16:1207007. [PMID: 37448959 PMCID: PMC10338073 DOI: 10.3389/fnmol.2023.1207007] [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: 04/16/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023] Open
Abstract
Remyelination biology and the therapeutic potential of restoring myelin sheaths to prevent neurodegeneration and disability in multiple sclerosis (MS) has made considerable gains over the past decade with many regeneration strategies undergoing tested in MS clinical trials. Animal models used to investigate oligodendroglial responses and regeneration of myelin vary considerably in the mechanism of demyelination, involvement of inflammatory cells, neurodegeneration and capacity for remyelination. The investigation of remyelination in the context of aging and an inflammatory environment are of considerable interest for the potential translation to progressive multiple sclerosis. Here we review how remyelination is assessed in mouse models of demyelination, differences and advantages of these models, therapeutic strategies that have emerged and current pro-remyelination clinical trials.
Collapse
|
38
|
Mandato C, Colucci A, Lanzillo R, Staiano A, Scarpato E, Schiavo L, Operto FF, Serra MR, Di Monaco C, Napoli JS, Massa G, Vajro P. Multiple Sclerosis-Related Dietary and Nutritional Issues: An Updated Scoping Review with a Focus on Pediatrics. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1022. [PMID: 37371254 PMCID: PMC10297186 DOI: 10.3390/children10061022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/03/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023]
Abstract
PURPOSE Lifestyle/dietetic habits play an important role in the development and progression of multiple sclerosis (MS) disease. Here, we examine the basic pathomechanisms underlying intestinal and brain barrier modifications in MS and consider diets and dietary supplementations proposed over time to complement pharmacological therapies for improving disease outcome both in adults and in children. METHODS Scoping literature search about evidence-based findings in MS-related gut-brain axis (GBA) pathophysiology and nutritional issues at all ages. FINDINGS Data show that (1) no universal best diet exists, (2) healthy/balanced diets are, however, necessary to safeguard the adequate intake of all essential nutrients, (3) diets with high intakes of fruits, vegetables, whole grains, and lean proteins that limit processed foods, sugar, and saturated fat appear beneficial for their antioxidant and anti-inflammatory properties and their ability to shape a gut microbiota that respects the gut and brain barriers, (4) obesity may trigger MS onset and/or its less favorable course, especially in pediatric-onset MS. Vitamin D and polyunsaturated fatty acids are the most studied supplements for reducing MS-associated inflammation. CONCLUSIONS Pending results from other and/or newer approaches targeting the GBA (e.g., pre- and probiotics, engineered probiotics, fecal-microbiota transplantation), accurate counseling in choosing adequate diet and maintaining physical activity remains recommended for MS prevention and management both in adults and children.
Collapse
Affiliation(s)
- Claudia Mandato
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Pediatrics Section, University of Salerno, 84081 Baronissi, Salerno, Italy (P.V.)
| | - Angelo Colucci
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Pediatrics Section, University of Salerno, 84081 Baronissi, Salerno, Italy (P.V.)
| | - Roberta Lanzillo
- Multiple Sclerosis Clinical Care and Research Centre, Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, 80138 Naples, Naples, Italy
| | - Annamaria Staiano
- Department of Translational Medical Science, Section of Pediatrics, University of Naples Federico II, 80138 Naples, Naples, Italy
| | - Elena Scarpato
- Department of Translational Medical Science, Section of Pediatrics, University of Naples Federico II, 80138 Naples, Naples, Italy
| | - Luigi Schiavo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Nutrition Section, University of Salerno, 84081 Baronissi, Salerno, Italy
| | - Francesca Felicia Operto
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Pediatric Psychiatry Section, University of Salerno, 84081 Baronissi, Salerno, Italy
| | - Maria Rosaria Serra
- Department of Translational Medical Science, Section of Pediatrics, University of Naples Federico II, 80138 Naples, Naples, Italy
| | - Cristina Di Monaco
- Multiple Sclerosis Clinical Care and Research Centre, Department of Neuroscience, Reproductive Science and Odontostomatology, University of Naples Federico II, 80138 Naples, Naples, Italy
| | - Julia Sara Napoli
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Pediatrics Section, University of Salerno, 84081 Baronissi, Salerno, Italy (P.V.)
| | - Grazia Massa
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Pediatrics Section, University of Salerno, 84081 Baronissi, Salerno, Italy (P.V.)
| | - Pietro Vajro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, Pediatrics Section, University of Salerno, 84081 Baronissi, Salerno, Italy (P.V.)
| |
Collapse
|
39
|
Zheng B, He Y, Yin S, Zhu X, Zhao Q, Yang H, Wang Z, Zhu R, Cheng L. Unresolved Excess Accumulation of Myelin-Derived Cholesterol Contributes to Scar Formation after Spinal Cord Injury. RESEARCH (WASHINGTON, D.C.) 2023; 6:0135. [PMID: 37223476 PMCID: PMC10202378 DOI: 10.34133/research.0135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 04/13/2023] [Indexed: 05/25/2023]
Abstract
Spinal cord injury triggers complex pathological cascades, resulting in destructive tissue damage and incomplete tissue repair. Scar formation is generally considered a barrier for regeneration in the central nervous system. However, the intrinsic mechanism of scar formation after spinal cord injury has not been fully elucidated. Here, we report that excess cholesterol accumulates in phagocytes and is inefficiently removed from spinal cord lesions in young adult mice. Interestingly, we observed that excessive cholesterol also accumulates in injured peripheral nerves but is subsequently removed by reverse cholesterol transport. Meanwhile, preventing reverse cholesterol transport leads to macrophage accumulation and fibrosis in injured peripheral nerves. Furthermore, the neonatal mouse spinal cord lesions are devoid of myelin-derived lipids and can heal without excess cholesterol accumulation. We found that transplantation of myelin into neonatal lesions disrupts healing with excessive cholesterol accumulation, persistent macrophage activation, and fibrosis. Myelin internalization suppresses macrophage apoptosis mediated by CD5L expression, indicating that myelin-derived cholesterol plays a critical role in impaired wound healing. Taken together, our data suggest that the central nervous system lacks an efficient approach for cholesterol clearance, resulting in excessive accumulation of myelin-derived cholesterol, thereby inducing scar formation after injury.
Collapse
Affiliation(s)
- Bolin Zheng
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
| | - Yijing He
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
| | - Shuai Yin
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
| | - Xu Zhu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
| | - Qing Zhao
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
| | - Huiyi Yang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
| | - Zhaojie Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
- Frontier Science Center for Stem Cell Research, School of Life Science and Technology,
Tongji University, Shanghai 200092, China
| | - Rongrong Zhu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
- Frontier Science Center for Stem Cell Research, School of Life Science and Technology,
Tongji University, Shanghai 200092, China
| | - Liming Cheng
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration, Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Medicine,
Tongji University, Shanghai 200092, China
- Clinical Center for Brain and Spinal Cord Research,
Tongji University, Shanghai 200092, China
| |
Collapse
|
40
|
Devanand M, V N S, Madhu K. Signaling mechanisms involved in the regulation of remyelination in multiple sclerosis: a mini review. J Mol Med (Berl) 2023:10.1007/s00109-023-02312-9. [PMID: 37084092 DOI: 10.1007/s00109-023-02312-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 02/22/2023] [Accepted: 03/28/2023] [Indexed: 04/22/2023]
Abstract
Multiple sclerosis is an autoimmune neurodegenerative disease of the CNS that causes progressive disabilities, owing to CNS axon degeneration as a late result of demyelination. In the search for the prevention of axonal loss, mitigating inflammatory attacks in the CNS and myelin restoration are two possible approaches. As a result, therapies that target diverse signaling pathways involved in neuroprotection and remyelination have the potential to overcome the challenges in the development of multiple sclerosis treatments. LINGO1 (Leucine rich repeat and Immunoglobulin domain containing, Nogo receptor- interaction protein), AKT/PIP3/mTOR, Notch, Wnt, RXR (Retinoid X receptor gamma), and Nrf2 (nuclear factor erythroid 2-related factor 2) signaling pathways are highlighted in this section. This article reviews the present knowledge regarding numerous signaling pathways and their functions in regulating remyelination in multiple sclerosis pathogenesis. These pathways are potential biomarkers and therapeutic targets in MS.
Collapse
Affiliation(s)
- Midhuna Devanand
- Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - Saiprabha V N
- Department of Pharmaceutical Chemistry and Analysis, Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India.
| | - Krishnadas Madhu
- Department of Pharmacology, Amrita School of Pharmacy, AIMS Health Science Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India.
| |
Collapse
|
41
|
Zahaf A, Kassoussi A, Hutteau-Hamel T, Mellouk A, Marie C, Zoupi L, Tsouki F, Mattern C, Bobé P, Schumacher M, Williams A, Parras C, Traiffort E. Androgens show sex-dependent differences in myelination in immune and non-immune murine models of CNS demyelination. Nat Commun 2023; 14:1592. [PMID: 36949062 PMCID: PMC10033728 DOI: 10.1038/s41467-023-36846-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 02/20/2023] [Indexed: 03/24/2023] Open
Abstract
Neuroprotective, anti-inflammatory, and remyelinating properties of androgens are well-characterized in demyelinated male mice and men suffering from multiple sclerosis. However, androgen effects mediated by the androgen receptor (AR), have been only poorly studied in females who make low androgen levels. Here, we show a predominant microglial AR expression in demyelinated lesions from female mice and women with multiple sclerosis, but virtually undetectable AR expression in lesions from male animals and men with multiple sclerosis. In female mice, androgens and estrogens act in a synergistic way while androgens drive microglia response towards regeneration. Transcriptomic comparisons of demyelinated mouse spinal cords indicate that, regardless of the sex, androgens up-regulate genes related to neuronal function integrity and myelin production. Depending on the sex, androgens down-regulate genes related to the immune system in females and lipid catabolism in males. Thus, androgens are required for proper myelin regeneration in females and therapeutic approaches of demyelinating diseases need to consider male-female differences.
Collapse
Affiliation(s)
- Amina Zahaf
- U1195 Inserm, Paris-Saclay University, Kremlin-Bicêtre, France
| | | | | | - Amine Mellouk
- UMR996 Inserm, Paris-Saclay University, Clamart, France
| | | | - Lida Zoupi
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, UK
| | - Foteini Tsouki
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, UK
| | | | - Pierre Bobé
- UMR996 Inserm, Paris-Saclay University, Clamart, France
| | | | - Anna Williams
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, UK
| | - Carlos Parras
- Paris Brain Institute, Sorbonne University, Paris, France
| | | |
Collapse
|
42
|
Xu T, Liu C, Deng S, Gan L, Zhang Z, Yang GY, Tian H, Tang Y. The roles of microglia and astrocytes in myelin phagocytosis in the central nervous system. J Cereb Blood Flow Metab 2023; 43:325-340. [PMID: 36324281 PMCID: PMC9941857 DOI: 10.1177/0271678x221137762] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022]
Abstract
Myelination is an important process in the central nervous system (CNS). Oligodendrocytes (OLs) extend multiple layers to densely sheath on axons, composing the myelin to achieve efficient electrical signal conduction. The myelination during developmental stage maintains a balanced state. However, numerous CNS diseases including neurodegenerative and cerebrovascular diseases cause demyelination and disrupt the homeostasis, resulting in inflammation and white matter deficits. Effective clearance of myelin debris is needed in the region of demyelination, which is a key step for remyelination and tissue regeneration. Microglia and astrocytes are the major resident phagocytic cells in the brain, which may play different or collaborative roles in myelination. Microglia and astrocytes participate in developmental myelination through engulfing excessive unneeded myelin. They are also involved in the clearance of degenerated myelin debris for accelerating remyelination, or engulfing healthy myelin sheath for inhibiting remyelination. This review focuses on the roles of microglia and astrocytes in phagocytosing myelin in the developmental brain and diseased brain. In addition, the interaction between microglia and astrocytes to mediate myelin engulfment is also summarized.
Collapse
Affiliation(s)
- Tongtong Xu
- Shanghai Jiao Tong Affiliated Sixth People’s
Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University,
Shanghai, China
| | - Chang Liu
- Shanghai Jiao Tong Affiliated Sixth People’s
Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University,
Shanghai, China
| | - Shiyu Deng
- Shanghai Jiao Tong Affiliated Sixth People’s
Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University,
Shanghai, China
| | - Lin Gan
- Shanghai Jiao Tong Affiliated Sixth People’s
Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University,
Shanghai, China
| | - Zhijun Zhang
- Shanghai Jiao Tong Affiliated Sixth People’s
Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University,
Shanghai, China
| | - Guo-Yuan Yang
- Shanghai Jiao Tong Affiliated Sixth People’s
Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University,
Shanghai, China
| | - Hengli Tian
- Shanghai Jiao Tong Affiliated Sixth People’s
Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University,
Shanghai, China
| | - Yaohui Tang
- Shanghai Jiao Tong Affiliated Sixth People’s
Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University,
Shanghai, China
| |
Collapse
|
43
|
Jia J, Zheng L, Ye L, Chen J, Shu S, Xu S, Bao X, Xia S, Liu R, Xu Y, Zhang M. CD11c + microglia promote white matter repair after ischemic stroke. Cell Death Dis 2023; 14:156. [PMID: 36828819 PMCID: PMC9958101 DOI: 10.1038/s41419-023-05689-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/26/2023]
Abstract
Ischemic stroke leads to white matter damage and neurological deficits. However, the characteristics of white matter injury and repair after stroke are unclear. Additionally, the precise molecular communications between microglia and white matter repair during the stroke rehabilitation phase remain elusive. In this current study, MRI DTI scan and immunofluorescence staining were performed to trace white matter and microglia in the mouse transient middle cerebral artery occlusion (tMCAO) stroke model. We found that the most serious white matter damage was on Day 7 after the ischemic stroke, then it recovered gradually from Day 7 to Day 30. Parallel to white matter recovery, we observed that microglia centered around the damaged myelin sheath and swallowed myelin debris in the ischemic areas. Then, microglia of the ischemic hemisphere were sorted by flow cytometry for RNA sequencing and subpopulation analysis. We found that CD11c+ microglia increased from Day 7 to Day 30, demonstrating high phagocytotic capabilities, myelin-supportive genes, and lipid metabolism associated genes. CD11c+ microglia population was partly depleted by the stereotactic injecting of rAAV2/6M-taCasp3 (rAAV2/6M-CMV-DIO-taCasp3-TEVp) into CD11c-cre mice. Selective depletion of CD11c+ microglia disrupted white matter repair, oligodendrocyte maturation, and functional recovery after stroke by Rotarod test, Adhesive Removal test, and Morris Water Maze test. These findings suggest that spontaneous white matter repair occurs after ischemic stroke, while CD11c+ microglia play critical roles in this white matter restorative progress.
Collapse
Affiliation(s)
- Junqiu Jia
- Department of Neurology, Drum Tower Hospital, The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Medical School, Nanjing University, Nanjing, China
| | - Lili Zheng
- Department of Neurology, Drum Tower Hospital, The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Medical School, Nanjing University, Nanjing, China
| | - Lei Ye
- Department of Neurology, Drum Tower Hospital, The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Medical School, Nanjing University, Nanjing, China
| | - Jian Chen
- Department of Neurology, Drum Tower Hospital, The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Medical School, Nanjing University, Nanjing, China
| | - Shu Shu
- Department of Neurology, Drum Tower Hospital, The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Medical School, Nanjing University, Nanjing, China
| | - Siyi Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Zhenjiang, China
| | - Xinyu Bao
- Department of Neurology, Drum Tower Hospital, The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Medical School, Nanjing University, Nanjing, China
| | - Shengnan Xia
- Department of Neurology, Drum Tower Hospital, The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Medical School, Nanjing University, Nanjing, China
| | - Renyuan Liu
- Department of Radiology, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yun Xu
- Department of Neurology, Drum Tower Hospital, The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Medical School, Nanjing University, Nanjing, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China.
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China.
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China.
| | - Meijuan Zhang
- Department of Neurology, Drum Tower Hospital, The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Medical School, Nanjing University, Nanjing, China.
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China.
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China.
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China.
| |
Collapse
|
44
|
Papiri G, D’Andreamatteo G, Cacchiò G, Alia S, Silvestrini M, Paci C, Luzzi S, Vignini A. Multiple Sclerosis: Inflammatory and Neuroglial Aspects. Curr Issues Mol Biol 2023; 45:1443-1470. [PMID: 36826039 PMCID: PMC9954863 DOI: 10.3390/cimb45020094] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Multiple sclerosis (MS) represents the most common acquired demyelinating disorder of the central nervous system (CNS). Its pathogenesis, in parallel with the well-established role of mechanisms pertaining to autoimmunity, involves several key functions of immune, glial and nerve cells. The disease's natural history is complex, heterogeneous and may evolve over a relapsing-remitting (RRMS) or progressive (PPMS/SPMS) course. Acute inflammation, driven by infiltration of peripheral cells in the CNS, is thought to be the most relevant process during the earliest phases and in RRMS, while disruption in glial and neural cells of pathways pertaining to energy metabolism, survival cascades, synaptic and ionic homeostasis are thought to be mostly relevant in long-standing disease, such as in progressive forms. In this complex scenario, many mechanisms originally thought to be distinctive of neurodegenerative disorders are being increasingly recognized as crucial from the beginning of the disease. The present review aims at highlighting mechanisms in common between MS, autoimmune diseases and biology of neurodegenerative disorders. In fact, there is an unmet need to explore new targets that might be involved as master regulators of autoimmunity, inflammation and survival of nerve cells.
Collapse
Affiliation(s)
- Giulio Papiri
- Neurology Unit, Ospedale Provinciale “Madonna del Soccorso”, 63074 San Benedetto del Tronto, Italy
| | - Giordano D’Andreamatteo
- Neurology Unit, Ospedale Provinciale “Madonna del Soccorso”, 63074 San Benedetto del Tronto, Italy
| | - Gabriella Cacchiò
- Neurology Unit, Ospedale Provinciale “Madonna del Soccorso”, 63074 San Benedetto del Tronto, Italy
| | - Sonila Alia
- Section of Biochemistry, Biology and Physics, Department of Clinical Sciences, Università Politecnica delle Marche, 60100 Ancona, Italy
| | - Mauro Silvestrini
- Neurology Unit, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60100 Ancona, Italy
| | - Cristina Paci
- Neurology Unit, Ospedale Provinciale “Madonna del Soccorso”, 63074 San Benedetto del Tronto, Italy
| | - Simona Luzzi
- Neurology Unit, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60100 Ancona, Italy
| | - Arianna Vignini
- Section of Biochemistry, Biology and Physics, Department of Clinical Sciences, Università Politecnica delle Marche, 60100 Ancona, Italy
| |
Collapse
|
45
|
Dimovasili C, Fair AE, Garza IR, Batterman KV, Mortazavi F, Moore TL, Rosene DL. Aging compromises oligodendrocyte precursor cell maturation and efficient remyelination in the monkey brain. GeroScience 2023; 45:249-264. [PMID: 35930094 PMCID: PMC9886778 DOI: 10.1007/s11357-022-00621-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/07/2022] [Indexed: 02/03/2023] Open
Abstract
Age-associated cognitive decline is common among otherwise healthy elderly people, even in the absence of Alzheimer's disease and neuron loss. Instead, white matter loss and myelin damage are strongly associated with cognitive decline. Myelin is subject to lifelong oxidative stress that damages the myelin sheath, which is repaired by cells of the oligodendrocyte lineage. This process is mediated by oligodendrocyte precursor cells (OPCs) that sense the damage and respond by proliferating locally and migrating to the region, where they differentiate into mature myelinating oligodendrocytes. In aging, extensive myelin damage, in combination with inefficient remyelination, leads to chronically damaged myelin and loss of efficient neuronal conduction. This study used the rhesus monkey model of normal aging to examine how myelin regeneration capacity is affected by age. Results show that older subjects have reduced numbers of new BCAS1 + myelinating oligodendrocytes, which are newly formed cells, and that this reduction is associated with poorer cognitive performance. Interestingly, this does not result from limited proliferation of progenitor OPCs. Instead, the transcription factor NKX2.2, which regulates OPCs differentiation, is significantly decreased in aged OPCs. This suggests that these OPCs have a diminished potential for differentiation into mature oligodendrocytes. In addition, mature oligodendrocytes have reduced RNA expression of two essential myelin protein markers, MBP and PLP. These data collectively suggest that in the normal aging brain, there is a reduction in regenerative OPCs as well as myelin production that impairs the capacity for remyelination.
Collapse
Affiliation(s)
- Christina Dimovasili
- Laboratory for Cognitive Neurobiology, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, USA.
| | - Ashley E Fair
- Laboratory for Cognitive Neurobiology, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Isabella R Garza
- Laboratory for Cognitive Neurobiology, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Katelyn V Batterman
- Laboratory for Cognitive Neurobiology, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Farzad Mortazavi
- Laboratory for Cognitive Neurobiology, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, USA
| | - Tara L Moore
- Laboratory for Cognitive Neurobiology, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, USA
| | - Douglas L Rosene
- Laboratory for Cognitive Neurobiology, Department of Anatomy & Neurobiology, Boston University School of Medicine, Boston, MA, USA
- Center for Systems Neuroscience, Boston University, Boston, MA, USA
| |
Collapse
|
46
|
Harder JW, Ma J, Alard P, Sokoloski KJ, Mathiowitz E, Furtado S, Egilmez NK, Kosiewicz MM. Male microbiota-associated metabolite restores macrophage efferocytosis in female lupus-prone mice via activation of PPARγ/LXR signaling pathways. J Leukoc Biol 2023; 113:41-57. [PMID: 36822162 DOI: 10.1093/jleuko/qiac002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Indexed: 01/11/2023] Open
Abstract
Systemic lupus erythematosus development is influenced by both sex and the gut microbiota. Metabolite production is a major mechanism by which the gut microbiota influences the immune system, and we have previously found differences in the fecal metabolomic profiles of lupus-prone female and lupus-resistant male BWF1 mice. Here we determine how sex and microbiota metabolite production may interact to affect lupus. Transcriptomic analysis of female and male splenocytes showed genes that promote phagocytosis were upregulated in BWF1 male mice. Because patients with systemic lupus erythematosus exhibit defects in macrophage-mediated phagocytosis of apoptotic cells (efferocytosis), we compared splenic macrophage efferocytosis in vitro between female and male BWF1 mice. Macrophage efferocytosis was deficient in female compared to male BWF1 mice but could be restored by feeding male microbiota. Further transcriptomic analysis of the genes upregulated in male BWF1 mice revealed enrichment of genes stimulated by PPARγ and LXR signaling. Our previous fecal metabolomics analyses identified metabolites in male BWF1 mice that can activate PPARγ and LXR signaling and identified one in particular, phytanic acid, that is a very potent agonist. We show here that treatment of female BWF1 splenic macrophages with phytanic acid restores efferocytic activity via activation of the PPARγ and LXR signaling pathways. Furthermore, we found phytanic acid may restore female BWF1 macrophage efferocytosis through upregulation of the proefferocytic gene CD36. Taken together, our data indicate that metabolites produced by BWF1 male microbiota can enhance macrophage efferocytosis and, through this mechanism, could potentially influence lupus progression.
Collapse
Affiliation(s)
- James W Harder
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock St, Rm 609, Louisville, KY 40202, USA
| | - Jing Ma
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock St, Rm 609, Louisville, KY 40202, USA
| | - Pascale Alard
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock St, Rm 609, Louisville, KY 40202, USA
| | - Kevin J Sokoloski
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock St, Rm 609, Louisville, KY 40202, USA
| | - Edith Mathiowitz
- Department of Medical Science and Engineering, Brown University, 222 Richmond Street, Providence, RI 02903, USA
| | - Stacia Furtado
- Department of Medical Science and Engineering, Brown University, 222 Richmond Street, Providence, RI 02903, USA
| | - Nejat K Egilmez
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock St, Rm 609, Louisville, KY 40202, USA
| | - Michele M Kosiewicz
- Department of Microbiology and Immunology, University of Louisville, 505 South Hancock St, Rm 609, Louisville, KY 40202, USA
| |
Collapse
|
47
|
Rawji KS, Neumann B, Franklin RJM. Glial aging and its impact on central nervous system myelin regeneration. Ann N Y Acad Sci 2023; 1519:34-45. [PMID: 36398864 DOI: 10.1111/nyas.14933] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Aging is a major risk factor for several neurodegenerative diseases and is associated with cognitive decline. In addition to affecting neuronal function, the aging process significantly affects the functional phenotype of the glial cell compartment, comprising oligodendrocyte lineage cells, astrocytes, and microglia. These changes result in a more inflammatory microenvironment, resulting in a condition that is favorable for neuron and synapse loss. In addition to facilitating neurodegeneration, the aging glial cell population has negative implications for central nervous system remyelination, a regenerative process that is of particular importance to the chronic demyelinating disease multiple sclerosis. This review will discuss the changes that occur with aging in the three main glial populations and provide an overview of the studies documenting the impact these changes have on remyelination.
Collapse
Affiliation(s)
- Khalil S Rawji
- Altos Labs, Cambridge Institute of Science, Cambridge, UK
| | - Björn Neumann
- Altos Labs, Cambridge Institute of Science, Cambridge, UK
| | | |
Collapse
|
48
|
Graves JS, Krysko KM, Hua LH, Absinta M, Franklin RJM, Segal BM. Ageing and multiple sclerosis. Lancet Neurol 2023; 22:66-77. [PMID: 36216015 DOI: 10.1016/s1474-4422(22)00184-3] [Citation(s) in RCA: 103] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 03/09/2022] [Accepted: 04/20/2022] [Indexed: 11/07/2022]
Abstract
The factor that is most relevant and strongly associated with the clinical course of multiple sclerosis is chronological age. Very young patients exclusively have relapsing remitting disease, whereas those with later onset disease face a more rapid development of permanent disability. For people with progressive multiple sclerosis, the poor response to current disease modifying therapies might be related to ageing in the immune system and CNS. Ageing is also associated with increased risks of side-effects caused by some multiple sclerosis therapies. Both somatic and reproductive ageing processes might contribute to development of progressive multiple sclerosis. Understanding the role of ageing in immune and neural cell function in patients with multiple sclerosis might be key to halting non-relapse-related progression. The growing literature on potential therapies that target senescent cells and ageing processes might provide effective strategies for remyelination and neuroprotection.
Collapse
Affiliation(s)
- Jennifer S Graves
- Department of Neurosciences, University of California, San Diego, CA, USA; Pediatric Multiple Sclerosis Center, Rady Children's Hospital, San Diego, CA, USA; Department of Neurology, San Diego VA Hospital, San Diego, CA, USA.
| | - Kristen M Krysko
- Division of Neurology, Department of Medicine, Li Ka Shing Knowledge Institute, St Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Le H Hua
- Department of Neurology, Cleveland Clinic, Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Martina Absinta
- Department of Neurology, Johns Hopkins University, Baltimore, MD, USA; Division of Neuroscience, IRCCS San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milan, Italy
| | - Robin J M Franklin
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Benjamin M Segal
- Department of Neurology and the Neuroscience Research Institute, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
49
|
Ming X, Cai W, Li Z, Yang X, Yang M, Pan D, Chen X. CD40LG and GZMB were correlated with adipose tissue macrophage infiltration and involved in obstructive sleep apnea related metabolic dysregulation: Evidence from bioinformatics analysis. Front Genet 2023; 14:1128139. [PMID: 36923793 PMCID: PMC10009156 DOI: 10.3389/fgene.2023.1128139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/16/2023] [Indexed: 03/03/2023] Open
Abstract
Both obesity and obstructive sleep apnea (OSA) can lead to metabolic dysregulation and systemic inflammation. Similar to obesity, increasing evidence has revealed that immune infiltration in the visceral adipose tissue (VAT) is associated with obstructive sleep apnea-related morbidity. However, the pathological changes and potential molecular mechanisms in visceral adipose tissue of obstructive sleep apnea patients need to be further studied. Herein, by bioinformatics analysis and clinical validation methods, including the immune-related differentially expressed genes (IRDEGs) analysis, protein-protein interaction network (PPI), functional enrichment analysis, a devolution algorithm (CIBERSORT), spearman's correlation analysis, polymerase chain reaction (PCR), Enzyme-linked immunosorbent assay (ELISA) and immunohistochemistry (IHC), we identified and validated 10 hub IRDEGs, the relative mRNA expression of four hub genes (CRP, CD40LG, CCL20, and GZMB), and the protein expression level of two hub genes (CD40LG and GZMB) were consistent with the bioinformatics analysis results. Immune infiltration results further revealed that obstructive sleep apnea patients contained a higher proportion of pro-inflammatory M1 macrophages and a lower proportion of M2 macrophages. Spearman's correlation analysis showed that CD40LG was positively correlated with M1 macrophages and GZMB was negatively correlated with M2 macrophages. CD40LG and GZMB might play a vital role in the visceral adipose tissue homeostasis of obstructive sleep apnea patients. Their interaction with macrophages and involved pathways not only provides new insights for understanding molecular mechanisms but also be of great significance in discovering novel small molecules or other promising candidates as immunotherapies of OSA-associated metabolic complications.
Collapse
Affiliation(s)
- Xiaoping Ming
- Department of Otorhinolaryngology, Head, and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.,Sleep Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Weisong Cai
- Department of Otorhinolaryngology, Head, and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.,Sleep Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhen Li
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.,Bariatric and Metabolic Disease Surgery Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiuping Yang
- Department of Otorhinolaryngology, Head, and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.,Sleep Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Minlan Yang
- Department of Otorhinolaryngology, Head, and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.,Sleep Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Dingyu Pan
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.,Bariatric and Metabolic Disease Surgery Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiong Chen
- Department of Otorhinolaryngology, Head, and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China.,Sleep Medicine Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
50
|
Leo H, Kipp M. Remyelination in Multiple Sclerosis: Findings in the Cuprizone Model. Int J Mol Sci 2022; 23:ijms232416093. [PMID: 36555733 PMCID: PMC9783537 DOI: 10.3390/ijms232416093] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Remyelination therapies, which are currently under development, have a great potential to delay, prevent or even reverse disability in multiple sclerosis patients. Several models are available to study the effectiveness of novel compounds in vivo, among which is the cuprizone model. This model is characterized by toxin-induced demyelination, followed by endogenous remyelination after cessation of the intoxication. Due to its high reproducibility and ease of use, this model enjoys high popularity among various research and industrial groups. In this review article, we will summarize recent findings using this model and discuss the potential of some of the identified compounds to promote remyelination in multiple sclerosis patients.
Collapse
Affiliation(s)
| | - Markus Kipp
- Correspondence: ; Tel.: +49-(0)-381-494-8400
| |
Collapse
|