1
|
Walter E, Angst G, Bollinger J, Truong L, Ware E, Wohleb ES, Fan Y, Wang C. Atg5 in microglia regulates sex-specific effects on postnatal neurogenesis in Alzheimer's disease. NPJ AGING 2025; 11:18. [PMID: 40091054 PMCID: PMC11911432 DOI: 10.1038/s41514-025-00209-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025]
Abstract
Female Alzheimer's disease (AD) patients display greater cognitive deficits and worse AD pathology as compared to male AD patients. In this study, we found that conditional knockout (cKO) of Atg5 in female microglia failed to obtain disease-associated microglia (DAM) gene signatures in familiar AD mouse model (5xFAD). Next, we analyzed the maintenance and neurogenesis of neural stem cells (NSCs) in the hippocampus and subventricular zone (SVZ) from 5xFAD mice with Atg5 cKO. Our data indicated that Atg5 cKO reduced the NSC number in hippocampus of female but not male 5xFAD mice. However, in the SVZ, Atg5 cKO only impaired NSCs in male 5xFAD mice. Interestingly, female 5xFAD;Fip200 cKO mice and 5xFAD;Atg14 cKO mice did not show NSC defects. These autophagy genes cKO 5xFAD mice exhibited a higher neurogenesis activity in their SVZ. Together, our data indicate a sex-specific role for microglial Atg5 in postnatal neurogenesis in AD mice.
Collapse
Affiliation(s)
- Ellen Walter
- Department of Cancer Biology, University of Cincinnati College Medicine, Cincinnati, OH, USA
| | - Gabrielle Angst
- Department of Cancer Biology, University of Cincinnati College Medicine, Cincinnati, OH, USA
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute and College of Medicine at The Ohio State University, Columbus, OH, USA
- Center for Cancer Metabolism, James Comprehensive Cancer Center at The Ohio State University, Columbus, OH, USA
| | - Justin Bollinger
- Department of Pharmacology & Systems Physiology, University of Cincinnati College Medicine, Cincinnati, OH, USA
| | - Linh Truong
- Department of Cancer Biology, University of Cincinnati College Medicine, Cincinnati, OH, USA
| | - Elena Ware
- Department of Cancer Biology, University of Cincinnati College Medicine, Cincinnati, OH, USA
| | - Eric S Wohleb
- Department of Pharmacology & Systems Physiology, University of Cincinnati College Medicine, Cincinnati, OH, USA
| | - Yanbo Fan
- Department of Cancer Biology, University of Cincinnati College Medicine, Cincinnati, OH, USA
| | - Chenran Wang
- Department of Cancer Biology, University of Cincinnati College Medicine, Cincinnati, OH, USA.
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute and College of Medicine at The Ohio State University, Columbus, OH, USA.
- Center for Cancer Metabolism, James Comprehensive Cancer Center at The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
2
|
Fu J, Wang R, He J, Liu X, Wang X, Yao J, Liu Y, Ran C, Ye Q, He Y. Pathogenesis and therapeutic applications of microglia receptors in Alzheimer's disease. Front Immunol 2025; 16:1508023. [PMID: 40028337 PMCID: PMC11867950 DOI: 10.3389/fimmu.2025.1508023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Microglia, the resident immune cells of the central nervous system, continuously monitor the brain's microenvironment through their array of specific receptors. Once brain function is altered, microglia are recruited to specific sites to perform their immune functions, including phagocytosis of misfolded proteins, cellular debris, and apoptotic cells to maintain homeostasis. When toxic substances are overproduced, microglia are over-activated to produce large amounts of pro-inflammatory cytokines, which induce chronic inflammatory responses and lead to neurotoxicity. Additionally, microglia can also monitor and protect neuronal function through microglia-neuron crosstalk. Microglia receptors are important mediators for microglia to receive external stimuli, regulate the functional state of microglia, and transmit signals between cells. In this paper, we first review the role of microglia-expressed receptors in the pathogenesis and treatment of Alzheimer's disease; moreover, we emphasize the complexity of targeting microglia for therapeutic interventions in neurodegenerative disorders to inform the discovery of new biomarkers and the development of innovative therapeutics.
Collapse
Affiliation(s)
- Jiao Fu
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - RuoXuan Wang
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - JiHui He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - XiaoJing Liu
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - XinXin Wang
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - JuMing Yao
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
| | - Ye Liu
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - ChongZhao Ran
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - QingSong Ye
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yan He
- Institute of Regenerative and Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
- First Clinical College, Wuhan University of Science and Technology, Wuhan, China
- Department of Stomatology, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
3
|
Tamatta R, Pai V, Jaiswal C, Singh I, Singh AK. Neuroinflammaging and the Immune Landscape: The Role of Autophagy and Senescence in Aging Brain. Biogerontology 2025; 26:52. [PMID: 39907842 PMCID: PMC11799035 DOI: 10.1007/s10522-025-10199-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: 12/14/2024] [Accepted: 01/29/2025] [Indexed: 02/06/2025]
Abstract
Neuroinflammation is closely linked to aging, which damages the structure and function of the brain. It is caused by the intricate interactions of immune cells in the aged brain, such as the dysregulated glial cells and the dysfunctional astrocytes. Aging-associated chronic low inflammation, referred to as neuroinflammaging, shows an upregulated proinflammatory response. Autophagy and senescence play crucial roles as moderators of aging and neuroinflammatory responses. The dysregulated neuroimmune system, dystrophic glial cells, and release of proinflammatory factors alter blood-brain barrier, causing a neuroinflammatory landscape. Chronic inflammation combined with deteriorating neurons exacerbate neurological disorders and decline in cognitive function. This review highlights the neuroinflammaging and mechanism associated with immune cells interplay with central nervous system and aging, cellular senescence, and autophagy regulation in the brain's immune system under neuroinflammatory conditions. Moreover, the roles of microglia and peripheral immune cells in the neuroinflammatory process in the aging brain have also been discussed. Determining treatment targets and comprehending mechanisms that influence immune cells in the aged brain is necessary to decrease neuroinflammation.
Collapse
Affiliation(s)
- Rajesh Tamatta
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal, 576104, India
| | - Varsha Pai
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal, 576104, India
| | - Charu Jaiswal
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal, 576104, India
| | - Ishika Singh
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal, 576104, India
| | - Abhishek Kumar Singh
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal, 576104, India.
| |
Collapse
|
4
|
Cai L, Fan Q, Pang R, Chen C, Zhang Y, Xie H, Huang J, Wang Y, Li P, Huang D, Jin X, Zhou Y, Li Y. Microglia programmed cell death in neurodegenerative diseases and CNS injury. Apoptosis 2025; 30:446-465. [PMID: 39656359 PMCID: PMC11799081 DOI: 10.1007/s10495-024-02041-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2024] [Indexed: 02/06/2025]
Abstract
Programmed cell death (PCD) has emerged as a critical regulatory mechanism in the initiation and progression of various pathological conditions. PCD in microglia, including necroptosis, pyroptosis, apoptosis, ferroptosis, and autophagy, occurs in a variety of central nervous system (CNS) diseases. Dysregulation of microglia can lead to excessive tissue damage or neuronal death in CNS injury. Various injury stimuli trigger aberrant activation of the PCD pathway of microglia, which then further leads to inflammatory cascades that exacerbates CNS pathology in a vicious cycle. Therefore, targeting PCD in microglia is considered an important avenue for the treatment of various neurodegenerative diseases and CNS injury. In this review, we summarize the major and recent findings focusing on the mechanisms of PCD in microglia modulating functions in neurodegenerative diseases and CNS injury and provide a systematic overview of the current inhibitors targeting various PCD pathways, which may provide important therapeutic targets that merit further investigation.
Collapse
Affiliation(s)
- Ling Cai
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qiuyue Fan
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Pang
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Chen
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yueman Zhang
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiyi Xie
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingyi Huang
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Wang
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peiying Li
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Clinical Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dan Huang
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xia Jin
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yuxi Zhou
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yan Li
- Department of Anesthesiology, Key Laboratory of the Ministry of Education, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
5
|
Hamagami N, Kapadia D, Abduljawad N, Cheng Z, McLaughlin L, Singhania D, Barclay KM, Yang J, Sun Z, Bayguinov P, Yu G, Gabel HW, Li Q. Microglial plasticity governed by state-specific enhancer landscapes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635595. [PMID: 39975390 PMCID: PMC11838276 DOI: 10.1101/2025.01.30.635595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Single-cell transcriptomic studies have identified distinct microglial subpopulations with shared and divergent gene signatures across development, aging and disease. Whether these microglial subsets represent ontogenically separate lineages of cells, or they are manifestations of plastic changes of microglial states downstream of some converging signals is unknown. Furthermore, despite the well-established role of enhancer landscapes underlying the identity of microglia, to what extent histone modifications and DNA methylation regulate microglial state switches at enhancers have not been defined. Here, using genetic fate mapping, we demonstrate the common embryonic origin of proliferative-region-associated microglia (PAM) enriched in developing white matter, and track their dynamic transitions into disease-associated microglia (DAM) and white matter-associated microglia (WAM) states in disease and aging contexts, respectively. This study links spatiotemporally discrete microglial states through their transcriptomic and epigenomic plasticity, while revealing state-specific histone modification profiles that govern state switches in health and disease.
Collapse
Affiliation(s)
- Nicole Hamagami
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO 63110, USA
- These authors contributed equally
| | - Dvita Kapadia
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- These authors contributed equally
| | - Nora Abduljawad
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Neuroscience Graduate Program, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Zuolin Cheng
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Liam McLaughlin
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Darsh Singhania
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Kia M. Barclay
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Neuroscience Graduate Program, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jin Yang
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Zhixin Sun
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Peter Bayguinov
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Guoqiang Yu
- Department of Automation, Tsinghua University, Beijing 100084, China
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China
| | - Harrison W. Gabel
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- These authors contributed equally
| | - Qingyun Li
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
- Department of Genetics, Washington University School of Medicine in St. Louis, School of Medicine, St. Louis, MO 63110, USA
- These authors contributed equally
- Lead contact
| |
Collapse
|
6
|
Meimei C, Fei Z, Wen X, Huangwei L, Zhenqiang H, Rongjun Y, Qiang Z, Qiuyang L, Xiaozhen L, Yuan Y, Zhaoyang Y, Candong L. Taxus chinensis (Pilg.) Rehder fruit attenuates aging behaviors and neuroinflammation by inhibiting microglia activation via TLR4/NF-κB/NLRP3 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118943. [PMID: 39413938 DOI: 10.1016/j.jep.2024.118943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/20/2024] [Accepted: 10/13/2024] [Indexed: 10/18/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE As one of the important by-products of Taxus chinensis (Pilg.) Rehder, its fruit (TCF) has a sweet taste, which is commonly used in folklore to make health care wine reputed for enhancing immune function and promoting anti-aging effects, especially popular in the longevity villages of China for a long history. Evidences had showed that Taxus chinensis fruit contained polysaccharides, flavonoids, amino acids and terpenoids, which all were free of toxic compounds, but its medicinal value has not been fully recognized. Our previous studies have found that TCF extract may reverse many biological events, including oxidative stress, inflammatory response, neuronal apoptosis, etc. by in silico methods, suggesting potential avenues for future pharmaceutical exploration in aging and age-related diseases. AIM OF THE STUDY Yet, the anti-aging properties of TCF have not been specifically studied, this study aims to fill this gap by investigating the effects of TCF extract (TCFE) in an aging mouse model, particularly focusing on its role in inhibiting microglial activation and elucidating its underlying anti-aging mechanisms. MATERIALS AND METHODS An aging mouse model was induced using D-galactose, with interventions involving high, medium, and low doses of TCFE compared to a positive control (2 mg/kg rapamycin combined with 100 mg/kg metformin). The methodology involved evaluating behavioral changes, serum oxidative and antioxidative markers, hypothalamic β-galactosidase activity, expression of the aging-related protein P63, serum inflammatory factors, and the TLR4/NF-κB/NLRP3 inflammatory pathway in hypothalamic tissues. Additionally, to strengthen our in vivo findings, we conducted in vitro experiments on LPS-stimulated BV2 microglial cells. Finally, UPLC-MS/MS for precise component analysis using compound standards, coupled with molecular docking analyses, were employed to discern and elucidate the anti-inflammatory mechanisms of TCF. RESULTS In vivo results revealed TCFE significantly ameliorated behavioral deficits, reduced oxidative stress markers (MDA) and pro-inflammatory cytokines (IL1-β, IL-6, IFNg, TNFα, IL-17), and increased in antioxidants (SOD, T-AOC) and anti-inflammatory factors (IL-10). TCFE also reduced hypothalamic senescence, improved cellular integrity, lowered p63, and inhibited microglia activation and inflammatory pathways (TLR4, NFKB, NLRP3). The overall effect of TCFE was better than that of the positive drug group (rapamycin combined with metformin). In vitro results further revealed that TCFE markedly decreased IL1-β, NFKB, and TLR4 levels in BV2 microglial cells, showing comparable efficacy to a TLR4 classic positive inhibitor C34, supporting its anti-inflammatory role. Through UPLC-MS/MS analysis coupled with compound standards, we identified ten bioactive compounds, including gallocatechin, epigallocatechin, catechin, procyanidin B2, kaempferol, quercetin, rutin, naringin, apigenin, ginkgetin. All these compounds showed strong binding affinity to TLR4, notably procyanidin B2 and rutin, potentially through hydrogen bonds, aromatic cation-π interactions, and hydrophobic interactions, suggesting a molecular basis for their anti-inflammatory action. CONCLUSION TCFE showed strong anti-aging effects by inhibiting microglia activation and lessening oxidative stress and modulating inflammatory pathways. This research supports TCF's use in anti-aging and sets a base for future drug development in the realms of neuroinflammation and aging.
Collapse
Affiliation(s)
- Chen Meimei
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China; Fujian Key Laboratory of TCM Health Status Identification, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Zhang Fei
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China; Fujian Key Laboratory of TCM Health Status Identification, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Xu Wen
- Science and Innovation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Lei Huangwei
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China; Fujian Key Laboratory of TCM Health Status Identification, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Hong Zhenqiang
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China; Fujian Key Laboratory of TCM Health Status Identification, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China; Key Laboratory of Orthopedics & Traumatology of Traditional Chinese Medicine and Rehabilitation Ministry of Education, Fujian University of TCM, China
| | - Yu Rongjun
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Zhao Qiang
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China; Fujian Key Laboratory of TCM Health Status Identification, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Li Qiuyang
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China; Fujian Key Laboratory of TCM Health Status Identification, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Liu Xiaozhen
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China; Fujian Key Laboratory of TCM Health Status Identification, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Yang Yuan
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China; Fujian Key Laboratory of TCM Health Status Identification, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Yang Zhaoyang
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China; Fujian Key Laboratory of TCM Health Status Identification, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China; Key Laboratory of Orthopedics & Traumatology of Traditional Chinese Medicine and Rehabilitation Ministry of Education, Fujian University of TCM, China.
| | - Li Candong
- College of Traditional Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China; Fujian Key Laboratory of TCM Health Status Identification, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China.
| |
Collapse
|
7
|
Penati S, Brioschi S, Cai Z, Han CZ, Colonna M. Mechanisms and environmental factors shaping the ecosystem of brain macrophages. Front Immunol 2025; 16:1539988. [PMID: 39925814 PMCID: PMC11802581 DOI: 10.3389/fimmu.2025.1539988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 01/03/2025] [Indexed: 02/11/2025] Open
Abstract
Brain macrophages encompass two major populations: microglia in the parenchyma and border-associated macrophages (BAMs) in the extra-parenchymal compartments. These cells play crucial roles in maintaining brain homeostasis and immune surveillance. Microglia and BAMs are phenotypically and epigenetically distinct and exhibit highly specialized functions tailored to their environmental niches. Intriguingly, recent studies have shown that both microglia and BAMs originate from the same myeloid progenitor during yolk sac hematopoiesis, but their developmental fates diverge within the brain. Several works have partially unveiled the mechanisms orchestrating the development of microglia and BAMs in both mice and humans; however, many questions remain unanswered. Defining the molecular underpinnings controlling the transcriptional and epigenetic programs of microglia and BAMs is one of the upcoming challenges for the field. In this review, we outline current knowledge on ontogeny, phenotypic diversity, and the factors shaping the ecosystem of brain macrophages. We discuss insights garnered from human studies, highlighting similarities and differences compared to mice. Lastly, we address current research gaps and potential future directions in the field. Understanding how brain macrophages communicate with their local environment and how the tissue instructs their developmental trajectories and functional features is essential to fully comprehend brain physiology in homeostasis and disease.
Collapse
Affiliation(s)
- Silvia Penati
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, United States
| | - Simone Brioschi
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, United States
| | - Zhangying Cai
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, United States
| | - Claudia Z. Han
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, United States
- Brain Immunology and Glia (BIG) Center, Washington University School of Medicine in Saint Louis, Saint Louis, MO, United States
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, United States
- Brain Immunology and Glia (BIG) Center, Washington University School of Medicine in Saint Louis, Saint Louis, MO, United States
| |
Collapse
|
8
|
Škandík M, Friess L, Vázquez-Cabrera G, Keane L, Grabert K, Cruz De Los Santos M, Posada-Pérez M, Baleviciute A, Cheray M, Joseph B. Age-associated microglial transcriptome leads to diminished immunogenicity and dysregulation of MCT4 and P2RY12/P2RY13 related functions. Cell Death Discov 2025; 11:16. [PMID: 39828750 PMCID: PMC11743796 DOI: 10.1038/s41420-025-02295-1] [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: 11/20/2024] [Revised: 12/17/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025] Open
Abstract
The aging process is marked by a time-dependent deterioration in cellular functions, particularly the immune and neural systems. Understanding the phenotype acquisition of microglia, the sentinel immune cells of the brain, is crucial for understanding the nature of age-related neurological diseases. However, the specific phenotype adopted by microglia during aging remains a subject of debate and is contingent on the chosen experimental model. To address these unresolved questions, we employed a novel and highly controlled approach utilizing long-term cultivated BV-2 microglia, exempted from additional external stimuli. Our findings revealed that aged microglial cells, in comparison to their younger counterparts, acquire a distinct gene expression profile, primarily characterized by alterations in microglial immune response. Indeed, pro-inflammatory stimulated aged and young BV-2 microglia exhibited similar transcriptomic profiles, yet the response intensity to the stimulus was markedly muted in the aged microglia. Functional neurotoxic assays confirmed diminished neuronal death in coculture with aged, activated microglia, underscoring a compromised immune response. Furthermore, a subsequent comparative analysis of aged BV-2 microglia with established transcriptomic microglial datasets from aged mice and humans identified 13 overlapping genes, laying the foundation for identifying core microglial aging signature. Particularly noteworthy were SLC16A3 and P2RY13, which consistently exhibited upregulation and downregulation, respectively, across all datasets. Additionally, four other genes-CAPG, LGALS3BP, NRIP1, and P2RY12-were found to share regulatory patterns in response to both aging and extrinsic activation. An in-depth investigation focused on SLC16A3, encoding the high-affinity lactate transporter MCT4, revealed disruptions in extracellular acidification rate and lactate concentration with age. Microglial purine sensing and motility capacities, regulated by P2RY12/P2RY13, displayed age-related alterations. Remarkably, protein analysis in human brain tissue validated the observed upregulation of MCT4 and downregulation of P2RY12 in aged microglia. In conclusion, our study unveils a distinct phenotype in aged microglia characterized by compromised immune responsiveness. Through the integration of in vitro cultured BV-2 microglia with primary microglia datasets, we identify critical molecular determinants of microglial cellular aging confirmed in human-aged brain tissue. This comprehensive approach offers potential insights for understanding and potentially reprogramming aged microglia, with implications for combating age-related neurological disorders.
Collapse
Affiliation(s)
- Martin Škandík
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lara Friess
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Lily Keane
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kathleen Grabert
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mireia Cruz De Los Santos
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Mercedes Posada-Pérez
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Center for Neuromusculoskeletal Restorative Medicine, Hong-Kong, China
| | - Austeja Baleviciute
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mathilde Cheray
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bertrand Joseph
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden.
- Center for Neuromusculoskeletal Restorative Medicine, Hong-Kong, China.
| |
Collapse
|
9
|
Gobbo D, Kirchhoff F. Animal-based approaches to understanding neuroglia physiology in vitro and in vivo. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:229-263. [PMID: 40122627 DOI: 10.1016/b978-0-443-19104-6.00012-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
This chapter describes the pivotal role of animal models for unraveling the physiology of neuroglial cells in the central nervous system (CNS). The two rodent species Mus musculus (mice) and Rattus norvegicus (rats) have been indispensable in scientific research due to their remarkable resemblance to humans anatomically, physiologically, and genetically. Their ease of maintenance, short gestation times, and rapid development make them ideal candidates for studying the physiology of astrocytes, oligodendrocyte-lineage cells, and microglia. Moreover, their genetic similarity to humans facilitates the investigation of molecular mechanisms governing neural physiology. Mice are largely the predominant model of neuroglial research, owing to advanced genetic manipulation techniques, whereas rats remain invaluable for applications requiring larger CNS structures for surgical manipulations. Next to rodents, other animal models, namely, Danio rerio (zebrafish) and Drosophila melanogaster (fruit fly), will be discussed to emphasize their critical role in advancing our understanding of glial physiology. Each animal model provides distinct advantages and disadvantages. By combining the strengths of each of them, researchers can gain comprehensive insights into glial function across species, ultimately promoting the understanding of glial physiology in the human CNS and driving the development of novel therapeutic interventions for CNS disorders.
Collapse
Affiliation(s)
- Davide Gobbo
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany.
| | - Frank Kirchhoff
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany; Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, Homburg, Germany.
| |
Collapse
|
10
|
Heneka MT, van der Flier WM, Jessen F, Hoozemanns J, Thal DR, Boche D, Brosseron F, Teunissen C, Zetterberg H, Jacobs AH, Edison P, Ramirez A, Cruchaga C, Lambert JC, Laza AR, Sanchez-Mut JV, Fischer A, Castro-Gomez S, Stein TD, Kleineidam L, Wagner M, Neher JJ, Cunningham C, Singhrao SK, Prinz M, Glass CK, Schlachetzki JCM, Butovsky O, Kleemann K, De Jaeger PL, Scheiblich H, Brown GC, Landreth G, Moutinho M, Grutzendler J, Gomez-Nicola D, McManus RM, Andreasson K, Ising C, Karabag D, Baker DJ, Liddelow SA, Verkhratsky A, Tansey M, Monsonego A, Aigner L, Dorothée G, Nave KA, Simons M, Constantin G, Rosenzweig N, Pascual A, Petzold GC, Kipnis J, Venegas C, Colonna M, Walter J, Tenner AJ, O'Banion MK, Steinert JR, Feinstein DL, Sastre M, Bhaskar K, Hong S, Schafer DP, Golde T, Ransohoff RM, Morgan D, Breitner J, Mancuso R, Riechers SP. Neuroinflammation in Alzheimer disease. Nat Rev Immunol 2024:10.1038/s41577-024-01104-7. [PMID: 39653749 DOI: 10.1038/s41577-024-01104-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2024] [Indexed: 02/20/2025]
Abstract
Increasing evidence points to a pivotal role of immune processes in the pathogenesis of Alzheimer disease, which is the most prevalent neurodegenerative and dementia-causing disease of our time. Multiple lines of information provided by experimental, epidemiological, neuropathological and genetic studies suggest a pathological role for innate and adaptive immune activation in this disease. Here, we review the cell types and pathological mechanisms involved in disease development as well as the influence of genetics and lifestyle factors. Given the decade-long preclinical stage of Alzheimer disease, these mechanisms and their interactions are driving forces behind the spread and progression of the disease. The identification of treatment opportunities will require a precise understanding of the cells and mechanisms involved as well as a clear definition of their temporal and topographical nature. We will also discuss new therapeutic strategies for targeting neuroinflammation, which are now entering the clinic and showing promise for patients.
Collapse
Affiliation(s)
- Michael T Heneka
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette/Belvaux, Luxembourg.
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Neurology, Vrije Universiteit Amsterdam, Amsterdam UMC location VUmc, Amsterdam, The Netherlands
| | - Frank Jessen
- Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
| | - Jeroen Hoozemanns
- Department of Pathology, Amsterdam Neuroscience, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Dietmar Rudolf Thal
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
- Laboratory for Neuropathology, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- Laboratory for Neuropathology, Department of Imaging and Pathology, Leuven Brain Institute (LBI), Leuven, Belgium
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | | | - Charlotte Teunissen
- Department of Laboratory Medicine, VUMC Amsterdam, Amsterdam, The Netherlands
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | - Andreas H Jacobs
- European Institute for Molecular Imaging, University of Münster, Münster, Germany
| | - Paul Edison
- Division of Neurology, Department of Brain Sciences, Imperial College London, London, UK
| | - Alfredo Ramirez
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University of Cologne, Cologne, Germany
- Cluster of Excellence Cellular Stress Response in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Carlos Cruchaga
- Department of Psychiatry, Washington School of Medicine in St. Louis, St. Louis, MO, USA
| | - Jean-Charles Lambert
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Agustin Ruiz Laza
- ACE Alzheimer Center Barcelona, Universitat Internacional de Catalunya (UIC), Barcelona, Spain
| | - Jose Vicente Sanchez-Mut
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), Alicante, Spain
| | - Andre Fischer
- Clinic for Psychiatry and Psychotherapy, University Medical Center, Georg-August-University Göttingen, Göttingen, Germany
- Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Centre for Neurodegenerative Disease (DZNE), Göttingen, Germany
| | - Sergio Castro-Gomez
- Center for Neurology, Clinic of Parkinson, Sleep and Movement Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
- Institute of Physiology II, University Hospital Bonn, University of Bonn, Bonn, Germany
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Thor D Stein
- Boston University Alzheimer's Disease Research Center and CTE Center, Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Luca Kleineidam
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Michael Wagner
- Department of Neurodegenerative Disease and Geriatric Psychiatry, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Jonas J Neher
- Biomedical Center Munich, Biochemistry, Medical Faculty, LMU Munich, Munich, Germany
- Neuroimmunology and Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Colm Cunningham
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute (TBSI), Trinity College Dublin, Dublin, Ireland
- Trinity College Institute of Neuroscience (TCIN), Trinity College Dublin, Dublin, Ireland
| | - Sim K Singhrao
- Brain and Behaviour Centre, Faculty of Clinical and Biomedical Sciences, School of Dentistry, University of Central Lancashire, Preston, UK
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
- Signalling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Oleg Butovsky
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kilian Kleemann
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Philip L De Jaeger
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Hannah Scheiblich
- Center for Neurology, Clinic of Parkinson, Sleep and Movement Disorders, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Guy C Brown
- Deparment of Biochemistry, University of Cambridge, Cambridge, UK
| | - Gary Landreth
- School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Miguel Moutinho
- School of Medicine, Indiana University, Indianapolis, IN, USA
| | - Jaime Grutzendler
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Diego Gomez-Nicola
- School of Biological Sciences, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Róisín M McManus
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Katrin Andreasson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Christina Ising
- Cluster of Excellence Cellular Stress Response in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Deniz Karabag
- Cluster of Excellence Cellular Stress Response in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Darren J Baker
- Department of Paediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York City, NY, USA
- Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York City, NY, USA
- Department of Ophthalmology, NYU Grossman School of Medicine, New York City, NY, USA
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Malu Tansey
- College of Medicine, University of Florida, Gainsville, FL, USA
| | - Alon Monsonego
- Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Guillaume Dorothée
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine (CRSA), Hôpital Saint-Antoine, Paris, France
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
| | - Gabriela Constantin
- Section of General Pathology, Department of Medicine, University of Verona, Verona, Italy
| | - Neta Rosenzweig
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alberto Pascual
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Gabor C Petzold
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Department of Vascular Neurology, University of Bonn, Bonn, Germany
| | - Jonathan Kipnis
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Center for Brain Immunology and Glia (BIG), Washington University School of Medicine, St. Louis, MO, USA
| | - Carmen Venegas
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette/Belvaux, Luxembourg
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
- Instituto Biosanitario de Granada (ibs.Granada), Granada, Spain
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jochen Walter
- Center of Neurology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Andrea J Tenner
- Department of Molecular Biology & Biochemistry, University of California Irvine, Irvine, CA, USA
- Department of Neurobiology and Behaviour, University of California Irvine, Irvine, CA, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - M Kerry O'Banion
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, USA
- Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| | - Joern R Steinert
- Faculty of Medicine and Health Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK
| | - Douglas L Feinstein
- Department of NeuroAnesthesia, University of Illinois at Chicago, Chicago, IL, USA
| | - Magdalena Sastre
- Department of Brain Sciences, Imperial College London, Hammersmith Hospital, London, UK
| | - Kiran Bhaskar
- Department of Molecular Genetics & Microbiology and Neurology, University of New Mexico, Albuquerque, NM, USA
| | - Soyon Hong
- UK Dementia Research Institute, Institute of Neurology, University College London, London, UK
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Todd Golde
- Department of Pharmacology and Chemical Biology, Emory Center for Neurodegenerative Disease, Emory University, Atlanta, GA, USA
- Department of Neurology, Emory Center for Neurodegenerative Disease, Emory University, Atlanta, GA, USA
| | | | - David Morgan
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - John Breitner
- Department of Psychiatry, McGill University Faculty of Medicine, Montreal, Québec, Canada
| | - Renzo Mancuso
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, University of Antwerp, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Sean-Patrick Riechers
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette/Belvaux, Luxembourg
| |
Collapse
|
11
|
Huang KC, Tawfik M, Samuel MA. Retinal ganglion cell circuits and glial interactions in humans and mice. Trends Neurosci 2024; 47:994-1013. [PMID: 39455342 PMCID: PMC11631666 DOI: 10.1016/j.tins.2024.09.010] [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: 05/17/2024] [Revised: 08/30/2024] [Accepted: 09/26/2024] [Indexed: 10/28/2024]
Abstract
Retinal ganglion cells (RGCs) are the brain's gateway for vision, and their degeneration underlies several blinding diseases. RGCs interact with other neuronal cell types, microglia, and astrocytes in the retina and in the brain. Much knowledge has been gained about RGCs and glia from mice and other model organisms, often with the assumption that certain aspects of their biology may be conserved in humans. However, RGCs vary considerably between species, which could affect how they interact with their neuronal and glial partners. This review details which RGC and glial features are conserved between mice, humans, and primates, and which differ. We also discuss experimental approaches for studying human and primate RGCs. These strategies will help to bridge the gap between rodent and human RGC studies and increase study translatability to guide future therapeutic strategies.
Collapse
Affiliation(s)
- Kang-Chieh Huang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030. USA.
| | - Mohamed Tawfik
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030. USA
| | - Melanie A Samuel
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030. USA.
| |
Collapse
|
12
|
Tangavelou K, Bhaskar K. The Mechanistic Link Between Tau-Driven Proteotoxic Stress and Cellular Senescence in Alzheimer's Disease. Int J Mol Sci 2024; 25:12335. [PMID: 39596399 PMCID: PMC11595124 DOI: 10.3390/ijms252212335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/09/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
In Alzheimer's disease (AD), tau dissociates from microtubules (MTs) due to hyperphosphorylation and misfolding. It is degraded by various mechanisms, including the 20S proteasome, chaperone-mediated autophagy (CMA), 26S proteasome, macroautophagy, and aggrephagy. Neurofibrillary tangles (NFTs) form upon the impairment of aggrephagy, and eventually, the ubiquitin chaperone valosin-containing protein (VCP) and heat shock 70 kDa protein (HSP70) are recruited to the sites of NFTs for the extraction of tau for the ubiquitin-proteasome system (UPS)-mediated degradation. However, the impairment of tau degradation in neurons allows tau to be secreted into the extracellular space. Secreted tau can be monomers, oligomers, and paired helical filaments (PHFs), which are seeding competent pathological tau that can be endocytosed/phagocytosed by healthy neurons, microglia, astrocytes, oligodendrocyte progenitor cells (OPCs), and oligodendrocytes, often causing proteotoxic stress and eventually triggers senescence. Senescent cells secrete various senescence-associated secretory phenotype (SASP) factors, which trigger cellular atrophy, causing decreased brain volume in human AD. However, the molecular mechanisms of proteotoxic stress and cellular senescence are not entirely understood and are an emerging area of research. Therefore, this comprehensive review summarizes pertinent studies that provided evidence for the sequential tau degradation, failure, and the mechanistic link between tau-driven proteotoxic stress and cellular senescence in AD.
Collapse
Affiliation(s)
- Karthikeyan Tangavelou
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| | - Kiran Bhaskar
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| |
Collapse
|
13
|
Martinez MM, Walsh JR, Kamocka MM, Lee H, Dunn KW. Longitudinal intravital microscopy of the mouse kidney: inflammatory responses to abdominal imaging windows. Am J Physiol Renal Physiol 2024; 327:F845-F868. [PMID: 39323386 PMCID: PMC11563595 DOI: 10.1152/ajprenal.00071.2024] [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: 03/01/2024] [Revised: 09/10/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024] Open
Abstract
Intravital microscopy enables direct observation of cell biology and physiology at subcellular resolution in real time in living animals. Implanted windows extend the scope of intravital microscopy to processes extending for weeks or even months, such as disease progression or tumor development. However, a question that must be addressed in such studies is whether the imaging window, like any foreign body, triggers an inflammatory response, and whether that response alters the biological process under investigation. To directly evaluate this question, we conducted large-scale intravital microscopy of the kidney of LysM-EGFP mice over time after implantation of abdominal imaging windows. These studies demonstrate that windows stimulated a variety of changes consistent with a foreign body response. Within a few days of implantation, leukocytes were recruited to the window and the region between the window and kidney where, over the next 16 days, they increased in number in an expanding volume that developed a new vascular network. These changes were accompanied by a dramatic increase in glomerular albumin permeability within 2-5 days of implantation. Similar results were obtained from mice implanted with windows coated with poly(l-lysine)-graft-polyethylene glycol (PLL-g-PEG), but not from immune-deficient mice. These studies demonstrate the importance of evaluating whether implanted windows induce an inflammatory response, and whether that response impacts the processes under evaluation in longitudinal intravital microscopy studies.NEW & NOTEWORTHY Intravital microscopy studies of LysM-EGFP mice demonstrate that abdominal imaging windows placed over the kidney stimulated a variety of changes consistent with a foreign body response. Within a day of implantation, leukocytes were recruited to the window where, over the next 16 days, they increased in number in an expanding volume that developed a new vascular network. These changes were accompanied by a dramatic increase in glomerular permeability to albumin.
Collapse
Affiliation(s)
- Michelle M Martinez
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Julia R Walsh
- Weldon School of Biomedical Engineering, Center for Implantable Devices, Purdue University, West Lafayette, Indiana, United States
| | - Malgorzata M Kamocka
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Hyowon Lee
- Weldon School of Biomedical Engineering, Center for Implantable Devices, Purdue University, West Lafayette, Indiana, United States
| | - Kenneth W Dunn
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Weldon School of Biomedical Engineering, Center for Implantable Devices, Purdue University, West Lafayette, Indiana, United States
| |
Collapse
|
14
|
Nikodemova M, Oberto JR, Kaye EL, Berschel MR, Michaelson AL, Watters JJ, Mitchell GS. Acute postnatal inflammation alters adult microglial responses to LPS that are sex-, region- and timing of postnatal inflammation-dependent. J Neuroinflammation 2024; 21:256. [PMID: 39390483 PMCID: PMC11465935 DOI: 10.1186/s12974-024-03245-x] [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: 06/11/2024] [Accepted: 09/26/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Adverse events in early life can have impact lasting into adulthood. We investigated the long-term effects of systemic inflammation during postnatal development on adult microglial responses to lipopolysaccharide (LPS) in two CNS regions (cortex, cervical spinal cord) in male and female rats. METHODS Inflammation was induced in Sprague-Dawley rats by LPS (1 mg/kg) administered intraperitoneally during postnatal development at P7, P12 or P18. As adults (12 weeks of age), the rats received a second LPS dose (1 mg/kg). Control rats received saline. Microglia were isolated 3 h post-LPS followed by gene expression analysis via qRT-PCR for pro-inflammatory (IL-6, iNOS, Ptgs2, C/EBPb, CD14, CXCL10), anti-inflammatory (CD68, Arg-1), and homeostatic genes (P2Y12, Tmemm119). CSF-1 and CX3CL1 mRNAs were analyzed in microglia-free homogenates. RESULTS Basal gene expression in adult microglia was largely unaffected by postnatal inflammation. Adult cortical microglial pro-inflammatory gene responses to LPS were either unchanged or attenuated in rats exposed to LPS during postnatal development. Ptgs2, C/EBPb, CXCL10 and Arg-1 were the most affected genes, with expression significantly downregulated vs. rats without postnatal LPS. Spinal microglia were affected most by LPS at P18, with mixed and sometimes opposing effects on proinflammatory genes in males vs. females. Overall, male cortical vs. spinal microglia were more affected by postnatal LPS. Females were affected in both cortex and spinal cord, but the effect was dependent on timing of postnatal LPS. Overall, inflammatory challenge at P18 had greater effect on adult microglia vs. challenge at P12 or P7. CONCLUSIONS Long-lasting effects of postnatal inflammation on adult microglia depend on postnatal timing, CNS region and sex.
Collapse
Affiliation(s)
- Maria Nikodemova
- Breathing Research and Therapeutic Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA.
| | - Jose R Oberto
- Breathing Research and Therapeutic Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Ethan L Kaye
- Breathing Research and Therapeutic Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Mackenzie R Berschel
- Breathing Research and Therapeutic Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Alysha L Michaelson
- Breathing Research and Therapeutic Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| | - Jyoti J Watters
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin, Madison, WI, 53706, USA
| | - Gordon S Mitchell
- Breathing Research and Therapeutic Center, Department of Physical Therapy and McKnight Brain Institute, University of Florida, Gainesville, FL, 32610, USA
| |
Collapse
|
15
|
Lopez-Ortiz AO, Eyo UB. Astrocytes and microglia in the coordination of CNS development and homeostasis. J Neurochem 2024; 168:3599-3614. [PMID: 37985374 PMCID: PMC11102936 DOI: 10.1111/jnc.16006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/20/2023] [Accepted: 10/20/2023] [Indexed: 11/22/2023]
Abstract
Glia have emerged as important architects of central nervous system (CNS) development and maintenance. While traditionally glial contributions to CNS development and maintenance have been studied independently, there is growing evidence that either suggests or documents that glia may act in coordinated manners to effect developmental patterning and homeostatic functions in the CNS. In this review, we focus on astrocytes, the most abundant glia in the CNS, and microglia, the earliest glia to colonize the CNS highlighting research that documents either suggestive or established coordinated actions by these glial cells in various CNS processes including cell and/or debris clearance, neuronal survival and morphogenesis, synaptic maturation, and circuit function, angio-/vasculogenesis, myelination, and neurotransmission. Some molecular mechanisms underlying these processes that have been identified are also described. Throughout, we categorize the available evidence as either suggestive or established interactions between microglia and astrocytes in the regulation of the respective process and raise possible avenues for further research. We conclude indicating that a better understanding of coordinated astrocyte-microglial interactions in the developing and mature brain holds promise for developing effective therapies for brain pathologies where these processes are perturbed.
Collapse
Affiliation(s)
- Aída Oryza Lopez-Ortiz
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Neuroscience Graduate Program, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Ukpong B Eyo
- Center for Brain Immunology and Glia, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Department of Neuroscience, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Neuroscience Graduate Program, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
16
|
Jiao H, Kalsbeek A, Yi CX. Microglia, circadian rhythm and lifestyle factors. Neuropharmacology 2024; 257:110029. [PMID: 38852838 DOI: 10.1016/j.neuropharm.2024.110029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/11/2024]
Abstract
Microglia, a vital homeostasis-keeper of the central nervous system, perform critical functions such as synaptic pruning, clearance of cellular debris, and participation in neuroinflammatory processes. Recent research has shown that microglia exhibit strong circadian rhythms that not only actively regulate their own immune activity, but also affect neuronal function. Disruptions of the circadian clock have been linked to a higher risk of developing a variety of diseases. In this article we will provide an overview of how lifestyle factors impact microglial function, with a focus on disruptions caused by irregular sleep-wake patterns, reduced physical activity, and eating at the wrong time-of-day. We will also discuss the potential connection between these lifestyle factors, disrupted circadian rhythms, and the role of microglia in keeping brain health. This article is part of the Special Issue on "Microglia".
Collapse
Affiliation(s)
- Han Jiao
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, location AMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam, the Netherlands; Department of Clinical Chemistry, Laboratory of Endocrinology, Amsterdam University Medical Center, location AMC, Amsterdam, the Netherlands; Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, location AMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam, the Netherlands; Department of Clinical Chemistry, Laboratory of Endocrinology, Amsterdam University Medical Center, location AMC, Amsterdam, the Netherlands; Netherlands Institute for Neuroscience, Amsterdam, the Netherlands
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Amsterdam University Medical Center, location AMC, University of Amsterdam, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam, the Netherlands; Department of Clinical Chemistry, Laboratory of Endocrinology, Amsterdam University Medical Center, location AMC, Amsterdam, the Netherlands; Netherlands Institute for Neuroscience, Amsterdam, the Netherlands.
| |
Collapse
|
17
|
Henningfield CM, Kwang N, Tsourmas KI, Neumann J, Kawauchi S, Swarup V, MacGregor GR, Green KN. Generation of an inducible destabilized-domain Cre mouse line to target disease associated microglia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.18.613773. [PMID: 39345513 PMCID: PMC11429805 DOI: 10.1101/2024.09.18.613773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
The function of microglia during progression of Alzheimer's disease (AD) can be investigated using mouse models that enable genetic manipulation of microglial subpopulations in a temporal manner. We developed a mouse strain that expresses destabilized-domain Cre recombinase (DD-Cre) from the Cst7 locus ( Cst7 DD-Cre ) and tested this in 5xFAD amyloidogenic, Ai14 tdTomato cre-reporter line mice. Dietary administration of trimethoprim to induce DD-Cre activity produces long-term labeling in disease associated microglia (DAM) without evidence of leakiness, with tdTomato-expression restricted to cells surrounding plaques. Using this model, we found that DAMs are a subset of plaque-associated microglia (PAMs) and their transition to DAM increases with age and disease stage. Spatial transcriptomic analysis revealed that tdTomato+ cells show higher expression of disease and inflammatory genes compared to other microglial populations, including non-labeled PAMs. This model should allow inducible cre-loxP targeting of DAMs, without leakiness. Highlights We developed a new mouse strain which specifically enables recombination of loxP sites in disease associated microglia (DAMs) and can be used to manipulate DAM-gene expression.DAMs represent a subset of plaque associated microglia (PAMs), and DAM expression increases with disease progression.Spatial transcriptomic analyses reveal that DAMs have higher expression of disease and inflammatory genes compared to other PAMs.
Collapse
|
18
|
Tiwari V, Prajapati B, Asare Y, Damkou A, Ji H, Liu L, Naser N, Gouna G, Leszczyńska KB, Mieczkowski J, Dichgans M, Wang Q, Kawaguchi R, Shi Z, Swarup V, Geschwind DH, Prinz M, Gokce O, Simons M. Innate immune training restores pro-reparative myeloid functions to promote remyelination in the aged central nervous system. Immunity 2024; 57:2173-2190.e8. [PMID: 39053462 DOI: 10.1016/j.immuni.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 11/21/2023] [Accepted: 07/01/2024] [Indexed: 07/27/2024]
Abstract
The reduced ability of the central nervous system to regenerate with increasing age limits functional recovery following demyelinating injury. Previous work has shown that myelin debris can overwhelm the metabolic capacity of microglia, thereby impeding tissue regeneration in aging, but the underlying mechanisms are unknown. In a model of demyelination, we found that a substantial number of genes that were not effectively activated in aged myeloid cells displayed epigenetic modifications associated with restricted chromatin accessibility. Ablation of two class I histone deacetylases in microglia was sufficient to restore the capacity of aged mice to remyelinate lesioned tissue. We used Bacillus Calmette-Guerin (BCG), a live-attenuated vaccine, to train the innate immune system and detected epigenetic reprogramming of brain-resident myeloid cells and functional restoration of myelin debris clearance and lesion recovery. Our results provide insight into aging-associated decline in myeloid function and how this decay can be prevented by innate immune reprogramming.
Collapse
Affiliation(s)
- Vini Tiwari
- Institute of Neuronal Cell Biology, Technical University Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Bharat Prajapati
- Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 41390 Gothenburg, Sweden
| | - Yaw Asare
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Alkmini Damkou
- Institute of Neuronal Cell Biology, Technical University Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Hao Ji
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Lu Liu
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Nawraa Naser
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Garyfallia Gouna
- Institute of Neuronal Cell Biology, Technical University Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Katarzyna B Leszczyńska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 02093 Warsaw, Poland
| | - Jakub Mieczkowski
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 02093 Warsaw, Poland; 3P-Medicine Laboratory, Medical University of Gdańsk, 80211 Gdańsk, Poland
| | - Martin Dichgans
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), 81377 Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Qing Wang
- Departments of Neurology and Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Riki Kawaguchi
- Departments of Neurology and Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Psychiatry, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zechuan Shi
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Daniel H Geschwind
- Departments of Neurology and Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79085 Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Ozgun Gokce
- Munich Cluster of Systems Neurology (SyNergy), 81377 Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany; Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, 53127 Bonn, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), 81377 Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany.
| |
Collapse
|
19
|
Netzahualcoyotzi C, Santillán-Cigales JJ, Adalid-Peralta LV, Velasco I. Infiltration of immune cells to the brain and its relation to the pathogenesis of Alzheimer's and Parkinson's diseases. J Neurochem 2024; 168:2316-2334. [PMID: 38549444 DOI: 10.1111/jnc.16106] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 02/22/2024] [Accepted: 03/13/2024] [Indexed: 10/04/2024]
Abstract
The neurovascular unit, composed of vascular endothelium, vascular smooth muscle, extracellular matrix components, pericytes, astrocytes, microglia, and neurons, allows the highly regulated exchange of molecules and the limited trafficking of cells to the brain through coordinated signaling activity. The passage of peripheral immune cells to the brain parenchyma is observed when there is clear damage to the barriers of this neurovascular unit, as occurs in traumatic brain injury. The possibility of leukocyte infiltration to the brain in neurodegenerative conditions has been proposed. In this review, we focus on describing the evidence for peripheral immune cell infiltration to the brain in the two most frequent neurodegenerative diseases: Alzheimer's and Parkinson's diseases. In particular, we address the mechanisms that promote the passage of these cells into the brain under such pathological conditions. We also discuss the relevance of the resulting cellular interactions, which provide evidence that the presence of peripheral immune cells in the brain is a key point in these neurodegenerative diseases.
Collapse
Affiliation(s)
- Citlalli Netzahualcoyotzi
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
| | - Juan Jair Santillán-Cigales
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Laura Virginia Adalid-Peralta
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
| | - Iván Velasco
- Instituto de Fisiología Celular-Neurociencias, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Ciudad de México, Mexico
| |
Collapse
|
20
|
Botella Lucena P, Heneka MT. Inflammatory aspects of Alzheimer's disease. Acta Neuropathol 2024; 148:31. [PMID: 39196440 DOI: 10.1007/s00401-024-02790-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024]
Abstract
Alzheimer´s disease (AD) stands out as the most common chronic neurodegenerative disorder. AD is characterized by progressive cognitive decline and memory loss, with neurodegeneration as its primary pathological feature. The role of neuroinflammation in the disease course has become a focus of intense research. While microglia, the brain's resident macrophages, have been pivotal to study central immune inflammation, recent evidence underscores the contributions of other cellular entities to the neuroinflammatory process. In this article, we review the inflammatory role of microglia and astrocytes, focusing on their interactions with AD's core pathologies, amyloid beta deposition, and tau tangle formation. Additionally, we also discuss how different modes of regulated cell death in AD may impact the chronic neuroinflammatory environment. This review aims to highlight the evolving landscape of neuroinflammatory research in AD and underscores the importance of considering multiple cellular contributors when developing new therapeutic strategies.
Collapse
Affiliation(s)
- Pablo Botella Lucena
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6, Avenue du Swing, Belvaux, L-4367, Esch-Belval, Luxembourg
| | - Michael T Heneka
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 6, Avenue du Swing, Belvaux, L-4367, Esch-Belval, Luxembourg.
- Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
21
|
Wang W, Ma L, Liu B, Ouyang L. The role of trained immunity in sepsis. Front Immunol 2024; 15:1449986. [PMID: 39221248 PMCID: PMC11363069 DOI: 10.3389/fimmu.2024.1449986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Sepsis is defined as a life-threatening organ dysfunction syndrome caused by dysregulated host response to infection, characterized by a systemic inflammatory response to infection. The use of antibiotics, fluid resuscitation, and organ support therapy has limited prognostic benefit in patients with sepsis, and its incidence is not diminishing, which is attracting increased attention in medicine. Sepsis remains one of the most debilitating and expensive illnesses. One of the main reasons of septic mortality is now understood to be disruption of immune homeostasis. Immunotherapy is revolutionizing the treatment of illnesses in which dysregulated immune responses play a significant role. This "trained immunity", which is a potent defense against infection regardless of the type of bacteria, fungus, or virus, is attributed to the discovery that the innate immune cells possess immune memory via metabolic and epigenetic reprogramming. Here we reviewed the immunotherapy of innate immune cells in sepsis, the features of trained immunity, and the relationship between trained immunity and sepsis.
Collapse
Affiliation(s)
| | | | | | - Liangliang Ouyang
- Department of Medical Laboratory, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
| |
Collapse
|
22
|
Camacho-Morales A, Cárdenas-Tueme M. Prenatal Programming of Monocyte Chemotactic Protein-1 Signaling in Autism Susceptibility. Mol Neurobiol 2024; 61:6119-6134. [PMID: 38277116 DOI: 10.1007/s12035-024-03940-z] [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: 09/12/2023] [Accepted: 01/11/2024] [Indexed: 01/27/2024]
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder that involves functional and structural defects in selective central nervous system (CNS) regions, harming the individual capability to process and respond to external stimuli, including impaired verbal and non-verbal communications. Etiological causes of ASD have not been fully clarified; however, prenatal activation of the innate immune system by external stimuli might infiltrate peripheral immune cells into the fetal CNS and activate cytokine secretion by microglia and astrocytes. For instance, genomic and postmortem histological analysis has identified proinflammatory gene signatures, microglia-related expressed genes, and neuroinflammatory markers in the brain during ASD diagnosis. Active neuroinflammation might also occur during the developmental stage, promoting the establishment of a defective brain connectome and increasing susceptibility to ASD after birth. While still under investigation, we tested the hypothesis whether the monocyte chemoattractant protein-1 (MCP-1) signaling is prenatally programmed to favor peripheral immune cell infiltration and activate microglia into the fetal CNS, setting susceptibility to autism-like behavior. In this review, we will comprehensively provide the current understanding of the prenatal activation of MCP-1 signaling by external stimuli during the developmental stage as a new selective node to promote neuroinflammation, brain structural alterations, and behavioral defects associated to ASD diagnosis.
Collapse
Affiliation(s)
- Alberto Camacho-Morales
- College of Medicine, Department of Biochemistry, Universidad Autónoma de Nuevo Leon, Monterrey, NL, Mexico.
- Center for Research and Development in Health Sciences, Neurometabolism Unit, Universidad Autónoma de Nuevo Leon, San Nicolás de los Garza, Monterrey, NL, Mexico.
| | - Marcela Cárdenas-Tueme
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de La Salud and The Institute for Obesity Research, 64710, Monterrey, Mexico
- Nutrition Unit, Center for Research and Development in Health Sciences, Universidad Autonoma de Nuevo Leon, 64460, Monterrey, Mexico
| |
Collapse
|
23
|
Sierra A, Miron VE, Paolicelli RC, Ransohoff RM. Microglia in Health and Diseases: Integrative Hubs of the Central Nervous System (CNS). Cold Spring Harb Perspect Biol 2024; 16:a041366. [PMID: 38438189 PMCID: PMC11293550 DOI: 10.1101/cshperspect.a041366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Microglia are usually referred to as "the innate immune cells of the brain," "the resident macrophages of the central nervous system" (CNS), or "CNS parenchymal macrophages." These labels allude to their inherent immune function, related to their macrophage lineage. However, beyond their classic innate immune responses, microglia also play physiological roles crucial for proper brain development and maintenance of adult brain homeostasis. Microglia sense both external and local stimuli through a variety of surface receptors. Thus, they might serve as integrative hubs at the interface between the external environment and the CNS, able to decode, filter, and buffer cues from outside, with the aim of preserving and maintaining brain homeostasis. In this perspective, we will cast a critical look at how these multiple microglial functions are acquired and coordinated, and we will speculate on their impact on human brain physiology and pathology.
Collapse
Affiliation(s)
- Amanda Sierra
- Achucarro Basque Center for Neuroscience, Glial Cell Biology Laboratory, Science Park of UPV/EHU, E-48940 Leioa, Bizkaia, Spain
- Department of Biochemistry and Molecular Biology, University of the Basque Country EHU/UPV, 48940 Leioa, Spain
- Ikerbasque Foundation, Bilbao 48009, Spain
| | - Veronique E Miron
- BARLO Multiple Sclerosis Centre, Keenan Research Centre for Biomedical Science at St. Michael's Hospital, Toronto M5B 1T8, Canada
- Department of Immunology, University of Toronto, Toronto M5S 1A8, Canada
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh BioQuarter, Edinburgh EH16 4TJ, United Kingdom
| | - Rosa C Paolicelli
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, CH-1005 Lausanne, Switzerland
| | | |
Collapse
|
24
|
Wang Y, Kuca K, You L, Nepovimova E, Heger Z, Valko M, Adam V, Wu Q, Jomova K. The role of cellular senescence in neurodegenerative diseases. Arch Toxicol 2024; 98:2393-2408. [PMID: 38744709 PMCID: PMC11272704 DOI: 10.1007/s00204-024-03768-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 04/24/2024] [Indexed: 05/16/2024]
Abstract
Increasing evidence has revealed that cellular senescence drives NDs, including Alzheimer's disease (AD) and Parkinson's disease. Different senescent cell populations secrete senescence-associated secretory phenotypes (SASP), including matrix metalloproteinase-3, interleukin (IL)-1α, IL-6, and IL-8, which can harm adjacent microglia. Moreover, these cells possess high expression levels of senescence hallmarks (p16 and p21) and elevated senescence-associated β-galactosidase activity in in vitro and in vivo ND models. These senescence phenotypes contribute to the deposition of β-amyloid and tau-protein tangles. Selective clearance of senescent cells and SASP regulation by inhibiting p38/mitogen-activated protein kinase and nuclear factor kappa B signaling attenuate β-amyloid load and prevent tau-protein tangle deposition, thereby improving cognitive performance in AD mouse models. In addition, telomere shortening, a cellular senescence biomarker, is associated with increased ND risks. Telomere dysfunction causes cellular senescence, stimulating IL-6, tumor necrosis factor-α, and IL-1β secretions. The forced expression of telomerase activators prevents cellular senescence, yielding considerable neuroprotective effects. This review elucidates the mechanism of cellular senescence in ND pathogenesis, suggesting strategies to eliminate or restore senescent cells to a normal phenotype for treating such diseases.
Collapse
Affiliation(s)
- Yating Wang
- College of Life Science, Yangtze University, Jingzhou, 434025, China
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Králové, 500 03, Hradec Králové, Czech Republic
- Biomedical Research Center, University Hospital Hradec Kralove, 500 05, Hradec Kralove, Czech Republic
- Andalusian Research Institute in Data Science and Computational Intelligence (DaSCI), University of Granada, Granada, Spain
| | - Li You
- College of Physical Education and Health, Chongqing College of International Business and Economics, Chongqing, 401520, China
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, 500 03, Hradec Králové, Czech Republic
| | - Zbynek Heger
- Department of Chemistry and Biochemistry, Mendel University in Brno, 613 00, Brno, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, 812 37, Bratislava, Slovakia
| | - Vojtech Adam
- Department of Chemistry and Biochemistry, Mendel University in Brno, 613 00, Brno, Czech Republic
| | - Qinghua Wu
- College of Life Science, Yangtze University, Jingzhou, 434025, China.
- Department of Chemistry, Faculty of Science, University of Hradec Králové, 500 03, Hradec Králové, Czech Republic.
| | - Klaudia Jomova
- Department of Chemistry, Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, 949 74, Nitra, Slovakia.
| |
Collapse
|
25
|
Stavgiannoudaki I, Goulielmaki E, Garinis GA. Broken strands, broken minds: Exploring the nexus of DNA damage and neurodegeneration. DNA Repair (Amst) 2024; 140:103699. [PMID: 38852477 DOI: 10.1016/j.dnarep.2024.103699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/15/2024] [Accepted: 05/28/2024] [Indexed: 06/11/2024]
Abstract
Neurodegenerative disorders are primarily characterized by neuron loss progressively leading to cognitive decline and the manifestation of incurable and debilitating conditions, such as Alzheimer's, Parkinson's, and Huntington's diseases. Loss of genome maintenance causally contributes to age-related neurodegeneration, as exemplified by the premature appearance of neurodegenerative features in a growing family of human syndromes and mice harbouring inborn defects in DNA repair. Here, we discuss the relevance of persistent DNA damage, key DNA repair mechanisms and compromised genome integrity in age-related neurodegeneration highlighting the significance of investigating these connections to pave the way for the development of rationalized intervention strategies aimed at delaying the onset of neurodegenerative disorders and promoting healthy aging.
Collapse
Affiliation(s)
- Ioanna Stavgiannoudaki
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology-Hellas, Crete, Heraklion, Greece; Department of Biology, University of Crete, Crete, Heraklion, Greece
| | - Evi Goulielmaki
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology-Hellas, Crete, Heraklion, Greece
| | - George A Garinis
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology-Hellas, Crete, Heraklion, Greece; Department of Biology, University of Crete, Crete, Heraklion, Greece.
| |
Collapse
|
26
|
Yang D, Sun Y, Lin D, Li S, Zhang Y, Wu A, Wei C. Interleukin-33 ameliorates perioperative neurocognitive disorders by modulating microglial state. Neuropharmacology 2024; 253:109982. [PMID: 38701943 DOI: 10.1016/j.neuropharm.2024.109982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/16/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
Perioperative neurocognitive disorders (PND) are cognitive dysfunctions that usually occur in elderly patients after anesthesia and surgery. Microglial overactivation is a key underlying mechanism. Interleukin-33 (IL-33) is a member of the IL-1 family that orchestrates microglial function. In the present study, we explored how IL-33, which regulates microglia, contributes to cognitive improvement in a male mouse model of PND. An exploratory laparotomy was performed to establish a PND model. The expression levels of IL-33 and its receptor ST2 were evaluated using Western blot. IL-33/ST2 secretion, microglial density, morphology, phagocytosis of synapse, and proliferation, and dystrophic microglia were assessed using immunofluorescence. Synaptic plasticity was measured using Golgi staining and long-term potentiation. The Morris water maze and open field test were used to evaluate cognitive function and anxiety. Hippocampal expression of IL-33 and ST2 were elevated on postoperative day 3. We confirmed that IL-33 was secreted by astrocytes and neurons, whereas ST2 mainly colocalized with microglia. IL-33 treatment induced microgliosis after anesthesia and surgery. These microglia had larger soma sizes and shorter and fragmented branches. Compared to the Surgery group, IL-33 treatment reduced the synaptic phagocytosis of microglia and increased microglial proliferation and dystrophic microglia. IL-33 treatment also reversed the impaired synaptic plasticity and cognitive function caused by anesthesia and surgery. In conclusion, these results indicate that IL-33 plays a key role in regulating microglial state and synaptic phagocytosis in a PND mouse model. IL-33 treatment has a therapeutic potential for improving cognitive dysfunction in PND.
Collapse
Affiliation(s)
- Di Yang
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yi Sun
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Dandan Lin
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Sijie Li
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yan Zhang
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing, China.
| | - Anshi Wu
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Changwei Wei
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
27
|
Sun ED, Zhou OY, Hauptschein M, Rappoport N, Xu L, Navarro Negredo P, Liu L, Rando TA, Zou J, Brunet A. Spatiotemporal transcriptomic profiling and modeling of mouse brain at single-cell resolution reveals cell proximity effects of aging and rejuvenation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.603809. [PMID: 39071282 PMCID: PMC11275735 DOI: 10.1101/2024.07.16.603809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Old age is associated with a decline in cognitive function and an increase in neurodegenerative disease risk1. Brain aging is complex and accompanied by many cellular changes2-20. However, the influence that aged cells have on neighboring cells and how this contributes to tissue decline is unknown. More generally, the tools to systematically address this question in aging tissues have not yet been developed. Here, we generate spatiotemporal data at single-cell resolution for the mouse brain across lifespan, and we develop the first machine learning models based on spatial transcriptomics ('spatial aging clocks') to reveal cell proximity effects during brain aging and rejuvenation. We collect a single-cell spatial transcriptomics brain atlas of 4.2 million cells from 20 distinct ages and across two rejuvenating interventions-exercise and partial reprogramming. We identify spatial and cell type-specific transcriptomic fingerprints of aging, rejuvenation, and disease, including for rare cell types. Using spatial aging clocks and deep learning models, we find that T cells, which infiltrate the brain with age, have a striking pro-aging proximity effect on neighboring cells. Surprisingly, neural stem cells have a strong pro-rejuvenating effect on neighboring cells. By developing computational tools to identify mediators of these proximity effects, we find that pro-aging T cells trigger a local inflammatory response likely via interferon-γ whereas pro-rejuvenating neural stem cells impact the metabolism of neighboring cells possibly via growth factors (e.g. vascular endothelial growth factor) and extracellular vesicles, and we experimentally validate some of these predictions. These results suggest that rare cells can have a drastic influence on their neighbors and could be targeted to counter tissue aging. We anticipate that these spatial aging clocks will not only allow scalable assessment of the efficacy of interventions for aging and disease but also represent a new tool for studying cell-cell interactions in many spatial contexts.
Collapse
Affiliation(s)
- Eric D. Sun
- Department of Biomedical Data Science, Stanford University, CA, USA
- Department of Genetics, Stanford University, CA, USA
| | - Olivia Y. Zhou
- Department of Genetics, Stanford University, CA, USA
- Stanford Biophysics Program, Stanford University, CA, USA
- Stanford Medical Scientist Training Program, Stanford University, CA, USA
| | | | | | - Lucy Xu
- Department of Genetics, Stanford University, CA, USA
- Department of Biology, Stanford University, CA, USA
| | | | - Ling Liu
- Department of Neurology, Stanford University, CA, USA
- Department of Neurology, UCLA, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology, UCLA, Los Angeles, CA, USA
| | - Thomas A. Rando
- Department of Neurology, Stanford University, CA, USA
- Department of Neurology, UCLA, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology, UCLA, Los Angeles, CA, USA
| | - James Zou
- Department of Biomedical Data Science, Stanford University, CA, USA
- These authors contributed equally: James Zou, Anne Brunet
| | - Anne Brunet
- Department of Genetics, Stanford University, CA, USA
- Glenn Center for the Biology of Aging, Stanford University, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, CA, USA
- These authors contributed equally: James Zou, Anne Brunet
| |
Collapse
|
28
|
Le Thuc O, García-Cáceres C. Obesity-induced inflammation: connecting the periphery to the brain. Nat Metab 2024; 6:1237-1252. [PMID: 38997442 DOI: 10.1038/s42255-024-01079-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 06/11/2024] [Indexed: 07/14/2024]
Abstract
Obesity is often associated with a chronic, low-grade inflammatory state affecting the entire body. This sustained inflammatory state disrupts the coordinated communication between the periphery and the brain, which has a crucial role in maintaining homeostasis through humoural, nutrient-mediated, immune and nervous signalling pathways. The inflammatory changes induced by obesity specifically affect communication interfaces, including the blood-brain barrier, glymphatic system and meninges. Consequently, brain areas near the third ventricle, including the hypothalamus and other cognition-relevant regions, become susceptible to impairments, resulting in energy homeostasis dysregulation and an elevated risk of cognitive impairments such as Alzheimer's disease and dementia. This Review explores the intricate communication between the brain and the periphery, highlighting the effect of obesity-induced inflammation on brain function.
Collapse
Affiliation(s)
- Ophélia Le Thuc
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Cristina García-Cáceres
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
29
|
Thorp EB, Filipp M, Dima M, Tan C, Feinstein M, Popko B, DeBerge M. CCR2 + monocytes promote white matter injury and cognitive dysfunction after myocardial infarction. Brain Behav Immun 2024; 119:818-835. [PMID: 38735403 PMCID: PMC11574971 DOI: 10.1016/j.bbi.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 04/17/2024] [Accepted: 05/02/2024] [Indexed: 05/14/2024] Open
Abstract
Survivors of myocardial infarction are at increased risk for vascular dementia. Neuroinflammation has been implicated in the pathogenesis of vascular dementia, yet little is known about the cellular and molecular mediators of neuroinflammation after myocardial infarction. Using a mouse model of myocardial infarction coupled with flow cytometric analyses and immunohistochemistry, we discovered increased monocyte abundance in the brain after myocardial infarction, which was associated with increases in brain-resident perivascular macrophages and microglia. Myeloid cell recruitment and activation was also observed in post-mortem brains of humans that died after myocardial infarction. Spatial and single cell transcriptomic profiling of brain-resident myeloid cells after experimental myocardial infarction revealed increased expression of monocyte chemoattractant proteins. In parallel, myocardial infarction increased crosstalk between brain-resident myeloid cells and oligodendrocytes, leading to neuroinflammation, white matter injury, and cognitive dysfunction. Inhibition of monocyte recruitment preserved white matter integrity and cognitive function, linking monocytes to neurodegeneration after myocardial infarction. Together, these preclinical and clinical results demonstrate that monocyte infiltration into the brain after myocardial infarction initiate neuropathological events that lead to vascular dementia.
Collapse
Affiliation(s)
- Edward B Thorp
- Department of Pathology, Northwestern University, Chicago, IL, United States.
| | - Mallory Filipp
- Department of Pathology, Northwestern University, Chicago, IL, United States
| | - Maria Dima
- Department of Neurology, Division of Multiple Sclerosis and Neuroimmunology, Northwestern University, Chicago, IL, United States
| | - Chunfeng Tan
- Department of Neurology, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Matthew Feinstein
- Department of Pathology, Northwestern University, Chicago, IL, United States; Department of Medicine, Division of Cardiology, Northwestern University, Chicago, IL, United States
| | - Brian Popko
- Department of Neurology, Division of Multiple Sclerosis and Neuroimmunology, Northwestern University, Chicago, IL, United States
| | - Matthew DeBerge
- Department of Pathology, Northwestern University, Chicago, IL, United States; Department of Anesthesiology, Critical Care and Pain Medicine, The University of Texas Health Science Center at Houston, Houston, TX, United States.
| |
Collapse
|
30
|
Zhou M, Li R, Hua H, Dai Y, Yin Z, Li L, Zeng J, Yang M, Zhao J, Tan R. The role of tetrahydrocurcumin in disease prevention and treatment. Food Funct 2024; 15:6798-6824. [PMID: 38836693 DOI: 10.1039/d3fo05739a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
In recent decades, natural compounds derived from herbal medicine or dietary sources have played important roles in prevention and treatment of various diseases and have attracted more and more attention. Curcumin, extracted from the Curcumae Longae Rhizoma and widely used as food spice and coloring agent, has been proven to possess high pharmacological value. However, the pharmacological application of curcumin is limited due to its poor systemic bioavailability. As a major active metabolite of curcumin, tetrahydrocurcumin (THC) has higher bioavailability and stability than curcumin. Increasing evidence confirmed that THC had a wide range of biological activities and significant treatment effects on diseases. In this paper, we reviewed the research progress on the biological activities and therapeutic potential of THC on different diseases such as neurological disorders, metabolic syndromes, cancers, and inflammatory diseases. The extensive pharmacological effects of THC involve the modulation of various signaling transduction pathways including MAPK, JAK/STAT, NF-κB, Nrf2, PI3K/Akt/mTOR, AMPK, Wnt/β-catenin. In addition, the pharmacokinetics, drug combination and toxicology of THC were discussed, thus providing scientific basis for the safe application of THC and the development of its dietary supplements and drugs.
Collapse
Affiliation(s)
- Mengting Zhou
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Translational Chinese Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China.
| | - Rui Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Hua Hua
- Translational Chinese Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China.
| | - Ying Dai
- Translational Chinese Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China.
| | - Zhujun Yin
- Translational Chinese Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China.
| | - Li Li
- Translational Chinese Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China.
| | - Jin Zeng
- Translational Chinese Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China.
| | - Mengni Yang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Translational Chinese Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China.
| | - Junning Zhao
- Translational Chinese Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China.
- National Key Laboratory of Drug Regulatory Science, National Medical Products Administration (NMPA), Beijing 100038, China.
| | - Ruirong Tan
- Translational Chinese Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Quality Evaluation of Traditional Chinese Medicine, Sichuan Engineering Technology Research Center of Genuine Regional Drug, Sichuan Provincial Engineering Research Center of Formation Principle and Quality Evaluation of Genuine Medicinal Materials, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China.
| |
Collapse
|
31
|
Linsley JW, Reisine T, Finkbeiner S. Three dimensional and four dimensional live imaging to study mechanisms of progressive neurodegeneration. J Biol Chem 2024; 300:107433. [PMID: 38825007 PMCID: PMC11261153 DOI: 10.1016/j.jbc.2024.107433] [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: 08/30/2023] [Revised: 05/20/2024] [Accepted: 05/26/2024] [Indexed: 06/04/2024] Open
Abstract
Neurodegenerative diseases are complex and progressive, posing challenges to their study and understanding. Recent advances in microscopy imaging technologies have enabled the exploration of neurons in three spatial dimensions (3D) over time (4D). When applied to 3D cultures, tissues, or animals, these technologies can provide valuable insights into the dynamic and spatial nature of neurodegenerative diseases. This review focuses on the use of imaging techniques and neurodegenerative disease models to study neurodegeneration in 4D. Imaging techniques such as confocal microscopy, two-photon microscopy, miniscope imaging, light sheet microscopy, and robotic microscopy offer powerful tools to visualize and analyze neuronal changes over time in 3D tissue. Application of these technologies to in vitro models of neurodegeneration such as mouse organotypic culture systems and human organoid models provide versatile platforms to study neurodegeneration in a physiologically relevant context. Additionally, use of 4D imaging in vivo, including in mouse and zebrafish models of neurodegenerative diseases, allows for the investigation of early dysfunction and behavioral changes associated with neurodegeneration. We propose that these studies have the power to overcome the limitations of two-dimensional monolayer neuronal cultures and pave the way for improved understanding of the dynamics of neurodegenerative diseases and the development of effective therapeutic strategies.
Collapse
Affiliation(s)
- Jeremy W Linsley
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, California, USA; Operant Biopharma, San Francisco, California, USA
| | - Terry Reisine
- Independent Scientific Consultant, Santa Cruz, California, USA
| | - Steven Finkbeiner
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, California, USA; Operant Biopharma, San Francisco, California, USA; Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes, San Francisco, California, USA; Departments of Neurology and Physiology, University of California, San Francisco, California, USA; Neuroscience Graduate Program, University of California, San Francisco, California, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, California, USA.
| |
Collapse
|
32
|
Barclay KM, Abduljawad N, Cheng Z, Kim MW, Zhou L, Yang J, Rustenhoven J, Mazzitelli JA, Smyth LCD, Kapadia D, Brioschi S, Beatty W, Hou J, Saligrama N, Colonna M, Yu G, Kipnis J, Li Q. An inducible genetic tool to track and manipulate specific microglial states reveals their plasticity and roles in remyelination. Immunity 2024; 57:1394-1412.e8. [PMID: 38821054 PMCID: PMC11299637 DOI: 10.1016/j.immuni.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 02/14/2024] [Accepted: 05/07/2024] [Indexed: 06/02/2024]
Abstract
Recent single-cell RNA sequencing studies have revealed distinct microglial states in development and disease. These include proliferative-region-associated microglia (PAMs) in developing white matter and disease-associated microglia (DAMs) prevalent in various neurodegenerative conditions. PAMs and DAMs share a similar core gene signature. However, the extent of the dynamism and plasticity of these microglial states, as well as their functional significance, remains elusive, partly due to the lack of specific tools. Here, we generated an inducible Cre driver line, Clec7a-CreERT2, that targets PAMs and DAMs in the brain parenchyma. Utilizing this tool, we profiled labeled cells during development and in several disease models, uncovering convergence and context-dependent differences in PAM and DAM gene expression. Through long-term tracking, we demonstrated microglial state plasticity. Lastly, we specifically depleted DAMs in demyelination, revealing their roles in disease recovery. Together, we provide a versatile genetic tool to characterize microglial states in CNS development and disease.
Collapse
Affiliation(s)
- Kia M Barclay
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA; Neuroscience Graduate Program, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nora Abduljawad
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA; Neuroscience Graduate Program, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Zuolin Cheng
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Min Woo Kim
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA; Immunology Graduate Program, Washington University School of Medicine, St. Louis, MO 63110, USA; Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lu Zhou
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jin Yang
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Justin Rustenhoven
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Jose A Mazzitelli
- Neuroscience Graduate Program, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA; Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Leon C D Smyth
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Dvita Kapadia
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Simone Brioschi
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Wandy Beatty
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - JinChao Hou
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Naresha Saligrama
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine in St. Louis, School of Medicine, St. Louis, MO 63110, USA; Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine in St. Louis, St. Louis, MO 63112, USA
| | - Marco Colonna
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Guoqiang Yu
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Jonathan Kipnis
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Qingyun Li
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Genetics, Washington University School of Medicine in St. Louis, School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
33
|
Shukla H, John D, Banerjee S, Tiwari AK. Drug repurposing for neurodegenerative diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 207:249-319. [PMID: 38942541 DOI: 10.1016/bs.pmbts.2024.03.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
Neurodegenerative diseases (NDDs) are neuronal problems that include the brain and spinal cord and result in loss of sensory and motor dysfunction. Common NDDs include Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), Multiple Sclerosis (MS), and Amyotrophic Lateral Sclerosis (ALS) etc. The occurrence of these diseases increases with age and is one of the challenging problems among elderly people. Though, several scientific research has demonstrated the key pathologies associated with NDDs still the underlying mechanisms and molecular details are not well understood and need to be explored and this poses a lack of effective treatments for NDDs. Several lines of evidence have shown that NDDs have a high prevalence and affect more than a billion individuals globally but still, researchers need to work forward in identifying the best therapeutic target for NDDs. Thus, several researchers are working in the directions to find potential therapeutic targets to alter the disease pathology and treat the diseases. Several steps have been taken to identify the early detection of the disease and drug repurposing for effective treatment of NDDs. Moreover, it is logical that current medications are being evaluated for their efficacy in treating such disorders; therefore, drug repurposing would be an efficient, safe, and cost-effective way in finding out better medication. In the current manuscript we discussed the utilization of drugs that have been repurposed for the treatment of AD, PD, HD, MS, and ALS.
Collapse
Affiliation(s)
- Halak Shukla
- Department of Biotechnology and Bioengineering, Institute of Advanced Research (IAR), Gandhinagar, Gujarat, India
| | - Diana John
- Department of Biotechnology and Bioengineering, Institute of Advanced Research (IAR), Gandhinagar, Gujarat, India
| | - Shuvomoy Banerjee
- Department of Biotechnology and Bioengineering, Institute of Advanced Research (IAR), Gandhinagar, Gujarat, India
| | - Anand Krishna Tiwari
- Genetics and Developmental Biology Laboratory, Department of Biotechnology and Bioengineering, Institute of Advanced Research (IAR), Gandhinagar, Gujarat, India.
| |
Collapse
|
34
|
Green TRF, Rowe RK. Quantifying microglial morphology: an insight into function. Clin Exp Immunol 2024; 216:221-229. [PMID: 38456795 PMCID: PMC11097915 DOI: 10.1093/cei/uxae023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 01/17/2024] [Accepted: 03/06/2024] [Indexed: 03/09/2024] Open
Abstract
Microglia are specialized immune cells unique to the central nervous system (CNS). Microglia have a highly plastic morphology that changes rapidly in response to injury or infection. Qualitative and quantitative measurements of ever-changing microglial morphology are considered a cornerstone of many microglia-centric research studies. The distinctive morphological variations seen in microglia are a useful marker of inflammation and severity of tissue damage. Although a wide array of damage-associated microglial morphologies has been documented, the exact functions of these distinct morphologies are not fully understood. In this review, we discuss how microglia morphology is not synonymous with microglia function, however, morphological outcomes can be used to make inferences about microglial function. For a comprehensive examination of the reactive status of a microglial cell, both histological and genetic approaches should be combined. However, the importance of quality immunohistochemistry-based analyses should not be overlooked as they can succinctly answer many research questions.
Collapse
Affiliation(s)
- Tabitha R F Green
- Department of Integrative Physiology, The University of Colorado Boulder, Boulder, CO, USA
| | - Rachel K Rowe
- Department of Integrative Physiology, The University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
35
|
Liampas A, Tseriotis VS, Artemiadis A, Zis P, Argyropoulou C, Grigoriadis N, Hadjigeorgiou GM, Vavougyios G. Adult Neoneurogenesis and Oligodendrogenesis in Multiple Sclerosis: A Systematic Review of Human and Animal Studies. Brain Connect 2024; 14:209-225. [PMID: 38534961 DOI: 10.1089/brain.2023.0081] [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: 04/17/2024] Open
Abstract
Introduction: The subventricular zone promotes remyelination through activation differentiation of oligodendroglial precursor cells (OPCs) and neural stem cells (NSCs) into mature oligodendrocytes and thus in the adult brain. In multiple sclerosis (MS) this regenerative capability is halted resulting in neurodegeneration. We aimed to systematically search and synthesize evidence on mechanisms and phenomena associated with subventricular zone (SVZ) dysfunction in MS. Materials and Methods: Our systematic review was reported according to the PRISMA-ScR statement. MEDLINE, SCOPUS, ProQuest, and Google Scholar were searched using the terms "subventricular zone" and "multiple sclerosis," including English-written in vivo and postmortem studies. Results: Twenty studies were included. Thirteen studies on models of experimental autoimmune encephalomyelitis (EAE) reported among others strong stathmin immunoreactivity in the SVZ of EAE models, the role of MOG immunization in neurogenesis impairment, the effect of parenchymal OPCs and NSCs in myelin repair, and the importance of ependymal cells (E1/E2) and ciliated B1 cells in SVZ stem cell signaling. CXCR4 signaling and transcriptional profiles of SVZ microglia, Gli1 pathway, and galactin-3 were also explored. Studies in humans demonstrated microstructural SVZ damage in progressive MS and the persistence of black holes near the SVZ, whereas postmortem confirmed the generation of polysialic acid-neural cell adhesion molecule and NG2-positive progenitors through SVZ activation, SVZ stathmin immunoreactivity, Shh pathway, and Gal-3 upregulation. Discussion: Oligodendrogenesis defects translate to reduced remyelination, a hallmark of MS that determines its end-phenotype and disease course. Conclusion: The role of inflammation and subsequent SVZ microenvironment disruption is evident in MS pathology.
Collapse
Affiliation(s)
- Andreas Liampas
- Department of Neurology, Nicosia General Hospital, Nicosia, Cyprus
| | | | | | | | | | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology and the Multiple Sclerosis Center, 2nd Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - George Vavougyios
- Medical School, University of Cyprus, Nicosia, Cyprus
- University of Thessaly School of Health Sciences, Thessaloniki, Greece
| |
Collapse
|
36
|
Xie C, Zhang Q, Ye X, Wu W, Cheng X, Ye X, Ruan J, Pan X. Periodontitis-induced neuroinflammation impacts dendritic spine immaturity and cognitive impairment. Oral Dis 2024; 30:2558-2569. [PMID: 37455416 DOI: 10.1111/odi.14674] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 06/03/2023] [Accepted: 06/27/2023] [Indexed: 07/18/2023]
Abstract
OBJECTIVE This study investigated the spinal changes in ligature-induced periodontitis and the role of periodontitis in cognitive impairment. METHODS Twenty mice were randomized into the control and chronic periodontitis (CP) groups, with the latter receiving ligature-induced periodontitis. Cognitive performance was assessed by fear conditioning test. Periodontal inflammation and alveolar bone resorption were evaluated by micro-computed tomography and histopathology. The hippocampal microglial activation was evaluated by immunohistochemistry (IHC). The expressions of hippocampal cytokines (TNF-α, iNOS, IL-1β, IL-4, IL-10, and TREM2) were measured by reverse transcription-polymerase chain reaction. The morphology and density of the dendritic spines were determined by Golgi-Cox staining. RESULTS The CP mice reported significant inflammatory cell infiltration and alveolar bone resorption, with marked increases in cytokine levels (TNF-α, iNOS, IL-1β, and TREM2) in the brain. Moreover, the CP mice showed significantly reduced freezing to the conditioned stimulus in the cued and contextual tests, indicating impaired memory. Further analyses revealed, in the hippocampus of the CP mice, enhanced microglial activation, decreased dendritic spine density, and increased proportion of thin dendritic spines. CONCLUSIONS Periodontitis-induced neuroinflammation may impair the cognitive function by activating hippocampal microglia and inducing dendritic spine immaturity.
Collapse
Affiliation(s)
- Changfu Xie
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Qiuyang Zhang
- The Third Clinical Medical College, Fujian Medical University, Fuzhou, China
- Fuzhou Second Hospital, Fuzhou, China
| | - Xinyi Ye
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Weiliang Wu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Xiaojuan Cheng
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
- Institute of Clinical Neurology, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Xiaoan Ye
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Jianyong Ruan
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Xiaodong Pan
- Department of Neurology, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
- Institute of Clinical Neurology, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| |
Collapse
|
37
|
Strohm AO, Johnston C, Hernady E, Marples B, O'Banion MK, Majewska AK. Cranial irradiation disrupts homeostatic microglial dynamic behavior. J Neuroinflammation 2024; 21:82. [PMID: 38570852 PMCID: PMC10993621 DOI: 10.1186/s12974-024-03073-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 03/22/2024] [Indexed: 04/05/2024] Open
Abstract
Cranial irradiation causes cognitive deficits that are in part mediated by microglia, the resident immune cells of the brain. Microglia are highly reactive, exhibiting changes in shape and morphology depending on the function they are performing. Additionally, microglia processes make dynamic, physical contacts with different components of their environment to monitor the functional state of the brain and promote plasticity. Though evidence suggests radiation perturbs homeostatic microglia functions, it is unknown how cranial irradiation impacts the dynamic behavior of microglia over time. Here, we paired in vivo two-photon microscopy with a transgenic mouse model that labels cortical microglia to follow these cells and determine how they change over time in cranial irradiated mice and their control littermates. We show that a single dose of 10 Gy cranial irradiation disrupts homeostatic cortical microglia dynamics during a 1-month time course. We found a lasting loss of microglial cells following cranial irradiation, coupled with a modest dysregulation of microglial soma displacement at earlier timepoints. The homogeneous distribution of microglia was maintained, suggesting microglia rearrange themselves to account for cell loss and maintain territorial organization following cranial irradiation. Furthermore, we found cranial irradiation reduced microglia coverage of the parenchyma and their surveillance capacity, without overtly changing morphology. Our results demonstrate that a single dose of radiation can induce changes in microglial behavior and function that could influence neurological health. These results set the foundation for future work examining how cranial irradiation impacts complex cellular dynamics in the brain which could contribute to the manifestation of cognitive deficits.
Collapse
Affiliation(s)
- Alexandra O Strohm
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Carl Johnston
- Department of Pediatrics, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Eric Hernady
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Brian Marples
- Department of Radiation Oncology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - M Kerry O'Banion
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Ania K Majewska
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA.
- Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, 14642, USA.
- Center for Visual Science, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| |
Collapse
|
38
|
Sun R, Jiang H. Border-associated macrophages in the central nervous system. J Neuroinflammation 2024; 21:67. [PMID: 38481312 PMCID: PMC10938757 DOI: 10.1186/s12974-024-03059-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/05/2024] [Indexed: 03/17/2024] Open
Abstract
Tissue-resident macrophages play an important role in the local maintenance of homeostasis and immune surveillance. In the central nervous system (CNS), brain macrophages are anatomically divided into parenchymal microglia and non-parenchymal border-associated macrophages (BAMs). Among these immune cell populations, microglia have been well-studied for their roles during development as well as in health and disease. BAMs, mostly located in the choroid plexus, meningeal and perivascular spaces, are now gaining increased attention due to advancements in multi-omics technologies and genetic methodologies. Research on BAMs over the past decade has focused on their ontogeny, immunophenotypes, involvement in various CNS diseases, and potential as therapeutic targets. Unlike microglia, BAMs display mixed origins and distinct self-renewal capacity. BAMs are believed to regulate neuroimmune responses associated with brain barriers and contribute to immune-mediated neuropathology. Notably, BAMs have been observed to function in diverse cerebral pathologies, including Alzheimer's disease, Parkinson's disease, multiple sclerosis, ischemic stroke, and gliomas. The elucidation of the heterogeneity and diverse functions of BAMs during homeostasis and neuroinflammation is mesmerizing, since it may shed light on the precision medicine that emphasizes deep insights into programming cues in the unique brain immune microenvironment. In this review, we delve into the latest findings on BAMs, covering aspects like their origins, self-renewal capacity, adaptability, and implications in different brain disorders.
Collapse
Affiliation(s)
- Rui Sun
- Department of Neurological Surgery, Washington University School of Medicine in St. Louis, 660 S. Euclid Ave., Box 8057, St. Louis, MO, 63110, USA.
| | - Haowu Jiang
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine in St. Louis, 660 S. Euclid Ave., CB 8054, St. Louis, MO, 63110, USA.
| |
Collapse
|
39
|
Lv Z, Chen L, Chen P, Peng H, Rong Y, Hong W, Zhou Q, Li N, Li B, Paolicelli RC, Zhan Y. Clearance of β-amyloid and synapses by the optogenetic depolarization of microglia is complement selective. Neuron 2024; 112:740-754.e7. [PMID: 38295790 DOI: 10.1016/j.neuron.2023.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 10/31/2023] [Accepted: 12/07/2023] [Indexed: 02/03/2024]
Abstract
Microglia actively monitor the neighboring brain microenvironments and constantly contact synapses with their unique ramified processes. In neurodegenerative diseases, including Alzheimer's disease (AD), microglia undergo morphological and functional alterations. Whether the direct manipulation of microglia can selectively or concurrently modulate synaptic function and the response to disease-associated factors remains elusive. Here, we employ optogenetic methods to stimulate microglia in vitro and in vivo. Membrane depolarization rapidly changes microglia morphology and leads to enhanced phagocytosis. We found that the optogenetic stimulation of microglia can efficiently promote β-amyloid (Aβ) clearance in the brain parenchyma, but it can also enhance synapse elimination. Importantly, the inhibition of C1q selectively prevents synapse loss induced by microglia depolarization but does not affect Aβ clearance. Our data reveal independent microglia-mediated phagocytosis pathways toward Aβ and synapses. Our results also shed light on a synergistic strategy of depolarizing microglia and inhibiting complement functions for the clearance of Aβ while sparing synapses.
Collapse
Affiliation(s)
- Zezhong Lv
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lixi Chen
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Neuroscience Program, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Ping Chen
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Huipai Peng
- Shenzhen Institute of Synthetic Biology, CAS Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yi Rong
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Wei Hong
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Qiang Zhou
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Nan Li
- Shenzhen Institute of Synthetic Biology, CAS Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Boxing Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Neuroscience Program, Zhongshan School of Medicine and the Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Rosa C Paolicelli
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland
| | - Yang Zhan
- Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China.
| |
Collapse
|
40
|
Rao Y, Peng B. Allogenic microglia replacement: A novel therapeutic strategy for neurological disorders. FUNDAMENTAL RESEARCH 2024; 4:237-245. [PMID: 38933508 PMCID: PMC11197774 DOI: 10.1016/j.fmre.2023.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/17/2022] [Accepted: 02/19/2023] [Indexed: 03/29/2023] Open
Abstract
Microglia are resident immune cells in the central nervous system (CNS) that play vital roles in CNS development, homeostasis and disease pathogenesis. Genetic defects in microglia lead to microglial dysfunction, which in turn leads to neurological disorders. The correction of the specific genetic defects in microglia in these disorders can lead to therapeutic effects. Traditional genetic defect correction approaches are dependent on viral vector-based genetic defect corrections. However, the viruses used in these approaches, including adeno-associated viruses, lentiviruses and retroviruses, do not primarily target microglia; therefore, viral vector-based genetic defect corrections are ineffective in microglia. Microglia replacement is a novel approach to correct microglial genetic defects via replacing microglia of genetic defects with allogenic healthy microglia. In this paper, we systematically review the history, rationale and therapeutic perspectives of microglia replacement, which would be a novel strategy for treating CNS disorders.
Collapse
Affiliation(s)
- Yanxia Rao
- Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Bo Peng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200000, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| |
Collapse
|
41
|
He D, Liu Q, Mi Y, Meng Q, Xu L, Hou C, Wang J, Li N, Liu Y, Chai H, Yang Y, Liu J, Wang L, Hou Y. De Novo Generation and Identification of Novel Compounds with Drug Efficacy Based on Machine Learning. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307245. [PMID: 38204214 PMCID: PMC10962488 DOI: 10.1002/advs.202307245] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/05/2023] [Indexed: 01/12/2024]
Abstract
One of the main challenges in small molecule drug discovery is finding novel chemical compounds with desirable activity. Traditional drug development typically begins with target selection, but the correlation between targets and disease remains to be further investigated, and drugs designed based on targets may not always have the desired drug efficacy. The emergence of machine learning provides a powerful tool to overcome the challenge. Herein, a machine learning-based strategy is developed for de novo generation of novel compounds with drug efficacy termed DTLS (Deep Transfer Learning-based Strategy) by using dataset of disease-direct-related activity as input. DTLS is applied in two kinds of disease: colorectal cancer (CRC) and Alzheimer's disease (AD). In each case, novel compound is discovered and identified in in vitro and in vivo disease models. Their mechanism of actionis further explored. The experimental results reveal that DTLS can not only realize the generation and identification of novel compounds with drug efficacy but also has the advantage of identifying compounds by focusing on protein targets to facilitate the mechanism study. This work highlights the significant impact of machine learning on the design of novel compounds with drug efficacy, which provides a powerful new approach to drug discovery.
Collapse
Affiliation(s)
- Dakuo He
- College of Information Science and EngineeringState Key Laboratory of Synthetical Automation for Process IndustriesNortheastern UniversityShenyang110819China
| | - Qing Liu
- College of Information Science and EngineeringState Key Laboratory of Synthetical Automation for Process IndustriesNortheastern UniversityShenyang110819China
| | - Yan Mi
- Key Laboratory of Bioresource Research and Development of Liaoning ProvinceCollege of Life and Health SciencesNational Frontiers Science Center for Industrial Intelligence and Systems OptimizationNortheastern UniversityShenyang110169China
- Key Laboratory of Data Analytics and Optimization for Smart IndustryMinistry of EducationNortheastern UniversityShenyang110169China
| | - Qingqi Meng
- Key Laboratory of Bioresource Research and Development of Liaoning ProvinceCollege of Life and Health SciencesNational Frontiers Science Center for Industrial Intelligence and Systems OptimizationNortheastern UniversityShenyang110169China
- Key Laboratory of Data Analytics and Optimization for Smart IndustryMinistry of EducationNortheastern UniversityShenyang110169China
| | - Libin Xu
- Key Laboratory of Bioresource Research and Development of Liaoning ProvinceCollege of Life and Health SciencesNational Frontiers Science Center for Industrial Intelligence and Systems OptimizationNortheastern UniversityShenyang110169China
- Key Laboratory of Data Analytics and Optimization for Smart IndustryMinistry of EducationNortheastern UniversityShenyang110169China
| | - Chunyu Hou
- College of Information Science and EngineeringState Key Laboratory of Synthetical Automation for Process IndustriesNortheastern UniversityShenyang110819China
| | - Jinpeng Wang
- College of Information Science and EngineeringState Key Laboratory of Synthetical Automation for Process IndustriesNortheastern UniversityShenyang110819China
| | - Ning Li
- School of Traditional Chinese Materia MedicaKey Laboratory for TCM Material Basis Study and Innovative Drug Development of Shenyang CityShenyang Pharmaceutical UniversityShenyang110016China
| | - Yang Liu
- Key Laboratory of Structure‐Based Drug Design & Discovery of Ministry of EducationShenyang Pharmaceutical UniversityShenyang110016China
| | - Huifang Chai
- School of PharmacyGuizhou University of Traditional Chinese MedicineGuiyang550025China
| | - Yanqiu Yang
- Key Laboratory of Bioresource Research and Development of Liaoning ProvinceCollege of Life and Health SciencesNational Frontiers Science Center for Industrial Intelligence and Systems OptimizationNortheastern UniversityShenyang110169China
- Key Laboratory of Data Analytics and Optimization for Smart IndustryMinistry of EducationNortheastern UniversityShenyang110169China
| | - Jingyu Liu
- Key Laboratory of Bioresource Research and Development of Liaoning ProvinceCollege of Life and Health SciencesNational Frontiers Science Center for Industrial Intelligence and Systems OptimizationNortheastern UniversityShenyang110169China
- Key Laboratory of Data Analytics and Optimization for Smart IndustryMinistry of EducationNortheastern UniversityShenyang110169China
| | - Lihui Wang
- Department of PharmacologyShenyang Pharmaceutical UniversityShenyang110016China
| | - Yue Hou
- Key Laboratory of Bioresource Research and Development of Liaoning ProvinceCollege of Life and Health SciencesNational Frontiers Science Center for Industrial Intelligence and Systems OptimizationNortheastern UniversityShenyang110169China
- Key Laboratory of Data Analytics and Optimization for Smart IndustryMinistry of EducationNortheastern UniversityShenyang110169China
| |
Collapse
|
42
|
Weathered C, Bardehle S, Yoon C, Kumar N, Leyns CEG, Kennedy ME, Bloomingdale P, Pienaar E. Microglial roles in Alzheimer's disease: An agent-based model to elucidate microglial spatiotemporal response to beta-amyloid. CPT Pharmacometrics Syst Pharmacol 2024; 13:449-463. [PMID: 38078626 PMCID: PMC10941569 DOI: 10.1002/psp4.13095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 11/16/2023] [Accepted: 11/20/2023] [Indexed: 02/03/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by beta-amyloid (Aβ) plaques in the brain and widespread neuronal damage. Because of the high drug attrition rates in AD, there is increased interest in characterizing neuroimmune responses to Aβ plaques. In response to AD pathology, microglia are innate phagocytotic immune cells that transition into a neuroprotective state and form barriers around plaques. We seek to understand the role of microglia in modifying Aβ dynamics and barrier formation. To quantify the influence of individual microglia behaviors (activation, chemotaxis, phagocytosis, and proliferation) on plaque size and barrier coverage, we developed an agent-based model to characterize the spatiotemporal interactions between microglia and Aβ. Our model qualitatively reproduces mouse data trends where the fraction of microglia coverage decreases as plaques become larger. In our model, the time to microglial arrival at the plaque boundary is significantly negatively correlated (p < 0.0001) with plaque size, indicating the importance of the time to microglial activation for regulating plaque size. In addition, in silico behavioral knockout simulations show that phagocytosis knockouts have the strongest impact on plaque size, but modest impacts on microglial coverage and activation. In contrast, the chemotaxis knockouts had a strong impact on microglial coverage with a more modest impact on plaque volume and microglial activation. These simulations suggest that phagocytosis, chemotaxis, and replication of activated microglia have complex impacts on plaque volume and coverage, whereas microglial activation remains fairly robust to perturbations of these functions. Thus, our work provides insights into the potential and limitations of targeting microglial activation as a pharmacological strategy for the treatment of AD.
Collapse
Affiliation(s)
- Catherine Weathered
- Weldon School of Biomedical EngineeringPurdue UniversityWest LafayetteIndianaUSA
| | - Sophia Bardehle
- NeuroimmunologyMerck & Co., Inc.RahwayNew JerseyUSA
- Present address:
Cerevel TherapeuticsCambridgeMassachusettsUSA
| | - Choya Yoon
- NeuroimmunologyMerck & Co., Inc.RahwayNew JerseyUSA
| | - Niyanta Kumar
- Pharmacokinetics and PharmacodynamicsMerck & Co., Inc.RahwayNew JerseyUSA
- Present address:
Mersana TherapeuticsCambridgeMassachusettsUSA
| | | | | | - Peter Bloomingdale
- Quantitative Pharmacology and PharmacometricsMerck & Co., Inc.RahwayNew JerseyUSA
- Present address:
Boehringer IngelheimIngelheim am RheinGermany
| | - Elsje Pienaar
- Weldon School of Biomedical EngineeringPurdue UniversityWest LafayetteIndianaUSA
- Regenstrief Center for Healthcare EngineeringWest LafayetteIndianaUSA
| |
Collapse
|
43
|
Rim C, You MJ, Nahm M, Kwon MS. Emerging role of senescent microglia in brain aging-related neurodegenerative diseases. Transl Neurodegener 2024; 13:10. [PMID: 38378788 PMCID: PMC10877780 DOI: 10.1186/s40035-024-00402-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 01/31/2024] [Indexed: 02/22/2024] Open
Abstract
Brain aging is a recognized risk factor for neurodegenerative diseases like Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis (ALS, Lou Gehrig's disease), but the intricate interplay between brain aging and the pathogenesis of these conditions remains inadequately understood. Cellular senescence is considered to contribute to cellular dysfunction and inflammaging. According to the threshold theory of senescent cell accumulation, the vulnerability to neurodegenerative diseases is associated with the rates of senescent cell generation and clearance within the brain. Given the role of microglia in eliminating senescent cells, the accumulation of senescent microglia may lead to the acceleration of brain aging, contributing to inflammaging and increased vulnerability to neurodegenerative diseases. In this review, we propose the idea that the senescence of microglia, which is notably vulnerable to aging, could potentially serve as a central catalyst in the progression of neurodegenerative diseases. The senescent microglia are emerging as a promising target for mitigating neurodegenerative diseases.
Collapse
Affiliation(s)
- Chan Rim
- Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA University, CHA Bio Complex, 335 Pangyo, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Min-Jung You
- Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA University, CHA Bio Complex, 335 Pangyo, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea
| | - Minyeop Nahm
- Dementia Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Min-Soo Kwon
- Department of Pharmacology, Research Institute for Basic Medical Science, School of Medicine, CHA University, CHA Bio Complex, 335 Pangyo, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.
- Brainimmunex Inc., 26 Yatap-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 13522, Republic of Korea.
| |
Collapse
|
44
|
Sun R, Jiang H. Border-associated macrophages in the central nervous system. Clin Immunol 2024:109921. [PMID: 38316202 DOI: 10.1016/j.clim.2024.109921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 01/31/2024] [Indexed: 02/07/2024]
Abstract
Tissue-resident macrophages play an important role in the local maintenance of homeostasis and immune surveillance. In the central nervous system (CNS), brain macrophages are anatomically divided into parenchymal microglia and non-parenchymal border-associated macrophages (BAMs). Among these immune cell populations, microglia have been well-studied for their roles in normal brain development, neurodegeneration, and brain cancers. BAMs, mostly located in the choroid plexus, meningeal and perivascular spaces, are now gaining increased attention due to advancements in multi-omics technologies and genetic methodologies. Research on BAMs over the past decade has focused on their ontogeny, immunophenotypes, involvement in various CNS diseases, and potential as therapeutic targets. Unlike microglia, BAMs display mixed origins and distinct self-renewal capacity. BAMs are believed to regulate neuroimmune responses associated with brain barriers and contribute to immune-mediated neuropathology. Notably, BAMs have been observed to function in diverse cerebral pathologies, including Alzheimer's disease, Parkinson's disease, multiple sclerosis, ischemic stroke, and gliomas. The elucidation of the heterogeneity and diverse functions of BAMs during homeostasis and neuroinflammation is mesmerizing, since it may shed light on the precision medicine that emphasizes deep insights into programming cues in the unique brain immune microenvironment. In this review, we delve into the latest findings on BAMs, covering aspects like their origins, self-renewal capacity, adaptability, and implications in different brain disorders.
Collapse
Affiliation(s)
- Rui Sun
- Department of Neurological Surgery, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA.
| | - Haowu Jiang
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine in St Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
45
|
Salvi J, Andreoletti P, Audinat E, Balland E, Ben Fradj S, Cherkaoui-Malki M, Heurtaux T, Liénard F, Nédélec E, Rovère C, Savary S, Véjux A, Trompier D, Benani A. Microgliosis: a double-edged sword in the control of food intake. FEBS J 2024; 291:615-631. [PMID: 35880408 DOI: 10.1111/febs.16583] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/30/2022] [Accepted: 07/25/2022] [Indexed: 02/16/2024]
Abstract
Maintaining energy balance is essential for survival and health. This physiological function is controlled by the brain, which adapts food intake to energy needs. Indeed, the brain constantly receives a multitude of biological signals that are derived from digested foods or that originate from the gastrointestinal tract, energy stores (liver and adipose tissues) and other metabolically active organs (muscles). These signals, which include circulating nutrients, hormones and neuronal inputs from the periphery, collectively provide information on the overall energy status of the body. In the brain, several neuronal populations can specifically detect these signals. Nutrient-sensing neurons are found in discrete brain areas and are highly enriched in the hypothalamus. In turn, specialized brain circuits coordinate homeostatic responses acting mainly on appetite, peripheral metabolism, activity and arousal. Accumulating evidence shows that hypothalamic microglial cells located at the vicinity of these circuits can influence the brain control of energy balance. However, microglial cells could have opposite effects on energy balance, that is homeostatic or detrimental, and the conditions for this shift are not totally understood yet. One hypothesis relies on the extent of microglial activation, and nutritional lipids can considerably change it.
Collapse
Affiliation(s)
- Juliette Salvi
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Pierre Andreoletti
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Etienne Audinat
- IGF, Université de Montpellier, CNRS, Inserm, Montpellier, France
| | - Eglantine Balland
- Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Notting Hill, Australia
| | - Selma Ben Fradj
- IPMC, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Valbonne, France
| | | | - Tony Heurtaux
- Luxembourg Center of Neuropathology (LCNP), Dudelange, Luxembourg
- Department of Life Sciences and Medicine, University of Luxembourg, Belvaux, Luxembourg
| | - Fabienne Liénard
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Emmanuelle Nédélec
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| | - Carole Rovère
- IPMC, Institut de Pharmacologie Moléculaire et Cellulaire, CNRS, Université Côte d'Azur, Valbonne, France
| | - Stéphane Savary
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Anne Véjux
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Doriane Trompier
- Laboratoire Bio-PeroxIL, Université Bourgogne Franche-Comté, Dijon, France
| | - Alexandre Benani
- CSGA, Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro Dijon, Université Bourgogne Franche-Comté, Dijon, France
| |
Collapse
|
46
|
Ma Q, Chen J, Kong X, Zeng Y, Chen Z, Liu H, Liu L, Lu S, Wang X. Interactions between CNS and immune cells in tuberculous meningitis. Front Immunol 2024; 15:1326859. [PMID: 38361935 PMCID: PMC10867975 DOI: 10.3389/fimmu.2024.1326859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/10/2024] [Indexed: 02/17/2024] Open
Abstract
The central nervous system (CNS) harbors its own special immune system composed of microglia in the parenchyma, CNS-associated macrophages (CAMs), dendritic cells, monocytes, and the barrier systems within the brain. Recently, advances in the immune cells in the CNS provided new insights to understand the development of tuberculous meningitis (TBM), which is the predominant form of Mycobacterium tuberculosis (M.tb) infection in the CNS and accompanied with high mortality and disability. The development of the CNS requires the protection of immune cells, including macrophages and microglia, during embryogenesis to ensure the accurate development of the CNS and immune response following pathogenic invasion. In this review, we summarize the current understanding on the CNS immune cells during the initiation and development of the TBM. We also explore the interactions of immune cells with the CNS in TBM. In the future, the combination of modern techniques should be applied to explore the role of immune cells of CNS in TBM.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Shuihua Lu
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Shenzhen, Guangdong, China
| | - Xiaomin Wang
- National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
47
|
Meng J, Han L, Xu H, Zhang L, Liu Z, Zhou Y, Zhang X, Luo H, Zhang YW. TREM2 regulates microglial phagocytosis of synapses in innate immune tolerance. Int Immunopharmacol 2024; 127:111445. [PMID: 38147777 DOI: 10.1016/j.intimp.2023.111445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 12/28/2023]
Abstract
Increasing evidence indicates that innate immune cells also possess immunological memory. Microglia are brain-resident innate immune cells and execute inflammatory and phagocytic functions upon environmental stimulation, during which processes triggering receptor expressed on myeloid cells 2 (TREM2) plays an important regulatory role. However, although microglia are known to exhibit innate immune memory related to inflammation when subjected to continuous inflammatory stimuli, whether microglia exhibit innate immune memory related to phagocytosis and whether TREM2 participates in innate immune memory of microglia remain unknown. Herein, we treated WT and Trem2 KO mice with peripheral injection of lipopolysaccharides (LPS) to induce microglial activation or microglial immune tolerance. We found that Tnfα and Il-1β expression levels in the hippocampi were significantly elevated after 1xLPS and then dramatically decreased after 4xLPS in both WT and Trem2 KO mice; and their level changes were indistinguishable between WT and Trem2 KO mice. Moreover, 1xLPS significantly promoted microglial phagocytosis of synapses and caused microglial morphology changes resembling activated status in both WT and Trem2 KO mice. However, 4xLPS significantly reduced synapse phagocytosis and largely reversed morphology changes in WT microglia. While 4xLPS had no effect on reducing synapse phagocytosis in Trem2 KO microglia. RNA-seq analysis revealed that TREM2 deficiency reprogrammed complement and phagosome-related transcriptional landscape during immune tolerance. Our results demonstrate that microglia also exhibit immune tolerance related to phagocytosis of synapses and that TREM2 plays a crucial role in this process possibly through regulating complement system and phagosome-related gene expressions.
Collapse
Affiliation(s)
- Jian Meng
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| | - Linkun Han
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Hui Xu
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Lingliang Zhang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhaoji Liu
- Department of Neurology, Zhongshan Hospital Xiamen University, Xiamen, Fujian 361004, China
| | - Yunqiang Zhou
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Xian Zhang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Hong Luo
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Yun-Wu Zhang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, and Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China.
| |
Collapse
|
48
|
Kent SA, Miron VE. Microglia regulation of central nervous system myelin health and regeneration. Nat Rev Immunol 2024; 24:49-63. [PMID: 37452201 DOI: 10.1038/s41577-023-00907-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2023] [Indexed: 07/18/2023]
Abstract
Microglia are resident macrophages of the central nervous system that have key functions in its development, homeostasis and response to damage and infection. Although microglia have been increasingly implicated in contributing to the pathology that underpins neurological dysfunction and disease, they also have crucial roles in neurological homeostasis and regeneration. This includes regulation of the maintenance and regeneration of myelin, the membrane that surrounds neuronal axons, which is required for axonal health and function. Myelin is damaged with normal ageing and in several neurodegenerative diseases, such as multiple sclerosis and Alzheimer disease. Given the lack of approved therapies targeting myelin maintenance or regeneration, it is imperative to understand the mechanisms by which microglia support and restore myelin health to identify potential therapeutic approaches. However, the mechanisms by which microglia regulate myelin loss or integrity are still being uncovered. In this Review, we discuss recent work that reveals the changes in white matter with ageing and neurodegenerative disease, how this relates to microglia dynamics during myelin damage and regeneration, and factors that influence the regenerative functions of microglia.
Collapse
Affiliation(s)
- Sarah A Kent
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK
| | - Veronique E Miron
- UK Dementia Research Institute at The University of Edinburgh, Edinburgh, UK.
- Centre for Discovery Brain Sciences, Chancellor's Building, The University of Edinburgh, Edinburgh, UK.
- Barlo Multiple Sclerosis Centre, St Michael's Hospital, Toronto, Ontario, Canada.
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, Toronto, Ontario, Canada.
- Department of Immunology, The University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
49
|
Strohm AO, O'Connor TN, Oldfield S, Young S, Hammond C, McCall M, Dirksen RT, Majewska AK. Cortical microglia dynamics are conserved during voluntary wheel running. J Appl Physiol (1985) 2024; 136:89-108. [PMID: 37969082 PMCID: PMC11212787 DOI: 10.1152/japplphysiol.00311.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 11/13/2023] [Accepted: 11/14/2023] [Indexed: 11/17/2023] Open
Abstract
We present the first demonstration of chronic in vivo imaging of microglia in mice undergoing voluntary wheel running. We find that healthy mice undergoing voluntary wheel running have similar microglia dynamics, morphologies, and responses to injury when compared to sedentary mice. This suggests that exercise over a period of 1 mo does not grossly alter cortical microglial phenotypes and that exercise may exert its beneficial effects on the brain through other mechanisms. Future work examining how microglia dynamics may be altered during exercise in disease or injury models could provide further insights into the therapeutic benefit of exercise.NEW & NOTEWORTHY We demonstrate the first use of chronic in vivo imaging of microglia over time during physical exercise. We found that microglia movement, morphology, and process motility were remarkably stable during voluntary wheel running (VWR). Additionally, microglia in running mice respond similarly to laser ablation injury compared to sedentary mice. These findings indicate that VWR does not induce changes in microglia dynamics in healthy adults. Exercise may elicit positive effects on the brain through other mechanisms.
Collapse
Affiliation(s)
- Alexandra O Strohm
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, New York, United States
| | - Thomas N O'Connor
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, New York, United States
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York, United States
| | - Sadie Oldfield
- Department of Neuroscience, University of Rochester Medical Center, Rochester, New York, United States
| | - Sala Young
- Department of Neuroscience, University of Rochester Medical Center, Rochester, New York, United States
| | - Christian Hammond
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, New York, United States
| | - Matthew McCall
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, New York, United States
| | - Robert T Dirksen
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York, United States
| | - Ania K Majewska
- Department of Neuroscience, University of Rochester Medical Center, Rochester, New York, United States
- Center for Visual Science, University of Rochester Medical Center, Rochester, New York, United States
| |
Collapse
|
50
|
Castro R, Lopes M, De Biase L, Valdez G. Aging spinal cord microglia become phenotypically heterogeneous and preferentially target motor neurons and their synapses. Glia 2024; 72:206-221. [PMID: 37737058 PMCID: PMC10773989 DOI: 10.1002/glia.24470] [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: 06/01/2023] [Revised: 08/07/2023] [Accepted: 09/03/2023] [Indexed: 09/23/2023]
Abstract
Microglia have been found to acquire unique region-dependent deleterious features with age and diseases that contribute to neuronal dysfunction and degeneration in the brain. However, it remains unknown whether microglia exhibit similar phenotypic heterogeneity in the spinal cord. Here, we performed a regional analysis of spinal cord microglia in 3-, 16-, 23-, and 30-month-old mice. Using light and electron microscopy, we discovered that spinal cord microglia acquire an increasingly activated phenotype during the course of aging regardless of regional location. However, aging causes microglia in the ventral but not dorsal horn to lose their spatial organization. Aged ventral horn microglia also aggregate around the somata of motor neurons and increase their contacts with motor synapses, which have been shown to be lost with age. These findings suggest that microglia may affect the ability of motor neurons to receive and relay motor commands during aging. To generate additional insights about aging spinal cord microglia, we performed RNA-sequencing on FACS-isolated microglia from 3-, 18-, 22-, and 29-month-old mice. We found that spinal cord microglia acquire a similar transcriptional identity as those in the brain during aging that includes altered expression of genes with roles in microglia-neuron interactions and inflammation. By 29 months of age, spinal cord microglia exhibit additional and unique transcriptional changes known and predicted to cause senescence and to alter lysosomal and ribosomal regulation. Altogether, this work provides the foundation to target microglia to ameliorate aged-related changes in the spinal cord, and particularly on the motor circuit.
Collapse
Affiliation(s)
- Ryan Castro
- Neuroscience Graduate Program, Brown University, Providence, Rhode Island, USA
| | - Mikayla Lopes
- Molecular Biology, Cell Biology & Biochemistry Graduate Program, Brown University, Providence, Rhode Island, USA
| | - Lindsay De Biase
- Department of Physiology, David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - Gregorio Valdez
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
- Center for Translational Neuroscience, Robert J. and Nancy D. Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, Rhode Island, USA
- Department of Neurology, Warren Alpert Medical School of Medicine, Brown University, Providence, USA
| |
Collapse
|