1
|
Lößlein L, Linnerbauer M, Zuber F, Tsaktanis T, Vandrey O, Peter A, Panier F, Zissler J, Riekher V, Bäuerle T, Hanspach J, Laun FB, Nagel L, Mészáros L, Zunke F, Winkler J, Naumann UJ, Schwingen N, Neumaier E, Liesz A, Quintana F, Rothhammer V. TGFα controls checkpoints in CNS resident and infiltrating immune cells to promote resolution of inflammation. Nat Commun 2025; 16:5344. [PMID: 40537468 DOI: 10.1038/s41467-025-60363-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/19/2025] [Indexed: 06/22/2025] Open
Abstract
After acute lesions in the central nervous system (CNS), the interaction of microglia, astrocytes, and infiltrating immune cells decides over their resolution or chronification. However, this CNS-intrinsic cross-talk is poorly characterized. Analyzing cerebrospinal fluid (CSF) samples of Multiple Sclerosis (MS) patients as well as CNS samples of female mice with experimental autoimmune encephalomyelitis (EAE), the animal model of MS, we identify microglia-derived TGFα as key factor driving recovery. Through mechanistic in vitro studies, in vivo treatment paradigms, scRNA sequencing, CRISPR-Cas9 genetic perturbation models and MRI in the EAE model, we show that together with other glial and non-glial cells, microglia secrete TGFα in a highly regulated temporospatial manner in EAE. Here, TGFα contributes to recovery by decreasing infiltrating T cells, pro-inflammatory myeloid cells, oligodendrocyte loss, demyelination, axonal damage and neuron loss even at late disease stages. In a therapeutic approach in EAE, blood-brain barrier penetrating intranasal application of TGFα attenuates pro-inflammatory signaling in astrocytes and CNS infiltrating immune cells while promoting neuronal survival and lesion resolution. Together, microglia-derived TGFα is an important mediator of glial-immune crosstalk, highlighting its therapeutic potential in resolving acute CNS inflammation.
Collapse
Affiliation(s)
- Lena Lößlein
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Mathias Linnerbauer
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Finnja Zuber
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Thanos Tsaktanis
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Oliver Vandrey
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Anne Peter
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Franziska Panier
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Julia Zissler
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Vivienne Riekher
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Tobias Bäuerle
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
- Department of Diagnostic and Interventional Radiology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Jannis Hanspach
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Frederik B Laun
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Lisa Nagel
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Lisa Mészáros
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Friederike Zunke
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Jürgen Winkler
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Ulrike J Naumann
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Nora Schwingen
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Emely Neumaier
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Arthur Liesz
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Francisco Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Veit Rothhammer
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany.
| |
Collapse
|
2
|
Li Y, Zhao Q, Wang Y, Du W, Yang R, Wu J, Li Y. Lipid droplet accumulation in microglia and their potential roles. Lipids Health Dis 2025; 24:215. [PMID: 40514678 PMCID: PMC12166618 DOI: 10.1186/s12944-025-02633-3] [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: 02/04/2025] [Accepted: 06/02/2025] [Indexed: 06/16/2025] Open
Abstract
Microglia are the resident immune cells of the central nervous system (CNS), where lipid metabolism is critical for maintaining homeostasis. In response to various external stimuli, they demonstrate a range of phenotypic expressions and lipid metabolic reprogramming. Lipid droplets (LDs) are dynamic organelles that function beyond energy storage, actively participating in neuropathological progress. Recent investigations have identified a subset of microglia characterized by the accumulation of LDs, referred to as "lipid-droplet-accumulating microglia" (LDAM). This review aims to investigate the processes involved in LD formation and degradation, the factors that modulate them, focusing particularly on the function of LDAM and their implications for CNS disorders. By synthesizing current evidence, we clarify the biological significance of LDs in these cells and their therapeutic targeting potential, providing new directions for future research.
Collapse
Affiliation(s)
- Yunxia Li
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Qi Zhao
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Yan Wang
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
- School of Health Medicine, Nantong Institute of Technology, Nantong, Jiangsu, China
| | - Wenyi Du
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Riyun Yang
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Jian Wu
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China.
| | - Yi Li
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
3
|
Prakash P, Manchanda P, Paouri E, Bisht K, Sharma K, Rajpoot J, Wendt V, Hossain A, Wijewardhane PR, Randolph CE, Chen Y, Stanko S, Gasmi N, Gjojdeshi A, Card S, Fine J, Jethava KP, Clark MG, Dong B, Ma S, Crockett A, Thayer EA, Nicolas M, Davis R, Hardikar D, Allende D, Prayson RA, Zhang C, Davalos D, Chopra G. Amyloid-β induces lipid droplet-mediated microglial dysfunction via the enzyme DGAT2 in Alzheimer's disease. Immunity 2025; 58:1536-1552.e8. [PMID: 40393454 PMCID: PMC12168635 DOI: 10.1016/j.immuni.2025.04.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/30/2024] [Accepted: 04/22/2025] [Indexed: 05/22/2025]
Abstract
Microglial phagocytosis genes have been linked to increased risk for Alzheimer's disease (AD), but the mechanisms translating genetic association to cellular dysfunction remain unknown. Here, we showed that microglia formed lipid droplets (LDs) upon amyloid-β (Aβ) exposure and that LD loads increased with proximity to amyloid plaques in brains from individuals with AD and the 5xFAD mouse model. LD-laden microglia exhibited defects in Aβ phagocytosis, and unbiased lipidomic analyses identified a parallel decrease in free fatty acids (FFAs) and increase in triacylglycerols (TGs) as the key metabolic transition underlying LD formation. Diacylglycerol O-acyltransferase 2 (DGAT2)-a key enzyme that converts FFAs to TGs-promoted microglial LD formation and was increased in mouse 5xFAD and human AD brains. Pharmacologically targeting DGAT2 improved microglial uptake of Aβ and reduced plaque load and neuronal damage in 5xFAD mice. These findings identify a lipid-mediated mechanism underlying microglial dysfunction that could become a therapeutic target for AD.
Collapse
Affiliation(s)
- Priya Prakash
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Palak Manchanda
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Evi Paouri
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Kanchan Bisht
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Kaushik Sharma
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Jitika Rajpoot
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Victoria Wendt
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Ahad Hossain
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | | | - Caitlin E Randolph
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Yihao Chen
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Sarah Stanko
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Nadia Gasmi
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Anxhela Gjojdeshi
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Sophie Card
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Jonathan Fine
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Krupal P Jethava
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Matthew G Clark
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Bin Dong
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Seohee Ma
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Alexis Crockett
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Elizabeth A Thayer
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Marlo Nicolas
- Division of Pathology, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Ryann Davis
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Dhruv Hardikar
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Daniela Allende
- Division of Pathology, Cleveland Clinic, Cleveland, OH 44106, USA
| | | | - Chi Zhang
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Dimitrios Davalos
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
| | - Gaurav Chopra
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA; Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
4
|
Lee Y, Jung I, Lee DW, Seo Y, Kim S, Park HC. Transforming growth factor-β receptor I kinase plays a crucial role in oligodendrocyte regeneration after demyelination. Biomed Pharmacother 2025; 187:118094. [PMID: 40315672 DOI: 10.1016/j.biopha.2025.118094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/08/2025] [Accepted: 04/24/2025] [Indexed: 05/04/2025] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by the loss of oligodendrocytes (OLs) and axon demyelination in the central nervous system. Most therapeutic agents focus on regulating the immune response by suppressing autoimmune reactions. Therefore, developing therapeutic agents that promote remyelination by OLs at disease sites that have already undergone demyelination is necessary. In this study, we generated a new transgenic zebrafish with high efficiency for OL ablation and established a high-throughput screening (HTS)-based platform to identify therapeutic candidates that promote remyelination. Next, we screened a library of kinase inhibitors and identified one candidate, a transforming growth factor-β receptor I (TGF-βRI) kinase inhibitor. Treatment with this kinase inhibitor rapidly recruited microglia to induce clearance of myelin debris, early after OL removal. It also increased the proliferation of OL progenitor cells in demyelinating zebrafish larvae, resulting in restored OL numbers and reduced locomotor activity. Based on these results, we expect our HTS-based platform, along with our newly developed zebrafish model, to be very useful for identifying therapeutic agents that promote remyelination. Furthermore, since the candidate TGF-βRI kinase inhibitor identified in this study restored the phenotype following demyelination, we suggest that TGF-βRI kinase may potentially be a therapeutic target for the treatment of demyelinating diseases.
Collapse
Affiliation(s)
- Yunkyoung Lee
- Core Research and Development Center, Korea University Ansan Hospital, Ansan, Gyeonggi-do 15588, Republic of Korea; Zebrafish Translational Medical Research Center, College of Medicine, Korea University, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Inyoung Jung
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Erling Skalgssons gate 1, Trondheim 7030, Norway
| | - Dong-Won Lee
- Core Research and Development Center, Korea University Ansan Hospital, Ansan, Gyeonggi-do 15588, Republic of Korea; Zebrafish Translational Medical Research Center, College of Medicine, Korea University, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Yongbo Seo
- Zebrafish Translational Medical Research Center, College of Medicine, Korea University, Ansan, Gyeonggi-do 15588, Republic of Korea
| | - Suhyun Kim
- Zebrafish Translational Medical Research Center, College of Medicine, Korea University, Ansan, Gyeonggi-do 15588, Republic of Korea; Department of Convergence Medicine, Korea University College of Medicine, Seoul 04763, Republic of Korea
| | - Hae-Chul Park
- Core Research and Development Center, Korea University Ansan Hospital, Ansan, Gyeonggi-do 15588, Republic of Korea; Zebrafish Translational Medical Research Center, College of Medicine, Korea University, Ansan, Gyeonggi-do 15588, Republic of Korea; Department of Convergence Medicine, Korea University College of Medicine, Seoul 04763, Republic of Korea.
| |
Collapse
|
5
|
Zhang Y, Shi Y, Wang L, Li Z, Wang Y, Yan J, Sun X, Luo Q, Li L. TREM2 activation reduces white matter injury via PI3K/Akt/GSK-3β signalling after intracerebral haemorrhage. Br J Pharmacol 2025; 182:2542-2559. [PMID: 39965632 DOI: 10.1111/bph.17475] [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: 09/07/2024] [Revised: 12/06/2024] [Accepted: 12/30/2024] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND White matter injury (WMI) considerably exacerbates the prognosis following intracerebral haemorrhage (ICH). While the triggering receptor on myeloid cells 2 (TREM2) is recognized for its neuroprotective roles in a range of neurological disorders through the modulation of neuroinflammation, phagocytosis, promoting cell survival, its specific function in WMI after ICH has yet to be fully elucidated. METHODS This study involved inducing ICH in mice through autologous blood injection. Neurological functions were tested via behavioural assessments and electrophysiological recordings. WMI was examined using immunofluorescence, Luxol fast blue staining, MRI and transmission electron microscopy. Microglia were isolated and analysed using real-time polymerase chain reaction (PCR). Microglia depletion was achieved with PLX3397, primary cultures of microglia and oligodendrocytes were investigated. RESULTS The activation of TREM2 resulted in improved neurological outcomes after ICH, correlated with reduced WMI, demonstrated by decreased white matter loss in the corpus striatum, reduced damage to the nodes of Ranvier, and better preservation of myelin and white matter tract integrity. These neuroprotective effects were attributed to changes in microglial states mediated via the PI3K/Akt/GSK-3β signalling pathway. However, the neuroprotective advantages conferred by TREM2 activation were negated in TREM2 KO mice, either through microglia depletion or inhibition of PI3K. CONCLUSIONS This research is the first to illustrate that TREM2 activation mitigates WMI following ICH through a microglia-dependent mechanism involving the PI3K/Akt/GSK-3β pathway. TREM2 represents a potential therapeutic target for ICH.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Neurosurgery, Nanchong Central Hospital, Nanchong, China
| | - You Shi
- Department of Neuro-oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Lin Wang
- Department of Neurosurgery, Nanchong Central Hospital, Nanchong, China
| | - Zhao Li
- Emergency Department, Chengdu First People's Hospital, Chengdu, China
| | - Yingwen Wang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jin Yan
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaochuan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qing Luo
- Department of Ultrasound, Chong Gang General Hospital, Chongqing, China
| | - Lin Li
- Department of Neurosurgery, chong gang general hospital, Chongqing, China
| |
Collapse
|
6
|
Burtscher J, Motl RW, Berek K, Ehrenreich H, Kopp M, Hohenauer E. Hypoxia in multiple sclerosis. Redox Biol 2025; 83:103666. [PMID: 40347693 DOI: 10.1016/j.redox.2025.103666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 05/05/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025] Open
Abstract
Low oxygen availability (hypoxia) is a prominent but poorly understood feature in multiple sclerosis (MS). Whether hypoxia causes or drives MS pathology and symptoms or whether it is a consequence of other pathological events, such as inflammation and vascular dysfunction, is unknown. Here, we summarize the available literature on the interplay between hypoxia and both pathological and symptomatic features of MS. Severe environmental hypoxia (i.e., altitude) may trigger or facilitate MS-related events, possibly by exacerbating tissue hypoxia in the central nervous system. Accordingly, increasing oxygen supply can mitigate pathological and clinical parameters in MS models. In contrast, stimulating the endogenous hypoxia response and adaptation systems by controlled exposure to hypoxia (hypoxia conditioning) renders the central nervous system more resistant to hypoxic insults, thereby attenuating pathology and symptomatology in MS models. Overlapping mechanisms likely play a role in the benefits conferred by physical activity in MS. We provide an integrative model to explain the paradoxically beneficial outcomes of both increased and decreased ambient oxygen conditions. In conclusion, controlled exposure to hypoxia, perhaps in combination with exercise, is a promising, possibly disease-course modifying therapeutic approach for MS. However, many open questions remain.
Collapse
Affiliation(s)
- Johannes Burtscher
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria.
| | - Robert W Motl
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL, USA
| | - Klaus Berek
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Hannelore Ehrenreich
- Experimental Medicine, Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, J 5, Mannheim, Germany
| | - Martin Kopp
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
| | - Erich Hohenauer
- Rehabilitation and Exercise Science Laboratory, Department of Business Economics, Health and Social Care, University of Applied Sciences and Arts of Southern Switzerland, Landquart, Switzerland; Department of Neurosciences and Movement Science, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
7
|
Xie Y, Wang X, Liu S, He Z, Zhang H, Yu Z, Xie M, Wang W. Transforming Growth Factor β1 Protects Against Ischemic Demyelination via Regulating Microglial Lipid Metabolism Pathway. Stroke 2025; 56:1554-1568. [PMID: 40160039 DOI: 10.1161/strokeaha.124.048206] [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: 06/17/2024] [Revised: 01/13/2025] [Accepted: 02/10/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Chronic cerebral hypoperfusion-induced white matter lesions are an important cause of vascular cognitive impairment in aging life. TGF-β1 (transforming growth factor β1) is widely recognized as a multifunctional cytokine participating in numerous pathophysiological processes in the central nervous system. In this study, we aimed to evaluate the neuroprotective potentials of TGF-β1 in ischemic white matter lesions. METHODS A mouse model of bilateral common carotid artery stenosis was established to imitate the ischemic white matter lesions. The agonist of the TGF-β1 pathway was continuously applied via intraperitoneal injection. The Morris water maze test and gait analysis system were used to assess the cognitive and gait disorders in modeling mice. The Luxol fast blue staining, immunofluorescence, and electron microscopy were conducted to determine the severity of demyelinating lesions, microglial activation, and dysfunction of the autophagy-lysosomal pathway in microglia. Furthermore, primary cultured microglia were exposed to extracted myelin debris and TGF-β1 in vitro to explore the underlying mechanisms. RESULTS As evaluated by behavioral tests, TGF-β1 significantly alleviated the cognitive dysfunction and gait disorder in bilateral common carotid artery stenosis-modeling mice. The demyelinating lesion and remyelination process were also found to be highly improved by activation of the TGF-β1 pathway. The results of immunostaining and electron microscopy showed that TGF-β1 could ameliorate microglial activation and the dysfunction of lipid metabolism in myelin-engulfed microglia. Mechanistically, in primary cultured microglia exposed to myelin debris, administration of TGF-β1 notably mitigated the inflammatory response and accumulation of intracellular lipid droplets via promoting the lipid droplets degradation in the autophagy-lysosomal pathway, as quantified by flow cytometry, immunostaining, Western blot, etc. Yet, the application of autophagy inhibitor 3-methyladenine significantly reversed the above anti-inflammatory effects of TGF-β1. CONCLUSIONS TGF-β1 relieved cognitive deficit, demyelinating lesions, and microglia-mediated neuroinflammation in bilateral common carotid artery stenosis modeling by reducing abnormal lipid droplet accumulation and dysfunction of the autophagy-lysosomal pathway in microglia. Clinically, staged activation of the TGF-β1 pathway may become a potential target and promising treatment for ischemic white matter lesions and vascular cognitive impairment.
Collapse
Affiliation(s)
- Yi Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.), Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology, Wuhan, China (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.)
| | - Xinyue Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.), Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology, Wuhan, China (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.)
| | - Shuai Liu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College (S.L.), Huazhong University of Science and Technology, Wuhan, China
| | - Ziyu He
- Department of Neurology, Tongji Hospital, Tongji Medical College (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.), Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology, Wuhan, China (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.)
| | - Hang Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.), Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology, Wuhan, China (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.)
| | - Zhiyuan Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.), Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology, Wuhan, China (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.)
| | - Minjie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.), Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology, Wuhan, China (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.)
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.), Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College (W.W.), Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction Huazhong University of Science and Technology, Wuhan, China (Y.X., X.W., Z.H., H.Z., Z.Y., M.X., W.W.)
| |
Collapse
|
8
|
Yu JB, Hong C, Ren XW, Guo W, Chen YF, Ji J, Zhang XY, Sun XL. FTY720 Modulating Microglia-Mediated Cholesterol Recycling via TREM2 Promotes Remyelination Following Ischemic Damage. Stroke 2025. [PMID: 40260538 DOI: 10.1161/strokeaha.124.049745] [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: 10/19/2024] [Revised: 03/15/2025] [Accepted: 04/01/2025] [Indexed: 04/23/2025]
Abstract
BACKGROUND Following ischemic white matter damage, microglia are responsible for phagocytosing and degrading cholesterol-rich myelin debris, storing them as lipid droplets. However, our understanding of how microglia process this engulfed material remains limited. Our previous findings identified FTY720 as a high-affinity ligand for microglial TREM2 (triggering receptor expressed on myeloid cells 2). Therefore, we aimed to reveal the role of FTY720 targeting TREM2 in regulating microglial cholesterol metabolism during remyelination. METHODS Chronic ischemic white matter damage was induced by bilateral carotid artery stenosis in male wild-type and TREM2-/- mice. FTY720 was administered daily via intraperitoneal injection for 28 days following bilateral carotid artery stenosis surgery. Cognitive function, white matter integrity, accumulation of cholesterol and lipid droplets in microglia, and oligodendrocyte differentiation were evaluated using behavioral tests, transmission electron microscopy, immunofluorescence, and biochemical analyses. In vitro coculture systems were used to evaluate cholesterol transfer and remyelination efficacy. RESULTS FTY720 significantly alleviated cognitive deficits and promoted remyelination in bilateral carotid artery stenosis mice, as evidenced by enhanced performance in the Morris water maze and reduced demyelination observed via transmission electron microscopy and immunofluorescence. This therapeutic effect was absent in TREM2-/- bilateral carotid artery stenosis mice. Mechanistically, FTY720 promoted the redistribution of ABCA1 (ATP-binding cassette transporter A1) from lysosomes to the cell membrane in microglia via TREM2, which facilitated cholesterol efflux and reduced the accumulation of intracellular cholesterol and lipid droplets. Additionally, in vitro coculture experiments revealed that FTY720 enhanced cholesterol transfer from microglia to oligodendrocytes through TREM2, thereby promoting oligodendrocyte myelination. CONCLUSIONS Our study suggested that FTY720 regulated the recycling of myelin-derived cholesterol from microglia through TREM2, supplying cholesterol to oligodendrocytes and supporting remyelination, thus offering a novel therapeutic target for ischemic white matter damage.
Collapse
Affiliation(s)
- Jian-Bing Yu
- Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, China (J.-B.Y., C.H., X.-W.R., W.G., Y.-F.C., J.J., X.-Y.Z., X.-L.S.)
| | - Chen Hong
- Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, China (J.-B.Y., C.H., X.-W.R., W.G., Y.-F.C., J.J., X.-Y.Z., X.-L.S.)
| | - Xue-Wei Ren
- Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, China (J.-B.Y., C.H., X.-W.R., W.G., Y.-F.C., J.J., X.-Y.Z., X.-L.S.)
| | - Wei Guo
- Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, China (J.-B.Y., C.H., X.-W.R., W.G., Y.-F.C., J.J., X.-Y.Z., X.-L.S.)
| | - Ye-Fan Chen
- Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, China (J.-B.Y., C.H., X.-W.R., W.G., Y.-F.C., J.J., X.-Y.Z., X.-L.S.)
| | - Juan Ji
- Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, China (J.-B.Y., C.H., X.-W.R., W.G., Y.-F.C., J.J., X.-Y.Z., X.-L.S.)
| | - Xi-Yue Zhang
- Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, China (J.-B.Y., C.H., X.-W.R., W.G., Y.-F.C., J.J., X.-Y.Z., X.-L.S.)
| | - Xiu-Lan Sun
- Neuroprotective Drug Discovery Key Laboratory, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, China (J.-B.Y., C.H., X.-W.R., W.G., Y.-F.C., J.J., X.-Y.Z., X.-L.S.)
- Nanjing University of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, China (X.-L.S.)
| |
Collapse
|
9
|
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
|
10
|
Hou J, Magliozzi R, Chen Y, Wu J, Wulf J, Strout G, Fang X, Colonna M. Acute TREM2 inhibition depletes MAFB-high microglia and hinders remyelination. Proc Natl Acad Sci U S A 2025; 122:e2426786122. [PMID: 40131948 PMCID: PMC12002275 DOI: 10.1073/pnas.2426786122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 02/18/2025] [Indexed: 03/27/2025] Open
Abstract
We investigated the role of Triggering Receptor Expressed on Myeloid cells 2 (TREM2) in myelin regeneration in the brain. TREM2 is a receptor that activates microglia, which are crucial for clearing myelin debris and promoting remyelination. Previous studies in a mouse model of demyelination induced by the copper-chelating agent Cuprizone (CPZ) have shown that stimulation of TREM2 with a monoclonal antibody reduces demyelination, while deleting the Trem2 gene in mice impairs remyelination. Here, we blocked TREM2 function acutely with an antibody during both the demyelination and remyelination phases of the CPZ model and analyzed the impact of the antibody treatment on myelination and gene expression in single cells. We found that blocking TREM2 depleted a distinct population of microglia with high expression of the transcription factor MAFB during remyelination. The loss of these MAFB-high microglia was linked to impaired generation of myelinating oligodendrocytes. Importantly, we identified MAFB+ microglia in acute and acute-chronic brain lesions from individuals with multiple sclerosis (MS), but not in inactive lesions. We conclude that TREM2 is essential for maintaining a population of MAFB-high microglia that is associated with myelin repair. This finding has significant implications for understanding demyelinating diseases like MS and suggests that stimulating TREM2 could be a promising therapeutic approach for myelin repair.
Collapse
Affiliation(s)
- Jinchao Hou
- Department of Anesthesiology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou310052, China
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO63110
| | - Roberta Magliozzi
- Department of Brain Sciences, Faculty of Medicine, Imperial College London, LondonW12 0NN, United Kingdom
- Neurology Section of Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona371734, Italy
| | - Yun Chen
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO63110
- Department of Neurology, Washington University School of Medicine, St. Louis, MO63110
| | - Junjie Wu
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO63110
- Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, MO63110
| | - John Wulf
- Department of Neuroscience, Washington University Center for Cellular Imaging, Washington University School of Medicine, St. Louis, MO63110
| | - Gregory Strout
- Department of Neuroscience, Washington University Center for Cellular Imaging, Washington University School of Medicine, St. Louis, MO63110
| | - Xiangming Fang
- Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou310003, China
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO63110
| |
Collapse
|
11
|
Montilla A, Zabala A, Calvo I, Bosch-Juan M, Tomé-Velasco I, Mata P, Koster M, Sierra A, Kooistra SM, Soria FN, Eggen BJL, Fresnedo O, Fernández JA, Tepavcevic V, Matute C, Domercq M. Microglia regulate myelin clearance and cholesterol metabolism after demyelination via interferon regulatory factor 5. Cell Mol Life Sci 2025; 82:131. [PMID: 40137979 PMCID: PMC11947375 DOI: 10.1007/s00018-025-05648-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 12/16/2024] [Accepted: 01/08/2025] [Indexed: 03/29/2025]
Abstract
Interferon regulatory factor 5 (IRF5) is a transcription factor that plays a role in orchestrating innate immune responses, particularly in response to viral infections. Notably, IRF5 has been identified as a microglia risk gene linked to multiple sclerosis (MS), but its specific role in MS pathogenesis remains unclear. Through the use of Irf5-/- mice, our study uncovers a non-canonical function of IRF5 in MS recovery. Irf5-/- mice exhibited increased damage in an experimental autoimmune encephalomyelitis (EAE) model and demonstrated impaired oligodendrocyte recruitment into the lesion core following lysolecithin-induced demyelination. Transcriptomic and lipidomic analyses revealed that IRF5 has a role in microglia-mediated myelin phagocytosis, lipid metabolism, and cholesterol homeostasis. Indeed, Irf5-/- microglia phagocytose myelin, but myelin debris is not adequately degraded, leading to an accumulation of lipid droplets, cholesterol esters, and cholesterol crystals within demyelinating lesions. This abnormal buildup can hinder remyelination processes. Importantly, treatments that promote cholesterol transport were found to reduce lipid droplet accumulation and mitigate the exacerbated damage in Irf5-/- mice with EAE. Altogether, our study identified the antiviral transcription factor IRF5 as a key transcriptional regulator of lipid degradation and cholesterol homeostasis and suggest that loss of IRF5 function leads to pathogenic lipid accumulation in microglia, thereby obstructing remyelination. These data and the fact that Irf5 polymorphisms are significantly associated with MS, highlight IRF5 as a potential therapeutic target to promote regenerative responses.
Collapse
Affiliation(s)
- Alejandro Montilla
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain.
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain.
| | - Alazne Zabala
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Ibai Calvo
- Department of Physical Chemistry, Faculty of Sciences, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Marina Bosch-Juan
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Irene Tomé-Velasco
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Paloma Mata
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Mirjam Koster
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Amanda Sierra
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Ikerbasque Foundation, E-48009, Bilbao, Spain
- Department of Biochemistry and Molecular Biology, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Susanne M Kooistra
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Federico N Soria
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Ikerbasque Foundation, E-48009, Bilbao, Spain
| | - Bart J L Eggen
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Olatz Fresnedo
- Lipids & Liver Research Group, Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - José Andrés Fernández
- Department of Physical Chemistry, Faculty of Sciences, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Vanja Tepavcevic
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Carlos Matute
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain.
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain.
| | - María Domercq
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain.
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain.
| |
Collapse
|
12
|
Wang S, Cao C, Peng D. The various roles of TREM2 in cardiovascular disease. Front Immunol 2025; 16:1462508. [PMID: 40083551 PMCID: PMC11903262 DOI: 10.3389/fimmu.2025.1462508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025] Open
Abstract
Triggering receptor expressed on myeloid cells-2 (TREM2) is a transmembrane immune receptor that is expressed mainly on macrophages. As a pathology-induced immune signaling hub, TREM2 senses tissue damage and activates immune remodeling in response. Previous studies have predominantly focused on the TREM2 signaling pathway in Alzheimer's disease, metabolic syndrome, and cancer. Recent research has indicated that TREM2 signaling is also activated in various cardiovascular diseases. In this review, we summarize the current understanding and the unanswered questions regarding the role of TREM2 signaling in mediating the metabolism and function of macrophages in atherosclerosis and various models of heart failure. In the context of atherosclerosis, TREM2 signaling promotes foam cell formation and is crucial for maintaining macrophage survival and plaque stability through efferocytosis and cholesterol efflux. Recent studies on myocardial infarction, sepsis-induced cardiomyopathy, and hypertensive heart failure also implicated the protective role of TREM2 signaling in cardiac macrophages through efferocytosis and paracrine functions. Additionally, we discuss the clinical significance of elevated soluble TREM2 (sTREM2) in cardiovascular disease and propose potential therapies targeting TREM2. The overall aim of this review is to highlight the various roles of TREM2 in cardiovascular diseases and to provide a framework for therapeutic strategies targeting TREM2.
Collapse
Affiliation(s)
| | | | - Daoquan Peng
- Second Xiangya Hospital of Central South University, Cardiovascular Medicine, Changsha, China
| |
Collapse
|
13
|
Guo D, Wu J, Shen C, Zhang A, Zou T, Chen K, Huang W, Pan Y, Shen Y, Ji P, Zhong Y, Wen Q, Kong B, Xiang M, Ye B. Upregulation of LXRβ/ABCA1 pathway alleviates cochlear hair cell senescence of C57BL/6 J mice via reducing lipid droplet accumulation. Biogerontology 2025; 26:49. [PMID: 39890652 DOI: 10.1007/s10522-025-10192-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 01/20/2025] [Indexed: 02/03/2025]
Abstract
Senescence and loss of cochlear hair cells is an important pathologic basis of age-related hearing loss. Lipid droplet accumulation has previously been shown to play an important role in neurodegeneration; however, its role in age-related hearing loss has not yet been investigated. LXRβ/ABCA1 is a key pathway that regulates lipid metabolism, while its dysfunction can cause abnormal accumulation of lipid droplets in neurons, leading to neurodegeneration. In this study, we found that decreased expression of LXRβ/ABCA1, elevated levels of lipid droplet accumulation, and increased activation of the NLRP3 inflammasome were demonstrated in senescent cochlear hair cells in both animal and cellular models of age-related hearing loss. We then manipulated the LXRβ/ABCA1 pathway transduction of cochlear hair cells. Upregulation of LXRβ/ABCA1 in senescent hair cells was found to reduce the accumulation of lipid droplets, inhibit NLRP3 inflammasome activation, and ultimately alleviate cochlear hair cell senescence. In our study, we also found that NLRP3 inflammasome activation can abrogate the alleviated effect of LXRβ/ABCA1 pathway on the senescence of cochlear hair cells but did not affect the expression of LXRβ/ABCA1.Our study are the first to demonstrate that abnormal lipid droplet accumulation and decreased LXRβ/ABCA1 pathway are observed in cochlear hair cells following the occurrence of age-related hearing loss. Upregulation of LXRβ/ABCA1 in senescent cochlear hair cells can reduce lipid droplet accumulation in cochlear hair cells and alleviate their senescence, which may be related to the inhibition of NLRP3 inflammasome activation. These findings provide potential targets for the treatment of age-related hearing loss.
Collapse
Affiliation(s)
- Dongye Guo
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jichang Wu
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenling Shen
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Andi Zhang
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianyuan Zou
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kaili Chen
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiyi Huang
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi Pan
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yilin Shen
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peilin Ji
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiming Zhong
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Audiology & Speech-Language Pathology, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing Wen
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bing Kong
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingliang Xiang
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China.
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Audiology & Speech-Language Pathology, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Bin Ye
- Department of Otolaryngology & Head and Neck Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Translational Medicine On Ear and Nose Diseases, Shanghai, China.
- Ear Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Audiology & Speech-Language Pathology, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
14
|
Liu X, Zhao X, Qiu M, Yang J. Cell surface receptor-mediated signaling in CNS regeneration. Neuroscience 2024; 562:198-208. [PMID: 39486572 DOI: 10.1016/j.neuroscience.2024.10.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/21/2024] [Accepted: 10/25/2024] [Indexed: 11/04/2024]
Abstract
Degenerative diseases and injuries of central nervous system (CNS) often cause nerve cell apoptosis and neural dysfunction. Protection of surviving cells or inducing the differentiation of stem cells into functional cells is considered to be an important way of neurorepair. In addition, transdifferentiation technology emerged recently is expected to provide new solutions for nerve regeneration. Cell surface receptors are transmembrane proteins embedded in cytoplasmic membrane, and play crucial roles in maintaining communication between extracellular signals and intracellular signaling processes. The extracellular microenvironment changed dramatically upon neural lesion, exploring the biological function of signals mediated by cell surface receptors will help to develop molecular strategies for nerve regeneration. An increasing number of studies have reported that cell surface receptor-mediated signaling affects the survival, differentiation, and functioning of neural cells, and even regulate their trans-lineage reprogramming. Here, we provide a review on the roles of cell surface receptors in CNS regeneration, thus providing new cues for better treatment of neurodegenerative diseases or nerve injury.
Collapse
Affiliation(s)
- Xinyu Liu
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; Department of Immunology and International Cancer Center, Shenzhen University Medical School, Shenzhen 518000, China
| | - Xiaofeng Zhao
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; Key Laboratory of Organ Development and Regeneration of Zhejiang Province, Hangzhou 311121, China
| | - Mengsheng Qiu
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; Key Laboratory of Organ Development and Regeneration of Zhejiang Province, Hangzhou 311121, China.
| | - Junlin Yang
- College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China; Key Laboratory of Organ Development and Regeneration of Zhejiang Province, Hangzhou 311121, China.
| |
Collapse
|
15
|
Shafer C, Di Lucente J, Mendiola UR, Maezawa I, Jin LW, Neumann EK. Effects of Sex and Western Diet on Spatial Lipidomic Profiles for the Hippocampus, Cortex, and Corpus Callosum in Mice Using MALDI MSI. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2024; 35:2554-2563. [PMID: 38456419 PMCID: PMC11544704 DOI: 10.1021/jasms.3c00446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 03/09/2024]
Abstract
Diet is inextricably linked to human health and biological functionality. Reduced cognitive function among other health issues has been correlated with a western diet (WD) in mouse models, indicating that increases in neurodegeneration could be fueled in part by a poor diet. In this study, we use matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) to spatially map the lipidomic profiles of male and female mice that were fed a high-fat, high-sucrose WD for a period of 7 weeks. Our findings concluded that the cortex and corpus callosum showed significant lipid variation by WD in female mice, while there was little to no variation in the hippocampus, regardless of sex. On the other hand, lipid profiles were significantly affected by sex in all regions. Overall, 83 lipids were putatively identified in the mouse brain; among them, HexCer(40:1;O3) and PE(34:0) were found to have the largest statistical difference based on diet for female mice in the cortex and corpus callosum, respectively. Additional lipid changes are noted and can serve as a metric for understanding the brain's metabolomic response to changes in diet, particularly as it relates to disease.
Collapse
Affiliation(s)
- Catelynn
C. Shafer
- Department
of Chemistry, University of California,
Davis. Davis, California 95616, United States
| | - Jacopo Di Lucente
- Department
of Pathology and Laboratory Medicine, University
of California Davis, Sacramento, California 95817, United States
| | - Ulises Ruiz Mendiola
- Department
of Pathology and Laboratory Medicine, University
of California Davis, Sacramento, California 95817, United States
| | - Izumi Maezawa
- Department
of Pathology and Laboratory Medicine, University
of California Davis, Sacramento, California 95817, United States
| | - Lee-Way Jin
- Department
of Pathology and Laboratory Medicine, University
of California Davis, Sacramento, California 95817, United States
| | - Elizabeth K. Neumann
- Department
of Chemistry, University of California,
Davis. Davis, California 95616, United States
| |
Collapse
|
16
|
Harlina PW, Maritha V, Geng F, Nawaz A, Yuliana T, Subroto E, Dahlan HJ, Lembong E, Huda S. Comprehensive review on the application of omics analysis coupled with Chemometrics in gelatin authentication of food and pharmaceutical products. Food Chem X 2024; 23:101710. [PMID: 39206450 PMCID: PMC11350464 DOI: 10.1016/j.fochx.2024.101710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/16/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Gelatin is a protein molecule that can be hydrolyzed from collagen, animal bones, skin and it easily soluble in water. Source animals for gelatin ingredients must be evaluated, as well as their halal status. The omics method towards gelatin authentication in food and pharmaceutical products has several advantages, including high sensitivity and reliable data. Omics investigation employs the process of breaking down substances into small particles, hence enhancing the ability to detect a greater number of compounds. Omics study has the capability to identify substances at the subclass level, which makes it highly suitable for gelatin authentication. Gelatin lipids, metabolites, proteins, and volatile chemicals can be utilized as references to authenticate gelatin. In adopting gelatin authentication, lipidomics, metabolomics, proteomics, and volatilomics must be combined with chemometrics for data interpretation. Chemometrics can convert omics analysis data into easily viewable data. Chemometric approaches capable of presenting omics analysis data for gelatin authentication include PCA, HCA, PLS-DA, PLSR, SIMCA, and FACS. Visually chemometrically explain the differences in gelatin from different animal sources. The combination of omics analysis and chemometrics is a very promising technology for gelatin authentication in food and pharmaceutical products.
Collapse
Affiliation(s)
- Putri Widyanti Harlina
- Department of Food Industrial Technology, Faculty of Agro-Industrial Technology, Universitas Padjadjaran, 45363 Bandung, Indonesia
- Padjadjaran Halal Center, Universitas Padjadjaran, 45363 Bandung, Indonesia
| | - Vevi Maritha
- Pharmacy Study Program, Faculty of Health and Science, Universitas PGRI, Madiun, Indonesia
| | - Fang Geng
- Meat Processing Key Laboratory of Sichuan Province, School of Food and Biological Engineering, Chengdu University, Chengdu, 610106, China
| | - Asad Nawaz
- Hunan Engineering Technology Research Center for Comprehensive Development and Utilization of Biomass Resources, College of Chemistry and Bioengineering, Hunan University of Science and Engineering, 425199 Yongzhou, China
| | - Tri Yuliana
- Department of Food Industrial Technology, Faculty of Agro-Industrial Technology, Universitas Padjadjaran, 45363 Bandung, Indonesia
| | - Edy Subroto
- Department of Food Industrial Technology, Faculty of Agro-Industrial Technology, Universitas Padjadjaran, 45363 Bandung, Indonesia
| | - Havilah Jemima Dahlan
- Department of Food Industrial Technology, Faculty of Agro-Industrial Technology, Universitas Padjadjaran, 45363 Bandung, Indonesia
| | - Elazmanawati Lembong
- Department of Food Industrial Technology, Faculty of Agro-Industrial Technology, Universitas Padjadjaran, 45363 Bandung, Indonesia
| | - Syamsul Huda
- Department of Food Industrial Technology, Faculty of Agro-Industrial Technology, Universitas Padjadjaran, 45363 Bandung, Indonesia
| |
Collapse
|
17
|
Li L, Sun B, Harris OA, Luo J. TGF-β Signaling in Microglia: A Key Regulator of Development, Homeostasis and Reactivity. Biomedicines 2024; 12:2468. [PMID: 39595034 PMCID: PMC11592028 DOI: 10.3390/biomedicines12112468] [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: 09/27/2024] [Revised: 10/18/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
Microglia, the resident immune cells of the central nervous system (CNS), are crucial for normal brain development and function. They become reactive in response to brain injury and disease, a process known as microglial reactivity. This reactivity, along with microglial homeostasis, is tightly regulated by the local microenvironment and interactions with surrounding cells. The TGF-β signaling pathway plays an essential role in this regulation. Recent genetic studies employing microglia-specific manipulation of the TGF-β signaling pathway have shed light on its significance in microglial development, homeostasis and reactivity. This review provides an updated overview of how TGF-β signaling modulates microglial function and reactivity, contributing to our understanding of microglial biology in health and disease.
Collapse
Affiliation(s)
- Lulin Li
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Bryan Sun
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Odette A. Harris
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Polytrauma System of Care, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Jian Luo
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
- Polytrauma System of Care, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| |
Collapse
|
18
|
Tiwari V, Prajapati B, Asare Y, Damkou A, Ji H, Liu L, Naser N, Gouna G, Leszczyńska KB, Mieczkowski J, Dichgans M, Wang Q, Kawaguchi R, Shi Z, Swarup V, Geschwind DH, Prinz M, Gokce O, Simons M. Innate immune training restores pro-reparative myeloid functions to promote remyelination in the aged central nervous system. Immunity 2024; 57:2173-2190.e8. [PMID: 39053462 DOI: 10.1016/j.immuni.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 11/21/2023] [Accepted: 07/01/2024] [Indexed: 07/27/2024]
Abstract
The reduced ability of the central nervous system to regenerate with increasing age limits functional recovery following demyelinating injury. Previous work has shown that myelin debris can overwhelm the metabolic capacity of microglia, thereby impeding tissue regeneration in aging, but the underlying mechanisms are unknown. In a model of demyelination, we found that a substantial number of genes that were not effectively activated in aged myeloid cells displayed epigenetic modifications associated with restricted chromatin accessibility. Ablation of two class I histone deacetylases in microglia was sufficient to restore the capacity of aged mice to remyelinate lesioned tissue. We used Bacillus Calmette-Guerin (BCG), a live-attenuated vaccine, to train the innate immune system and detected epigenetic reprogramming of brain-resident myeloid cells and functional restoration of myelin debris clearance and lesion recovery. Our results provide insight into aging-associated decline in myeloid function and how this decay can be prevented by innate immune reprogramming.
Collapse
Affiliation(s)
- Vini Tiwari
- Institute of Neuronal Cell Biology, Technical University Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Bharat Prajapati
- Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 41390 Gothenburg, Sweden
| | - Yaw Asare
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Alkmini Damkou
- Institute of Neuronal Cell Biology, Technical University Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Hao Ji
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Lu Liu
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Nawraa Naser
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Garyfallia Gouna
- Institute of Neuronal Cell Biology, Technical University Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Katarzyna B Leszczyńska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 02093 Warsaw, Poland
| | - Jakub Mieczkowski
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 02093 Warsaw, Poland; 3P-Medicine Laboratory, Medical University of Gdańsk, 80211 Gdańsk, Poland
| | - Martin Dichgans
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), 81377 Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Qing Wang
- Departments of Neurology and Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Riki Kawaguchi
- Departments of Neurology and Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Psychiatry, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zechuan Shi
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Daniel H Geschwind
- Departments of Neurology and Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79085 Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Ozgun Gokce
- Munich Cluster of Systems Neurology (SyNergy), 81377 Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany; Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, 53127 Bonn, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), 81377 Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany.
| |
Collapse
|
19
|
Shang G, Shao Q, Lv K, Xu W, Ji J, Fan S, Kang X, Cheng F, Wang X, Wang Q. Hypercholesterolemia and the Increased Risk of Vascular Dementia: a Cholesterol Perspective. Curr Atheroscler Rep 2024; 26:435-449. [PMID: 38814418 DOI: 10.1007/s11883-024-01217-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2024] [Indexed: 05/31/2024]
Abstract
PURPOSE OF REVIEW Vascular dementia (VaD) is the second most prevalent type of dementia after Alzheimer's disease.Hypercholesterolemia may increase the risk of dementia, but the association between cholesterol and cognitive function is very complex. From the perspective of peripheral and brain cholesterol, we review the relationship between hypercholesterolemia and increased risk of VaD and how the use of lipid-lowering therapies affects cognition. RECENT FINDINGS Epidemiologic studies show since 1980, non-HDL-C levels of individuals has increased rapidly in Asian countries.The study has suggested that vascular risk factors increase the risk of VaD, such as disordered lipid metabolism. Dyslipidemia has been found to interact with chronic cerebral hypoperfusion to promote inflammation resulting in cognitive dysfunction in the brain.Hypercholesterolemia may be a risk factor for VaD. Inflammation could potentially serve as a link between hypercholesterolemia and VaD. Additionally, the potential impact of lipid-lowering therapy on cognitive function is also worth considering. Finding strategies to prevent and treat VaD is critical given the aging of the population to lessen the load on society. Currently, controlling underlying vascular risk factors is considered one of the most effective methods of preventing VaD. Understanding the relationship between abnormal cholesterol levels and VaD, as well as discovering potential serum biomarkers, is important for the early prevention and treatment of VaD.
Collapse
Affiliation(s)
- Guojiao Shang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No.11 East Beisanhuan Road, Chaoyang District, Beijing, China
| | - Qi Shao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No.11 East Beisanhuan Road, Chaoyang District, Beijing, China
| | - Kai Lv
- Department of Geratology, The Third Affiliated Hospital of Beijing University of Traditional Chinese Medicine, No.51 Xiaoguan Street, Andingmenwai, Chaoyang District, Beijing, China
| | - Wenxiu Xu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No.11 East Beisanhuan Road, Chaoyang District, Beijing, China
| | - Jing Ji
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No.11 East Beisanhuan Road, Chaoyang District, Beijing, China
| | - Shuning Fan
- Dongzhimen Hospital of Beijing University of Chinese Medicine, No.5 Haiyuncang, Dongcheng District, Beijing, China
| | - Xiangdong Kang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No.11 East Beisanhuan Road, Chaoyang District, Beijing, China
| | - Fafeng Cheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No.11 East Beisanhuan Road, Chaoyang District, Beijing, China.
| | - Xueqian Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No.11 East Beisanhuan Road, Chaoyang District, Beijing, China.
| | - Qingguo Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, No.11 East Beisanhuan Road, Chaoyang District, Beijing, China.
| |
Collapse
|
20
|
Etxeberria A, Shen YAA, Vito S, Silverman SM, Imperio J, Lalehzadeh G, Soung AL, Du C, Xie L, Choy MK, Hsiao YC, Ngu H, Cho CH, Ghosh S, Novikova G, Rezzonico MG, Leahey R, Weber M, Gogineni A, Elstrott J, Xiong M, Greene JJ, Stark KL, Chan P, Roth GA, Adrian M, Li Q, Choi M, Wong WR, Sandoval W, Foreman O, Nugent AA, Friedman BA, Sadekar S, Hötzel I, Hansen DV, Chih B, Yuen TJ, Weimer RM, Easton A, Meilandt WJ, Bohlen CJ. Neutral or Detrimental Effects of TREM2 Agonist Antibodies in Preclinical Models of Alzheimer's Disease and Multiple Sclerosis. J Neurosci 2024; 44:e2347232024. [PMID: 38830764 PMCID: PMC11255434 DOI: 10.1523/jneurosci.2347-23.2024] [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: 12/18/2023] [Revised: 05/06/2024] [Accepted: 05/25/2024] [Indexed: 06/05/2024] Open
Abstract
Human genetics and preclinical studies have identified key contributions of TREM2 to several neurodegenerative conditions, inspiring efforts to modulate TREM2 therapeutically. Here, we characterize the activities of three TREM2 agonist antibodies in multiple mixed-sex mouse models of Alzheimer's disease (AD) pathology and remyelination. Receptor activation and downstream signaling are explored in vitro, and active dose ranges are determined in vivo based on pharmacodynamic responses from microglia. For mice bearing amyloid-β (Aβ) pathology (PS2APP) or combined Aβ and tau pathology (TauPS2APP), chronic TREM2 agonist antibody treatment had limited impact on microglia engagement with pathology, overall pathology burden, or downstream neuronal damage. For mice with demyelinating injuries triggered acutely with lysolecithin, TREM2 agonist antibodies unexpectedly disrupted injury resolution. Likewise, TREM2 agonist antibodies limited myelin recovery for mice experiencing chronic demyelination from cuprizone. We highlight the contributions of dose timing and frequency across models. These results introduce important considerations for future TREM2-targeting approaches.
Collapse
Affiliation(s)
- Ainhoa Etxeberria
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Yun-An A Shen
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Stephen Vito
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Sean M Silverman
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Jose Imperio
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Guita Lalehzadeh
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Allison L Soung
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Changchun Du
- Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California 94080
| | - Luke Xie
- Translational Imaging, Genentech, Inc., South San Francisco, California 94080
| | - Man Kin Choy
- Translational Imaging, Genentech, Inc., South San Francisco, California 94080
| | - Yi-Chun Hsiao
- Antibody Engineering, Genentech, Inc., South San Francisco, California 94080
| | - Hai Ngu
- Pathology, Genentech, Inc., South San Francisco, California 94080
| | - Chang Hoon Cho
- Human Pathobiology and OMNI Reverse Translation, Genentech, Inc., South San Francisco, California 94080
| | - Soumitra Ghosh
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Gloriia Novikova
- Bioinformatics, Genentech, Inc., South San Francisco, California 94080
| | | | - Rebecca Leahey
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Martin Weber
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Alvin Gogineni
- Translational Imaging, Genentech, Inc., South San Francisco, California 94080
| | - Justin Elstrott
- Translational Imaging, Genentech, Inc., South San Francisco, California 94080
| | - Monica Xiong
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Jacob J Greene
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Kimberly L Stark
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Pamela Chan
- Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California 94080
| | - Gillie A Roth
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, Inc., South San Francisco, California 94080
| | - Max Adrian
- Pathology, Genentech, Inc., South San Francisco, California 94080
| | - Qingling Li
- Microchemistry Lipidomics and Proteomics, Genentech, Inc., South San Francisco, California 94080
| | - Meena Choi
- Microchemistry Lipidomics and Proteomics, Genentech, Inc., South San Francisco, California 94080
| | - Weng Ruh Wong
- Microchemistry Lipidomics and Proteomics, Genentech, Inc., South San Francisco, California 94080
| | - Wendy Sandoval
- Microchemistry Lipidomics and Proteomics, Genentech, Inc., South San Francisco, California 94080
| | - Oded Foreman
- Pathology, Genentech, Inc., South San Francisco, California 94080
| | - Alicia A Nugent
- Human Pathobiology and OMNI Reverse Translation, Genentech, Inc., South San Francisco, California 94080
| | - Brad A Friedman
- Bioinformatics, Genentech, Inc., South San Francisco, California 94080
| | - Shraddha Sadekar
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech, Inc., South San Francisco, California 94080
| | - Isidro Hötzel
- Antibody Engineering, Genentech, Inc., South San Francisco, California 94080
| | - David V Hansen
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Ben Chih
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
- Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California 94080
| | - Tracy J Yuen
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Robby M Weimer
- Translational Imaging, Genentech, Inc., South San Francisco, California 94080
| | - Amy Easton
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - William J Meilandt
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| | - Christopher J Bohlen
- Departments of Neuroscience, Genentech, Inc., South San Francisco, California 94080
| |
Collapse
|
21
|
Ou Z, Cheng Y, Ma H, Chen K, Lin Q, Chen J, Guo R, Huang Z, Cheng Q, Alaeiilkhchi N, Zhu Q, Huang Z, Jiang H. miR-223 accelerates lipid droplets clearance in microglia following spinal cord injury by upregulating ABCA1. J Transl Med 2024; 22:659. [PMID: 39010173 PMCID: PMC11247820 DOI: 10.1186/s12967-024-05480-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/03/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Spinal cord injury (SCI) is characterized by extensive demyelination and inflammatory responses. Facilitating the clearance of lipid droplets (LDs) within microglia contributes to creating a microenvironment that favors neural recovery and provides essential materials for subsequent remyelination. Therefore, investigating MicroRNAs (miRNAs) that regulate lipid homeostasis after SCI and elucidating their potential mechanisms in promoting LDs clearance in microglia have become focal points of SCI research. METHODS We established a subacute C5 hemicontusion SCI model in mice and performed transcriptomic sequencing on the injury epicenter to identify differentially expressed genes and associated pathways. Confocal imaging was employed to observe LDs accumulation. Multi-omics analyses were conducted to identify differentially expressed mRNA and miRNA post-SCI. Pathway enrichment analysis and protein-protein interaction network construction were performed using bioinformatics methods, revealing miR-223-Abca1 as a crucial miRNA-mRNA pair in lipid metabolism regulation. BV2 microglia cell lines overexpressing miR-223 were engineered, and immunofluorescence staining, western blot, and other techniques were employed to assess LDs accumulation, relevant targets, and inflammatory factor expression, confirming its role in regulating lipid homeostasis in microglia. RESULTS Histopathological results of our hemicontusion SCI model confirmed LDs aggregation at the injury epicenter, predominantly within microglia. Our transcriptomic analysis during the subacute phase of SCI in mice implicated ATP-binding cassette transporter A1 (Abca1) as a pivotal gene in lipid homeostasis, cholesterol efflux and microglial activation. Integrative mRNA-miRNA multi-omics analysis highlighted the crucial role of miR-223 in the neuroinflammation process following SCI, potentially through the regulation of lipid metabolism via Abca1. In vitro experiments using BV2 cells overexpressing miR-223 demonstrated that elevated levels of miR-223 enhance ABCA1 expression in myelin debris and LPS-induced BV2 cells. This promotes myelin debris degradation and LDs clearance, and induces a shift toward an anti-inflammatory M2 phenotype. CONCLUSIONS In summary, our study unveils the critical regulatory role of miR-223 in lipid homeostasis following SCI. The mechanism by which this occurs involves the upregulation of ABCA1 expression, which facilitates LDs clearance and myelin debris degradation, consequently alleviating the lipid burden, and inhibiting inflammatory polarization of microglia. These findings suggest that strategies to enhance miR-223 expression and target ABCA1, thereby augmenting LDs clearance, may emerge as appealing new clinical targets for SCI treatment.
Collapse
Affiliation(s)
- Zhilin Ou
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Yongquan Cheng
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Hao Ma
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Kai Chen
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Qiong Lin
- School of Anesthesiology, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Jiayu Chen
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Ruqin Guo
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zhiping Huang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Qixian Cheng
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Nima Alaeiilkhchi
- International Collaboration on Repair Discoveries (ICORD), Blusson Spinal Cord Centre, University of British Columbia, Vancouver, Canada
| | - Qingan Zhu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Zucheng Huang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Hui Jiang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
22
|
Zveik O, Rechtman A, Ganz T, Vaknin-Dembinsky A. The interplay of inflammation and remyelination: rethinking MS treatment with a focus on oligodendrocyte progenitor cells. Mol Neurodegener 2024; 19:53. [PMID: 38997755 PMCID: PMC11245841 DOI: 10.1186/s13024-024-00742-8] [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: 05/29/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Multiple sclerosis (MS) therapeutic goals have traditionally been dichotomized into two distinct avenues: immune-modulatory-centric interventions and pro-regenerative strategies. Oligodendrocyte progenitor cells (OPCs) were regarded for many years solely in concern to their potential to generate oligodendrocytes and myelin in the central nervous system (CNS). However, accumulating data elucidate the multifaceted roles of OPCs, including their immunomodulatory functions, positioning them as cardinal constituents of the CNS's immune landscape. MAIN BODY In this review, we will discuss how the two therapeutic approaches converge. We present a model by which (1) an inflammation is required for the appropriate pro-myelinating immune function of OPCs in the chronically inflamed CNS, and (2) the immune function of OPCs is crucial for their ability to differentiate and promote remyelination. This model highlights the reciprocal interactions between OPCs' pro-myelinating and immune-modulating functions. Additionally, we review the specific effects of anti- and pro-inflammatory interventions on OPCs, suggesting that immunosuppression adversely affects OPCs' differentiation and immune functions. CONCLUSION We suggest a multi-systemic therapeutic approach, which necessitates not a unidimensional focus but a harmonious balance between OPCs' pro-myelinating and immune-modulatory functions.
Collapse
Affiliation(s)
- Omri Zveik
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel
| | - Ariel Rechtman
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel
| | - Tal Ganz
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel
| | - Adi Vaknin-Dembinsky
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel.
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel.
| |
Collapse
|
23
|
McManus RM, Latz E. NLRP3 inflammasome signalling in Alzheimer's disease. Neuropharmacology 2024; 252:109941. [PMID: 38565393 DOI: 10.1016/j.neuropharm.2024.109941] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/20/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
Every year, 10 million people develop dementia, the most common of which is Alzheimer's disease (AD). To date, there is no way to prevent cognitive decline and therapies are limited. This review provides a neuroimmunological perspective on the progression of AD, and discusses the immune-targeted therapies that are in preclinical and clinical trials that may impact the development of this disease. Specifically, we look to the role of the NLRP3 inflammasome, its triggers in the brain and how its activation can contribute to the progression of dementia. We summarise the range of inhibitors targeting the NLRP3 inflammasome and its downstream pathways that are under investigation, and discuss future therapeutic perspectives for this devastating condition.
Collapse
Affiliation(s)
- Róisín M McManus
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127, Bonn, Germany; Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany.
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany; Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, 7491, Trondheim, Norway; Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, 01605, USA; Deutsches Rheuma-Forschungszentrum (DRFZ), Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|
24
|
Zhang R, Wuerch E, Yong VW, Xue M. LXR agonism for CNS diseases: promises and challenges. J Neuroinflammation 2024; 21:97. [PMID: 38627787 PMCID: PMC11022383 DOI: 10.1186/s12974-024-03056-0] [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: 01/09/2024] [Accepted: 02/27/2024] [Indexed: 04/19/2024] Open
Abstract
The unfavorable prognosis of many neurological conditions could be attributed to limited tissue regeneration in central nervous system (CNS) and overwhelming inflammation, while liver X receptor (LXR) may regulate both processes due to its pivotal role in cholesterol metabolism and inflammatory response, and thus receives increasing attentions from neuroscientists and clinicians. Here, we summarize the signal transduction of LXR pathway, discuss the therapeutic potentials of LXR agonists based on preclinical data using different disease models, and analyze the dilemma and possible resolutions for clinical translation to encourage further investigations of LXR related therapies in CNS disorders.
Collapse
Affiliation(s)
- Ruiyi Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Emily Wuerch
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada.
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
25
|
Xiao YL, Gong Y, Qi YJ, Shao ZM, Jiang YZ. Effects of dietary intervention on human diseases: molecular mechanisms and therapeutic potential. Signal Transduct Target Ther 2024; 9:59. [PMID: 38462638 PMCID: PMC10925609 DOI: 10.1038/s41392-024-01771-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 02/05/2024] [Accepted: 02/18/2024] [Indexed: 03/12/2024] Open
Abstract
Diet, serving as a vital source of nutrients, exerts a profound influence on human health and disease progression. Recently, dietary interventions have emerged as promising adjunctive treatment strategies not only for cancer but also for neurodegenerative diseases, autoimmune diseases, cardiovascular diseases, and metabolic disorders. These interventions have demonstrated substantial potential in modulating metabolism, disease trajectory, and therapeutic responses. Metabolic reprogramming is a hallmark of malignant progression, and a deeper understanding of this phenomenon in tumors and its effects on immune regulation is a significant challenge that impedes cancer eradication. Dietary intake, as a key environmental factor, can influence tumor metabolism. Emerging evidence indicates that dietary interventions might affect the nutrient availability in tumors, thereby increasing the efficacy of cancer treatments. However, the intricate interplay between dietary interventions and the pathogenesis of cancer and other diseases is complex. Despite encouraging results, the mechanisms underlying diet-based therapeutic strategies remain largely unexplored, often resulting in underutilization in disease management. In this review, we aim to illuminate the potential effects of various dietary interventions, including calorie restriction, fasting-mimicking diet, ketogenic diet, protein restriction diet, high-salt diet, high-fat diet, and high-fiber diet, on cancer and the aforementioned diseases. We explore the multifaceted impacts of these dietary interventions, encompassing their immunomodulatory effects, other biological impacts, and underlying molecular mechanisms. This review offers valuable insights into the potential application of these dietary interventions as adjunctive therapies in disease management.
Collapse
Affiliation(s)
- Yu-Ling Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yue Gong
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ying-Jia Qi
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
26
|
Linnerbauer M, Lößlein L, Vandrey O, Peter A, Han Y, Tsaktanis T, Wogram E, Needhamsen M, Kular L, Nagel L, Zissler J, Andert M, Meszaros L, Hanspach J, Zuber F, Naumann UJ, Diebold M, Wheeler MA, Beyer T, Nirschl L, Cirac A, Laun FB, Günther C, Winkler J, Bäuerle T, Jagodic M, Hemmer B, Prinz M, Quintana FJ, Rothhammer V. The astrocyte-produced growth factor HB-EGF limits autoimmune CNS pathology. Nat Immunol 2024; 25:432-447. [PMID: 38409259 PMCID: PMC10907300 DOI: 10.1038/s41590-024-01756-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 01/12/2024] [Indexed: 02/28/2024]
Abstract
Central nervous system (CNS)-resident cells such as microglia, oligodendrocytes and astrocytes are gaining increasing attention in respect to their contribution to CNS pathologies including multiple sclerosis (MS). Several studies have demonstrated the involvement of pro-inflammatory glial subsets in the pathogenesis and propagation of inflammatory events in MS and its animal models. However, it has only recently become clear that the underlying heterogeneity of astrocytes and microglia can not only drive inflammation, but also lead to its resolution through direct and indirect mechanisms. Failure of these tissue-protective mechanisms may potentiate disease and increase the risk of conversion to progressive stages of MS, for which currently available therapies are limited. Using proteomic analyses of cerebrospinal fluid specimens from patients with MS in combination with experimental studies, we here identify Heparin-binding EGF-like growth factor (HB-EGF) as a central mediator of tissue-protective and anti-inflammatory effects important for the recovery from acute inflammatory lesions in CNS autoimmunity. Hypoxic conditions drive the rapid upregulation of HB-EGF by astrocytes during early CNS inflammation, while pro-inflammatory conditions suppress trophic HB-EGF signaling through epigenetic modifications. Finally, we demonstrate both anti-inflammatory and tissue-protective effects of HB-EGF in a broad variety of cell types in vitro and use intranasal administration of HB-EGF in acute and post-acute stages of autoimmune neuroinflammation to attenuate disease in a preclinical mouse model of MS. Altogether, we identify astrocyte-derived HB-EGF and its epigenetic regulation as a modulator of autoimmune CNS inflammation and potential therapeutic target in MS.
Collapse
Affiliation(s)
- Mathias Linnerbauer
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Lena Lößlein
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Oliver Vandrey
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Anne Peter
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Yanan Han
- Department of Clinical Neuroscience, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Thanos Tsaktanis
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Emile Wogram
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Maria Needhamsen
- Department of Clinical Neuroscience, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Lara Kular
- Department of Clinical Neuroscience, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Lisa Nagel
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Julia Zissler
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Marie Andert
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Lisa Meszaros
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Jannis Hanspach
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Finnja Zuber
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Ulrike J Naumann
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Martin Diebold
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Michael A Wheeler
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Tobias Beyer
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Lucy Nirschl
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Ana Cirac
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Frederik B Laun
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Claudia Günther
- Department of Medicine 1, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Jürgen Winkler
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tobias Bäuerle
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany
| | - Maja Jagodic
- Department of Clinical Neuroscience, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden
| | - Bernhard Hemmer
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Veit Rothhammer
- Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nuremberg, Erlangen, Germany.
- Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Erlangen, Germany.
| |
Collapse
|
27
|
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
|
Xie Y, Chen X, Wang X, Liu S, Chen S, Yu Z, Wang W. Transforming growth factor-β1 protects against white matter injury and reactive astrogliosis via the p38 MAPK pathway in rodent demyelinating model. J Neurochem 2024; 168:83-99. [PMID: 38183677 DOI: 10.1111/jnc.16037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/25/2023] [Accepted: 11/28/2023] [Indexed: 01/08/2024]
Abstract
In central nervous system (CNS), demyelination is a pathological process featured with a loss of myelin sheaths around axons, which is responsible for the diseases of multiple sclerosis, neuromyelitis optica, and so on. Transforming growth factor-beta1 (TGF-β1) is a multifunctional cytokine participating in abundant physiological and pathological processes in CNS. However, the effects of TGF-β1 on CNS demyelinating disease and its underlying mechanisms are controversial and not well understood. Herein, we evaluated the protective potential of TGF-β1 in a rodent demyelinating model established by lysophosphatidylcholine (LPC) injection. It was identified that supplement of TGF-β1 evidently rescued the cognitive deficit and motor dysfunction in LPC modeling mice assessed by novel object recognition and balance beam behavioral tests. Besides, quantified by luxol fast blue staining, immunofluorescence, and western blot, administration of TGF-β1 was found to significantly ameliorate the demyelinating lesion and reactive astrogliosis by suppressing p38 MAPK pathway. Mechanistically, the results of in vitro experiments indicated that treatment of TGF-β1 could directly promote the differentiation and migration of cultured oligodendrocytes. Our study revealed that modulating TGF-β1 activity might serve as a promising and innovative therapeutic strategy in CNS demyelinating diseases.
Collapse
Affiliation(s)
- Yi Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Xuejiao Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyue Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Shuai Liu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medicine College, Huazhong University of Science and Technology, Wuhan, China
| | - Simiao Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Medical College, Zhejiang University, Hangzhou, China
| | - Zhiyuan Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Neurological Diseases of the Chinese Ministry of Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
29
|
Liu X, Cai YD, Chiu JC. Regulation of protein O-GlcNAcylation by circadian, metabolic, and cellular signals. J Biol Chem 2024; 300:105616. [PMID: 38159854 PMCID: PMC10810748 DOI: 10.1016/j.jbc.2023.105616] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024] Open
Abstract
O-linked β-N-acetylglucosamine (O-GlcNAcylation) is a dynamic post-translational modification that regulates thousands of proteins and almost all cellular processes. Aberrant O-GlcNAcylation has been associated with numerous diseases, including cancer, neurodegenerative diseases, cardiovascular diseases, and type 2 diabetes. O-GlcNAcylation is highly nutrient-sensitive since it is dependent on UDP-GlcNAc, the end product of the hexosamine biosynthetic pathway (HBP). We previously observed daily rhythmicity of protein O-GlcNAcylation in a Drosophila model that is sensitive to the timing of food consumption. We showed that the circadian clock is pivotal in regulating daily O-GlcNAcylation rhythms given its control of the feeding-fasting cycle and hence nutrient availability. Interestingly, we reported that the circadian clock also modulates daily O-GlcNAcylation rhythm by regulating molecular mechanisms beyond the regulation of food consumption time. A large body of work now indicates that O-GlcNAcylation is likely a generalized cellular status effector as it responds to various cellular signals and conditions, such as ER stress, apoptosis, and infection. In this review, we summarize the metabolic regulation of protein O-GlcNAcylation through nutrient availability, HBP enzymes, and O-GlcNAc processing enzymes. We discuss the emerging roles of circadian clocks in regulating daily O-GlcNAcylation rhythm. Finally, we provide an overview of other cellular signals or conditions that impact O-GlcNAcylation. Many of these cellular pathways are themselves regulated by the clock and/or metabolism. Our review highlights the importance of maintaining optimal O-GlcNAc rhythm by restricting eating activity to the active period under physiological conditions and provides insights into potential therapeutic targets of O-GlcNAc homeostasis under pathological conditions.
Collapse
Affiliation(s)
- Xianhui Liu
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California, Davis, California, USA
| | - Yao D Cai
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California, Davis, California, USA
| | - Joanna C Chiu
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California, Davis, California, USA.
| |
Collapse
|
30
|
Kent SA, Miron VE. Microglia regulation of central nervous system myelin health and regeneration. Nat Rev Immunol 2024; 24:49-63. [PMID: 37452201 DOI: 10.1038/s41577-023-00907-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2023] [Indexed: 07/18/2023]
Abstract
Microglia are resident macrophages of the central nervous system that have key functions in its development, homeostasis and response to damage and infection. Although microglia have been increasingly implicated in contributing to the pathology that underpins neurological dysfunction and disease, they also have crucial roles in neurological homeostasis and regeneration. This includes regulation of the maintenance and regeneration of myelin, the membrane that surrounds neuronal axons, which is required for axonal health and function. Myelin is damaged with normal ageing and in several neurodegenerative diseases, such as multiple sclerosis and Alzheimer disease. Given the lack of approved therapies targeting myelin maintenance or regeneration, it is imperative to understand the mechanisms by which microglia support and restore myelin health to identify potential therapeutic approaches. However, the mechanisms by which microglia regulate myelin loss or integrity are still being uncovered. In this Review, we discuss recent work that reveals the changes in white matter with ageing and neurodegenerative disease, how this relates to microglia dynamics during myelin damage and regeneration, and factors that influence the regenerative functions of microglia.
Collapse
Affiliation(s)
- Sarah A Kent
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Veronique E Miron
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK.
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK.
- Barlo Multiple Sclerosis Centre, St Michael's Hospital, Toronto, Ontario, Canada.
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, Toronto, Ontario, Canada.
- Department of Immunology, The University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
31
|
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
|
32
|
Zhang R, Dong Y, Liu Y, Moezzi D, Ghorbani S, Mirzaei R, Lozinski BM, Dunn JF, Yong VW, Xue M. Enhanced liver X receptor signalling reduces brain injury and promotes tissue regeneration following experimental intracerebral haemorrhage: roles of microglia/macrophages. Stroke Vasc Neurol 2023; 8:486-502. [PMID: 37137522 PMCID: PMC10800269 DOI: 10.1136/svn-2023-002331] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/20/2023] [Indexed: 05/05/2023] Open
Abstract
BACKGROUND Inflammation-exacerbated secondary brain injury and limited tissue regeneration are barriers to favourable prognosis after intracerebral haemorrhage (ICH). As a regulator of inflammation and lipid metabolism, Liver X receptor (LXR) has the potential to alter microglia/macrophage (M/M) phenotype, and assist tissue repair by promoting cholesterol efflux and recycling from phagocytes. To support potential clinical translation, the benefits of enhanced LXR signalling are examined in experimental ICH. METHODS Collagenase-induced ICH mice were treated with the LXR agonist GW3965 or vehicle. Behavioural tests were conducted at multiple time points. Lesion and haematoma volume, and other brain parameters were assessed using multimodal MRI with T2-weighted, diffusion tensor imaging and dynamic contrast-enhanced MRI sequences. The fixed brain cryosections were stained and confocal microscopy was applied to detect LXR downstream genes, M/M phenotype, lipid/cholesterol-laden phagocytes, oligodendrocyte lineage cells and neural stem cells. Western blot and real-time qPCR were also used. CX3CR1CreER: Rosa26iDTR mice were employed for M/M-depletion experiments. RESULTS GW3965 treatment reduced lesion volume and white matter injury, and promoted haematoma clearance. Treated mice upregulated LXR downstream genes including ABCA1 and Apolipoprotein E, and had reduced density of M/M that apparently shifted from proinflammatory interleukin-1β+ to Arginase1+CD206+ regulatory phenotype. Fewer cholesterol crystal or myelin debris-laden phagocytes were observed in GW3965 mice. LXR activation increased the number of Olig2+PDGFRα+ precursors and Olig2+CC1+ mature oligodendrocytes in perihaematomal regions, and elevated SOX2+ or nestin+ neural stem cells in lesion and subventricular zone. MRI results supported better lesion recovery by GW3965, and this was corroborated by return to pre-ICH values of functional rotarod activity. The therapeutic effects of GW3965 were abrogated by M/M depletion in CX3CR1CreER: Rosa26iDTR mice. CONCLUSIONS LXR agonism using GW3965 reduced brain injury, promoted beneficial properties of M/M and facilitated tissue repair correspondent with enhanced cholesterol recycling.
Collapse
Affiliation(s)
- Ruiyi Zhang
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Yifei Dong
- Department of Biochemistry, Microbiology, & Immunology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Yang Liu
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
| | - Dorsa Moezzi
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Samira Ghorbani
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Reza Mirzaei
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Brian M Lozinski
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Jeff F Dunn
- Department of Radiology, University of Calgary, Calgary, Alberta, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
33
|
Schlepckow K, Morenas-Rodríguez E, Hong S, Haass C. Stimulation of TREM2 with agonistic antibodies-an emerging therapeutic option for Alzheimer's disease. Lancet Neurol 2023; 22:1048-1060. [PMID: 37863592 DOI: 10.1016/s1474-4422(23)00247-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 06/29/2023] [Accepted: 06/29/2023] [Indexed: 10/22/2023]
Abstract
Neurodegenerative disorders, including Alzheimer's disease, are associated with microgliosis. Microglia have long been considered to have detrimental roles in Alzheimer's disease. However, functional analyses of genes encoding risk factors that are linked to late-onset Alzheimer's disease, and that are enriched or exclusively expressed in microglia, have revealed unexpected protective functions. One of the major risk genes for Alzheimer's disease is TREM2. Risk variants of TREM2 are loss-of-function mutations affecting chemotaxis, phagocytosis, lipid and energy metabolism, and survival and proliferation. Agonistic anti-TREM2 antibodies have been developed to boost these protective functions in patients with intact TREM2 alleles. Several anti-TREM2 antibodies are in early clinical trials, and current efforts aim to achieve more efficient transport of these antibodies across the blood-brain barrier. PET imaging could be used to monitor target engagement. Data from animal models, and biomarker studies in patients, further support a rationale for boosting TREM2 functions during the preclinical stage of Alzheimer's disease.
Collapse
Affiliation(s)
- Kai Schlepckow
- German Centre for Neurodegenerative Diseases, Munich, Germany
| | - Estrella Morenas-Rodríguez
- Memory Unit, Department of Neurology, Hospital Universitario 12 de Octubre, Madrid, Spain; Group of Neurogenerative Diseases, Hospital Universitario 12 de Octubre Research Institute (imas12), Madrid, Spain
| | - Soyon Hong
- UK Dementia Research Institute, Institute of Neurology, University College London, London, UK
| | - Christian Haass
- German Centre for Neurodegenerative Diseases, Munich, Germany; Metabolic Biochemistry, Biomedical Centre, Faculty of Medicine, Ludwig-Maximilians University, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
34
|
Androvic P, Schifferer M, Perez Anderson K, Cantuti-Castelvetri L, Jiang H, Ji H, Liu L, Gouna G, Berghoff SA, Besson-Girard S, Knoferle J, Simons M, Gokce O. Spatial Transcriptomics-correlated Electron Microscopy maps transcriptional and ultrastructural responses to brain injury. Nat Commun 2023; 14:4115. [PMID: 37433806 DOI: 10.1038/s41467-023-39447-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 06/14/2023] [Indexed: 07/13/2023] Open
Abstract
Understanding the complexity of cellular function within a tissue necessitates the combination of multiple phenotypic readouts. Here, we developed a method that links spatially-resolved gene expression of single cells with their ultrastructural morphology by integrating multiplexed error-robust fluorescence in situ hybridization (MERFISH) and large area volume electron microscopy (EM) on adjacent tissue sections. Using this method, we characterized in situ ultrastructural and transcriptional responses of glial cells and infiltrating T-cells after demyelinating brain injury in male mice. We identified a population of lipid-loaded "foamy" microglia located in the center of remyelinating lesion, as well as rare interferon-responsive microglia, oligodendrocytes, and astrocytes that co-localized with T-cells. We validated our findings using immunocytochemistry and lipid staining-coupled single-cell RNA sequencing. Finally, by integrating these datasets, we detected correlations between full-transcriptome gene expression and ultrastructural features of microglia. Our results offer an integrative view of the spatial, ultrastructural, and transcriptional reorganization of single cells after demyelinating brain injury.
Collapse
Affiliation(s)
- Peter Androvic
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Martina Schifferer
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Katrin Perez Anderson
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Ludovico Cantuti-Castelvetri
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Hanyi Jiang
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Hao Ji
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Lu Liu
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Garyfallia Gouna
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Stefan A Berghoff
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Simon Besson-Girard
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Johanna Knoferle
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany
| | - Mikael Simons
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Ozgun Gokce
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany.
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany.
| |
Collapse
|
35
|
Schäffner E, Bosch-Queralt M, Edgar JM, Lehning M, Strauß J, Fleischer N, Kungl T, Wieghofer P, Berghoff SA, Reinert T, Krueger M, Morawski M, Möbius W, Barrantes-Freer A, Stieler J, Sun T, Saher G, Schwab MH, Wrede C, Frosch M, Prinz M, Reich DS, Flügel A, Stadelmann C, Fledrich R, Nave KA, Stassart RM. Myelin insulation as a risk factor for axonal degeneration in autoimmune demyelinating disease. Nat Neurosci 2023; 26:1218-1228. [PMID: 37386131 PMCID: PMC10322724 DOI: 10.1038/s41593-023-01366-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 05/17/2023] [Indexed: 07/01/2023]
Abstract
Axonal degeneration determines the clinical outcome of multiple sclerosis and is thought to result from exposure of denuded axons to immune-mediated damage. Therefore, myelin is widely considered to be a protective structure for axons in multiple sclerosis. Myelinated axons also depend on oligodendrocytes, which provide metabolic and structural support to the axonal compartment. Given that axonal pathology in multiple sclerosis is already visible at early disease stages, before overt demyelination, we reasoned that autoimmune inflammation may disrupt oligodendroglial support mechanisms and hence primarily affect axons insulated by myelin. Here, we studied axonal pathology as a function of myelination in human multiple sclerosis and mouse models of autoimmune encephalomyelitis with genetically altered myelination. We demonstrate that myelin ensheathment itself becomes detrimental for axonal survival and increases the risk of axons degenerating in an autoimmune environment. This challenges the view of myelin as a solely protective structure and suggests that axonal dependence on oligodendroglial support can become fatal when myelin is under inflammatory attack.
Collapse
Affiliation(s)
- Erik Schäffner
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Paul Flechsig Institute of Neuropathology, University Clinic Leipzig, Leipzig, Germany
| | - Mar Bosch-Queralt
- Paul Flechsig Institute of Neuropathology, University Clinic Leipzig, Leipzig, Germany
| | - Julia M Edgar
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Maria Lehning
- Paul Flechsig Institute of Neuropathology, University Clinic Leipzig, Leipzig, Germany
| | - Judith Strauß
- Institute of Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Niko Fleischer
- Paul Flechsig Institute of Neuropathology, University Clinic Leipzig, Leipzig, Germany
| | - Theresa Kungl
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | - Peter Wieghofer
- Institute of Anatomy, Leipzig University, Leipzig, Germany
- Cellular Neuroanatomy, Institute of Theoretical Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Stefan A Berghoff
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Tilo Reinert
- Paul Flechsig Institute of Neuropathology, University Clinic Leipzig, Leipzig, Germany
- Department of Neurophysics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Martin Krueger
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | - Markus Morawski
- Paul Flechsig Institute of Brain Research, Leipzig University, Leipzig, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | | | - Jens Stieler
- Paul Flechsig Institute of Brain Research, Leipzig University, Leipzig, Germany
| | - Ting Sun
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Gesine Saher
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Markus H Schwab
- Paul Flechsig Institute of Neuropathology, University Clinic Leipzig, Leipzig, Germany
| | - Christoph Wrede
- Institute of Functional and Applied Anatomy, Research Core Unit Electron Microscopy, Hannover Medical School, Hannover, Germany
| | - Maximilian Frosch
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
- Centre for NeuroModulation (NeuroModBasics), University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Alexander Flügel
- Institute of Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Christine Stadelmann
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Robert Fledrich
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
- Institute of Anatomy, Leipzig University, Leipzig, Germany.
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
| | - Ruth M Stassart
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
- Paul Flechsig Institute of Neuropathology, University Clinic Leipzig, Leipzig, Germany.
| |
Collapse
|
36
|
Distéfano-Gagné F, Bitarafan S, Lacroix S, Gosselin D. Roles and regulation of microglia activity in multiple sclerosis: insights from animal models. Nat Rev Neurosci 2023:10.1038/s41583-023-00709-6. [PMID: 37268822 DOI: 10.1038/s41583-023-00709-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2023] [Indexed: 06/04/2023]
Abstract
As resident macrophages of the CNS, microglia are critical immune effectors of inflammatory lesions and associated neural dysfunctions. In multiple sclerosis (MS) and its animal models, chronic microglial inflammatory activity damages myelin and disrupts axonal and synaptic activity. In contrast to these detrimental effects, the potent phagocytic and tissue-remodelling capabilities of microglia support critical endogenous repair mechanisms. Although these opposing capabilities have long been appreciated, a precise understanding of their underlying molecular effectors is only beginning to emerge. Here, we review recent advances in our understanding of the roles of microglia in animal models of MS and demyelinating lesions and the mechanisms that underlie their damaging and repairing activities. We also discuss how the structured organization and regulation of the genome enables complex transcriptional heterogeneity within the microglial cell population at demyelinating lesions.
Collapse
Affiliation(s)
- Félix Distéfano-Gagné
- Axe Neuroscience, Centre de Recherche du CHU de Québec - Université Laval, Québec, Québec, Canada
- Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Québec, Québec, Canada
| | - Sara Bitarafan
- Axe Neuroscience, Centre de Recherche du CHU de Québec - Université Laval, Québec, Québec, Canada
- Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Québec, Québec, Canada
| | - Steve Lacroix
- Axe Neuroscience, Centre de Recherche du CHU de Québec - Université Laval, Québec, Québec, Canada
- Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Québec, Québec, Canada
| | - David Gosselin
- Axe Neuroscience, Centre de Recherche du CHU de Québec - Université Laval, Québec, Québec, Canada.
- Département de Médecine Moléculaire de la Faculté de Médecine, Université Laval, Québec, Québec, Canada.
| |
Collapse
|
37
|
Qin C, Yang S, Chen M, Dong MH, Zhou LQ, Chu YH, Shen ZX, Bosco DB, Wu LJ, Tian DS, Wang W. Modulation of microglial metabolism facilitates regeneration in demyelination. iScience 2023; 26:106588. [PMID: 37138776 PMCID: PMC10149336 DOI: 10.1016/j.isci.2023.106588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/28/2023] [Accepted: 03/30/2023] [Indexed: 05/05/2023] Open
Abstract
Microglia exhibit diverse phenotypes in various central nervous system disorders and metabolic pathways exert crucial effects on microglial activation and effector functions. Here, we discovered two novel distinct microglial clusters, functionally associated with enhanced phagocytosis (PEMs) and myelination (MAMs) respectively, in human patients with multiple sclerosis by integrating public snRNA-seq data. Microglia adopt a PEMs phenotype during the early phase of demyelinated lesions, predominated in pro-inflammatory responses and aggravated glycolysis, while MAMs mainly emerged during the later phase, with regenerative signatures and enhanced oxidative phosphorylation. In addition, microglial triggering receptor expressed on myeloid cells 2 (Trem2) was greatly involved in the phenotype transition in demyelination, but not indispensable for microglia transition toward PEMs. Rosiglitazone could promote microglial phenotype conversion from PEMs to MAMs, thus favoring myelin repair. Taken together, these findings provide insights into therapeutic interventions targeting immunometabolism to switch microglial phenotypes and facilitate regenerative capacity in demyelination.
Collapse
Affiliation(s)
- Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Sheng Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Man Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ming-Hao Dong
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Luo-Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yun-Hui Chu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhu-Xia Shen
- Department of Cardiology, Jing’an District Centre Hospital of Shanghai, Fudan University, Shanghai 200040, China
| | - Dale B. Bosco
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Corresponding author
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Corresponding author
| |
Collapse
|
38
|
Esmaeilzadeh A, Mohammadi V, Elahi R. Transforming growth factor β (TGF-β) pathway in the immunopathogenesis of multiple sclerosis (MS); molecular approaches. Mol Biol Rep 2023:10.1007/s11033-023-08419-z. [PMID: 37204543 DOI: 10.1007/s11033-023-08419-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/30/2023] [Indexed: 05/20/2023]
Abstract
INTRODUCTION Multiple sclerosis (MS) is an acute demyelinating disease with an autoimmune nature, followed by gradual neurodegeneration and enervating scar formation. Dysregulated immune response is a crucial dilemma contributing to the pathogenesis of MS. The role of chemokines and cytokines, such as transforming growth factor-β (TGF-β), have been recently highlighted regarding their altered expressions in MS. TGF-β has three isoforms, TGF-β1, TGF-β2, and TGF-β3, that are structurally similar; however, they can show different functions. RESULTS All three isoforms are known to induce immune tolerance by modifying Foxp3+ regulatory T cells. Nevertheless, there are controversial reports concerning the role of TGF-β1 and 2 in the progression of scar formation in MS. At the same time, these proteins also improve oligodendrocyte differentiation and have shown neuroprotective behavior, two cellular processes that suppress the pathogenesis of MS. TGF-β3 shares the same properties but is less likely contributes to scar formation, and its direct role in MS remains elusive. DISCUSSION To develop novel neuroimmunological treatment strategies for MS, the optimal strategy could be the one that causes immune modulation, induces neurogenesis, stimulates remyelination, and prevents excessive scar formation. Therefore, regarding its immunological properties, TGF-β could be an appropriate candidate; however, contradictory results of previous studies have questioned its role and therapeutic potential in MS. In this review article, we provide an overview of the role of TGF-β in immunopathogenesis of MS, related clinical and animal studies, and the treatment potential of TGF-β in MS, emphasizing the role of different TGF-β isoforms.
Collapse
Affiliation(s)
- Abdolreza Esmaeilzadeh
- Department of Immunology, Zanjan University of Medical Sciences, Zanjan, Iran.
- Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Vahid Mohammadi
- School of Medicine, Zanjan University of medical sciences, Zanjan, Iran
| | - Reza Elahi
- School of Medicine, Zanjan University of medical sciences, Zanjan, Iran
| |
Collapse
|
39
|
Wang Y, Kyauk RV, Shen YAA, Xie L, Reichelt M, Lin H, Jiang Z, Ngu H, Shen K, Greene JJ, Sheng M, Yuen TJ. TREM2-dependent microglial function is essential for remyelination and subsequent neuroprotection. Glia 2023; 71:1247-1258. [PMID: 36625077 DOI: 10.1002/glia.24335] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 01/11/2023]
Abstract
Disability in multiple sclerosis (MS) is driven in part by the failure of remyelination and progressive neurodegeneration. Microglia, and specifically triggering receptor expressed on myeloid cells 2 (TREM2), a factor highly expressed in microglia, have been shown to play an important role in remyelination. Here, using a focal demyelination model in the brain, we demonstrate that demyelination is persistent in TREM2 knockout mice, lasting more than 6 weeks after lysolecithin injection and resulting in substantial neurodegeneration. We also find that TREM2 knockout mice exhibit an altered glial response following demyelination. TREM2 knockout microglia demonstrate defects in migration and phagocytosis of myelin debris. In addition, human monocyte-derived macrophages from subjects with a TREM2 mutation prevalent in human disease also show a defect in myelin debris phagocytosis. Together, we highlight the central role of TREM2 signaling in remyelination and neuroprotection. These findings provide insights into how chronic demyelination might lead to axonal damage and could help identify novel neuroprotective therapeutic targets for MS.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Department of Neuroscience, Genentech Inc., South San Francisco, California, USA
| | - Roxanne V Kyauk
- Department of Neuroscience, Genentech Inc., South San Francisco, California, USA
| | - Yun-An A Shen
- Department of Neuroscience, Genentech Inc., South San Francisco, California, USA
| | - Luke Xie
- Department of Biomedical Imaging, Genentech Inc., South San Francisco, California, USA
| | - Mike Reichelt
- Department of Pathology, Genentech Inc., South San Francisco, California, USA
| | - Han Lin
- Department of Neuroscience, Genentech Inc., South San Francisco, California, USA
| | - Zhiyu Jiang
- Department of Neuroscience, Genentech Inc., South San Francisco, California, USA
| | - Hai Ngu
- Department of Pathology, Genentech Inc., South San Francisco, California, USA
| | - Kimberle Shen
- Department of Neuroscience, Genentech Inc., South San Francisco, California, USA
| | - Jacob J Greene
- Department of Neuroscience, Genentech Inc., South San Francisco, California, USA
| | - Morgan Sheng
- Department of Neuroscience, Genentech Inc., South San Francisco, California, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Tracy J Yuen
- Department of Neuroscience, Genentech Inc., South San Francisco, California, USA
| |
Collapse
|
40
|
van Lengerich B, Zhan L, Xia D, Chan D, Joy D, Park JI, Tatarakis D, Calvert M, Hummel S, Lianoglou S, Pizzo ME, Prorok R, Thomsen E, Bartos LM, Beumers P, Capell A, Davis SS, de Weerd L, Dugas JC, Duque J, Earr T, Gadkar K, Giese T, Gill A, Gnörich J, Ha C, Kannuswamy M, Kim DJ, Kunte ST, Kunze LH, Lac D, Lechtenberg K, Leung AWS, Liang CC, Lopez I, McQuade P, Modi A, Torres VO, Nguyen HN, Pesämaa I, Propson N, Reich M, Robles-Colmenares Y, Schlepckow K, Slemann L, Solanoy H, Suh JH, Thorne RG, Vieira C, Wind-Mark K, Xiong K, Zuchero YJY, Diaz D, Dennis MS, Huang F, Scearce-Levie K, Watts RJ, Haass C, Lewcock JW, Di Paolo G, Brendel M, Sanchez PE, Monroe KM. A TREM2-activating antibody with a blood-brain barrier transport vehicle enhances microglial metabolism in Alzheimer's disease models. Nat Neurosci 2023; 26:416-429. [PMID: 36635496 PMCID: PMC9991924 DOI: 10.1038/s41593-022-01240-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 11/29/2022] [Indexed: 01/13/2023]
Abstract
Loss-of-function variants of TREM2 are associated with increased risk of Alzheimer's disease (AD), suggesting that activation of this innate immune receptor may be a useful therapeutic strategy. Here we describe a high-affinity human TREM2-activating antibody engineered with a monovalent transferrin receptor (TfR) binding site, termed antibody transport vehicle (ATV), to facilitate blood-brain barrier transcytosis. Upon peripheral delivery in mice, ATV:TREM2 showed improved brain biodistribution and enhanced signaling compared to a standard anti-TREM2 antibody. In human induced pluripotent stem cell (iPSC)-derived microglia, ATV:TREM2 induced proliferation and improved mitochondrial metabolism. Single-cell RNA sequencing and morphometry revealed that ATV:TREM2 shifted microglia to metabolically responsive states, which were distinct from those induced by amyloid pathology. In an AD mouse model, ATV:TREM2 boosted brain microglial activity and glucose metabolism. Thus, ATV:TREM2 represents a promising approach to improve microglial function and treat brain hypometabolism found in patients with AD.
Collapse
Affiliation(s)
| | - Lihong Zhan
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | - Dan Xia
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | - Darren Chan
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | - David Joy
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | - Joshua I Park
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | | | | | - Selina Hummel
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilians University, Munich, Germany
| | | | | | - Rachel Prorok
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | | | - Laura M Bartos
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilians University, Munich, Germany
| | - Philipp Beumers
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilians University, Munich, Germany
| | - Anja Capell
- Biomedical Center (BMC), Division of Metabolic Biochemistry, Faculty of Medicine, Ludwig Maximilians University, Munich, Germany
| | | | - Lis de Weerd
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Jason C Dugas
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | - Joseph Duque
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | - Timothy Earr
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | - Kapil Gadkar
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | - Tina Giese
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | - Audrey Gill
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | - Johannes Gnörich
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilians University, Munich, Germany
| | - Connie Ha
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | | | - Do Jin Kim
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | - Sebastian T Kunte
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilians University, Munich, Germany
| | - Lea H Kunze
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilians University, Munich, Germany
| | - Diana Lac
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | | | | | | | - Isabel Lopez
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | - Paul McQuade
- Takeda Pharmaceutical Company, Cambridge, MA, USA
| | - Anuja Modi
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | | | | | - Ida Pesämaa
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Graduate School of Systemic Neurosciences, Ludwig Maximilians University Munich, Planegg-Martinsried, Germany
| | | | - Marvin Reich
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Graduate School of Systemic Neurosciences, Ludwig Maximilians University Munich, Planegg-Martinsried, Germany
| | | | - Kai Schlepckow
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Luna Slemann
- Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilians University, Munich, Germany
| | - Hilda Solanoy
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | - Jung H Suh
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | | | | | - Karin Wind-Mark
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilians University, Munich, Germany
| | - Ken Xiong
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | | | - Dolo Diaz
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | - Mark S Dennis
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | - Fen Huang
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | | | - Ryan J Watts
- Denali Therapeutics, Inc., South San Francisco, CA, USA
| | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Biomedical Center (BMC), Division of Metabolic Biochemistry, Faculty of Medicine, Ludwig Maximilians University, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | | | | | - Matthias Brendel
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.,Department of Nuclear Medicine, University Hospital of Munich, Ludwig Maximilians University, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | | | | |
Collapse
|
41
|
Attfield KE, Jensen LT, Kaufmann M, Friese MA, Fugger L. The immunology of multiple sclerosis. Nat Rev Immunol 2022; 22:734-750. [PMID: 35508809 DOI: 10.1038/s41577-022-00718-z] [Citation(s) in RCA: 172] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2022] [Indexed: 12/11/2022]
Abstract
Our incomplete understanding of the causes and pathways involved in the onset and progression of multiple sclerosis (MS) limits our ability to effectively treat this complex neurological disease. Recent studies explore the role of immune cells at different stages of MS and how they interact with cells of the central nervous system (CNS). The findings presented here begin to question the exclusivity of an antigen-specific cause and highlight how seemingly distinct immune cell types can share common functions that drive disease. Innovative techniques further expose new disease-associated immune cell populations and reinforce how environmental context is critical to their phenotype and subsequent role in disease. Importantly, the differentiation of immune cells into a pathogenic state is potentially reversible through therapeutic manipulation. As such, understanding the mechanisms that provide plasticity to causal cell types is likely key to uncoupling these disease processes and may identify novel therapeutic targets that replace the need for cell ablation.
Collapse
Affiliation(s)
- Kathrine E Attfield
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, Oxford University Hospitals, University of Oxford, Oxford, UK
| | - Lise Torp Jensen
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Max Kaufmann
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel A Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Fugger
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, Oxford University Hospitals, University of Oxford, Oxford, UK.
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
42
|
Wang N, Wang M, Jeevaratnam S, Rosenberg C, Ikezu TC, Shue F, Doss SV, Alnobani A, Martens YA, Wren M, Asmann YW, Zhang B, Bu G, Liu CC. Opposing effects of apoE2 and apoE4 on microglial activation and lipid metabolism in response to demyelination. Mol Neurodegener 2022; 17:75. [PMID: 36419137 PMCID: PMC9682675 DOI: 10.1186/s13024-022-00577-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 10/21/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Abnormal lipid accumulation has been recognized as a key element of immune dysregulation in microglia whose dysfunction contributes to neurodegenerative diseases. Microglia play essential roles in the clearance of lipid-rich cellular debris upon myelin damage or demyelination, a common pathogenic event in neuronal disorders. Apolipoprotein E (apoE) plays a pivotal role in brain lipid homeostasis; however, the apoE isoform-dependent mechanisms regulating microglial response upon demyelination remain unclear. METHODS To determine how apoE isoforms impact microglial response to myelin damage, 2-month-old apoE2-, apoE3-, and apoE4-targeted replacement (TR) mice were fed with normal diet (CTL) or 0.2% cuprizone (CPZ) diet for four weeks to induce demyelination in the brain. To examine the effects on subsequent remyelination, the cuprizone diet was switched back to regular chow for an additional two weeks. After treatment, brains were collected and subjected to immunohistochemical and biochemical analyses to assess the myelination status, microglial responses, and their capacity for myelin debris clearance. Bulk RNA sequencing was performed on the corpus callosum (CC) to address the molecular mechanisms underpinning apoE-mediated microglial activation upon demyelination. RESULTS We demonstrate dramatic isoform-dependent differences in the activation and function of microglia upon cuprizone-induced demyelination. ApoE2 microglia were hyperactive and more efficient in clearing lipid-rich myelin debris, whereas apoE4 microglia displayed a less activated phenotype with reduced clearance efficiency, compared with apoE3 microglia. Transcriptomic profiling revealed that key molecules known to modulate microglial functions had differential expression patterns in an apoE isoform-dependent manner. Importantly, apoE4 microglia had excessive buildup of lipid droplets, consistent with an impairment in lipid metabolism, whereas apoE2 microglia displayed a superior ability to metabolize myelin enriched lipids. Further, apoE2-TR mice had a greater extent of remyelination; whereas remyelination was compromised in apoE4-TR mice. CONCLUSIONS Our findings provide critical mechanistic insights into how apoE isoforms differentially regulate microglial function and the maintenance of myelin dynamics, which may inform novel therapeutic avenues for targeting microglial dysfunctions in neurodegenerative diseases.
Collapse
Affiliation(s)
- Na Wang
- grid.417467.70000 0004 0443 9942Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Minghui Wang
- grid.59734.3c0000 0001 0670 2351Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Suren Jeevaratnam
- grid.417467.70000 0004 0443 9942Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Cassandra Rosenberg
- grid.417467.70000 0004 0443 9942Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Tadafumi C. Ikezu
- grid.417467.70000 0004 0443 9942Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Francis Shue
- grid.417467.70000 0004 0443 9942Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Sydney V. Doss
- grid.417467.70000 0004 0443 9942Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Alla Alnobani
- grid.417467.70000 0004 0443 9942Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Yuka A. Martens
- grid.417467.70000 0004 0443 9942Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Melissa Wren
- grid.417467.70000 0004 0443 9942Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Yan W. Asmann
- grid.417467.70000 0004 0443 9942Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL 32224 USA
| | - Bin Zhang
- grid.59734.3c0000 0001 0670 2351Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA.
| | - Chia-Chen Liu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA.
| |
Collapse
|
43
|
Franklin RJM, Simons M. CNS remyelination and inflammation: From basic mechanisms to therapeutic opportunities. Neuron 2022; 110:3549-3565. [PMID: 36228613 DOI: 10.1016/j.neuron.2022.09.023] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/09/2022] [Accepted: 09/19/2022] [Indexed: 11/06/2022]
Abstract
Remyelination, the myelin regenerative response that follows demyelination, restores saltatory conduction and function and sustains axon health. Its declining efficiency with disease progression in the chronic autoimmune disease multiple sclerosis (MS) contributes to the currently untreatable progressive phase of the disease. Although some of the bona fide myelin regenerative medicine clinical trials have succeeded in demonstrating proof-of-principle, none of these compounds have yet proceeded toward approval. There therefore remains a need to increase our understanding of the fundamental biology of remyelination so that existing targets can be refined and new ones discovered. Here, we review the role of inflammation, in particular innate immunity, in remyelination, describing its many and complex facets and discussing how our evolving understanding can be harnessed to translational goals.
Collapse
Affiliation(s)
- Robin J M Franklin
- Altos Labs - Cambridge Institute of Science, Granta Park, Cambridge CB21 6GP, UK.
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases, Munich, Germany; Cluster of Systems Neurology (SyNergy), Munich, Germany; Institute for Stroke and Dementia Research, Munich, Germany.
| |
Collapse
|
44
|
Düking T, Spieth L, Berghoff SA, Piepkorn L, Schmidke AM, Mitkovski M, Kannaiyan N, Hosang L, Scholz P, Shaib AH, Schneider LV, Hesse D, Ruhwedel T, Sun T, Linhoff L, Trevisiol A, Köhler S, Pastor AM, Misgeld T, Sereda M, Hassouna I, Rossner MJ, Odoardi F, Ischebeck T, de Hoz L, Hirrlinger J, Jahn O, Saher G. Ketogenic diet uncovers differential metabolic plasticity of brain cells. SCIENCE ADVANCES 2022; 8:eabo7639. [PMID: 36112685 PMCID: PMC9481126 DOI: 10.1126/sciadv.abo7639] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 08/01/2022] [Indexed: 06/15/2023]
Abstract
To maintain homeostasis, the body, including the brain, reprograms its metabolism in response to altered nutrition or disease. However, the consequences of these challenges for the energy metabolism of the different brain cell types remain unknown. Here, we generated a proteome atlas of the major central nervous system (CNS) cell types from young and adult mice, after feeding the therapeutically relevant low-carbohydrate, high-fat ketogenic diet (KD) and during neuroinflammation. Under steady-state conditions, CNS cell types prefer distinct modes of energy metabolism. Unexpectedly, the comparison with KD revealed distinct cell type-specific strategies to manage the altered availability of energy metabolites. Astrocytes and neurons but not oligodendrocytes demonstrated metabolic plasticity. Moreover, inflammatory demyelinating disease changed the neuronal metabolic signature in a similar direction as KD. Together, these findings highlight the importance of the metabolic cross-talk between CNS cells and between the periphery and the brain to manage altered nutrition and neurological disease.
Collapse
Affiliation(s)
- Tim Düking
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Lena Spieth
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Stefan A. Berghoff
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Lars Piepkorn
- Neuroproteomics Group, Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Translational Neuroproteomics Group, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Annika M. Schmidke
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Miso Mitkovski
- City Campus Light Microscopy Facility, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Nirmal Kannaiyan
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Leon Hosang
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Patricia Scholz
- Department of Plant Biochemistry, Albrecht-von-Haller-Institute for Plant Sciences and Göttingen Center for Molecular Biosciences (GZMB), University of Göttingen, Göttingen, Germany
| | - Ali H. Shaib
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Institute for Neuro- and Sensory Physiology, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Göttingen, Göttingen, Germany
| | - Lennart V. Schneider
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Dörte Hesse
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Neuroproteomics Group, Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Torben Ruhwedel
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Electron Microscopy Core Unit, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Ting Sun
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Lisa Linhoff
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
- Translational Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Andrea Trevisiol
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Susanne Köhler
- Carl-Ludwig-Institute for Physiology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Adrian Marti Pastor
- Institute of Neuronal Cell Biology, Technische Universität München, Cluster for Systems Neurology (SyNergy), German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technische Universität München, Cluster for Systems Neurology (SyNergy), German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Michael Sereda
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
- Translational Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Imam Hassouna
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Moritz J. Rossner
- Department of Psychiatry and Psychotherapy, University Hospital, LMU Munich, Munich, Germany
| | - Francesca Odoardi
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Center Göttingen, Göttingen, Germany
| | - Till Ischebeck
- Department of Plant Biochemistry, Albrecht-von-Haller-Institute for Plant Sciences and Göttingen Center for Molecular Biosciences (GZMB), University of Göttingen, Göttingen, Germany
- Institute of Plant Biology and Biotechnology (IBBP), Green Biotechnology, University of Münster, Münster, Germany
| | - Livia de Hoz
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Neurowissenschafliches Forschungszentrum, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Johannes Hirrlinger
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Carl-Ludwig-Institute for Physiology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Olaf Jahn
- Neuroproteomics Group, Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Translational Neuroproteomics Group, Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Gesine Saher
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| |
Collapse
|
45
|
Raffaele S, Fumagalli M. Dynamics of Microglia Activation in the Ischemic Brain: Implications for Myelin Repair and Functional Recovery. Front Cell Neurosci 2022; 16:950819. [PMID: 35899017 PMCID: PMC9309466 DOI: 10.3389/fncel.2022.950819] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Ischemic stroke is a neurological disorder representing a leading cause of death and permanent disability world-wide, for which effective regenerative treatments are missing. Oligodendrocyte degeneration and consequent myelin disruption are considered major contributing factors to stroke-associated neurological deficits. Therefore, fostering myelin reconstruction by oligodendrocyte precursor cells (OPCs) has emerged as a promising therapeutic approach to enhance functional recovery in stroke patients. A pivotal role in regulating remyelination is played by microglia, the resident immune cells of the brain. Early after stroke, microglial cells exert beneficial functions, promoting OPC recruitment toward the ischemic lesion and preserving myelin integrity. However, the protective features of microglia are lost during disease progression, contributing to remyelination failure. Unveiling the mechanisms driving the pro-remyelination properties of microglia may provide important opportunities for both reducing myelin damage and promoting its regeneration. Here, we summarize recent evidence describing microglia activation kinetics in experimental models of ischemic injury, focusing on the contribution of these innate immune cells to myelin damage and repair. Some molecular signals regulating the pro-regenerative functions of microglia after stroke have been highlighted to provide new possible therapeutic targets involved in the protective functions of these cells. Finally, we analyzed the impact of microglia-to-OPCs communication via extracellular vesicles on post-stroke remyelination and functional recovery. The results collected in this review underline the importance of supporting the pro-remyelination functions of microglial cells after stroke.
Collapse
|
46
|
Dong Y, Yong VW. Oxidized phospholipids as novel mediators of neurodegeneration. Trends Neurosci 2022; 45:419-429. [PMID: 35393134 DOI: 10.1016/j.tins.2022.03.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/19/2022] [Accepted: 03/08/2022] [Indexed: 10/18/2022]
Abstract
Neurodegeneration drives the progression of many neurological diseases. Inflammation and oxidative stress occurring in the CNS promote lipid peroxidation, leading to the generation of oxidized phospholipids such as oxidized phosphatidylcholines (OxPCs). OxPCs have been proposed as biomarkers of oxidative stress, where their detection in lesions in multiple sclerosis (MS), frontotemporal lobe dementia, spinal cord injury, and amyotrophic lateral sclerosis (ALS) implies that oxidative insult had occurred. However, recent findings highlight OxPCs as potent neurotoxic species requiring neutralization by microglia. Here, we summarize the science of OxPCs, including lessons from non-CNS diseases. We discuss the potential of OxPCs as common drivers of injury across neurological conditions and encourage investigations of OxPCs as novel neurotoxins.
Collapse
Affiliation(s)
- Yifei Dong
- Hotchkiss Brain Institute and the Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and the Department of Clinical Neuroscience, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
47
|
Bosch-Queralt M, Tiwari V, Damkou A, Vaculčiaková L, Alexopoulos I, Simons M. A fluorescence microscopy-based protocol for volumetric measurement of lysolecithin lesion-associated de- and re-myelination in mouse brain. STAR Protoc 2022; 3:101141. [PMID: 35141565 PMCID: PMC8810560 DOI: 10.1016/j.xpro.2022.101141] [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] [Indexed: 11/28/2022] Open
Abstract
Lysolecithin injections into the white matter tracts of the central nervous system are a valuable tool to study remyelination, but evaluating the resulting demyelinating lesion size is challenging. Here, we present a protocol to consistently measure the volume of demyelination and remyelination in mice following brain lysolecithin injections. We describe serial sectioning of the lesion, followed by the evaluation of the demyelinated area in two-dimensional images. We then detail the computation of the volume using our own automated iPython script. For complete details on the use and execution of this profile, please refer to Bosch-Queralt et al. (2021).
Collapse
Affiliation(s)
- Mar Bosch-Queralt
- Institute of Neuronal Cell Biology, Technical University Munich, 80802 Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Vini Tiwari
- Institute of Neuronal Cell Biology, Technical University Munich, 80802 Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Alkmini Damkou
- Institute of Neuronal Cell Biology, Technical University Munich, 80802 Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Lenka Vaculčiaková
- Department of Neurophysics, Max Planck Institute for Human Cognitive and Brain Sciences, Leipzig, Germany
| | - Ioannis Alexopoulos
- Institute of Neuronal Cell Biology, Technical University Munich, 80802 Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, 80802 Munich, Germany.,German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany.,Munich Cluster of Systems Neurology (SyNergy), 81377 Munich, Germany
| |
Collapse
|
48
|
Reifschneider A, Robinson S, van Lengerich B, Gnörich J, Logan T, Heindl S, Vogt MA, Weidinger E, Riedl L, Wind K, Zatcepin A, Pesämaa I, Haberl S, Nuscher B, Kleinberger G, Klimmt J, Götzl JK, Liesz A, Bürger K, Brendel M, Levin J, Diehl‐Schmid J, Suh J, Di Paolo G, Lewcock JW, Monroe KM, Paquet D, Capell A, Haass C. Loss of TREM2 rescues hyperactivation of microglia, but not lysosomal deficits and neurotoxicity in models of progranulin deficiency. EMBO J 2022; 41:e109108. [PMID: 35019161 PMCID: PMC8844989 DOI: 10.15252/embj.2021109108] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 12/13/2021] [Accepted: 12/16/2021] [Indexed: 12/13/2022] Open
Abstract
Haploinsufficiency of the progranulin (PGRN)-encoding gene (GRN) causes frontotemporal lobar degeneration (GRN-FTLD) and results in microglial hyperactivation, TREM2 activation, lysosomal dysfunction, and TDP-43 deposition. To understand the contribution of microglial hyperactivation to pathology, we used genetic and pharmacological approaches to suppress TREM2-dependent transition of microglia from a homeostatic to a disease-associated state. Trem2 deficiency in Grn KO mice reduced microglia hyperactivation. To explore antibody-mediated pharmacological modulation of TREM2-dependent microglial states, we identified antagonistic TREM2 antibodies. Treatment of macrophages from GRN-FTLD patients with these antibodies led to reduced TREM2 signaling due to its enhanced shedding. Furthermore, TREM2 antibody-treated PGRN-deficient microglia derived from human-induced pluripotent stem cells showed reduced microglial hyperactivation, TREM2 signaling, and phagocytic activity, but lysosomal dysfunction was not rescued. Similarly, lysosomal dysfunction, lipid dysregulation, and glucose hypometabolism of Grn KO mice were not rescued by TREM2 ablation. Synaptic loss and neurofilament light-chain (NfL) levels, a biomarker for neurodegeneration, were further elevated in the Grn/Trem2 KO cerebrospinal fluid (CSF). These findings suggest that TREM2-dependent microglia hyperactivation in models of GRN deficiency does not promote neurotoxicity, but rather neuroprotection.
Collapse
Affiliation(s)
- Anika Reifschneider
- Division of Metabolic BiochemistryFaculty of MedicineBiomedical Center (BMC)Ludwig‐Maximilians‐Universität MünchenMunichGermany
| | - Sophie Robinson
- Institute for Stroke and Dementia ResearchUniversity Hospital MunichLudwig‐Maximilians‐Universität MünchenMunichGermany
- German Center for Neurodegenerative Diseases (DZNE) MunichMunichGermany
| | | | - Johannes Gnörich
- German Center for Neurodegenerative Diseases (DZNE) MunichMunichGermany
- Department of Nuclear MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Todd Logan
- Denali Therapeutics Inc.South San FranciscoCAUSA
| | - Steffanie Heindl
- Institute for Stroke and Dementia ResearchUniversity Hospital MunichLudwig‐Maximilians‐Universität MünchenMunichGermany
| | | | - Endy Weidinger
- Department of NeurologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Lina Riedl
- Department of Psychiatry and PsychotherapySchool of MedicineTechnical University of MunichMunichGermany
| | - Karin Wind
- German Center for Neurodegenerative Diseases (DZNE) MunichMunichGermany
- Department of Nuclear MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Artem Zatcepin
- German Center for Neurodegenerative Diseases (DZNE) MunichMunichGermany
- Department of Nuclear MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Ida Pesämaa
- German Center for Neurodegenerative Diseases (DZNE) MunichMunichGermany
| | | | - Brigitte Nuscher
- Division of Metabolic BiochemistryFaculty of MedicineBiomedical Center (BMC)Ludwig‐Maximilians‐Universität MünchenMunichGermany
| | | | - Julien Klimmt
- Institute for Stroke and Dementia ResearchUniversity Hospital MunichLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Julia K Götzl
- Division of Metabolic BiochemistryFaculty of MedicineBiomedical Center (BMC)Ludwig‐Maximilians‐Universität MünchenMunichGermany
| | - Arthur Liesz
- Institute for Stroke and Dementia ResearchUniversity Hospital MunichLudwig‐Maximilians‐Universität MünchenMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Katharina Bürger
- Institute for Stroke and Dementia ResearchUniversity Hospital MunichLudwig‐Maximilians‐Universität MünchenMunichGermany
- German Center for Neurodegenerative Diseases (DZNE) MunichMunichGermany
| | - Matthias Brendel
- German Center for Neurodegenerative Diseases (DZNE) MunichMunichGermany
- Department of Nuclear MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE) MunichMunichGermany
- Department of Nuclear MedicineUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Department of NeurologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
| | - Janine Diehl‐Schmid
- Department of NeurologyUniversity HospitalLudwig‐Maximilians‐Universität MünchenMunichGermany
- Department of Psychiatry and PsychotherapySchool of MedicineTechnical University of MunichMunichGermany
| | - Jung Suh
- Denali Therapeutics Inc.South San FranciscoCAUSA
| | | | | | | | - Dominik Paquet
- Institute for Stroke and Dementia ResearchUniversity Hospital MunichLudwig‐Maximilians‐Universität MünchenMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Anja Capell
- Division of Metabolic BiochemistryFaculty of MedicineBiomedical Center (BMC)Ludwig‐Maximilians‐Universität MünchenMunichGermany
| | - Christian Haass
- Division of Metabolic BiochemistryFaculty of MedicineBiomedical Center (BMC)Ludwig‐Maximilians‐Universität MünchenMunichGermany
- German Center for Neurodegenerative Diseases (DZNE) MunichMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| |
Collapse
|
49
|
Vogel A, Brunner JS, Hajto A, Sharif O, Schabbauer G. Lipid scavenging macrophages and inflammation. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159066. [PMID: 34626791 DOI: 10.1016/j.bbalip.2021.159066] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 08/18/2021] [Indexed: 12/11/2022]
Abstract
Macrophages are professional phagocytes, indispensable for maintenance of tissue homeostasis and integrity. Depending on their resident tissue, macrophages are exposed to highly diverse metabolic environments. Adapted to their niche, they can contribute to local metabolic turnover through metabolite uptake, conversion, storage and release. Disturbances in tissue homeostasis caused by infection, inflammation or damage dramatically alter the local milieu, impacting macrophage activation status and metabolism. In the case of persisting stimuli, defective macrophage responses ensue, which can promote tissue damage and disease. Especially relevant herein are disbalances in lipid rich environments, where macrophages are crucially involved in lipid uptake and turnover, preventing lipotoxicity. Lipid uptake is to a large extent facilitated by macrophage expressed scavenger receptors that are dynamically regulated and important in many metabolic diseases. Here, we review the receptors mediating lipid uptake and summarize recent findings on their role in health and disease. We further highlight the underlying pathways driving macrophage lipid acquisition and their impact on myeloid metabolic remodelling.
Collapse
Affiliation(s)
- Andrea Vogel
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University Vienna, Vienna, Austria; Christian Doppler Laboratory for Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Julia Stefanie Brunner
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University Vienna, Vienna, Austria; Christian Doppler Laboratory for Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Alexander Hajto
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University Vienna, Vienna, Austria; Christian Doppler Laboratory for Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria
| | - Omar Sharif
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University Vienna, Vienna, Austria; Christian Doppler Laboratory for Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria.
| | - Gernot Schabbauer
- Institute for Vascular Biology, Centre for Physiology and Pharmacology, Medical University Vienna, Vienna, Austria; Christian Doppler Laboratory for Arginine Metabolism in Rheumatoid Arthritis and Multiple Sclerosis, Vienna, Austria.
| |
Collapse
|
50
|
Cantuti-Castelvetri L, Gokce O, Simons M. Reparative inflammation in multiple sclerosis. Semin Immunol 2022; 59:101630. [PMID: 35750551 DOI: 10.1016/j.smim.2022.101630] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 04/09/2022] [Accepted: 06/13/2022] [Indexed: 01/15/2023]
Affiliation(s)
- Ludovico Cantuti-Castelvetri
- Institute of Neuronal Cell Biology, Technical University Munich, 80802 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Ozgun Gokce
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, 80802 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), 81377 Munich, Germany.
| |
Collapse
|