1
|
Zhang W, Zhang X, Wang K, Liu Z, Zhang L, Liu S, He K, Wang H, Wang J, Wang Y, Wang Y, Yang Y, Wu H. Single-nucleus transcriptome profiling provides insights into the pathophysiology of adhesive arachnoiditis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167655. [PMID: 39755217 DOI: 10.1016/j.bbadis.2024.167655] [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: 05/15/2024] [Revised: 12/08/2024] [Accepted: 12/28/2024] [Indexed: 01/06/2025]
Abstract
Adhesive arachnoiditis (AA) is a rare form of chronic degenerative pathology associated with persistent inflammation in the arachnoid matter of the spinal cord. Despite the existing knowledge, the detailed pathological mechanisms underlying AA are not fully understood. This study aimed to elucidate through comprehensive single nuclei RNA sequencing (snRNA-seq) to delineate the transcriptomic landscape of AA. From six arachnoid membrane samples, a total of 52,886 cells met the quality control standards for analysis. The main cell populations identified with specific gene markers were as follows: fibroblasts, glial cells, microglial cells, endothelial cells, mural cells, plasma cells, and T cells. Downstream analysis of fibroblasts, glial cells, and microglial cells was performed. Notably, fibroblast subsets 1 and 3 demonstrated a strong association with AA. Among them, subcluster 3 demonstrated elevated expression of genes COL1A1, COL3A1, and FN1, indicative of enhanced Wnt/β-catenin and extracellular matrix (ECM) synthesis pathways. Subcluster 3 was predicted to progressively transform into subcluster 1. In subcluster 1, there was a significant upregulation of genes such as BMP and ALPL, signaling enhanced activation of calcification-related pathways. This was highly relevant to end-stage arachnoid ossification formation. After being activated, microglial cells transformed into inflammatory disease-associated microglial cells and continued to express high levels of chemokines CCL2, CCL4, IL-1β, and other inflammatory factors NAMPT, INPP5D and NLRP3. This might be the main reason why AA recurrence is frequently observed in patients. These insights enhance our understanding of the pathological progression of AA and may contribute to the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Weikang Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xiangyu Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Kai Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Zhenlei Liu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Lei Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Shaocheng Liu
- Beijing Mentougou District Hospital, Beijing 102300, China
| | - Kun He
- Department of Neurosurgery, Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China; Department of Neurosurgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - He Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Junyi Wang
- Beijing Science and Technology Innovation Group, Beijing 100101, China
| | - Yaobin Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yutian Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yuhua Yang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Hao Wu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
2
|
Moriya A, Inoue SI, Saitow F, Keitoku M, Suzuki N, Oike E, Urano E, Matsumoto E, Suzuki H, Aoki Y, Ohnishi H. Q241R mutation of Braf causes neurological abnormalities in a mouse model of cardio-facio-cutaneous syndrome, independent of developmental malformations. Hum Mol Genet 2025; 34:418-434. [PMID: 39774818 DOI: 10.1093/hmg/ddae196] [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: 07/10/2024] [Revised: 09/25/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Constitutively active mutants of BRAF cause cardio-facio-cutaneous (CFC) syndrome, characterized by growth and developmental defects, cardiac malformations, facial features, cutaneous manifestations, and mental retardation. An animal model of human CFC syndrome, the systemic BrafQ241R/+ mutant mouse, has been reported to exhibit multiple CFC syndrome-like phenotypes. In this study, we analyzed the effects of Braf mutations on neural function, separately from their effects on developmental processes. To this end, we generated Braf mutant mice expressing BRAFQ241R specifically in mature excitatory neurons (n-BrafQ241R/+). We found no growth retardation or cardiac malformations in n-BrafQ241R/+ mice, indicating normal development. Behavioral analysis revealed that n-BrafQ241R/+ mice exhibited reduced home cage activity and learning disability, which were similar to those of systemic BrafQ241R/+ mice. The active form of ERK was increased in the hippocampus of n-BrafQ241R/+ mice, whereas basal synaptic transmission and synaptic plasticity in hippocampal Schaffer collateral-CA1 synapses seems to be normal. Transcriptome analysis of the hippocampal tissue revealed significant changes in the expression of genes involved in regulation of the RAS/mitogen-activated protein kinase (MAPK) signaling pathway, synaptic function and memory formation. These data suggest that the neuronal dysfunction observed in the systemic CFC mouse model is due to the disruption of homeostasis of the RAS/MAPK signaling pathway by the activated Braf mutant after maturation, rather than abnormal development of the brain. A similar mechanism may be possible in human CFC syndrome.
Collapse
Affiliation(s)
- Akira Moriya
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-machi, Maebashi, Gunma 371-8514, Japan
| | - Shin-Ichi Inoue
- Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8575, Japan
| | - Fumihito Saitow
- Department of Pharmacology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan
| | - Moe Keitoku
- School of Science and Technology, Gunma University, 1-5-1 Tenjin-chou, Kiryu, Gunma 376-8515, Japan
| | - Noato Suzuki
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-machi, Maebashi, Gunma 371-8514, Japan
| | - Etsumi Oike
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-machi, Maebashi, Gunma 371-8514, Japan
| | - Eriko Urano
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-machi, Maebashi, Gunma 371-8514, Japan
| | - Eiko Matsumoto
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-machi, Maebashi, Gunma 371-8514, Japan
| | - Hidenori Suzuki
- Department of Pharmacology, Graduate School of Medicine, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo 113-8602, Japan
| | - Yoko Aoki
- Department of Medical Genetics, Tohoku University School of Medicine, 1-1 Seiryo-machi, Aoba-ku, Sendai 980-8574, Japan
| | - Hiroshi Ohnishi
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, 3-39-22 Showa-machi, Maebashi, Gunma 371-8514, Japan
- Gunma University Center for Food Science and Wellness, 4-2 Aramaki-machi, Maebashi, Gunma 371-8510, Japan
| |
Collapse
|
3
|
Moon CY, Belabed M, Park MD, Mattiuz R, Puleston D, Merad M. Dendritic cell maturation in cancer. Nat Rev Cancer 2025:10.1038/s41568-024-00787-3. [PMID: 39920276 DOI: 10.1038/s41568-024-00787-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 02/09/2025]
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that are present at low abundance in the circulation and tissues; they serve as crucial immune sentinels by continually sampling their environment, migrating to secondary lymphoid organs and shaping adaptive immune responses through antigen presentation. Owing to their ability to orchestrate tolerogenic or immunogenic responses to a specific antigen, DCs have a pivotal role in antitumour immunity and the response to immune checkpoint blockade and other immunotherapeutic approaches. The multifaceted functions of DCs are acquired through a complex, multistage process called maturation. Although the role of inflammatory triggers in driving DC maturation was established decades ago, less is known about DC maturation in non-inflammatory contexts, such as during homeostasis and in cancer. The advent of single-cell technologies has enabled an unbiased, high-dimensional characterization of various DC states, including mature DCs. This approach has clarified the molecular programmes associated with DC maturation and also revealed how cancers exploit these pathways to subvert immune surveillance. In this Review, we discuss the mechanisms by which cancer disrupts DC maturation and highlight emerging therapeutic opportunities to modulate DC states. These insights could inform the development of DC-centric immunotherapies, expanding the arsenal of strategies to enhance antitumour immunity.
Collapse
Affiliation(s)
- Chang Yoon Moon
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meriem Belabed
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D Park
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raphaël Mattiuz
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel Puleston
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
4
|
Zhang T, Wang W, Li J, Ye X, Wang Z, Cui S, Shen S, Liang X, Chen YQ, Zhu S. Free fatty acid receptor 4 modulates dietary sugar preference via the gut microbiota. Nat Microbiol 2025; 10:348-361. [PMID: 39805952 DOI: 10.1038/s41564-024-01902-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 12/05/2024] [Indexed: 01/16/2025]
Abstract
Sugar preference is a key contributor to the overconsumption of sugar and the concomitant increase in the incidence of diabetes. However, the exact mechanism of its development remains ambiguous. Here we show that the expression of free fatty acid receptor Ffar4, a receptor for long-chain fatty acids, is decreased in patients and mouse models with diabetes, which is associated with high sugar intake. Deletion of intestinal Ffar4 in mice resulted in reduced gut Bacteroides vulgatus and its metabolite pantothenate, leading to dietary sugar preference. Pantothenate promoted the secretion of GLP-1 which inhibited sugar preference by stimulating hepatic FGF21 release, which in turn regulates energy metabolism. These findings uncover a previously unappreciated role of Ffar4 in negatively regulating sugar preference and suggest B. vulgatus-derived pantothenate as a potential therapeutic target for diabetes.
Collapse
Affiliation(s)
- Tingting Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi, China
- Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Ministry of Education, Wuxi, China
| | - Wei Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- The Second Clinical Medical School, Xuzhou Medical University, Xuzhou, China
| | - Jiayu Li
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi, China
- Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Ministry of Education, Wuxi, China
| | - Xianlong Ye
- Ganjiang Chinese Medicine Innovation Center, Nanchang, China
| | - Zhe Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Siyuan Cui
- Wuxi No.2 People's Hospital Affiliated to Jiangnan University, Wuxi, China
| | - Shiwei Shen
- Wuxi No.2 People's Hospital Affiliated to Jiangnan University, Wuxi, China
| | - Xinmiao Liang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Key Laboratory of Separation Science for Analytical Chemistry, Dalian, China.
| | - Yong Q Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, China.
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi, China.
- Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, Ministry of Education, Wuxi, China.
| | - Shenglong Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi, China.
| |
Collapse
|
5
|
Ghena N, Anderson SR, Roberts JM, Irvin E, Schwakopf J, Bosco A, Vetter ML. CD11c-Expressing Microglia Are Transient, Driven by Interactions With Apoptotic Cells. Glia 2025. [PMID: 39828972 DOI: 10.1002/glia.24674] [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: 07/16/2024] [Revised: 12/24/2024] [Accepted: 01/05/2025] [Indexed: 01/22/2025]
Abstract
Microglia, the parenchymal macrophage of the central nervous system, serve crucial remodeling functions throughout development. Microglia are transcriptionally heterogenous, suggesting that distinct microglial states confer discrete roles. Currently, little is known about how dynamic these states are, the cues that promote them, or how they impact microglial function. In the developing retina, we previously found a significant proportion of microglia express CD11c (Integrin αX, Itgax, subunit of complement receptor 4) which has also been reported in other developmental and disease contexts. Here, we sought to understand the regulation and function of CD11c+ microglia. We found that CD11c+ microglia track with prominent waves of neuronal apoptosis in postnatal retina. Using genetic fate mapping, we provide evidence that microglia transition out of the CD11c state to return to homeostasis. We show that CD11c+ microglia have elevated lysosomal content and contribute to the clearance of apoptotic neurons, and found that acquisition of CD11c expression is partially dependent upon the TAM receptor AXL. Using selective ablation, we found CD11c+ microglia are not uniquely critical for phagocytic clearance of apoptotic cells. Together, our data suggest that CD11c+ microglia are a transient state induced by developmental apoptosis rather than a specialized subset mediating phagocytic elimination.
Collapse
Affiliation(s)
- Nathaniel Ghena
- Department of Neurobiology, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, Utah, USA
| | - Sarah R Anderson
- Department of Neurobiology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Jacqueline M Roberts
- Department of Neurobiology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Emmalyn Irvin
- Department of Neurobiology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Joon Schwakopf
- Department of Neurobiology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Alejandra Bosco
- Department of Neurobiology, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Monica L Vetter
- Department of Neurobiology, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
6
|
Salminen A. The role of inhibitory immune checkpoint receptors in the pathogenesis of Alzheimer's disease. J Mol Med (Berl) 2025; 103:1-19. [PMID: 39601807 PMCID: PMC11739239 DOI: 10.1007/s00109-024-02504-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/16/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
There is mounting evidence that microglial cells have a key role in the pathogenesis of Alzheimer's disease (AD). In AD pathology, microglial cells not only are unable to remove β-amyloid (Aβ) plaques and invading pathogens but also are involved in synaptic pruning, chronic neuroinflammation, and neuronal degeneration. Microglial cells possess many different inhibitory immune checkpoint receptors, such as PD-1, LILRB2-4, Siglecs, and SIRPα receptors, which can be targeted by diverse cell membrane-bound and soluble ligand proteins to suppress the functions of microglia. Interestingly, in the brains of AD patients there are elevated levels of many of the inhibitory ligands acting via these inhibitory checkpoint receptors. For instance, Aβ oligomers, ApoE4, and fibronectin are able to stimulate the LILRB2-4 receptors. Increased deposition of sialoglycans, e.g., gangliosides, inhibits microglial function via Siglec receptors. AD pathology augments the accumulation of senescent cells, which are known to possess a high level of PD-L1 proteins, and thus, they can evade immune surveillance. A decrease in the expression of SIRPα receptor in microglia and its ligand CD47 in neurons enhances the phagocytic pruning of synapses in AD brains. Moreover, cerebral neurons contain inhibitory checkpoint receptors which can inhibit axonal growth, reduce synaptic plasticity, and impair learning and memory. It seems that inappropriate inhibitory immune checkpoint signaling impairs the functions of microglia and neurons thus promoting AD pathogenesis. KEY MESSAGES: Microglial cells have a major role in the pathogenesis of AD. A decline in immune activity of microglia promotes AD pathology. Microglial cells and neurons contain diverse inhibitory immune checkpoint receptors. The level of ligands for inhibitory checkpoint receptors is increased in AD pathology. Impaired signaling of inhibitory immune checkpoint receptors promotes AD pathology.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| |
Collapse
|
7
|
Deng Q, Wu C, Parker E, Liu TCY, Duan R, Yang L. Microglia and Astrocytes in Alzheimer's Disease: Significance and Summary of Recent Advances. Aging Dis 2024; 15:1537-1564. [PMID: 37815901 PMCID: PMC11272214 DOI: 10.14336/ad.2023.0907] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 09/07/2023] [Indexed: 10/12/2023] Open
Abstract
Alzheimer's disease, one of the most common forms of dementia, is characterized by a slow progression of cognitive impairment and neuronal loss. Currently, approved treatments for AD are hindered by various side effects and limited efficacy. Despite considerable research, practical treatments for AD have not been developed. Increasing evidence shows that glial cells, especially microglia and astrocytes, are essential in the initiation and progression of AD. During AD progression, activated resident microglia increases the ability of resting astrocytes to transform into reactive astrocytes, promoting neurodegeneration. Extensive clinical and molecular studies show the involvement of microglia and astrocyte-mediated neuroinflammation in AD pathology, indicating that microglia and astrocytes may be potential therapeutic targets for AD. This review will summarize the significant and recent advances of microglia and astrocytes in the pathogenesis of AD in three parts. First, we will review the typical pathological changes of AD and discuss microglia and astrocytes in terms of function and phenotypic changes. Second, we will describe microglia and astrocytes' physiological and pathological role in AD. These roles include the inflammatory response, "eat me" and "don't eat me" signals, Aβ seeding, propagation, clearance, synapse loss, synaptic pruning, remyelination, and demyelination. Last, we will review the pharmacological and non-pharmacological therapies targeting microglia and astrocytes in AD. We conclude that microglia and astrocytes are essential in the initiation and development of AD. Therefore, understanding the new role of microglia and astrocytes in AD progression is critical for future AD studies and clinical trials. Moreover, pharmacological, and non-pharmacological therapies targeting microglia and astrocytes, with specific studies investigating microglia and astrocyte-mediated neuronal damage and repair, may be a promising research direction for future studies regarding AD treatment and prevention.
Collapse
Affiliation(s)
- Qianting Deng
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| | - Chongyun Wu
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| | - Emily Parker
- Medical College of Georgia at Augusta University, Augusta, GA 30912, USA.
| | - Timon Cheng-Yi Liu
- Laboratory of Laser Sports Medicine, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| | - Rui Duan
- Laboratory of Regenerative Medicine in Sports Science, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| | - Luodan Yang
- Laboratory of Exercise and Neurobiology, School of Physical Education and Sports Science, South China Normal University, Guangzhou 510006, China.
| |
Collapse
|
8
|
Mayo F, González-Vinceiro L, Hiraldo-González L, Calle-Castillejo C, Torres-Rubio I, Mayo M, Ramírez-Lorca R, Echevarría M. Absence of Aquaporin-4 (AQP4) Prolongs the Presence of a CD11c+ Microglial Population during Postnatal Corpus Callosum Development. Int J Mol Sci 2024; 25:8332. [PMID: 39125902 PMCID: PMC11312288 DOI: 10.3390/ijms25158332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
Aquaporin-4 (AQP4) expression is associated with the development of congenital hydrocephalus due to its structural role in the ependymal membrane. Gene expression analysis of periaqueductal tissue in AQP4-knockout (KO) mice at 11 days of age (P11) showed a modification in ependymal cell adhesion and ciliary protein expression that could alter cerebrospinal fluid homeostasis. A microglial subpopulation of CD11c+ cells was overexpressed in the periaqueductal tissue of mice that did not develop hydrocephalus, suggesting a possible protective effect. Here, we verified the location of this CD11c+ expression in the corpus callosum (CC) and cerebellum of AQP4-KO mice and analysed its time course. Immunofluorescence labelling of the CD11c protein in the CC and cerebellum of WT and KO animals at P3, P5, P7 and P11 confirmed an expanded presence of these cells in both tissues of the KO animal; CD11c+ cells appeared at P3 and reached a peak at P11, whereas in the WT animal, they appeared at P5, reached their peak at P7 and were undetectable by P11. The gene expression analysis in the CC samples at P11 confirmed the presence of CD11c+ microglial cells in this tissue. Among the more than 4000 overexpressed genes, Spp1 stood out with the highest differential gene expression (≅600), with other genes, such as Gpnmb, Itgax, Cd68 and Atp6v0d2, also identified as overexpressed. Therefore, CD11c+ cells appear to be necessary for normal corpus callosum development during postnatal life, and the absence of AQP4 prolonged its expression in this tissue.
Collapse
Affiliation(s)
- Francisco Mayo
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
| | - Lourdes González-Vinceiro
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
| | - Laura Hiraldo-González
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
| | - Claudia Calle-Castillejo
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Ismael Torres-Rubio
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Manuel Mayo
- Física Teórica, Universidad de Sevilla, Apartado de Correos 1065, 41080 Sevilla, Spain
| | - Reposo Ramírez-Lorca
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
| | - Miriam Echevarría
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain
- Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, 41009 Sevilla, Spain
| |
Collapse
|
9
|
Ghena N, Anderson SR, Roberts JM, Irvin E, Schwakopf J, Bosco A, Vetter ML. CD11c-expressing microglia are transient, driven by interactions with apoptotic cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600082. [PMID: 38979153 PMCID: PMC11230207 DOI: 10.1101/2024.06.24.600082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Microglia, the parenchymal macrophage of the central nervous system serve crucial remodeling functions throughout development. Microglia are transcriptionally heterogenous, suggesting that distinct microglial states confer discrete roles. Currently, little is known about how dynamic these states are, the cues that promote them, or how they impact microglial function. In the developing retina, we previously found a significant proportion of microglia express CD11c (Integrin αX, complement receptor 4, Itgax) which has also been reported in other developmental and disease contexts. Here, we sought to understand the regulation and function of CD11c+ microglia. We found that CD11c+ microglia track with prominent waves of neuronal apoptosis in postnatal retina. Using genetic fate mapping, we provide evidence that microglia transition out of the CD11c state to return to homeostasis. We show that CD11c+ microglia have elevated lysosomal content and contribute to the clearance of apoptotic neurons, and found that acquisition of CD11c expression is, in part, dependent upon the TAM receptor Axl. Using selective ablation, we found CD11c+ microglia are not uniquely critical for phagocytic clearance of apoptotic cells. Together, our data suggest CD11c+ microglia are a transient state induced by developmental apoptosis rather than a specialized subset mediating phagocytic elimination.
Collapse
Affiliation(s)
- Nathaniel Ghena
- Department of Neurobiology, University of Utah School of Medicine
- Interdepartmental Program in Neuroscience, University of Utah
| | - Sarah R Anderson
- Department of Neurobiology, University of Utah School of Medicine
| | | | - Emmalyn Irvin
- Department of Neurobiology, University of Utah School of Medicine
| | - Joon Schwakopf
- Department of Neurobiology, University of Utah School of Medicine
| | - Alejandra Bosco
- Department of Neurobiology, University of Utah School of Medicine
| | - Monica L Vetter
- Department of Neurobiology, University of Utah School of Medicine
- Interdepartmental Program in Neuroscience, University of Utah
| |
Collapse
|
10
|
Szabó K, Makkai G, Konkoly J, Kormos V, Gaszner B, Berki T, Pintér E. TRPA1 Covalent Ligand JT010 Modifies T Lymphocyte Activation. Biomolecules 2024; 14:632. [PMID: 38927036 PMCID: PMC11202300 DOI: 10.3390/biom14060632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/25/2024] [Accepted: 05/26/2024] [Indexed: 06/28/2024] Open
Abstract
Transient Receptor Potential Ankyrin 1 (TRPA1) is a non-selective cation channel involved in sensitivity to a plethora of irritating agents and endogenous mediators of oxidative stress. TRPA1 influences neuroinflammation and macrophage and lymphocyte functions, but its role is controversial in immune cells. We reported earlier a detectable, but orders-of-magnitude-lower level of Trpa1 mRNA in monocytes and lymphocytes than in sensory neurons by qRT-PCR analyses of cells from lymphoid organs of mice. Our present goals were to (a) further elucidate the expression of Trpa1 mRNA in immune cells by RNAscope in situ hybridization (ISH) and (b) test the role of TRPA1 in lymphocyte activation. RNAscope ISH confirmed that Trpa1 transcripts were detectable in CD14+ and CD4+ cells from the peritoneal cavity of mice. A selective TRPA1 agonist JT010 elevated Ca2+ levels in these cells only at high concentrations. However, a concentration-dependent inhibitory effect of JT010 was observed on T-cell receptor (TcR)-induced Ca2+ signals in CD4+ T lymphocytes, while JT010 neither modified B cell activation nor ionomycin-stimulated Ca2+ level. Based on our present and past findings, TRPA1 activation negatively modulates T lymphocyte activation, but it does not appear to be a key regulator of TcR-stimulated calcium signaling.
Collapse
Affiliation(s)
- Katalin Szabó
- Institute of Pharmacology and Pharmacotherapy, University of Pécs Medical School, H-7624 Pécs, Hungary (E.P.)
| | - Géza Makkai
- Nano-Bio-Imaging Core Facility, University of Pécs Medical School, H-7624 Pécs, Hungary
| | - János Konkoly
- Institute of Pharmacology and Pharmacotherapy, University of Pécs Medical School, H-7624 Pécs, Hungary (E.P.)
| | - Viktória Kormos
- Institute of Pharmacology and Pharmacotherapy, University of Pécs Medical School, H-7624 Pécs, Hungary (E.P.)
| | - Balázs Gaszner
- Research Group for Mood Disorders, Department of Anatomy, University of Pécs Medical School, H-7624 Pécs, Hungary
| | - Tímea Berki
- Department of Immunology and Biotechnology, University of Pécs Clinical Center, H-7624 Pécs, Hungary
| | - Erika Pintér
- Institute of Pharmacology and Pharmacotherapy, University of Pécs Medical School, H-7624 Pécs, Hungary (E.P.)
| |
Collapse
|
11
|
Nomaki K, Fujikawa R, Masuda T, Tsuda M. Spatiotemporal dynamics of the CD11c + microglial population in the mouse brain and spinal cord from developmental to adult stages. Mol Brain 2024; 17:24. [PMID: 38762724 PMCID: PMC11102220 DOI: 10.1186/s13041-024-01098-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 05/08/2024] [Indexed: 05/20/2024] Open
Abstract
CD11c-positive (CD11c+) microglia have attracted considerable attention because of their potential implications in central nervous system (CNS) development, homeostasis, and disease. However, the spatiotemporal dynamics of the proportion of CD11c+ microglia in individual CNS regions are poorly understood. Here, we investigated the proportion of CD11c+ microglia in six CNS regions (forebrain, olfactory bulb, diencephalon/midbrain, cerebellum, pons/medulla, and spinal cord) from the developmental to adult stages by flow cytometry and immunohistochemical analyses using a CD11c reporter transgenic mouse line, Itgax-Venus. We found that the proportion of CD11c+ microglia in total microglia varied between CNS regions during postnatal development. Specifically, the proportion was high in the olfactory bulb and cerebellum at postnatal day P(4) and P7, respectively, and approximately half of the total microglia were CD11c+. The proportion declined sharply in all regions to P14, and the low percentage persisted over P56. In the spinal cord, the proportion of CD11c+ microglia was also high at P4 and declined to P14, but increased again at P21 and thereafter. Interestingly, the distribution pattern of CD11c+ microglia in the spinal cord markedly changed from gray matter at P4 to white matter at P21. Collectively, our findings reveal the differences in the spatiotemporal dynamics of the proportion of CD11c+ microglia among CNS regions from early development to adult stages in normal mice. These findings improve our understanding of the nature of microglial heterogeneity and its dynamics in the CNS.
Collapse
Affiliation(s)
- Kohei Nomaki
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Risako Fujikawa
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takahiro Masuda
- Division of Molecular Neuroimmunology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Makoto Tsuda
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
- Kyushu University Institute for Advanced Study, 744 Motooka Nishi-ku, Fukuoka, 819-0395, Japan.
| |
Collapse
|
12
|
Wang W, Li J, Cui S, Li J, Ye X, Wang Z, Zhang T, Jiang X, Kong Y, Chen X, Chen YQ, Zhu S. Microglial Ffar4 deficiency promotes cognitive impairment in the context of metabolic syndrome. SCIENCE ADVANCES 2024; 10:eadj7813. [PMID: 38306420 PMCID: PMC10836723 DOI: 10.1126/sciadv.adj7813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 01/03/2024] [Indexed: 02/04/2024]
Abstract
Metabolic syndrome (MetS) is closely associated with an increased risk of dementia and cognitive impairment, and a complex interaction of genetic and environmental dietary factors may be implicated. Free fatty acid receptor 4 (Ffar4) may bridge the genetic and dietary aspects of MetS development. However, the role of Ffar4 in MetS-related cognitive dysfunction is unclear. In this study, we found that Ffar4 expression is down-regulated in MetS mice and MetS patients with cognitive impairment. Conventional and microglial conditional knockout of Ffar4 exacerbated high-fat diet (HFD)-induced cognitive dysfunction and anxiety, whereas microglial Ffar4 overexpression improved HFD-induced cognitive dysfunction and anxiety. Mechanistically, we found that microglial Ffar4 regulated microglial activation through type I interferon signaling. Microglial depletion and NF-κB inhibition partially reversed cognitive dysfunction and anxiety in microglia-specific Ffar4 knockout MetS mice. Together, these findings uncover a previously unappreciated role of Ffar4 in negatively regulating the NF-κB-IFN-β signaling and provide an attractive therapeutic target for delaying MetS-associated cognitive decline.
Collapse
Affiliation(s)
- Wei Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Jinyou Li
- Affiliated Hospital of Jiangnan University, Wuxi 214122, China
| | - Siyuan Cui
- Jiangnan University Medical Center, Wuxi 214002, China
| | - Jiayu Li
- Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Xianlong Ye
- Ganjiang Chinese Medicine Innovation Center, Nanchang 330000, China
| | - Zhe Wang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Tingting Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Xuan Jiang
- Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Yulin Kong
- Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Xin Chen
- Jiangnan University Medical Center, Wuxi 214002, China
| | - Yong Q. Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
| | - Shenglong Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi 214000, China
- Jiangnan University Medical Center, Wuxi 214002, China
| |
Collapse
|
13
|
Hu Y, Wang X, Zhao Z, Liu M, Ren X, Xian X, Liu C, Wang Q. The Downregulation of ITGAX Exacerbates Amyloid-β Plaque Deposition in Alzheimer's Disease by Increasing Polarization of M1 Microglia. J Alzheimers Dis 2024; 100:657-673. [PMID: 38905043 DOI: 10.3233/jad-240118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/23/2024]
Abstract
Background Alzheimer's disease (AD) is the most common sort of neurodegenerative dementia, characterized by its challenging, diverse, and progressive nature. Despite significant progress in neuroscience, the current treatment strategies remain suboptimal. Objective Identifying a more accurate molecular target for the involvement of microglia in the pathogenic process of AD and exploring potential mechanisms via which it could influence disease. Methods We utilized single-cell RNA sequencing (scRNA-seq) analysis in conjunction with APP/PS1 mouse models to find out the molecular mechanism of AD. With the goal of investigating the cellular heterogeneity of AD, we downloaded the scRNA-seq data from the Gene Expression Omnibus (GEO) database and identified differentially expressed genes (DEGs). Additionally, we evaluated learning and memory capacity using the behavioral experiment. We also examined the expression of proteins associated with memory using western blotting. Immunofluorescence was employed to investigate alterations in amyloid plaques and microglia. Results Our findings revealed an upregulation of ITGAX expression in APP/PS1 transgenic mice, which coincided with a downregulation of synaptic plasticity-related proteins, an increase in amyloid-β (Aβ) plaques, and an elevation in the number of M1 microglia. Interestingly, deletion of ITGAX resulted in increased Aβ plaque deposition, a rise in the M1 microglial phenotype, and decreased production of synaptic plasticity-related proteins, all of which contributed to a decline in learning and memory. Conclusions This research suggested that ITGAX may have a beneficial impact on the APP/PS1 mice model, as its decreased expression could exacerbate the impairment of synaptic plasticity and worsen cognitive dysfunction.
Collapse
Affiliation(s)
- Yufei Hu
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xupeng Wang
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zijun Zhao
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Anesthesiology, Hebei Provincial Chest Hospital, Shijiazhuang, Hebei, China
| | - Menglin Liu
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaoqin Ren
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaohui Xian
- Department of Pathophysiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Chunxiao Liu
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qiujun Wang
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
14
|
Mimic S, Aru B, Pehlivanoğlu C, Sleiman H, Andjus PR, Yanıkkaya Demirel G. Immunology of amyotrophic lateral sclerosis - role of the innate and adaptive immunity. Front Neurosci 2023; 17:1277399. [PMID: 38105925 PMCID: PMC10723830 DOI: 10.3389/fnins.2023.1277399] [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: 08/14/2023] [Accepted: 11/07/2023] [Indexed: 12/19/2023] Open
Abstract
This review aims to summarize the latest evidence about the role of innate and adaptive immunity in Amyotrophic Lateral Sclerosis (ALS). ALS is a devastating neurodegenerative disease affecting upper and lower motor neurons, which involves essential cells of the immune system that play a basic role in innate or adaptive immunity, that can be neurotoxic or neuroprotective for neurons. However, distinguishing between the sole neurotoxic or neuroprotective function of certain cells such as astrocytes can be challenging due to intricate nature of these cells, the complexity of the microenvironment and the contextual factors. In this review, in regard to innate immunity we focus on the involvement of monocytes/macrophages, microglia, the complement, NK cells, neutrophils, mast cells, and astrocytes, while regarding adaptive immunity, in addition to humoral immunity the most important features and roles of T and B cells are highlighted, specifically different subsets of CD4+ as well as CD8+ T cells. The role of autoantibodies and cytokines is also discussed in distinct sections of this review.
Collapse
Affiliation(s)
- Stefan Mimic
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Jean Giaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Başak Aru
- Immunology Department, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Cemil Pehlivanoğlu
- Immunology Department, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Hadi Sleiman
- Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Pavle R. Andjus
- Centre for Laser Microscopy, Institute of Physiology and Biochemistry “Jean Giaja”, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | | |
Collapse
|
15
|
Zhu H, Guan A, Liu J, Peng L, Zhang Z, Wang S. Noteworthy perspectives on microglia in neuropsychiatric disorders. J Neuroinflammation 2023; 20:223. [PMID: 37794488 PMCID: PMC10548593 DOI: 10.1186/s12974-023-02901-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 09/22/2023] [Indexed: 10/06/2023] Open
Abstract
Microglia are so versatile that they not only provide immune surveillance for central nervous system, but participate in neural circuitry development, brain blood vessels formation, blood-brain barrier architecture, and intriguingly, the regulation of emotions and behaviors. Microglia have a profound impact on neuronal survival, brain wiring and synaptic plasticity. As professional phagocytic cells in the brain, they remove dead cell debris and neurotoxic agents via an elaborate mechanism. The functional profile of microglia varies considerately depending on age, gender, disease context and other internal or external environmental factors. Numerous studies have demonstrated a pivotal involvement of microglia in neuropsychiatric disorders, including negative affection, social deficit, compulsive behavior, fear memory, pain and other symptoms associated with major depression disorder, anxiety disorder, autism spectrum disorder and schizophrenia. In this review, we summarized the latest discoveries regarding microglial ontogeny, cell subtypes or state spectrum, biological functions and mechanistic underpinnings of emotional and behavioral disorders. Furthermore, we highlight the potential of microglia-targeted therapies of neuropsychiatric disorders, and propose outstanding questions to be addressed in future research of human microglia.
Collapse
Affiliation(s)
- Hongrui Zhu
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Ao Guan
- School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Jiayuan Liu
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Li Peng
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Zhi Zhang
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
- Hefei National Laboratory for Physical Sciences at the Microscale, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Sheng Wang
- Department of Anesthesiology, First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| |
Collapse
|
16
|
Tsuda M, Masuda T, Kohno K. Microglial diversity in neuropathic pain. Trends Neurosci 2023:S0166-2236(23)00124-8. [PMID: 37244781 DOI: 10.1016/j.tins.2023.05.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/17/2023] [Accepted: 05/02/2023] [Indexed: 05/29/2023]
Abstract
Microglia play pivotal roles in controlling CNS functions in diverse physiological and pathological contexts, including neuropathic pain, a chronic pain condition caused by lesions or diseases of the somatosensory nervous system. In this review article, we summarize evidence primarily from basic research on the role of microglia in the development and remission of neuropathic pain. The identification of a subset of microglia that emerged after pain development and that was necessary for remission of neuropathic pain highlights the highly divergent and dynamic nature of microglia in the course of neuropathic pain. Understanding microglial diversity in terms of gene expression, physiological states, and functional roles could lead to new strategies that aid in the diagnosis and management of neuropathic pain, and that may not have been anticipated from the viewpoint of targeting all microglia uniformly.
Collapse
Affiliation(s)
- Makoto Tsuda
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan; Kyushu University Institute for Advanced Study, Fukuoka, Japan.
| | - Takahiro Masuda
- Division of Molecular Neuroimmunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Keita Kohno
- Department of Molecular and System Pharmacology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
17
|
Schlett JS, Mettang M, Skaf A, Schweizer P, Errerd A, Mulugeta EA, Hein TM, Tsesmelis K, Tsesmelis M, Büttner UFG, Wendt H, Abaei A, Rasche V, Prex V, Nespoli E, Alami NO, Tews D, Walther P, Yilmazer-Hanke D, Oswald F, Dimou L, Wirth T, Baumann B. NF-κB is a critical mediator of post-mitotic senescence in oligodendrocytes and subsequent white matter loss. Mol Neurodegener 2023; 18:24. [PMID: 37069623 PMCID: PMC10108549 DOI: 10.1186/s13024-023-00616-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 03/25/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND Inflammaging represents an accepted concept where the immune system shifts to a low-grade chronic pro-inflammatory state without overt infection upon aging. In the CNS, inflammaging is mainly driven by glia cells and associated with neurodegenerative processes. White matter degeneration (WMD), a well-known process in the aging brain, manifests in myelin loss finally resulting in motor, sensory and cognitive impairments. Oligodendrocytes (OL) are responsible for homeostasis and maintenance of the myelin sheaths, which is a complex and highly energy demanding process sensitizing these cells to metabolic, oxidative and other forms of stress. Yet, the immediate impact of chronic inflammatory stress like inflammaging on OL homeostasis, myelin maintenance and WMD remains open. METHODS To functionally analyze the role of IKK/NF-κB signaling in the regulation of myelin homeostasis and maintenance in the adult CNS, we established a conditional mouse model allowing NF-κB activation in mature myelinating oligodendrocytes. IKK2-CAPLP-CreERT2 mice were characterized by biochemical, immunohistochemical, ultrastructural and behavioral analyses. Transcriptome data from isolated, primary OLs and microglia cells were explored by in silico pathway analysis and validated by complementary molecular approaches. RESULTS Chronic NF-κB activation in mature OLs leads to aggravated neuroinflammatory conditions phenocopying brain inflammaging. As a consequence, IKK2-CAPLP-CreERT2 mice showed specific neurological deficits and impaired motoric learning. Upon aging, persistent NF-κB signaling promotes WMD in these mice as ultrastructural analysis revealed myelination deficits in the corpus callosum accompanied by impaired myelin protein expression. RNA-Seq analysis of primary oligodendrocytes and microglia cells uncovers gene expression signatures associated with activated stress responses and increased post mitotic cellular senescence (PoMiCS) which was confirmed by elevated senescence-associated β-galactosidase activity and SASP gene expression profile. We identified an elevated integrated stress response (ISR) characterized by phosphorylation of eIF2α as a relevant molecular mechanism which is able to affect translation of myelin proteins. CONCLUSIONS Our findings demonstrate an essential role of IKK/NF-κB signaling in mature, post-mitotic OLs in regulating stress-induced senescence in these cells. Moreover, our study identifies PoMICS as an important driving force of age-dependent WMD as well as of traumatic brain injury induced myelin defects.
Collapse
Affiliation(s)
- Judith Stefanie Schlett
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Melanie Mettang
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Aladdin Skaf
- Molecular and Translational Neuroscience, Department of Neurology, University Medical Center Ulm, 89081, Ulm, Germany
| | - Pavel Schweizer
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Alina Errerd
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | | | - Tabea Melissa Hein
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Konstantinos Tsesmelis
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Miltiadis Tsesmelis
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Ulrike F G Büttner
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Heinrich Wendt
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Alireza Abaei
- Core Facility Small Animal Imaging (CF-SANI), Ulm University, 89081, Ulm, Germany
| | - Volker Rasche
- Core Facility Small Animal Imaging (CF-SANI), Ulm University, 89081, Ulm, Germany
| | - Vivien Prex
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Ester Nespoli
- Molecular and Translational Neuroscience, Department of Neurology, University Medical Center Ulm, 89081, Ulm, Germany
| | - Najwa Ouali Alami
- Institute of Clinical Neuroanatomy, Ulm University, Helmholtzstraße 8/1, 89081, Ulm, Germany
| | - Daniel Tews
- Core Facility Extracellular Flux Analyzer, Ulm University Medical Center, 89081, Ulm, Germany
| | - Paul Walther
- Central Facility for Electron Microscopy, Ulm University, 89081, Ulm, Germany
| | - Deniz Yilmazer-Hanke
- Institute of Clinical Neuroanatomy, Ulm University, Helmholtzstraße 8/1, 89081, Ulm, Germany
| | - Franz Oswald
- Department of Internal Medicine I, Center for Internal Medicine, University Medical Center Ulm, 89081, Ulm, Germany
| | - Leda Dimou
- Molecular and Translational Neuroscience, Department of Neurology, University Medical Center Ulm, 89081, Ulm, Germany
| | - Thomas Wirth
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany
| | - Bernd Baumann
- Institute of Physiological Chemistry, Ulm University, Albert- Einstein-Allee 11, 89081, Ulm, Germany.
| |
Collapse
|
18
|
Su X, Xie L, Li J, Tian X, Lin B, Chen M. Exploring molecular signatures related to the mechanism of aging in different brain regions by integrated bioinformatics. Front Mol Neurosci 2023; 16:1133106. [PMID: 37033380 PMCID: PMC10076559 DOI: 10.3389/fnmol.2023.1133106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 02/22/2023] [Indexed: 04/11/2023] Open
Abstract
The mechanism of brain aging is not fully understood. Few studies have attempted to identify molecular changes using bioinformatics at the subregional level in the aging brain. This study aimed to identify the molecular signatures and key genes involved in aging, depending on the brain region. Differentially expressed genes (DEGs) associated with aging of the cerebral cortex (CX), hippocampus (HC), and cerebellum (CB) were identified based on five datasets from the Gene Expression Omnibus (GEO). The molecular signatures of aging were explored using functional and pathway analyses. Hub genes of each brain region were determined by protein-protein interaction network analysis, and commonly expressed DEGs (co-DEGs) were also found. Gene-microRNAs (miRNAs) and gene-disease interactions were constructed using online databases. The expression levels and regional specificity of the hub genes and co-DEGs were validated using animal experiments. In total, 32, 293, and 141 DEGs were identified in aging CX, HC, and CB, respectively. Enrichment analysis indicated molecular changes related to leukocyte invasion, abnormal neurotransmission, and impaired neurogenesis due to inflammation as the major signatures of the CX, HC, and CB. Itgax is a hub gene of cortical aging. Zfp51 and Zfp62 were identified as hub genes involved in hippocampal aging. Itgax and Cxcl10 were identified as hub genes involved in cerebellar aging. S100a8 was the only co-DEG in all three regions. In addition, a series of molecular changes associated with inflammation was observed in all three brain regions. Several miRNAs interact with hub genes and S100a8. The change in gene levels was further validated in an animal experiment. Only the upregulation of Zfp51 and Zfp62 was restricted to the HC. The molecular signatures of aging exhibit regional differences in the brain and seem to be closely related to neuroinflammation. Itgax, Zfp51, Zfp62, Cxcl10, and S100a8 may be key genes and potential targets for the prevention of brain aging.
Collapse
Affiliation(s)
- Xie Su
- Department of Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lu Xie
- Department of Physiology, Pre-Clinical Science, Guangxi Medical University, Nanning, China
| | - Jing Li
- Department of Physiology, Pre-Clinical Science, Guangxi Medical University, Nanning, China
| | - Xinyue Tian
- Department of Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Bing Lin
- Department of Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Menghua Chen
- Department of Intensive Care Unit, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
- *Correspondence: Menghua Chen,
| |
Collapse
|
19
|
Li N, Deng M, Hu G, Li N, Yuan H, Zhou Y. New Insights into Microglial Mechanisms of Memory Impairment in Alzheimer's Disease. Biomolecules 2022; 12:1722. [PMID: 36421736 PMCID: PMC9687453 DOI: 10.3390/biom12111722] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 09/15/2023] Open
Abstract
Alzheimer's disease (AD) is the most common progressive and irreversible neurodegeneration characterized by the impairment of memory and cognition. Despite years of studies, no effective treatment and prevention strategies are available yet. Identifying new AD therapeutic targets is crucial for better elucidating the pathogenesis and establishing a valid treatment of AD. Growing evidence suggests that microglia play a critical role in AD. Microglia are resident macrophages in the central nervous system (CNS), and their core properties supporting main biological functions include surveillance, phagocytosis, and the release of soluble factors. Activated microglia not only directly mediate the central immune response, but also participate in the pathological changes of AD, including amyloid-beta (Aβ) aggregation, tau protein phosphorylation, synaptic dissection, neuron loss, memory function decline, etc. Based on these recent findings, we provide a new framework to summarize the role of microglia in AD memory impairment. This evidence suggests that microglia have the potential to become new targets for AD therapy.
Collapse
Affiliation(s)
- Na Li
- Department of Rehabilitation Medicine, Affiliated Hospital of Qingdao University, Qingdao 266000, China
- Department of Medicine, Qingdao Binhai University, Qingdao 266555, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao 266071, China
| | - Mingru Deng
- Department of Neurology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao 266042, China
| | - Gonghui Hu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao 266071, China
| | - Nan Li
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266000, China
| | - Haicheng Yuan
- Department of Neurology, Affiliated Qingdao Central Hospital of Qingdao University, Qingdao 266042, China
| | - Yu Zhou
- Department of Rehabilitation Medicine, Affiliated Hospital of Qingdao University, Qingdao 266000, China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Qingdao University, Qingdao 266071, China
- Department of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao 266000, China
- Institute of Brain Sciences and Related Disorders, Qingdao University, Qingdao 266071, China
| |
Collapse
|
20
|
Ortiz-Rivera J, Albors A, Kucheryavykh Y, Harrison JK, Kucheryavykh L. The Dynamics of Tumor-Infiltrating Myeloid Cell Activation and the Cytokine Expression Profile in a Glioma Resection Site during the Post-Surgical Period in Mice. Brain Sci 2022; 12:brainsci12070893. [PMID: 35884700 PMCID: PMC9313002 DOI: 10.3390/brainsci12070893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 02/01/2023] Open
Abstract
Glioblastoma is the most aggressive brain cancer and is highly infiltrated with cells of myeloid lineage (TIM) that support tumor growth and invasion. Tumor resection is the primary treatment for glioblastoma; however, the activation state of TIM at the site of tumor resection and its impact on glioma regrowth are poorly understood. Using the C57BL/6/GL261 mouse glioma implantation model, we investigated the state of TIM in the tumor resection area during the post-surgical period. TIM isolated from brain tissue at the resection site were analyzed at 0, 1, 4, 7, 14, and 21 days after tumor resection. An increase in expression of CD86 during the first 7 days after surgical resection and then upregulation of arginase 1 from the 14th to 21st days after resection were detected. Cytokine expression analysis combined with qRT-PCR revealed sustained upregulation of IL4, IL5, IL10, IL12, IL17, vascular endothelial growth factor (VEGF), and monocyte chemoattractant protein 1 (MCP1/CCL2) in TIM purified from regrown tumors compared with primary implanted tumors. Flow cytometry analysis revealed increased CD86+/CD206+ population in regrown tumors compared with primary implanted tumors. Overall, we found that TIM in primary implanted tumors and tumors regrown after resection exhibited different phenotypes and cytokine expression patterns.
Collapse
Affiliation(s)
- Jescelica Ortiz-Rivera
- Department of Biochemistry, School of Medicine, Universidad Central de Caribe, Bayamon, PR 00956, USA; (A.A.); (Y.K.); (L.K.)
- Correspondence:
| | - Alejandro Albors
- Department of Biochemistry, School of Medicine, Universidad Central de Caribe, Bayamon, PR 00956, USA; (A.A.); (Y.K.); (L.K.)
| | - Yuriy Kucheryavykh
- Department of Biochemistry, School of Medicine, Universidad Central de Caribe, Bayamon, PR 00956, USA; (A.A.); (Y.K.); (L.K.)
| | - Jeffrey K. Harrison
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL 32610, USA;
| | - Lilia Kucheryavykh
- Department of Biochemistry, School of Medicine, Universidad Central de Caribe, Bayamon, PR 00956, USA; (A.A.); (Y.K.); (L.K.)
| |
Collapse
|
21
|
Zhang Y, Lian L, Fu R, Liu J, Shan X, Jin Y, Xu S. Microglia: The Hub of Intercellular Communication in Ischemic Stroke. Front Cell Neurosci 2022; 16:889442. [PMID: 35518646 PMCID: PMC9062186 DOI: 10.3389/fncel.2022.889442] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/30/2022] [Indexed: 12/14/2022] Open
Abstract
Communication between microglia and other cells has recently been at the forefront of research in central nervous system (CNS) disease. In this review, we provide an overview of the neuroinflammation mediated by microglia, highlight recent studies of crosstalk between microglia and CNS resident and infiltrating cells in the context of ischemic stroke (IS), and discuss how these interactions affect the course of IS. The in-depth exploration of microglia-intercellular communication will be beneficial for therapeutic tools development and clinical translation for stroke control.
Collapse
Affiliation(s)
- Yunsha Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, China
| | - Lu Lian
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rong Fu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jueling Liu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoqian Shan
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine Tianjin, China.,Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yang Jin
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine Tianjin, China
| | - Shixin Xu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| |
Collapse
|
22
|
Velayudhan PS, Schwab N, Hazrati LN, Wheeler AL. Temporal patterns of microglial activation in white matter following experimental mild traumatic brain injury: a systematic literature review. Acta Neuropathol Commun 2021; 9:197. [PMID: 34924026 PMCID: PMC8684664 DOI: 10.1186/s40478-021-01297-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 11/23/2021] [Indexed: 12/18/2022] Open
Abstract
Mild traumatic brain injuries (mTBIs) are a prevalent form of injury that can result in persistent neurological impairments. Microglial activation has become increasingly recognized as a key process regulating the pathology of white matter in a wide range of brain injury and disease contexts. As white matter damage is known to be a major contributor to the impairments that follow mTBI, microglia have rightfully become a common target of investigation for the development of mTBI therapies and biomarkers. Recent work has demonstrated that the efficacy of microglial manipulation as a therapeutic intervention following injury or disease is highly time-sensitive, emphasizing the importance of advancing our understanding of the dynamics of post-mTBI microglial activation from onset to resolution. Current reporting of microglial activation in experimental studies of mTBI is non-standardized, which has limited our ability to identify concrete patterns of post-mTBI microglial activation over time. In this review, we examine preclinical studies of mTBI that report on microglial activation in white matter regions to summarize our current understanding of these patterns. Specifically, we summarize timecourses of post-mTBI microglial activation in white matter regions of the brain, identify factors that influence this activation, examine the temporal relationship between microglial activation and other post-mTBI assessments, and compare the relative sensitivities of various methods for detecting microglial activation. While the lack of replicated experimental conditions has limited the extent of conclusions that can confidently be drawn, we find that microglia are activated over a wide range of timecourses following mTBI and that microglial activation is a long-lasting outcome of mTBI that may resolve after most typical post-mTBI assessments, with the exception of those measuring oligodendrocyte lineage cell integrity. We identify several understudied parameters of post-mTBI microglial activation in white matter, such as the inclusion of female subjects. This review summarizes our current understanding of the progression of microglial activation in white matter structures following experimental mTBI and offers suggestions for important future research directions.
Collapse
Affiliation(s)
- Prashanth S Velayudhan
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Nicole Schwab
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Lili-Naz Hazrati
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Anne L Wheeler
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada.
| |
Collapse
|
23
|
TREM2 interacts with TDP-43 and mediates microglial neuroprotection against TDP-43-related neurodegeneration. Nat Neurosci 2021; 25:26-38. [PMID: 34916658 PMCID: PMC8741737 DOI: 10.1038/s41593-021-00975-6] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 11/02/2021] [Indexed: 12/22/2022]
Abstract
Triggering receptor expressed on myeloid cell 2 (TREM2) is linked to neurodegenerative disease risk. However, the function of TREM2 in neurodegeneration is still not fully understood. Here we investigated the role of microglial TREM2 in TAR-DNA binding protein 43 kDa (TDP-43)-related neurodegeneration using viral-mediated and transgenic mouse models. We found that TREM2 deficiency impaired phagocytic clearance of pathological TDP-43 by microglia, and enhanced neuronal damage and motor impairments. Mass cytometry analysis revealed that hTDP-43 induced a TREM2-dependent subpopulation of microglia with high CD11c expression and phagocytic ability. Using mass spectrometry and surface plasmon resonance analysis, we further demonstrated an interaction between TDP-43 and TREM2 in vitro and in vivo as well as in ALS patient tissues. We computationally identified regions within hTDP-43 that interact with TREM2. Our data highlights that TDP-43 is a possible ligand for microglial TREM2 and that this interaction mediates neuroprotection of microglia in TDP-43-related neurodegeneration.
Collapse
|
24
|
Wang H, Newton G, Wu L, Lin LL, Miracco AS, Natesan S, Luscinskas FW. CD47 antibody blockade suppresses microglia-dependent phagocytosis and monocyte transition to macrophages, impairing recovery in EAE. JCI Insight 2021; 6:148719. [PMID: 34591795 PMCID: PMC8663579 DOI: 10.1172/jci.insight.148719] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 09/24/2021] [Indexed: 11/24/2022] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a well-characterized animal model of multiple sclerosis. During the early phase of EAE, infiltrating monocytes and monocyte-derived macrophages contribute to T cell recruitment, especially CD4+ T cells, into the CNS, resulting in neuronal demyelination; however, in later stages, they promote remyelination and recovery by removal of myelin debris by phagocytosis. Signal regulatory protein α and CD47 are abundantly expressed in the CNS, and deletion of either molecule is protective in myelin oligodendrocyte glycoprotein–induced EAE because of failed effector T cell expansion and trafficking. Here we report that treatment with the function blocking CD47 Ab Miap410 substantially reduced the infiltration of pathogenic immune cells but impaired recovery from paresis. The underlying mechanism was by blocking the emergence of CD11chiMHCIIhi microglia at peak disease that expressed receptors for phagocytosis, scavenging, and lipid catabolism, which mediated clearance of myelin debris and the transition of monocytes to macrophages in the CNS. In the recovery phase of EAE, Miap410 Ab–treated mice had worsening paresis with sustained inflammation and limited remyelination as compared with control Ab–treated mice. In summary, Ab blockade of CD47 impaired resolution of CNS inflammation, thus worsening EAE.
Collapse
Affiliation(s)
- Huan Wang
- Center for Excellence in Vascular Biology, Department of Pathology, Mass General Brigham and Harvard Medical School, Boston, Massachusetts, USA
| | - Gail Newton
- Center for Excellence in Vascular Biology, Department of Pathology, Mass General Brigham and Harvard Medical School, Boston, Massachusetts, USA
| | - Liguo Wu
- Immunology & Inflammation Research Therapeutic Area, Sanofi US, Cambridge, Massachusetts, USA
| | - Lih-Ling Lin
- Immunology & Inflammation Research Therapeutic Area, Sanofi US, Cambridge, Massachusetts, USA
| | - Amy S Miracco
- Immunology & Inflammation Research Therapeutic Area, Sanofi US, Cambridge, Massachusetts, USA
| | - Sridaran Natesan
- Immunology & Inflammation Research Therapeutic Area, Sanofi US, Cambridge, Massachusetts, USA
| | - Francis W Luscinskas
- Center for Excellence in Vascular Biology, Department of Pathology, Mass General Brigham and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
25
|
Domowicz MS, Chan WC, Claudio-Vázquez P, Gonzalez T, Schwartz NB. Brain transcriptome analysis of a CLN2 mouse model as a function of disease progression. J Neuroinflammation 2021; 18:262. [PMID: 34749772 PMCID: PMC8576919 DOI: 10.1186/s12974-021-02302-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 10/20/2021] [Indexed: 12/13/2022] Open
Abstract
Background Neuronal ceroid lipofuscinoses, (NCLs or Batten disease) are a group of inherited, early onset, fatal neurodegenerative diseases associated with mutations in 13 genes. All forms of the disease are characterized by lysosomal accumulation of fluorescent storage material, as well as profound neurodegeneration, but the relationship of the various genes’ function to a single biological process is not obvious. In this study, we used a well-characterized mouse model of classical late infantile NCL (cLINCL) in which the tripeptidyl peptidase 1 (Tpp1) gene is disrupted by gene targeting, resulting in loss of detectable TPP1 activity and leading to progressive neurological phenotypes including ataxia, increased motor deficiency, and early death. Methods In order to identify genes and pathways that may contribute to progression of the neurodegenerative process, we analyzed forebrain/midbrain and cerebellar transcriptional differences at 1, 2, 3 and 4 months of age in control and TPP1-deficient mice by global RNA-sequencing. Results Progressive neurodegenerative inflammatory responses involving microglia, astrocytes and endothelial cells were observed, accompanied by activation of leukocyte extravasation signals and upregulation of nitric oxide production and reactive oxygen species. Several astrocytic (i.e., Gfap, C4b, Osmr, Serpina3n) and microglial (i.e., Ctss, Itgb2, Itgax, Lyz2) genes were identified as strong markers for assessing disease progression as they showed increased levels of expression in vivo over time. Furthermore, transient increased expression of choroid plexus genes was observed at 2 months in the lateral and fourth ventricle, highlighting an early role for the choroid plexus and cerebrospinal fluid in the disease pathology. Based on these gene expression changes, we concluded that neuroinflammation starts, for the most part, after 2 months in the Tpp1−/− brain and that activation of microglia and astrocytes occur more rapidly in cerebellum than in the rest of the brain; confirming increased severity of inflammation in this region. Conclusions These findings have led to a better understanding of cLINCL pathological onset and progression, which may aid in development of future therapeutic treatments for this disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02302-z.
Collapse
Affiliation(s)
- Miriam S Domowicz
- Department of Pediatrics, Biological Sciences Division, The University of Chicago Medical Center, 5841 S. Maryland Avenue, MC 5058, Chicago, IL, 60637, USA.
| | - Wen-Ching Chan
- Center for Research Informatics, Biological Sciences Division, The University of Chicago, Chicago, IL, 60637, USA
| | - Patricia Claudio-Vázquez
- Department of Pediatrics, Biological Sciences Division, The University of Chicago Medical Center, 5841 S. Maryland Avenue, MC 5058, Chicago, IL, 60637, USA
| | - Tatiana Gonzalez
- Department of Pediatrics, Biological Sciences Division, The University of Chicago Medical Center, 5841 S. Maryland Avenue, MC 5058, Chicago, IL, 60637, USA
| | - Nancy B Schwartz
- Department of Pediatrics, Biological Sciences Division, The University of Chicago Medical Center, 5841 S. Maryland Avenue, MC 5058, Chicago, IL, 60637, USA.,Department of Biochemistry and Molecular Biology, Biological Sciences Division, The University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
26
|
Mayrhofer F, Dariychuk Z, Zhen A, Daugherty DJ, Bannerman P, Hanson AM, Pleasure D, Soulika A, Deng W, Chechneva OV. Reduction in CD11c + microglia correlates with clinical progression in chronic experimental autoimmune demyelination. Neurobiol Dis 2021; 161:105556. [PMID: 34752925 DOI: 10.1016/j.nbd.2021.105556] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 11/01/2021] [Accepted: 11/04/2021] [Indexed: 12/25/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune demyelinating disease with high variability of clinical symptoms. In most cases MS appears as a relapsing-remitting disease course that at a later stage transitions into irreversible progressive decline of neurologic function. The mechanisms underlying MS progression remain poorly understood. Experimental autoimmune encephalomyelitis (EAE) is an animal model of MS. Here we demonstrate that mice that develop mild EAE after immunization with myelin oligodendrocyte glycoprotein 35-55 are prone to undergo clinical progression around 30 days after EAE induction. EAE progression was associated with reduction in CD11c+ microglia and dispersed coalescent parenchymal infiltration. We found sex-dependent differences mediated by p38α signaling, a key regulator of inflammation. Selective reduction of CD11c+ microglia in female mice with CD11c-promoter driven p38α knockout correlated with increased rate of EAE progression. In protected animals, we found CD11c+ microglia forming contacts with astrocyte processes at the glia limitans and immune cells retained within perivascular spaces. Together, our study identified pathological hallmarks of chronic EAE progression and suggests that CD11c+ microglia may regulate immune cell parenchymal infiltration in autoimmune demyelination.
Collapse
Affiliation(s)
- Florian Mayrhofer
- IPRM, Shriners Hospital for Children, Sacramento, CA 95817, United States of America
| | - Zhanna Dariychuk
- IPRM, Shriners Hospital for Children, Sacramento, CA 95817, United States of America
| | - Anthony Zhen
- IPRM, Shriners Hospital for Children, Sacramento, CA 95817, United States of America
| | - Daniel J Daugherty
- Department of Biochemistry and Molecular Medicine, UC Davis, Sacramento, CA 95817, United States of America; IPRM, Shriners Hospital for Children, Sacramento, CA 95817, United States of America
| | - Peter Bannerman
- IPRM, Shriners Hospital for Children, Sacramento, CA 95817, United States of America
| | - Angela M Hanson
- IPRM, Shriners Hospital for Children, Sacramento, CA 95817, United States of America
| | - David Pleasure
- IPRM, Shriners Hospital for Children, Sacramento, CA 95817, United States of America
| | - Athena Soulika
- IPRM, Shriners Hospital for Children, Sacramento, CA 95817, United States of America
| | - Wenbin Deng
- Department of Biochemistry and Molecular Medicine, UC Davis, Sacramento, CA 95817, United States of America; IPRM, Shriners Hospital for Children, Sacramento, CA 95817, United States of America
| | - Olga V Chechneva
- Department of Biochemistry and Molecular Medicine, UC Davis, Sacramento, CA 95817, United States of America; IPRM, Shriners Hospital for Children, Sacramento, CA 95817, United States of America.
| |
Collapse
|
27
|
Ott M, Prins RM, Heimberger AB. The immune landscape of common CNS malignancies: implications for immunotherapy. Nat Rev Clin Oncol 2021; 18:729-744. [PMID: 34117475 PMCID: PMC11090136 DOI: 10.1038/s41571-021-00518-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2021] [Indexed: 02/06/2023]
Abstract
Immunotherapy has enabled remarkable therapeutic responses across cancers of various lineages, albeit with some notable exceptions such as glioblastoma. Several previous misconceptions, which have impaired progress in the past, including the presence and role of the blood-brain barrier and a lack of lymphatic drainage, have been refuted. Nonetheless, a subset of patients with brain metastases but, paradoxically, not the vast majority of those with gliomas are able to respond to immune-checkpoint inhibitors. Immune profiling of samples obtained from patients with central nervous system malignancies using techniques such as mass cytometry and single-cell sequencing along with experimental data from genetically engineered mouse models have revealed fundamental differences in immune composition and immunobiology that not only explain the differences in responsiveness to these agents but also lay the foundations for immunotherapeutic strategies that are applicable to gliomas. Herein, we review the emerging data on the differences in immune cell composition, function and interactions within central nervous system tumours and provide guidance on the development of novel immunotherapies for these historically difficult-to-treat cancers.
Collapse
Affiliation(s)
- Martina Ott
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Robert M Prins
- Departments of Neurosurgery and Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Amy B Heimberger
- Department of Neurosurgery, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
28
|
Cornell J, Salinas S, Huang HY, Zhou M. Microglia regulation of synaptic plasticity and learning and memory. Neural Regen Res 2021; 17:705-716. [PMID: 34472455 PMCID: PMC8530121 DOI: 10.4103/1673-5374.322423] [Citation(s) in RCA: 209] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Microglia are the resident macrophages of the central nervous system. Microglia possess varied morphologies and functions. Under normal physiological conditions, microglia mainly exist in a resting state and constantly monitor their microenvironment and survey neuronal and synaptic activity. Through the C1q, C3 and CR3 “Eat Me” and CD47 and SIRPα “Don’t Eat Me” complement pathways, as well as other pathways such as CX3CR1 signaling, resting microglia regulate synaptic pruning, a process crucial for the promotion of synapse formation and the regulation of neuronal activity and synaptic plasticity. By mediating synaptic pruning, resting microglia play an important role in the regulation of experience-dependent plasticity in the barrel cortex and visual cortex after whisker removal or monocular deprivation, and also in the regulation of learning and memory, including the modulation of memory strength, forgetfulness, and memory quality. As a response to brain injury, infection or neuroinflammation, microglia become activated and increase in number. Activated microglia change to an amoeboid shape, migrate to sites of inflammation and secrete proteins such as cytokines, chemokines and reactive oxygen species. These molecules released by microglia can lead to synaptic plasticity and learning and memory deficits associated with aging, Alzheimer’s disease, traumatic brain injury, HIV-associated neurocognitive disorder, and other neurological or mental disorders such as autism, depression and post-traumatic stress disorder. With a focus mainly on recently published literature, here we reviewed the studies investigating the role of resting microglia in synaptic plasticity and learning and memory, as well as how activated microglia modulate disease-related plasticity and learning and memory deficits. By summarizing the function of microglia in these processes, we aim to provide an overview of microglia regulation of synaptic plasticity and learning and memory, and to discuss the possibility of microglia manipulation as a therapeutic to ameliorate cognitive deficits associated with aging, Alzheimer’s disease, traumatic brain injury, HIV-associated neurocognitive disorder, and mental disorders.
Collapse
Affiliation(s)
- Jessica Cornell
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Shelbi Salinas
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Hou-Yuan Huang
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Miou Zhou
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, USA
| |
Collapse
|
29
|
Delahaye-Duriez A, Dufour A, Bokobza C, Gressens P, Van Steenwinckel J. Targeting Microglial Disturbances to Protect the Brain From Neurodevelopmental Disorders Associated With Prematurity. J Neuropathol Exp Neurol 2021; 80:634-648. [PMID: 34363661 DOI: 10.1093/jnen/nlab049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Microglial activation during critical phases of brain development can result in short- and long-term consequences for neurological and psychiatric health. Several studies in humans and rodents have shown that microglial activation, leading to a transition from the homeostatic state toward a proinflammatory phenotype, has adverse effects on the developing brain and neurodevelopmental disorders. Targeting proinflammatory microglia may be an effective strategy for protecting the brain and attenuating neurodevelopmental disorders induced by inflammation. In this review we focus on the role of inflammation and the activation of immature microglia (pre-microglia) soon after birth in prematurity-associated neurodevelopmental disorders, and the specific features of pre-microglia during development. We also highlight the relevance of immunomodulatory strategies for regulating activated microglia in a rodent model of perinatal brain injury. An original neuroprotective approach involving a nanoparticle-based therapy and targeting microglia, with the aim of improving myelination and protecting the developing brain, is also addressed.
Collapse
Affiliation(s)
- Andrée Delahaye-Duriez
- From the NeuroDiderot, UMR 1141, Inserm, Université de Paris, Paris, France.,UFR SMBH, Université Sorbonne Paris Nord, Bobigny, France.,Assistance Publique des Hôpitaux de Paris, Hôpital Jean Verdier, Service d'Histologie-Embryologie-Cytogénétique, Bondy, France
| | - Adrien Dufour
- From the NeuroDiderot, UMR 1141, Inserm, Université de Paris, Paris, France
| | - Cindy Bokobza
- From the NeuroDiderot, UMR 1141, Inserm, Université de Paris, Paris, France
| | - Pierre Gressens
- From the NeuroDiderot, UMR 1141, Inserm, Université de Paris, Paris, France
| | | |
Collapse
|
30
|
Burns JC, Ransohoff RM, Mingueneau M. Isolation of Microglia and Analysis of Protein Expression by Flow Cytometry: Avoiding the Pitfall of Microglia Background Autofluorescence. Bio Protoc 2021; 11:e4091. [PMID: 34395729 DOI: 10.21769/bioprotoc.4091] [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: 11/23/2020] [Revised: 04/18/2021] [Accepted: 05/27/2021] [Indexed: 11/02/2022] Open
Abstract
Microglia are a unique type of tissue-resident innate immune cell found within the brain, spinal cord, and retina. In the healthy nervous system, their main functions are to defend the tissue against infectious microbes, support neuronal networks through synapse remodeling, and clear extracellular debris and dying cells through phagocytosis. Many existing microglia isolation protocols require the use of enzymatic tissue digestion or magnetic bead-based isolation steps, which increase both the time and cost of these procedures and introduce variability to the experiment. Here, we report a protocol to generate single-cell suspensions from freshly harvested murine brains or spinal cords, which efficiently dissociates tissue and removes myelin debris through simple mechanical dissociation and density centrifugation and can be applied to rat and non-human primate tissues. We further describe the importance of including empty channels in downstream flow cytometry analyses of microglia single-cell suspensions to accurately assess the expression of protein targets in this highly autofluorescent cell type. This methodology ensures that observed fluorescence signals are not incorrectly attributed to the protein target of interest by appropriately taking into account the unique autofluorescence of this cell type, a phenomenon already present in young animals and that increases with aging to levels that are comparable to those observed with antibodies against highly abundant antigens.
Collapse
Affiliation(s)
- Jeremy C Burns
- Multiple Sclerosis and Neurorepair Research Unit, Biogen, Cambridge, USA.,Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, USA
| | | | - Michaël Mingueneau
- Multiple Sclerosis and Neurorepair Research Unit, Biogen, Cambridge, USA
| |
Collapse
|
31
|
Rehman R, Tar L, Olamide AJ, Li Z, Kassubek J, Böckers T, Weishaupt J, Ludolph A, Wiesner D, Roselli F. Acute TBK1/IKK-ε Inhibition Enhances the Generation of Disease-Associated Microglia-Like Phenotype Upon Cortical Stab-Wound Injury. Front Aging Neurosci 2021; 13:684171. [PMID: 34326766 PMCID: PMC8313992 DOI: 10.3389/fnagi.2021.684171] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/21/2021] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury has a poorer prognosis in elderly patients, possibly because of the enhanced inflammatory response characteristic of advanced age, known as “inflammaging.” Recently, reduced activation of the TANK-Binding-Kinase 1 (Tbk1) pathway has been linked to age-associated neurodegeneration and neuroinflammation. Here we investigated how the blockade of Tbk1 and of the closely related IKK-ε by the small molecule Amlexanox could modify the microglial and immune response to cortical stab-wound injury in mice. We demonstrated that Tbk1/IKK-ε inhibition resulted in a massive expansion of microglial cells characterized by the TMEM119+/CD11c+ phenotype, expressing high levels of CD68 and CD317, and with the upregulation of Cst7a, Prgn and Ccl4 and the decrease in the expression levels of Tmem119 itself and P2yr12, thus a profile close to Disease-Associated Microglia (DAM, a subset of reactive microglia abundant in Alzheimer’s Disease and other neurodegenerative conditions). Furthermore, Tbk1/IKK-ε inhibition increased the infiltration of CD3+ lymphocytes, CD169+ macrophages and CD11c+/CD169+ cells. The enhanced immune response was associated with increased expression of Il-33, Ifn-g, Il-17, and Il-19. This upsurge in the response to the stab wound was associated with the expanded astroglial scars and increased deposition of chondroitin-sulfate proteoglycans at 7 days post injury. Thus, Tbk1/IKK-ε blockade results in a massive expansion of microglial cells with a phenotype resembling DAM and with the substantial enhancement of neuroinflammatory responses. In this context, the induction of DAM is associated with a detrimental outcome such as larger injury-related glial scars. Thus, the Tbk1/IKK-ε pathway is critical to repress neuroinflammation upon stab-wound injury and Tbk1/IKK-ε inhibitors may provide an innovative approach to investigate the consequences of DAM induction.
Collapse
Affiliation(s)
- Rida Rehman
- Department of Neurology, Ulm University, Ulm, Germany
| | - Lilla Tar
- Department of Neurology, Ulm University, Ulm, Germany.,German Center for Neurodegenerative Diseases (DZNE)-Ulm, Ulm, Germany
| | - Adeyemi Jubril Olamide
- Department of Neurology, Ulm University, Ulm, Germany.,Master in Translational and Molecular Neuroscience, Ulm University, Ulm, Germany
| | - Zhenghui Li
- Department of Neurology, Ulm University, Ulm, Germany.,Department of Neurosurgery, Kaifeng Central Hospital, Kaifeng, China
| | - Jan Kassubek
- Department of Neurology, Ulm University, Ulm, Germany
| | - Tobias Böckers
- Institute of Anatomy and Cell Biology, Ulm University, Ulm, Germany.,Neurozentrum Ulm, Ulm, Germany
| | - Jochen Weishaupt
- Department of Neurology, Ulm University, Ulm, Germany.,Neurozentrum Ulm, Ulm, Germany
| | - Albert Ludolph
- Department of Neurology, Ulm University, Ulm, Germany.,German Center for Neurodegenerative Diseases (DZNE)-Ulm, Ulm, Germany.,Neurozentrum Ulm, Ulm, Germany
| | - Diana Wiesner
- Department of Neurology, Ulm University, Ulm, Germany.,German Center for Neurodegenerative Diseases (DZNE)-Ulm, Ulm, Germany.,Neurozentrum Ulm, Ulm, Germany
| | - Francesco Roselli
- Department of Neurology, Ulm University, Ulm, Germany.,German Center for Neurodegenerative Diseases (DZNE)-Ulm, Ulm, Germany.,Neurozentrum Ulm, Ulm, Germany
| |
Collapse
|
32
|
An L, Shen Y, Chopp M, Zacharek A, Venkat P, Chen Z, Li W, Qian Y, Landschoot-Ward J, Chen J. Deficiency of Endothelial Nitric Oxide Synthase (eNOS) Exacerbates Brain Damage and Cognitive Deficit in A Mouse Model of Vascular Dementia. Aging Dis 2021; 12:732-746. [PMID: 34094639 PMCID: PMC8139201 DOI: 10.14336/ad.2020.0523] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 05/23/2020] [Indexed: 12/19/2022] Open
Abstract
Vascular Dementia (VaD) accounts for nearly 20% of all cases of dementia. eNOS plays an important role in neurovascular remodeling, anti-inflammation, and cognitive functional recovery after stroke. In this study, we investigated whether eNOS regulates brain damage, cognitive function in mouse model of bilateral common carotid artery stenosis (BCAS) induced VaD. Late-adult (6-8 months) C57BL/6J and eNOS knockout (eNOS-/-) mice were subjected to BCAS (n=12/group) or sham group (n=8/group). BCAS was performed by applying microcoils to both common carotid arteries. Cerebral blood flow (CBF) and blood pressure were measured. A battery of cognitive functional tests was performed, and mice were sacrificed 30 days after BCAS. Compared to corresponding sham mice, BCAS in wild-type (WT) and eNOS-/- mice significantly: 1) induces short term, long term memory loss, spatial learning and memory deficits; 2) decreases CBF, increases ischemic cell damage, including apoptosis, white matter (WM) and axonal damage; 3) increases blood brain barrier (BBB) leakage, decreases aquaporin-4 (AQP4) expression and vessel density; 4) increases microglial, astrocyte activation and oxidative stress in the brain; 5) increases inflammatory factor interleukin-1 receptor-associated kinase-1(IRAK-1) and amyloid beta (Aβ) expression in brain; 6) increases IL-6 and IRAK4 expression in brain. eNOS-/-sham mice exhibit increased blood pressure, decreased iNOS and nNOS in brain compared to WT-sham mice. Compared to WT-BCAS mice, eNOS-/-BCAS mice exhibit worse vascular and WM/axonal damage, increased BBB leakage and inflammatory response, increased cognitive deficit, decreased iNOS, nNOS in brain. eNOS deficit exacerbates BCAS induced brain damage and cognitive deficit.
Collapse
Affiliation(s)
- Lulu An
- 1Department of Neurology, Henry Ford Hospital, Detroit, MI-48202, USA
| | - Yi Shen
- 1Department of Neurology, Henry Ford Hospital, Detroit, MI-48202, USA.,2Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China (Current address)
| | - Michael Chopp
- 1Department of Neurology, Henry Ford Hospital, Detroit, MI-48202, USA.,3Department of Physics, Oakland University, Rochester, MI-48309, USA
| | - Alex Zacharek
- 1Department of Neurology, Henry Ford Hospital, Detroit, MI-48202, USA
| | - Poornima Venkat
- 1Department of Neurology, Henry Ford Hospital, Detroit, MI-48202, USA
| | - Zhili Chen
- 1Department of Neurology, Henry Ford Hospital, Detroit, MI-48202, USA
| | - Wei Li
- 1Department of Neurology, Henry Ford Hospital, Detroit, MI-48202, USA
| | - Yu Qian
- 1Department of Neurology, Henry Ford Hospital, Detroit, MI-48202, USA
| | | | - Jieli Chen
- 1Department of Neurology, Henry Ford Hospital, Detroit, MI-48202, USA
| |
Collapse
|
33
|
Keane L, Cheray M, Blomgren K, Joseph B. Multifaceted microglia - key players in primary brain tumour heterogeneity. Nat Rev Neurol 2021; 17:243-259. [PMID: 33692572 DOI: 10.1038/s41582-021-00463-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2021] [Indexed: 01/31/2023]
Abstract
Microglia are the resident innate immune cells of the immune-privileged CNS and, as such, represent the first line of defence against tissue injury and infection. Given their location, microglia are undoubtedly the first immune cells to encounter a developing primary brain tumour. Our knowledge of these cells is therefore important to consider in the context of such neoplasms. As the heterogeneous nature of the most aggressive primary brain tumours is thought to underlie their poor prognosis, this Review places a special emphasis on the heterogeneity of the tumour-associated microglia and macrophage populations present in primary brain tumours. Where available, specific information on microglial heterogeneity in various types and subtypes of brain tumour is included. Emerging evidence that highlights the importance of considering the heterogeneity of both the tumour and of microglial populations in providing improved treatment outcomes for patients is also discussed.
Collapse
Affiliation(s)
- Lily Keane
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm, Sweden
| | - Mathilde Cheray
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm, Sweden
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden.,Department of Paediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Bertrand Joseph
- Institute of Environmental Medicine, Toxicology Unit, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
34
|
IFN-γ regulates the transformation of microglia into dendritic-like cells via the ERK/c-myc signaling pathway during cerebral ischemia/reperfusion in mice. Neurochem Int 2020; 141:104860. [DOI: 10.1016/j.neuint.2020.104860] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/13/2020] [Accepted: 09/29/2020] [Indexed: 12/15/2022]
|
35
|
Owens T, Benmamar-Badel A, Wlodarczyk A, Marczynska J, Mørch MT, Dubik M, Arengoth DS, Asgari N, Webster G, Khorooshi R. Protective roles for myeloid cells in neuroinflammation. Scand J Immunol 2020; 92:e12963. [PMID: 32851668 DOI: 10.1111/sji.12963] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/14/2020] [Accepted: 08/19/2020] [Indexed: 12/20/2022]
Abstract
Myeloid cells represent the major cellular component of innate immune responses. Myeloid cells include monocytes and macrophages, granulocytes (neutrophils, basophils and eosinophils) and dendritic cells (DC). The role of myeloid cells has been broadly described both in physiological and in pathological conditions. All tissues or organs are equipped with resident myeloid cells, such as parenchymal microglia in the brain, which contribute to maintaining homeostasis. Moreover, in case of infection or tissue damage, other myeloid cells such as monocytes or granulocytes (especially neutrophils) can be recruited from the circulation, at first to promote inflammation and later to participate in repair and regeneration. This review aims to address the regulatory roles of myeloid cells in inflammatory diseases of the central nervous system (CNS), with a particular focus on recent work showing induction of suppressive function via stimulation of innate signalling in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE).
Collapse
Affiliation(s)
- Trevor Owens
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark
| | - Anouk Benmamar-Badel
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark.,Department of Neurology, Slagelse Hospital, Slagelse, Denmark
| | - Agnieszka Wlodarczyk
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark
| | - Joanna Marczynska
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark
| | - Marlene T Mørch
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark
| | - Magdalena Dubik
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark
| | - Dina S Arengoth
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark
| | - Nasrin Asgari
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark.,Department of Neurology, Slagelse Hospital, Slagelse, Denmark
| | - Gill Webster
- Innate Immunotherapeutics, Auckland, New Zealand
| | - Reza Khorooshi
- Neurobiology Research, Institute of Molecular Medicine, and BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense C, Denmark
| |
Collapse
|
36
|
Nishimura T, Saito Y, Washio K, Komori S, Respatika D, Kotani T, Murata Y, Ohnishi H, Mizobuchi S, Matozaki T. SIRPα on CD11c + cells induces Th17 cell differentiation and subsequent inflammation in the CNS in experimental autoimmune encephalomyelitis. Eur J Immunol 2020; 50:1560-1570. [PMID: 32438469 DOI: 10.1002/eji.201948410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 04/12/2020] [Indexed: 01/06/2023]
Abstract
Signal regulatory protein α (SIRPα) is expressed predominantly on type 2 conventional dendritic cells (cDC2s) and macrophages. We previously showed that mice systemically lacking SIRPα were resistant to experimental autoimmune encephalomyelitis (EAE). Here, we showed that deletion of SIRPα in CD11c+ cells of mice (SirpaΔDC mice) also markedly ameliorated the development of EAE. The frequency of cDCs and migratory DCs (mDCs), as well as that of Th17 cells, were significantly reduced in draining lymph nodes of SirpaΔDC mice at the onset of EAE. In addition, we found the marked reduction in the number of Th17 cells and DCs in the CNS of SirpaΔDC mice at the peak of EAE. Whereas inducible systemic ablation of SIRPα before the induction of EAE prevented disease development, that after EAE onset did not ameliorate the clinical signs of disease. We also found that EAE development was partially attenuated in mice with CD11c+ cell-specific ablation of CD47, a ligand of SIRPα. Collectively, our results suggest that SIRPα expressed on CD11c+ cells, such as cDC2s and mDCs, is indispensable for the development of EAE, being required for the priming of self-reactive Th17 cells in the periphery as well as for the inflammation in the CNS.
Collapse
Affiliation(s)
- Taichi Nishimura
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan.,Division of Anesthesiology, Department of Surgery Related, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | - Yasuyuki Saito
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | - Ken Washio
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | - Satomi Komori
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | - Datu Respatika
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan.,Division of Reconstruction, Oculoplasty, and Oncology, Department of Ophthalmology, Faculty of Medicine, Public Health, and Nursing, Gadjah Mada University, Yogyakarta, Indonesia
| | - Takenori Kotani
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | - Yoji Murata
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | - Hiroshi Ohnishi
- Department of Laboratory Sciences, Gunma University Graduate School of Health Sciences, Gunma, Japan
| | - Satoshi Mizobuchi
- Division of Anesthesiology, Department of Surgery Related, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| | - Takashi Matozaki
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Japan
| |
Collapse
|
37
|
Tribble JR, Harder JM, Williams PA, John SWM. Ocular hypertension suppresses homeostatic gene expression in optic nerve head microglia of DBA/2 J mice. Mol Brain 2020; 13:81. [PMID: 32450896 PMCID: PMC7249412 DOI: 10.1186/s13041-020-00603-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 04/13/2020] [Indexed: 12/18/2022] Open
Abstract
Glaucoma is the leading cause of irreversible vision loss. Ocular hypertension is a major risk factor for glaucoma and recent work has demonstrated critical early neuroinflammatory insults occur in the optic nerve head following ocular hypertension. Microglia and infiltrating monocytes are likely candidates to drive these neuroinflammatory insults. However, the exact molecular identity / transcriptomic profile of microglia following ocular hypertensive insults is unknown. To elucidate the molecular identity of microglia after long-term exposure to ocular hypertension, we used a mouse model of glaucoma (DBA/2 J). We performed RNA-sequencing of microglia mRNA from the optic nerve head at a time point following ocular hypertensive insults, but preceding detectable neurodegeneration (with microglia identified as being CD45lo/CD11b+/CD11c−). Furthermore, RNA-sequencing was performed on optic nerve head microglia from mice treated with radiation therapy, a potent therapy preventing neuroinflammatory insults. Transcriptomic profiling of optic nerve head microglia mRNA identifies metabolic priming with marked changes in mitochondrial gene expression, and changes to phagocytosis, inflammatory, and sensome pathways. The data predict that many functions of microglia that help maintain tissue homeostasis are affected. Comparative analysis of these data with data from previously published whole optic nerve head tissue or monocyte-only samples from DBA/2 J mice demonstrate that many of the neuroinflammatory signatures in these data sets arise from infiltrating monocytes and not reactive microglia. Finally, our data demonstrate that prophylactic radiation therapy of DBA/2 J mice potently abolishes these microglia metabolic transcriptomic changes at the same time points. Together, our data provide a unique resource for the community to help drive further hypothesis generation and testing in glaucoma.
Collapse
Affiliation(s)
- James R Tribble
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Jeffrey M Harder
- The Howard Hughes Medical Institute, The Jackson Laboratory, Bar Harbor, ME, USA
| | - Pete A Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden.
| | - Simon W M John
- The Howard Hughes Medical Institute, The Jackson Laboratory, Bar Harbor, ME, USA. .,Department of Ophthalmology and Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA.
| |
Collapse
|
38
|
Benmamar-Badel A, Owens T, Wlodarczyk A. Protective Microglial Subset in Development, Aging, and Disease: Lessons From Transcriptomic Studies. Front Immunol 2020; 11:430. [PMID: 32318054 PMCID: PMC7147523 DOI: 10.3389/fimmu.2020.00430] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/25/2020] [Indexed: 12/25/2022] Open
Abstract
Microglial heterogeneity has been the topic of much discussion in the scientific community. Elucidation of their plasticity and adaptability to disease states triggered early efforts to characterize microglial subsets. Over time, their phenotypes, and later on their homeostatic signature, were revealed, through the use of increasingly advanced transcriptomic techniques. Recently, an increasing number of these "microglial signatures" have been reported in various homeostatic and disease contexts. Remarkably, many of these states show similar overlapping microglial gene expression patterns, both in homeostasis and in disease or injury. In this review, we integrate information from these studies, and we propose a unique subset, for which we introduce a core signature, based on our own research and reports from the literature. We describe that this subset is found in development and in normal aging as well as in diverse diseases. We discuss the functions of this subset as well as how it is induced.
Collapse
Affiliation(s)
- Anouk Benmamar-Badel
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
- BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark
- Department of Neurology, Slagelse Hospital, Institute of Regional Health Research, Slagelse, Denmark
| | - Trevor Owens
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
- BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark
| | - Agnieszka Wlodarczyk
- Department of Neurobiology Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
- BRIDGE, Brain Research - Inter-Disciplinary Guided Excellence, Odense, Denmark
| |
Collapse
|
39
|
Deftu AF, Suter MR. Glia and Pain in Spinal Cord. THE SENSES: A COMPREHENSIVE REFERENCE 2020:235-248. [DOI: 10.1016/b978-0-12-809324-5.24214-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
40
|
Abstract
Microglia are increasingly shown to be key players in neuron development and synapse connectivity. However, the underlying mechanisms by which microglia regulate neuron function remain poorly understood in part because such analysis is challenging in the brain where neurons and synapses are intermingled and connectivity is only beginning to be mapped. Here, we discuss the features and function of microglia in the ordered mammalian retina where the laminar organization of neurons and synapses facilitates such molecular studies. We discuss microglia origins and consider the evidence for molecularly distinct microglia subpopulations and their potential for differential roles with a particular focus on the early stages of retina development. We then review the models and methods used for the study of these cells and discuss emerging data that link retina microglia to the genesis and survival of particular retina cell subtypes. We also highlight potential roles for microglia in shaping the development and organization of the vasculature and discuss cellular and molecular mechanisms involved in this process. Such insights may help resolve the mechanisms by which retinal microglia impact visual function and help guide studies of related features in brain development and disease.
Collapse
Affiliation(s)
- Fenge Li
- Department of Neuroscience, Huffington Center on Aging, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Danye Jiang
- Department of Neuroscience, Huffington Center on Aging, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Melanie A Samuel
- Department of Neuroscience, Huffington Center on Aging, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
41
|
Lee J, Hamanaka G, Lo EH, Arai K. Heterogeneity of microglia and their differential roles in white matter pathology. CNS Neurosci Ther 2019; 25:1290-1298. [PMID: 31733036 PMCID: PMC6887901 DOI: 10.1111/cns.13266] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/12/2022] Open
Abstract
Microglia are resident immune cells that play multiple roles in central nervous system (CNS) development and disease. Although the classical concept of microglia/macrophage activation is based on a biphasic beneficial‐versus‐deleterious polarization, growing evidence now suggests a much more heterogenous profile of microglial activation that underlie their complex roles in the CNS. To date, the majority of data are focused on microglia in gray matter. However, demyelination is a prominent pathologic finding in a wide range of diseases including multiple sclerosis, Alzheimer's disease, and vascular cognitive impairment and dementia. In this mini‐review, we discuss newly discovered functional subsets of microglia that contribute to white matter response in CNS disease onset and progression. Microglia show different molecular patterns and morphologies depending on disease type and brain region, especially in white matter. Moreover, in later stages of disease, microglia demonstrate unconventional immuno‐regulatory activities such as increased phagocytosis of myelin debris and secretion of trophic factors that stimulate oligodendrocyte lineage cells to facilitate remyelination and disease resolution. Further investigations of these multiple microglia subsets may lead to novel therapeutic approaches to treat white matter pathology in CNS injury and disease.
Collapse
Affiliation(s)
- Janice Lee
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Gen Hamanaka
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
42
|
Stratoulias V, Venero JL, Tremblay MÈ, Joseph B. Microglial subtypes: diversity within the microglial community. EMBO J 2019. [PMID: 31373067 DOI: 10.15252/embj.2019a101997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
Abstract
Microglia are brain-resident macrophages forming the first active immune barrier in the central nervous system. They fulfill multiple functions across development and adulthood and under disease conditions. Current understanding revolves around microglia acquiring distinct phenotypes upon exposure to extrinsic cues in their environment. However, emerging evidence suggests that microglia display differences in their functions that are not exclusively driven by their milieu, rather by the unique properties these cells possess. This microglial intrinsic heterogeneity has been largely overlooked, favoring the prevailing view that microglia are a single-cell type endowed with spectacular plasticity, allowing them to acquire multiple phenotypes and thereby fulfill their numerous functions in health and disease. Here, we review the evidence that microglia might form a community of cells in which each member (or "subtype") displays intrinsic properties and performs unique functions. Distinctive features and functional implications of several microglial subtypes are considered, across contexts of health and disease. Finally, we suggest that microglial subtype categorization shall be based on function and we propose ways for studying them. Hence, we advocate that plasticity (reaction states) and diversity (subtypes) should both be considered when studying the multitasking microglia.
Collapse
Affiliation(s)
- Vassilis Stratoulias
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jose Luis Venero
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain.,Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Marie-Ève Tremblay
- Department of Molecular Medicine, Université Laval, Quebec, QC, Canada.,Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Quebec, QC, Canada
| | - Bertrand Joseph
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
43
|
Stratoulias V, Venero JL, Tremblay M, Joseph B. Microglial subtypes: diversity within the microglial community. EMBO J 2019; 38:e101997. [PMID: 31373067 PMCID: PMC6717890 DOI: 10.15252/embj.2019101997] [Citation(s) in RCA: 357] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/29/2019] [Accepted: 05/03/2019] [Indexed: 12/03/2022] Open
Abstract
Microglia are brain-resident macrophages forming the first active immune barrier in the central nervous system. They fulfill multiple functions across development and adulthood and under disease conditions. Current understanding revolves around microglia acquiring distinct phenotypes upon exposure to extrinsic cues in their environment. However, emerging evidence suggests that microglia display differences in their functions that are not exclusively driven by their milieu, rather by the unique properties these cells possess. This microglial intrinsic heterogeneity has been largely overlooked, favoring the prevailing view that microglia are a single-cell type endowed with spectacular plasticity, allowing them to acquire multiple phenotypes and thereby fulfill their numerous functions in health and disease. Here, we review the evidence that microglia might form a community of cells in which each member (or "subtype") displays intrinsic properties and performs unique functions. Distinctive features and functional implications of several microglial subtypes are considered, across contexts of health and disease. Finally, we suggest that microglial subtype categorization shall be based on function and we propose ways for studying them. Hence, we advocate that plasticity (reaction states) and diversity (subtypes) should both be considered when studying the multitasking microglia.
Collapse
Affiliation(s)
- Vassilis Stratoulias
- Toxicology UnitInstitute of Environmental MedicineKarolinska InstitutetStockholmSweden
| | - Jose Luis Venero
- Departamento de Bioquímica y Biología MolecularFacultad de FarmaciaUniversidad de SevillaSevillaSpain
- Instituto de Biomedicina de Sevilla‐Hospital Universitario Virgen del Rocío/CSIC/Universidad de SevillaSevillaSpain
| | - Marie‐Ève Tremblay
- Department of Molecular MedicineUniversité LavalQuebecQCCanada
- Axe NeurosciencesCentre de Recherche du CHU de Québec‐Université LavalQuebecQCCanada
| | - Bertrand Joseph
- Toxicology UnitInstitute of Environmental MedicineKarolinska InstitutetStockholmSweden
| |
Collapse
|