1
|
Chen Y, Wei Y, Liu J, Zhu T, Zhou C, Zhang D. Spatial transcriptomics combined with single-nucleus RNA sequencing reveals glial cell heterogeneity in the human spinal cord. Neural Regen Res 2025; 20:3302-3316. [PMID: 38934400 PMCID: PMC11881709 DOI: 10.4103/nrr.nrr-d-23-01876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 03/04/2024] [Accepted: 04/30/2024] [Indexed: 06/28/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202511000-00032/figure1/v/2024-12-20T164640Z/r/image-tiff Glial cells play crucial roles in regulating physiological and pathological functions, including sensation, the response to infection and acute injury, and chronic neurodegenerative disorders. Glial cells include astrocytes, microglia, and oligodendrocytes in the central nervous system, and satellite glial cells and Schwann cells in the peripheral nervous system. Despite the greater understanding of glial cell types and functional heterogeneity achieved through single-cell and single-nucleus RNA sequencing in animal models, few studies have investigated the transcriptomic profiles of glial cells in the human spinal cord. Here, we used high-throughput single-nucleus RNA sequencing and spatial transcriptomics to map the cellular and molecular heterogeneity of astrocytes, microglia, and oligodendrocytes in the human spinal cord. To explore the conservation and divergence across species, we compared these findings with those from mice. In the human spinal cord, astrocytes, microglia, and oligodendrocytes were each divided into six distinct transcriptomic subclusters. In the mouse spinal cord, astrocytes, microglia, and oligodendrocytes were divided into five, four, and five distinct transcriptomic subclusters, respectively. The comparative results revealed substantial heterogeneity in all glial cell types between humans and mice. Additionally, we detected sex differences in gene expression in human spinal cord glial cells. Specifically, in all astrocyte subtypes, the levels of NEAT1 and CHI3L1 were higher in males than in females, whereas the levels of CST3 were lower in males than in females. In all microglial subtypes, all differentially expressed genes were located on the sex chromosomes. In addition to sex-specific gene differences, the levels of MT-ND4 , MT2A , MT-ATP6 , MT-CO3 , MT-ND2 , MT-ND3 , and MT-CO2 in all spinal cord oligodendrocyte subtypes were higher in females than in males. Collectively, the present dataset extensively characterizes glial cell heterogeneity and offers a valuable resource for exploring the cellular basis of spinal cord-related illnesses, including chronic pain, amyotrophic lateral sclerosis, and multiple sclerosis.
Collapse
Affiliation(s)
- Yali Chen
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yiyong Wei
- Department of Anesthesiology, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College), Shenzhen, Guangdong Province, China
- Department of Anesthesiology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Tao Zhu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Cheng Zhou
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Donghang Zhang
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
- Laboratory of Anesthesia and Critical Care Medicine, National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
2
|
Li JF, Chang HX, Zhao JN, Bao JH, Dai W, Li YF. The astrocytic sigma-1 receptor constitutes in the fast antidepressant action of hypidone hydrochloride (YL-0919) in rodents. Front Pharmacol 2025; 16:1564851. [PMID: 40206084 PMCID: PMC11980816 DOI: 10.3389/fphar.2025.1564851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/03/2025] [Indexed: 04/11/2025] Open
Abstract
Introduction: There is increasing evidence that astrocytes are involved in the therapeutic action of antidepressants. The fast antidepressant YL-0919 may interact with activation of astrocytic sigma-1 receptors (sigma-1R). Methods: In this study, function of astrocytic sigma-1R in ventral hippocampus (vHIP) mediating the rapid antidepressant effect of YL-0919 were investigated. Adeno-associated virus (AAV) expressing shRNA was constructed to knock down astrocytic sigma-1R in vHIP, and the role of astrocytic sigma-1R on the rapid antidepressant action of YL-0919 were tested in chronic restraint stress (CRS) model of mice. Small interfering RNA (siRNA) was used to knock down sigma-1R in primary astrocytes, and we explored the mitochondrial function and BDNF expression of primary astrocytes after YL-0919 and siRNA treatments. Result: The results indicated knocking down astrocytic sigma-1R in vHIP induced anxiety-like and depressive-like behavior in mice, and blocked the rapid anti-depressant and anxiolytic effects of YL-0919. Knocking down sigma-1R in primary astrocytes inhibited the YL-0919 induced enhancement of mitochondrial function and increased level of BDNF expression. In addition, increased BDNF in vHIP might play a role in fast antidepressant impact of YL-0919. Taken together, the data provide further evidence for a role of astrocyte receptors in the mechanisms of action of antidepressants. Conclusion: Taken together, these results reveal increased BDNF in vHIP by affecting glial cells might be one of the significant mechanisms of fast antidepressant effect of YL-0919. The data provide further evidence for a role of astrocyte receptors in the mechanisms of action of antidepressants.
Collapse
Affiliation(s)
- Jin-Feng Li
- Beijing Institute of Basic Medical Sciences, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Hai-Xia Chang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Jia-Ning Zhao
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
- Shenyang Pharmaceutical University, Shenyang, China
| | - Jin-Hao Bao
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wei Dai
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yun-Feng Li
- Beijing Institute of Basic Medical Sciences, Beijing, China
- Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
3
|
Yue Z, Zhou L, Sun P, Kang X, Huang F, Chen P. S2Map: a novel computational platform for identifying secretio-types through cell secretion-signal map. BIOINFORMATICS ADVANCES 2025; 5:vbaf059. [PMID: 40191548 PMCID: PMC11972122 DOI: 10.1093/bioadv/vbaf059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 02/20/2025] [Accepted: 03/17/2025] [Indexed: 04/09/2025]
Abstract
Motivation Cell communication is predominantly governed by secreted proteins, whose diverse secretion patterns often signify underlying physiological irregularities. Understanding these secreted signals at an individual cell level is crucial for gaining insights into regulatory mechanisms involving various molecular agents. To elucidate the array of cell secretion signals, which encompass different types of biomolecular secretion cues from individual immune cells, we introduce the secretion-signal map (S2Map). Results S2Map is an online interactive analytical platform designed to explore and interpret distinct cell secretion-signal patterns visually. It incorporates two innovative qualitative metrics, the signal inequality index and the signal coverage index, which are exquisitely sensitive in measuring dissymmetry and diffusion of signals in temporal data. S2Map's innovation lies in its depiction of signals through time-series analysis with multi-layer visualization. We tested the SII and SCI performance in distinguishing the simulated signal diffusion models. S2Map hosts a repository for the single-cell's secretion-signal data for exploring cell secretio-types, a new cell phenotyping based on the cell secretion signal pattern. We anticipate that S2Map will be a powerful tool to delve into the complexities of physiological systems, providing insights into the regulation of protein production, such as cytokines at the remarkable resolution of single cells. Availability and implementation The S2Map server is publicly accessible via https://au-s2map.streamlit.app/.
Collapse
Affiliation(s)
- Zongliang Yue
- Department of Health Outcomes Research and Policy, Harrison College of Pharmacy, Auburn University, Auburn, AL, 36849, United States
| | - Lang Zhou
- Department of Materials Engineering, Samuel Ginn College of Engineering, Auburn University, Auburn, AL, 36849, United States
| | - Peizhen Sun
- Department of Materials Engineering, Samuel Ginn College of Engineering, Auburn University, Auburn, AL, 36849, United States
| | - Xuejia Kang
- Department of Materials Engineering, Samuel Ginn College of Engineering, Auburn University, Auburn, AL, 36849, United States
| | - Fengyuan Huang
- Biomedical Research Department, Tuskegee University, Tuskegee, AL, 36083, United States
| | - Pengyu Chen
- Department of Materials Engineering, Samuel Ginn College of Engineering, Auburn University, Auburn, AL, 36849, United States
| |
Collapse
|
4
|
O'Dea MR, Hasel P. Are we there yet? Exploring astrocyte heterogeneity one cell at a time. Glia 2025; 73:619-631. [PMID: 39308429 PMCID: PMC11784854 DOI: 10.1002/glia.24621] [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: 05/13/2024] [Revised: 09/02/2024] [Accepted: 09/14/2024] [Indexed: 02/01/2025]
Abstract
Astrocytes are a highly abundant cell type in the brain and spinal cord. Like neurons, astrocytes can be molecularly and functionally distinct to fulfill specialized roles. Recent technical advances in sequencing-based single cell assays have driven an explosion of omics data characterizing astrocytes in the healthy, aged, injured, and diseased central nervous system. In this review, we will discuss recent studies which have furthered our understanding of astrocyte biology and heterogeneity, as well as discuss the limitations and challenges of sequencing-based single cell and spatial genomics methods and their potential future utility.
Collapse
Affiliation(s)
- Michael R. O'Dea
- Neuroscience InstituteNYU Grossman School of MedicineNew YorkNew YorkUSA
| | - Philip Hasel
- UK Dementia Research Institute at the University of EdinburghEdinburghScotlandUK
- Centre for Discovery Brain Sciences, School of Biomedical Sciences, College of Medicine and Veterinary MedicineThe University of EdinburghEdinburghScotlandUK
| |
Collapse
|
5
|
Gildea HK, Liddelow SA. Mechanisms of astrocyte aging in reactivity and disease. Mol Neurodegener 2025; 20:21. [PMID: 39979986 PMCID: PMC11844071 DOI: 10.1186/s13024-025-00810-7] [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: 10/22/2024] [Accepted: 02/06/2025] [Indexed: 02/22/2025] Open
Abstract
Normal aging alters brain functions and phenotypes. However, it is not well understood how astrocytes are impacted by aging, nor how they contribute to neuronal dysfunction and disease risk as organisms age. Here, we examine the transcriptional, cell biology, and functional differences in astrocytes across normal aging. Astrocytes at baseline are heterogenous, responsive to their environments, and critical regulators of brain microenvironments and neuronal function. With increasing age, astrocytes adopt different immune-related and senescence-associated states, which relate to organelle dysfunction and loss of homeostasis maintenance, both cell autonomously and non-cell autonomously. These perturbed states are increasingly associated with age-related dysfunction and the onset of neurodegeneration, suggesting that astrocyte aging is a compelling target for future manipulation in the prevention of disease.
Collapse
Affiliation(s)
- Holly K Gildea
- Institute for Translational Neuroscience, NYU Grossman School of Medicine, New York, USA.
| | - Shane A Liddelow
- Institute for Translational Neuroscience, NYU Grossman School of Medicine, New York, USA.
- Department of Neuroscience, NYU Grossman School of Medicine, New York, USA.
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, USA.
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, USA.
- Optimal Aging Institute, NYU Grossman School of Medicine, New York, USA.
| |
Collapse
|
6
|
Jiang H, Zhang M, Li XM, Zhang NN, Du YS, Xia CY, Wang HQ, Zhang YN, Yang XY, Chen AP, Lai HQ, Yan X, Chu SF, Wang ZZ, Chen NH. The pathogenesis of depression: Roles of connexin 43-based gap junctions and inflammation. Eur J Pharmacol 2025; 989:177260. [PMID: 39798913 DOI: 10.1016/j.ejphar.2025.177260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/13/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Depression is a leading chronic mental illness worldwide, characterized by anhedonia and pessimism. Connexin is a kind of widely distributed protein in the body. Connexin 43 (Cx43) plays an important role in the pathogenesis of depression. Growing evidence has indicated that inflammation is closely associated with neuropsychiatric diseases such as depression. Inflammation occurs in patients with mood disorders, and symptomatic relief after multiple treatments is often accompanied by proinflammatory restoration. In this study, we sought to determine the upstream and downstream relationship of Cx43 abnormalities and peripheral inflammation in inducing depression. METHODS AND RESULTS Gap27, as a Cx43 mimetic peptide, specifically blocks Cx43 gap junction channels in the brain. The mice were treated with injection of Gap27 into the prefrontal cortex (PFC), conditional knockout of Cx43 in the PFC, and lipopolysaccharide (LPS) intraperitoneal injection. Our results revealed that the treatments gave rise to the depressive-like behavior occurrence in mice, including reducing the sucrose preference and spontaneous activities, and increasing immobility time in the forced swimming test. Functional blockade of Cx43 induced abnormal expression of peripheral inflammatory cytokines including interleukin (IL)-1β, IL-6, tumor necrosis factor-α, IL-2, IL-10, and IL-18. Furthermore, depression associated with peripheral inflammation derived from LPS intraperitoneal injection significantly reduced the Cx43 expression and the diffusion distance of gap junction channel permeability dye in the PFC. CONCLUSION These results showed that blockade of Cx43 in the PFC and peripheral inflammation are complicatedly intertwined, and reinforcing each other during the induction of depression.
Collapse
Affiliation(s)
- Hong Jiang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Meng Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Xin-Mu Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Ning-Ning Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Yu-Sheng Du
- Hunan University of Traditional Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, Hunan, China
| | - Cong-Yuan Xia
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Hui-Qin Wang
- Hunan University of Traditional Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, Hunan, China
| | - Ya-Ni Zhang
- Institute of Clinical Pharmacology and Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xue-Ying Yang
- Institute of Clinical Pharmacology and Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Ai-Ping Chen
- China Three Gorges University College of Medicine and Health Sciences, Yichang, 443002, China
| | - Hua-Qing Lai
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Xu Yan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Shi-Feng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China
| | - Zhen-Zhen Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China.
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100050, China; Hunan University of Traditional Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, Hunan, China.
| |
Collapse
|
7
|
Shao X, Wu T, Li M, Zheng M, Lin H, Qi X. Enterococcus faecalis Exerts Neuroprotective Effects via the Vagus Nerve in a Mouse Model of Parkinson's Disease. Mol Neurobiol 2025:10.1007/s12035-025-04741-8. [PMID: 39954164 DOI: 10.1007/s12035-025-04741-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 02/02/2025] [Indexed: 02/17/2025]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease worldwide. Current treatment methods for PD are unable to halt disease progression. The gut microbiota contributes to the neurodevelopment of PD; however, the gut-brain connections and underlying neural bases that regulate this complex behavior are not yet clear. Enterococcus faecalis (EF) is a common commensal bacterium of the gut and a common pathogen associated with hospital-acquired infections. Here, we demonstrated the significant therapeutic effects of a non-pathogenic strain of EF (EF ATCC19433) on PD. In this study, we established a mouse model of PD by intraperitoneal injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). We found that EF treatment alleviated behavioral impairment, dopaminergic neuronal loss, blood-brain barrier damage, and neuroinflammation induced by MPTP in the mice. Additionally, 16S rRNA sequencing revealed that dysbiosis of PD-related microbial communities induced by MPTP was reversed by EF treatment. Moreover, EF treatment relieved gastrointestinal dysfunction in the mice. The therapeutic efficacy of EF in MPTP-induced PD mice is markedly diminished when the activity of EF is lost. Further mechanistic studies indicated that the neuroprotective effects of EF in PD were associated with the vagus nerve pathway. Following the surgical severance of the vagus nerve through subdiaphragmatic vagotomy, the protective effects of EF on PD were markedly diminished. Our study suggests that EF can alleviate neurofunctional impairments and gastrointestinal disorders associated with PD, indicating that gut-derived microbes influence brain function through the vagus nerve pathway.
Collapse
Affiliation(s)
- Xian Shao
- Department of Medical Research Center, Shaoxing People's Hospital, Zhejiang University Shaoxing Hospital, Shaoxing, 312000, Zhejiang, China
| | - Tao Wu
- School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Mengyun Li
- Department of Medical Research Center, Shaoxing People's Hospital, Zhejiang University Shaoxing Hospital, Shaoxing, 312000, Zhejiang, China
| | - Matao Zheng
- Department of Neurosurgery, Shaoxing People's Hospital, Zhejiang University Shaoxing Hospital, Shaoxing, 312000, Zhejiang, China
| | - Hui Lin
- Healthy Science Center, The Affiliated Lihuili Hospital of Ningbo University, Ningbo University, Ningbo, 315211, Zhejiang, China.
| | - Xuchen Qi
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310020, Zhejiang, China.
| |
Collapse
|
8
|
Li G, Huang L, Gu D, Wang P, Yi L, Kuang W, Zhang Y, Zhang J, Liu D, Shi Q, Tang H, Sun J, Zeng G, Peng X, Wang J. Activity-based chemical proteomics reveals caffeic acid ameliorates pentylenetetrazol-induced seizures by covalently targeting aconitate decarboxylase 1. Cell Commun Signal 2025; 23:62. [PMID: 39901156 PMCID: PMC11792687 DOI: 10.1186/s12964-024-01739-y] [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: 03/27/2024] [Accepted: 07/04/2024] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Epilepsy is a neurological disorder characterized by recurrent seizures, tightly associated with neuroinflammation. Activation of inflammatory cells and molecules in damaged nervous tissues plays a pivotal role in epilepsy. Caffeic acid, one of the most abundant polyphenols in coffee, has shown potent protective effects as a phytomedicine in various neurological disorders. However, the direct protein targets and exact molecular mechanisms of caffeic acid in epilepsy, remain largely elusive. PURPOSE This study aimed to explore the protective effects of caffeic acid in epilepsy and elucidate its underlying mechanism. METHODS In this study, we established pentylenetetrazol-induced acute and kindling models of seizures. Additionally, a BV2 microglial cellular inflammation model was established by lipopolysaccharide stimulation. The potential direct protein targets of caffeic acid in BV2 cells were analyzed using an activity-based protein profiling (ABPP) with a caffeic acid probe. Various methods such as pull-down assay, immunofluorescence and cellular heat transfer assays were used for experimental validation. The anti-inflammatory effects of caffeic acid in LPS-activated BV2 cells was proved by knocking down the target protein. RESULTS Here, we found that caffeic acid exhibits antiepileptic effects in pentylenetetrazol-induced epilepsy mice and exerts anti-neuroinflammation effect in vivo and in vitro. Besides, we discovered that caffeic acid directly binds to aconitate decarboxylase 1 and influenced its enzymatic activity. Moreover, we indicated that caffeic acid exhibits anti-neuroinflammation effect through aconitate decarboxylase 1 mediated PERK-NF-κB pathway in vitro. CONCLUSION In summary, this study elucidates, for the first time, the potential antiepileptic targets and mechanism of action of caffeic acid using the ABPP strategy. Our study provides evidence supporting the utilization of caffeic acid as a promising therapeutic agent for treating epilepsy and neuroinflammation-related disorders.
Collapse
Affiliation(s)
- Guanjun Li
- Department of Urology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Ling Huang
- Department of Urology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Di Gu
- Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510230, Guangdong, China
| | - Peili Wang
- Xiyuan Hospital, National Clinical Research Center for Chinese Medicine Cardiology, Academy of Chinese Medical Sciences, Beijing, China
| | - Letai Yi
- Inner Mongolia Medical University, 010000, Hohhot, Inner Mongolia, China
| | - Wenhua Kuang
- Department of Urology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Ying Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, 100700, Beijing, China
| | - Junzhe Zhang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, 100700, Beijing, China
| | - Dandan Liu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, 100700, Beijing, China
| | - Qiaoli Shi
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, 100700, Beijing, China
| | - Huan Tang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, 100700, Beijing, China
| | - Jichao Sun
- Department of Urology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
| | - Guohua Zeng
- Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510230, Guangdong, China.
| | - Xin Peng
- Ningbo Municipal Hospital of TCM, Affiliated Hospital of Zhejiang Chinese Medical University, 315000, Ningbo, Zhejiang, China.
| | - Jigang Wang
- Department of Urology, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
- Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510230, Guangdong, China.
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, 100700, Beijing, China.
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, 475004, Kaifeng, China.
| |
Collapse
|
9
|
Panganiban RP, McAninch C, Chulkina M, Pinchuk IV. Telocytes in inflammatory bowel diseases: contributions to pathology and therapeutic potentials. Front Cell Dev Biol 2025; 12:1452258. [PMID: 39872845 PMCID: PMC11770051 DOI: 10.3389/fcell.2024.1452258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 12/30/2024] [Indexed: 01/30/2025] Open
Abstract
Telocytes, a novel mesenchymal cell population, are characterized by their distinctive long and slender projections known as telopodes and have garnered significant interest since their formal introduction to the literature in 2010. These cells have been identified in various tissues, including the gastrointestinal (GI) tract, where they are suggested to play important roles in maintaining structural integrity, immune modulation, and barrier function. Inflammatory bowel diseases (IBD), which include Crohn's disease (CD) and ulcerative colitis (UC), are characterized by chronic inflammation and fibrosis. While limited information is available on the fate of telocytes in this group of diseases, it has been suggested that loss/plasticity of telocytes can be among the key factors contributing to their pathogenesis. This review focuses on the current understanding of telocytes, their structural features, and their distribution within the GI tract under gut homeostasis and IBD. We also discuss the roles of these cells in immune regulation and intestinal repair. We highlight evidence implicating telocytes in the pathogenesis of IBD and other chronic inflammatory diseases that share similar pathophysiological processes with IBD. Lastly, we discuss the current challenges in gut telocyte biology and the potential therapeutic implications of telocytes in IBD.
Collapse
Affiliation(s)
| | | | | | - Irina V. Pinchuk
- Division of Gastroenterology and Hepatology, Department of Medicine, Penn State College of Medicine, Hershey, PA, United States
| |
Collapse
|
10
|
Vo Q, Simon ZD, Park G, Nacionales DC, Gorski C, Barrios EL, Casadesus G, Efron PA, Moldawer LL, Nagpal R, Chakrabarty P, Febo M. Functional connectivity within sensorimotor cortical and striatal regions is regulated by sepsis in a sex-dependent manner. Neuroimage 2025; 305:120995. [PMID: 39753162 DOI: 10.1016/j.neuroimage.2024.120995] [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: 09/07/2024] [Revised: 11/29/2024] [Accepted: 12/31/2024] [Indexed: 01/07/2025] Open
Abstract
Sepsis is a state of systemic immune dysregulation and organ failure that is frequently associated with severe brain disability. Epidemiological studies have indicated that younger females have better prognosis and clinical outcomes relative to males, though the sex-dependent response of the brain to sepsis during post-sepsis recovery remains largely uncharacterized. Using a modified polymicrobial intra-abdominal murine model of surgical sepsis, we characterized the acute effects of intra-abdominal sepsis on peripheral inflammation, brain inflammation and brain functional connectivity in young adult mice of both sexes. Following sepsis, both male and female mice survived the procedure, regained body weight within 7 days post-sepsis and showed reduced diversity in their gut microbiome. Interestingly, compared to the sepsis-induced changes observed in female mice, the post-septic male mice exhibited a comparatively robust profile of splenic cell expansion and intracerebral glial proliferation relative to their healthy counterparts. Analysis of resting-state functional Magnetic Resonance Imaging (fMRI) data collected from the post-septic mice revealed that while connectivity to the somatosensory cortex were affected equally in both sexes, intra-network connectivity strength in the striatum preferentially increased in post-septic males but remained near baseline in post-septic female mice. Additionally, the female mice showed reduced network connectivity alterations in the projections from periaqueductal gray to the superior colliculus as also between the anterior cingulate cortex and the striatum. Coupled with the sustained intracerebral gliosis response, the intra-striatal fMRI response patterns in males could signify a delayed recovery from sepsis. Together, our study provides evidence that peripheral sepsis influences peripheral immunity, brain immunity and brain connectivity in a sex-dependent manner, with the fMRI response strongly indicating cognitive benefits in young females recovering from sepsis relative to their male counterparts.
Collapse
Affiliation(s)
- Quan Vo
- Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA.
| | - Zachary D Simon
- Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA; Department of Psychiatry, University of Florida, Gainesville, FL 32610, USA.
| | - Gwoncheol Park
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, FL 32304, USA.
| | - Dina C Nacionales
- Department of Surgery, University of Florida, Gainesville, FL 32610, USA; Sepsis & Critical Illness Research Center, University of Florida, Gainesville, FL 32610, USA.
| | - Carmelina Gorski
- Sepsis & Critical Illness Research Center, University of Florida, Gainesville, FL 32610, USA.
| | - Evan L Barrios
- Department of Surgery, University of Florida, Gainesville, FL 32610, USA; Sepsis & Critical Illness Research Center, University of Florida, Gainesville, FL 32610, USA.
| | - Gemma Casadesus
- Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA; Department of Pharmacology & Therapeutics, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| | - Philip A Efron
- Department of Surgery, University of Florida, Gainesville, FL 32610, USA; Sepsis & Critical Illness Research Center, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| | - Lyle L Moldawer
- Department of Surgery, University of Florida, Gainesville, FL 32610, USA; Sepsis & Critical Illness Research Center, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| | - Ravinder Nagpal
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, FL 32304, USA.
| | - Paramita Chakrabarty
- Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA; Sepsis & Critical Illness Research Center, University of Florida, Gainesville, FL 32610, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| | - Marcelo Febo
- Department of Psychiatry, University of Florida, Gainesville, FL 32610, USA; McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
11
|
Li J, Jin H, Zhao X, Sun X, Zhong J, Zhao J, Yan M. Effect of β-1,4-GalTI on the biological function of astrocytes treated by LPS. BIOMOLECULES & BIOMEDICINE 2024; 25:226-239. [PMID: 39284278 PMCID: PMC11647251 DOI: 10.17305/bb.2024.11088] [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: 08/05/2024] [Revised: 09/02/2024] [Accepted: 09/02/2024] [Indexed: 12/14/2024]
Abstract
Inflammation of the central nervous system (CNS) is a common feature of neurological disorders and infections, playing a crucial role in the development of CNS-related conditions. CNS inflammation is primarily regulated by glial cells, with astrocytes being the most abundant type in the mammalian CNS. Numerous studies have demonstrated that astrocytes, as immunocompetent cells, perform diverse and complex functions in both health and disease. Glycosylation, a critical post-translational modification of proteins, regulates numerous biological functions. The expression and activity of glycosyltransferases, the enzymes responsible for glycosylation, are closely associated with the pathogenesis of various diseases. β-1,4-GalTI, a mammalian glycosyltransferase, plays a significant role in cell-cell interactions, adhesion, and migration. Although many studies have focused on β-1,4-GalTI, few have explored its effects on astrocyte function. In this study, we constructed lentiviral vectors for both interference and overexpression of β-1,4-GalTI and discovered that β-1,4-GalTI knockdown inhibited astrocyte migration and proliferation, while its overexpression promoted these processes. Concurrently, β-1,4-GalTI knockdown reduced the expression of TNF-α, IL-1β, and IL-6, whereas overexpression enhanced the expression of these cytokines. These findings suggest that modulating β-1,4-GalTI activity can influence the molecular functions of astrocytes and provide a theoretical foundation for further research into β-1,4-GalTI as a potential therapeutic target in astrocyte-mediated inflammation.
Collapse
Affiliation(s)
- Jiyu Li
- Department of Orthopedic Oncology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Hui Jin
- The Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Xinmin Zhao
- The Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Xinran Sun
- The Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Jiyuan Zhong
- The Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| | - Jian Zhao
- Department of Orthopedic Oncology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Meijuan Yan
- The Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
12
|
Saglam-Metiner P, Yanasik S, Odabasi YC, Modamio J, Negwer M, Biray-Avci C, Guler A, Erturk A, Yildirim E, Yesil-Celiktas O. ICU patient-on-a-chip emulating orchestration of mast cells and cerebral organoids in neuroinflammation. Commun Biol 2024; 7:1627. [PMID: 39639082 PMCID: PMC11621364 DOI: 10.1038/s42003-024-07313-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: 03/08/2024] [Accepted: 11/22/2024] [Indexed: 12/07/2024] Open
Abstract
Propofol and midazolam are the current standard of care for prolonged sedation in Intensive Care Units (ICUs). However, the effects and mechanism of these sedatives in brain tissue are unclear. Herein, the development of an ICU patient-on-a-chip platform to elucidate those effects is reported. The humanized neural tissue compartment combines mast cells differentiated from human induced pluripotent stem cells (hiPSCs) with cerebral organoids in a three-dimensional (3D) matrix, which is covered with a membrane populated with human cerebral microvascular endothelial cells (hCMEC/D3) that separates the tissue chamber from the vascular lumen, where sedatives were infused for four days to evaluate neurotoxicity and cell-mediated immune responses. Subsequent to propofol administration, gene expressions of CD40 and TNF-α in mast cells, AIF1 in microglia and GFAP/S100B/OLIG2/MBP in macroglia were elevated, as well as NOS2, CD80, CD40, CD68, IL6 and TNF-α mediated proinflammation is noted in cerebral organoids, which resulted in higher expressions of GJB1, GABA-A and NMDAR1 in the tissue construct of the platform. Besides, midazolam administration stimulated expression of CD40 and CD203c+ reactivated mast cell proliferation and compromised BBB permeability and decreased TEER values with higher barrier disruption, whereas increased populations of CD11b+ microglia, higher expressions of GFAP/DLG4/GJB1 and GABA-A-/NMDAR1- identities, as well as glutamate related neurotoxicity and IL1B, IFNG, IFNA1, IL6 genes mediated proinflammation, resulting in increased apoptotic zones are observed in cerebral organoids. These results suggest that different sedatives cause variations in cell type activation that modulate different pathways related to neuroinflammation and neurotoxicity in the ICU patient-on-chip platform.
Collapse
Affiliation(s)
- Pelin Saglam-Metiner
- Department of Bioengineering, Faculty of Engineering, Ege University, Izmir, Türkiye
| | - Sena Yanasik
- Department of Bioengineering, Faculty of Engineering, Ege University, Izmir, Türkiye
| | - Yusuf Caglar Odabasi
- Department of Bioengineering, Faculty of Engineering, Ege University, Izmir, Türkiye
| | - Jennifer Modamio
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Zentrum München, Neuherberg, Germany
| | - Moritz Negwer
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Zentrum München, Neuherberg, Germany
| | - Cigir Biray-Avci
- Department of Medical Biology, Faculty of Medicine, Ege University, Bornova, Izmir, Türkiye
| | - Ayse Guler
- Department of Neuroscience, Faculty of Medicine, Ege University, Bornova, Izmir, Türkiye
| | - Ali Erturk
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Zentrum München, Neuherberg, Germany
| | - Ender Yildirim
- Department of Mechanical Engineering, Middle East Technical University, Ankara, Türkiye
- ODTÜ MEMS Center, Ankara, Türkiye
| | - Ozlem Yesil-Celiktas
- Department of Bioengineering, Faculty of Engineering, Ege University, Izmir, Türkiye.
- ODTÜ MEMS Center, Ankara, Türkiye.
| |
Collapse
|
13
|
Geloso MC, Zupo L, Corvino V. Crosstalk between peripheral inflammation and brain: Focus on the responses of microglia and astrocytes to peripheral challenge. Neurochem Int 2024; 180:105872. [PMID: 39362496 DOI: 10.1016/j.neuint.2024.105872] [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: 08/09/2024] [Revised: 09/18/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
A growing body of evidence supports the link between peripheral inflammation and impairment of neurologic functions, including mood and cognitive abilities. The pathogenic event connecting peripheral inflammation and brain dysfunction is represented by neuroinflammation, a pathogenic phenomenon that provides an important contribution to neurodegeneration and cognitive decline also in Alzheimer's, Parkinson's, Huntington's diseases, as well as in Multiple Sclerosis. It is driven by resident brain immune cells, microglia and astrocytes, that acquire an activated phenotype in response to proinflammatory molecules moving from the periphery to the brain parenchyma. Although a huge progress has been made in clarifying cellular and molecular mechanisms bridging peripheral and central inflammation, a clear picture has not been achieved so far. Therefore, experimental models are of crucial relevance to clarify knowledge gaps in this regard. Many findings demonstrate that systemic inflammation induced by pathogen-associated molecular patterns, such as lipopolysaccharide (LPS), is able to trigger neuroinflammation. Therefore, LPS-administration is widely considered a useful tool to study this phenomenon. On this basis, the present review will focus on in vivo studies based on acute and subacute effects of systemic administration of LPS, with special attention on the state of art of microglia and astrocyte response to peripheral challenge.
Collapse
Affiliation(s)
- Maria Concetta Geloso
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy; Gemelli Science and Technology Park (GSTeP)-Organoids Research Core Facility, Fondazione Policlinico Agostino Gemelli IRCCS, Rome, Italy.
| | - Luca Zupo
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| | - Valentina Corvino
- Department of Neuroscience, Section of Human Anatomy, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Rome, Italy
| |
Collapse
|
14
|
Penning A, Snoeck S, Garritsen O, Tosoni G, Hof A, de Boer F, van Hasenbroek J, Zhang L, Thrupp N, Craessaerts K, Fiers M, Salta E. NACC2, a molecular effector of miR-132 regulation at the interface between adult neurogenesis and Alzheimer's disease. Sci Rep 2024; 14:21163. [PMID: 39256511 PMCID: PMC11387632 DOI: 10.1038/s41598-024-72096-6] [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: 05/03/2024] [Accepted: 09/03/2024] [Indexed: 09/12/2024] Open
Abstract
The generation of new neurons at the hippocampal neurogenic niche, known as adult hippocampal neurogenesis (AHN), and its impairment, have been implicated in Alzheimer's disease (AD). MicroRNA-132 (miR-132), the most consistently downregulated microRNA (miRNA) in AD, was recently identified as a potent regulator of AHN, exerting multilayered proneurogenic effects in adult neural stem cells (NSCs) and their progeny. Supplementing miR-132 in AD mouse brain restores AHN and relevant memory deficits, yet the exact mechanisms involved are still unknown. Here, we identify NACC2 as a novel miR-132 target implicated in both AHN and AD. miR-132 deficiency in mouse hippocampus induces Nacc2 expression and inflammatory signaling in adult NSCs. We show that miR-132-dependent regulation of NACC2 is involved in the initial stages of human NSC differentiation towards astrocytes and neurons. Later, NACC2 function in astrocytic maturation becomes uncoupled from miR-132. We demonstrate that NACC2 is present in reactive astrocytes surrounding amyloid plaques in mouse and human AD hippocampus, and that there is an anticorrelation between miR-132 and NACC2 levels in AD and upon induction of inflammation. Unraveling the molecular mechanisms by which miR-132 regulates neurogenesis and cellular reactivity in AD, will provide valuable insights towards its possible application as a therapeutic target.
Collapse
Affiliation(s)
- Amber Penning
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Sarah Snoeck
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Oxana Garritsen
- UMC Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Giorgia Tosoni
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Amber Hof
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Fleur de Boer
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | | | - Lin Zhang
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Nicky Thrupp
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
| | | | - Mark Fiers
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
| | - Evgenia Salta
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands.
| |
Collapse
|
15
|
Megagiannis P, Mei Y, Yan RE, Yuan L, Wilde JJ, Eckersberg H, Suresh R, Tan X, Chen H, Farmer WT, Cha K, Le PU, Catoire H, Rochefort D, Kwan T, Yee BA, Dion P, Krishnaswamy A, Cloutier JF, Stifani S, Petrecca K, Yeo GW, Murai KK, Feng G, Rouleau GA, Ideker T, Sanjana NE, Zhou Y. Autism-associated CHD8 controls reactive gliosis and neuroinflammation via remodeling chromatin in astrocytes. Cell Rep 2024; 43:114637. [PMID: 39154337 DOI: 10.1016/j.celrep.2024.114637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 06/11/2024] [Accepted: 07/30/2024] [Indexed: 08/20/2024] Open
Abstract
Reactive changes of glial cells during neuroinflammation impact brain disorders and disease progression. Elucidating the mechanisms that control reactive gliosis may help us to understand brain pathophysiology and improve outcomes. Here, we report that adult ablation of autism spectrum disorder (ASD)-associated CHD8 in astrocytes attenuates reactive gliosis via remodeling chromatin accessibility, changing gene expression. Conditional Chd8 deletion in astrocytes, but not microglia, suppresses reactive gliosis by impeding astrocyte proliferation and morphological elaboration. Astrocyte Chd8 ablation alleviates lipopolysaccharide-induced neuroinflammation and septic-associated hypothermia in mice. Astrocytic CHD8 plays an important role in neuroinflammation by altering the chromatin landscape, regulating metabolic and lipid-associated pathways, and astrocyte-microglia crosstalk. Moreover, we show that reactive gliosis can be directly mitigated in vivo using an adeno-associated virus (AAV)-mediated Chd8 gene editing strategy. These findings uncover a role of ASD-associated CHD8 in the adult brain, which may warrant future exploration of targeting chromatin remodelers in reactive gliosis and neuroinflammation in injury and neurological diseases.
Collapse
Affiliation(s)
- Platon Megagiannis
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Yuan Mei
- Division of Genetics, Department of Medicine, University of California, San Diego, San Diego, CA, USA; Department of Cellular and Molecular Medicine, Stem Cell Program, Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Rachel E Yan
- New York Genome Center, New York, NY, USA; Department of Biology, New York University, New York, NY, USA
| | - Lin Yuan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Jonathan J Wilde
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hailey Eckersberg
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Rahul Suresh
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Xinzhu Tan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Hong Chen
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - W Todd Farmer
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Center, Montreal General Hospital, Montreal, QC, Canada
| | - Kuwook Cha
- Department of Physiology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Phuong Uyen Le
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Helene Catoire
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Daniel Rochefort
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Tony Kwan
- McGill Genome Center and Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Brian A Yee
- Department of Cellular and Molecular Medicine, Stem Cell Program, Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Patrick Dion
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Arjun Krishnaswamy
- Department of Physiology, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Jean-Francois Cloutier
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Stefano Stifani
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Kevin Petrecca
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, Stem Cell Program, Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Keith K Murai
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Center, Montreal General Hospital, Montreal, QC, Canada
| | - Guoping Feng
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Guy A Rouleau
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
| | - Trey Ideker
- Division of Genetics, Department of Medicine, University of California, San Diego, San Diego, CA, USA.
| | - Neville E Sanjana
- New York Genome Center, New York, NY, USA; Department of Biology, New York University, New York, NY, USA
| | - Yang Zhou
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada.
| |
Collapse
|
16
|
Franklin JP, Testen A, Mieczkowski PA, Hepperla A, Crynen G, Simon JM, Wood JD, Harder EV, Bellinger TJ, Witt EA, Powell NL, Reissner KJ. Investigating cocaine- and abstinence-induced effects on astrocyte gene expression in the nucleus accumbens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.05.606656. [PMID: 39149305 PMCID: PMC11326167 DOI: 10.1101/2024.08.05.606656] [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: 08/17/2024]
Abstract
In recent years, astrocytes have been increasingly implicated in cellular mechanisms of substance use disorders (SUD). Astrocytes are structurally altered following exposure to drugs of abuse; specifically, astrocytes within the nucleus accumbens (NAc) exhibit significantly decreased surface area, volume, and synaptic colocalization after operant self-administration of cocaine and extinction or protracted abstinence (45 days). However, the mechanisms that elicit these morphological modifications are unknown. The current study aims to elucidate the molecular modifications that lead to observed astrocyte structural changes in rats across cocaine abstinence using astrocyte-specific RiboTag and RNA-seq, as an unbiased, comprehensive approach to identify genes whose transcription or translation change within NAc astrocytes following cocaine self-administration and extended abstinence. Using this method, our data reveal cellular processes including cholesterol biosynthesis that are altered specifically by cocaine self-administration and abstinence, suggesting that astrocyte involvement in these processes is changed in cocaine-abstinent rats. Overall, the results of this study provide insight into astrocyte functional adaptations that occur due to cocaine exposure or during cocaine withdrawal, which may pinpoint further mechanisms that contribute to cocaine-seeking behavior.
Collapse
Affiliation(s)
- Janay P Franklin
- Neuroscience Center, University of North Carolina at Chapel Hill
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill
| | - Anze Testen
- Department of Neuroscience, Medical University of South Carolina
| | | | - Austin Hepperla
- Department of Genetics, University of North Carolina at Chapel Hill
| | - Gogce Crynen
- Bioinformatics and Statistics Core, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology
| | - Jeremy M Simon
- Department of Data Science, Dana-Farber Institute Department of Biostatistics, Harvard T.H. Chan School of Public Health
| | - Jonathan D Wood
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill
| | - Eden V Harder
- Neuroscience Center, University of North Carolina at Chapel Hill
| | - Tania J Bellinger
- Department of Pharmacology, University of North Carolina at Chapel Hill
| | - Emily A Witt
- Department of Medical Neuroscience, Dalhousie University
| | - N LaShae Powell
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill
| | - Kathryn J Reissner
- Neuroscience Center, University of North Carolina at Chapel Hill
- Department of Psychology & Neuroscience, University of North Carolina at Chapel Hill
| |
Collapse
|
17
|
Liddelow SA, Olsen ML, Sofroniew MV. Reactive Astrocytes and Emerging Roles in Central Nervous System (CNS) Disorders. Cold Spring Harb Perspect Biol 2024; 16:a041356. [PMID: 38316554 PMCID: PMC11216178 DOI: 10.1101/cshperspect.a041356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
In addition to their many functions in the healthy central nervous system (CNS), astrocytes respond to CNS damage and disease through a process called "reactivity." Recent evidence reveals that astrocyte reactivity is a heterogeneous spectrum of potential changes that occur in a context-specific manner. These changes are determined by diverse signaling events and vary not only with the nature and severity of different CNS insults but also with location in the CNS, genetic predispositions, age, and potentially also with "molecular memory" of previous reactivity events. Astrocyte reactivity can be associated with both essential beneficial functions as well as with harmful effects. The available information is rapidly expanding and much has been learned about molecular diversity of astrocyte reactivity. Emerging functional associations point toward central roles for astrocyte reactivity in determining the outcome in CNS disorders.
Collapse
Affiliation(s)
- Shane A Liddelow
- Neuroscience Institute, NYU School of Medicine, New York, New York 10016, USA
- Department of Neuroscience and Physiology, NYU School of Medicine, New York, New York 10016, USA
- Department of Ophthalmology, NYU School of Medicine, New York, New York 10016, USA
| | - Michelle L Olsen
- School of Neuroscience, Virginia Tech, Blacksburg, Virginia 24061, USA
| | - Michael V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, California 90095, USA
| |
Collapse
|
18
|
Baldwin KT, Murai KK, Khakh BS. Astrocyte morphology. Trends Cell Biol 2024; 34:547-565. [PMID: 38180380 PMCID: PMC11590062 DOI: 10.1016/j.tcb.2023.09.006] [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: 07/13/2023] [Revised: 09/18/2023] [Accepted: 09/29/2023] [Indexed: 01/06/2024]
Abstract
Astrocytes are predominant glial cells that tile the central nervous system (CNS). A cardinal feature of astrocytes is their complex and visually enchanting morphology, referred to as bushy, spongy, and star-like. A central precept of this review is that such complex morphological shapes evolved to allow astrocytes to contact and signal with diverse cells at a range of distances in order to sample, regulate, and contribute to the extracellular milieu, and thus participate widely in cell-cell signaling during physiology and disease. The recent use of improved imaging methods and cell-specific molecular evaluations has revealed new information on the structural organization and molecular underpinnings of astrocyte morphology, the mechanisms of astrocyte morphogenesis, and the contributions to disease states of reduced morphology. These insights have reignited interest in astrocyte morphological complexity as a cornerstone of fundamental glial biology and as a critical substrate for multicellular spatial and physiological interactions in the CNS.
Collapse
Affiliation(s)
- Katherine T Baldwin
- Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Keith K Murai
- Centre for Research in Neuroscience, Department of Neurology and Neurosurgery, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Avenue, Montreal, QC H3G 1A4, Canada.
| | - Baljit S Khakh
- Department of Physiology, David Geffen School of Medicine, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90034, USA; Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, 10833 Le Conte Avenue, Los Angeles, CA 90034, USA.
| |
Collapse
|
19
|
Jia Y, Zhang X, Wang Y, Liu Y, Dai J, Zhang L, Wu X, Zhang J, Xiang H, Yang Y, Zeng Z, Chen Y. Knocking out Selenium Binding Protein 1 Induces Depressive-Like Behavior in Mice. Biol Trace Elem Res 2024; 202:3149-3162. [PMID: 37801218 DOI: 10.1007/s12011-023-03894-8] [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: 05/04/2023] [Accepted: 09/27/2023] [Indexed: 10/07/2023]
Abstract
Selenium binding protein 1 (SELENBP1) is involved in neurologic disorders, such as multiple sclerosis, spinal cord injury, Parkinson's disease, epilepsy, and schizophrenia. However, the role of SELENBP1 in the neurogenesis of depression, which is a neurologic disorder, and the underlying mechanisms of oxidative stress and inflammation in depression remain unknown. In this study, we evaluated the changes in the expression levels of SELENBP1 in the hippocampus of a mouse model of depression and in the serum of human patients with depression using the Gene Expression Omnibus database. These changes were validated using blood samples from human patients with depression and mouse models with chronic unpredictable mild stress (CUMS)-induced depressive-like behavior. We also investigated the effects of SELENBP1 knockout (KO) on inflammation, oxidative stress, and hippocampal neurogenesis in mice with CUMS-induced depression. Our results revealed that SELENBP1 levels was decreased in the blood of human patients with depression and in the hippocampus of mice with CUMS-induced depression. SELENBP1 KO increased CUMS-induced depressive behavior in mice and caused dysregulation of inflammatory cytokines and oxidative stress. This led to a decrease in the numbers of doublecortin- and Ki67-positive cells, which might aggravate CUMS-induced depressive symptoms. These findings suggest that SELENBP1 might be involved in the regulation of neurogenesis in mice with depression and could be served as a potential target for diagnosing and treating depression.
Collapse
Affiliation(s)
- Yi Jia
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Cellular Immunotherapy Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550025, China.
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550025, China.
| | - Xin Zhang
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Cellular Immunotherapy Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550025, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550025, China
| | - Yongmei Wang
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Cellular Immunotherapy Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550025, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550025, China
| | - Yang Liu
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Cellular Immunotherapy Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550025, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550025, China
| | - Jie Dai
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Cellular Immunotherapy Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550025, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550025, China
| | - Liangliang Zhang
- Prenatal Diagnosis Center, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Xian Wu
- Prenatal Diagnosis Center, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Jie Zhang
- Department of Laboratory, the Second People's Hospital of Guizhou Province, Guiyang, 550004, Guizhou, China
| | - Hongxi Xiang
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Cellular Immunotherapy Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550025, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550025, China
| | - Yanping Yang
- Department of Histology and Embryology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550025, China
| | - Zhu Zeng
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Cellular Immunotherapy Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550025, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guiyang, 550025, China
| | - Yulian Chen
- Mental Health Education and Counseling Center for College Students, Guizhou Medical University, Guiyang, 550025, China
- Faculty of Psychology, Beijing Normal University, Beijing, 100875, China
| |
Collapse
|
20
|
Rezaei S, Prévot TD, Vieira E, Sibille E. LPS-induced inflammation reduces GABAergic interneuron markers and brain-derived neurotrophic factor in mouse prefrontal cortex and hippocampus. Brain Behav Immun Health 2024; 38:100761. [PMID: 38586282 PMCID: PMC10992730 DOI: 10.1016/j.bbih.2024.100761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 02/20/2024] [Accepted: 03/17/2024] [Indexed: 04/09/2024] Open
Abstract
Inflammation, reduced gamma-aminobutyric acidergic (GABAergic) function and altered neuroplasticity are co-occurring pathophysiologies in major depressive disorder (MDD). However, the link between these biological changes remains unclear. We hypothesized that inflammation induces deficits in GABAergic interneuron markers and that this effect is mediated by brain-derived neurotrophic factor (BDNF). We report here that systemic inflammation induced by intraperitoneal injection of lipopolysaccharide (LPS) (0.125, 0.25, 0.5, 1, 2 mg/kg) in the first cohort of C57BL/6 mice (n = 72; 10-11 weeks; 50% female) resulted in increased interleukin 1-beta and interleukin-6 in prefrontal cortex (PFC) and hippocampus (HPC), as measured using enzyme-linked immunosorbent assay (ELISA). Quantitative real-time polymerase reaction (qPCR) was used to explore the effect of LPS on the expression of GABAergic interneuron markers. In the PFC of the second cohort (n = 39; 10-11 weeks; 50% female), 2 mg/kg of LPS decreased the expression of somatostatin (Sst) (p = 0.0014), parvalbumin (Pv) (p = 0.0257), cortistatin (Cort) (p = 0.0003), neuropeptide Y (Npy) (p = 0.0033) and cholecystokinin (Cck) (p = 0.0041), and did not affect corticotropin-releasing hormone (Crh) and vasoactive intestinal peptide (Vip) expression. In the HPC, 2 mg/kg of LPS decreased the expression of Sst (p = 0.0543), Cort (p = 0.0011), Npy (p = 0.0001), and Cck (p < 0.0001), and did not affect Crh, Pv, and Vip expression. LPS decreased the expression of Bdnf in the PFC (p < 0.0001) and HPC (p = 0.0003), which significantly correlated with affected markers (Sst, Pv, Cort, Cck, and Npy). Collectively, these results suggest that inflammation may causally contribute to cortical cell microcircuit GABAergic deficits observed in MDD.
Collapse
Affiliation(s)
- Sara Rezaei
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, M5S 1A8, Canada
- Campbell Family Mental Health Research Institute at CAMH, Toronto, M5T 1R8, Canada
| | - Thomas D. Prévot
- Campbell Family Mental Health Research Institute at CAMH, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, M5T 1R8, Canada
| | - Erica Vieira
- Campbell Family Mental Health Research Institute at CAMH, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, M5T 1R8, Canada
| | - Etienne Sibille
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, M5S 1A8, Canada
- Campbell Family Mental Health Research Institute at CAMH, Toronto, M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, Toronto, M5T 1R8, Canada
| |
Collapse
|
21
|
Makarava N, Kushwaha R, Baskakov IV. Reactive astrocytes in prion diseases: Friend or foe? PLoS Pathog 2024; 20:e1012286. [PMID: 38900746 PMCID: PMC11189187 DOI: 10.1371/journal.ppat.1012286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024] Open
Affiliation(s)
- Natallia Makarava
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Rajesh Kushwaha
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Ilia V. Baskakov
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
22
|
Xu M, Wang H, Ren S, Wang B, Yang W, Lv L, Sha X, Li W, Wang Y. Identification of crucial inflammaging related risk factors in multiple sclerosis. Front Mol Neurosci 2024; 17:1398665. [PMID: 38836117 PMCID: PMC11148336 DOI: 10.3389/fnmol.2024.1398665] [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: 03/10/2024] [Accepted: 04/30/2024] [Indexed: 06/06/2024] Open
Abstract
Background Multiple sclerosis (MS) is an immune-mediated disease characterized by inflammatory demyelinating lesions in the central nervous system. Studies have shown that the inflammation is vital to both the onset and progression of MS, where aging plays a key role in it. However, the potential mechanisms on how aging-related inflammation (inflammaging) promotes MS have not been fully understood. Therefore, there is an urgent need to integrate the underlying mechanisms between inflammaging and MS, where meaningful prediction models are needed. Methods First, both aging and disease models were developed using machine learning methods, respectively. Then, an integrated inflammaging model was used to identify relative risk factors, by identifying essential "aging-inflammation-disease" triples. Finally, a series of bioinformatics analyses (including network analysis, enrichment analysis, sensitivity analysis, and pan-cancer analysis) were further used to explore the potential mechanisms between inflammaging and MS. Results A series of risk factors were identified, such as the protein homeostasis, cellular homeostasis, neurodevelopment and energy metabolism. The inflammaging indices were further validated in different cancer types. Therefore, various risk factors were integrated, and even both the theories of inflammaging and immunosenescence were further confirmed. Conclusion In conclusion, our study systematically investigated the potential relationships between inflammaging and MS through a series of computational approaches, and could present a novel thought for other aging-related diseases.
Collapse
Affiliation(s)
- Mengchu Xu
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, Liaoning, China
| | - Huize Wang
- Department of Nursing, First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Siwei Ren
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, Liaoning, China
| | - Bing Wang
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, Liaoning, China
| | - Wenyan Yang
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, Liaoning, China
| | - Ling Lv
- Department of Thorax, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xianzheng Sha
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, Liaoning, China
| | - Wenya Li
- Department of Thorax, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yin Wang
- Department of Biomedical Engineering, School of Intelligent Sciences, China Medical University, Shenyang, Liaoning, China
- Tumor Etiology and Screening Department of Cancer Institute and General Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
23
|
Sun R, Tang MY, Yang D, Zhang YY, Xu YH, Qiao Y, Yu B, Cao SX, Wang H, Huang HQ, Zhang H, Li XM, Lian H. C3aR in the medial prefrontal cortex modulates the susceptibility to LPS-induced depressive-like behaviors through glutamatergic neuronal excitability. Prog Neurobiol 2024; 236:102614. [PMID: 38641040 DOI: 10.1016/j.pneurobio.2024.102614] [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: 11/07/2023] [Revised: 03/18/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
Complement activation and prefrontal cortical dysfunction both contribute to the pathogenesis of major depressive disorder (MDD), but their interplay in MDD is unclear. We here studied the role of complement C3a receptor (C3aR) in the medial prefrontal cortex (mPFC) and its influence on depressive-like behaviors induced by systematic lipopolysaccharides (LPS) administration. C3aR knockout (KO) or intra-mPFC C3aR antagonism confers resilience, whereas C3aR expression in mPFC neurons makes KO mice susceptible to LPS-induced depressive-like behaviors. Importantly, the excitation and inhibition of mPFC neurons have opposing effects on depressive-like behaviors, aligning with increased and decreased excitability by C3aR deletion and activation in cortical neurons. In particular, inhibiting mPFC glutamatergic (mPFCGlu) neurons, the main neuronal subpopulation expresses C3aR, induces depressive-like behaviors in saline-treated WT and KO mice, but not in LPS-treated KO mice. Compared to hypoexcitable mPFCGlu neurons in LPS-treated WT mice, C3aR-null mPFCGlu neurons display hyperexcitability upon LPS treatment, and enhanced excitation of mPFCGlu neurons is anti-depressant, suggesting a protective role of C3aR deficiency in these circumstances. In conclusion, C3aR modulates susceptibility to LPS-induced depressive-like behaviors through mPFCGlu neuronal excitability. This study identifies C3aR as a pivotal intersection of complement activation, mPFC dysfunction, and depression and a promising therapeutic target for MDD.
Collapse
Affiliation(s)
- Rui Sun
- Department of Neurology and Department of Psychiatry of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Research Center of System Medicine, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China; Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, China
| | - Meng-Yu Tang
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Dan Yang
- Clinical Research Center, The second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yan-Yi Zhang
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Yi-Heng Xu
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Yong Qiao
- Department of Neurology and Department of Psychiatry of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Research Center of System Medicine, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China
| | - Bin Yu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
| | - Shu-Xia Cao
- Department of Neurology, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hao Wang
- Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui-Qian Huang
- Clinical Research Center, The second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hong Zhang
- Department of Nuclear Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao-Ming Li
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Hong Lian
- Department of Neurology and Department of Psychiatry of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Research Center of System Medicine, School of Basic Medical Sciences, Zhejiang University School of Medicine, Hangzhou, China; Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China.
| |
Collapse
|
24
|
Zimmer TS, Orr AL, Orr AG. Astrocytes in selective vulnerability to neurodegenerative disease. Trends Neurosci 2024; 47:289-302. [PMID: 38521710 PMCID: PMC11006581 DOI: 10.1016/j.tins.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 02/06/2024] [Accepted: 02/26/2024] [Indexed: 03/25/2024]
Abstract
Selective vulnerability of specific brain regions and cell populations is a hallmark of neurodegenerative disorders. Mechanisms of selective vulnerability involve neuronal heterogeneity, functional specializations, and differential sensitivities to stressors and pathogenic factors. In this review we discuss the growing body of literature suggesting that, like neurons, astrocytes are heterogeneous and specialized, respond to and integrate diverse inputs, and induce selective effects on brain function. In disease, astrocytes undergo specific, context-dependent changes that promote different pathogenic trajectories and functional outcomes. We propose that astrocytes contribute to selective vulnerability through maladaptive transitions to context-divergent phenotypes that impair specific brain regions and functions. Further studies on the multifaceted roles of astrocytes in disease may provide new therapeutic approaches to enhance resilience against neurodegenerative disorders.
Collapse
Affiliation(s)
- Till S Zimmer
- Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, USA; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Adam L Orr
- Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, USA; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Anna G Orr
- Appel Alzheimer's Disease Research Institute, Weill Cornell Medicine, New York, NY, USA; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA; Neuroscience Graduate Program, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
25
|
McCallum S, Suresh KB, Islam T, Saustad AW, Shelest O, Patil A, Lee D, Kwon B, Yenokian I, Kawaguchi R, Beveridge CH, Manchandra P, Randolph CE, Meares GP, Dutta R, Plummer J, Knott SRV, Chopra G, Burda JE. Lesion-remote astrocytes govern microglia-mediated white matter repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.15.585251. [PMID: 38558977 PMCID: PMC10979953 DOI: 10.1101/2024.03.15.585251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Spared regions of the damaged central nervous system undergo dynamic remodeling and exhibit a remarkable potential for therapeutic exploitation. Here, lesion-remote astrocytes (LRAs), which interact with viable neurons, glia and neural circuitry, undergo reactive transformations whose molecular and functional properties are poorly understood. Using multiple transcriptional profiling methods, we interrogated LRAs from spared regions of mouse spinal cord following traumatic spinal cord injury (SCI). We show that LRAs acquire a spectrum of molecularly distinct, neuroanatomically restricted reactivity states that evolve after SCI. We identify transcriptionally unique reactive LRAs in degenerating white matter that direct the specification and function of local microglia that clear lipid-rich myelin debris to promote tissue repair. Fueling this LRA functional adaptation is Ccn1 , which encodes for a secreted matricellular protein. Loss of astrocyte CCN1 leads to excessive, aberrant activation of local microglia with (i) abnormal molecular specification, (ii) dysfunctional myelin debris processing, and (iii) impaired lipid metabolism, culminating in blunted debris clearance and attenuated neurological recovery from SCI. Ccn1 -expressing white matter astrocytes are specifically induced by local myelin damage and generated in diverse demyelinating disorders in mouse and human, pointing to their fundamental, evolutionarily conserved role in white matter repair. Our findings show that LRAs assume regionally divergent reactivity states with functional adaptations that are induced by local context-specific triggers and influence disorder outcome. Astrocytes tile the central nervous system (CNS) where they serve vital roles that uphold healthy nervous system function, including regulation of synapse development, buffering of neurotransmitters and ions, and provision of metabolic substrates 1 . In response to diverse CNS insults, astrocytes exhibit disorder-context specific transformations that are collectively referred to as reactivity 2-5 . The characteristics of regionally and molecularly distinct reactivity states are incompletely understood. The mechanisms through which distinct reactivity states arise, how they evolve or resolve over time, and their consequences for local cell function and CNS disorder progression remain enigmatic. Immediately adjacent to CNS lesions, border-forming astrocytes (BFAs) undergo transcriptional reprogramming and proliferation to form a neuroprotective barrier that restricts inflammation and supports axon regeneration 6-9 . Beyond the lesion, spared but dynamic regions of the injured CNS exhibit varying degrees of synaptic circuit remodeling and progressive cellular responses to secondary damage that have profound consequences for neural repair and recovery 10,11 . Throughout these cytoarchitecturally intact, but injury-reactive regions, lesion-remote astrocytes (LRAs) intermingle with neurons and glia, undergo little to no proliferation, and exhibit varying degrees of cellular hypertrophy 7,12,13 . The molecular and functional properties of LRAs remain grossly undefined. Therapeutically harnessing spared regions of the injured CNS will require a clearer understanding of the accompanying cellular and molecular landscape. Here, we leveraged integrative transcriptional profiling methodologies to identify multiple spatiotemporally resolved, molecularly distinct states of LRA reactivity within the injured spinal cord. Computational modeling of LRA-mediated heterotypic cell interactions, astrocyte-specific conditional gene deletion, and multiple mouse models of acute and chronic CNS white matter degeneration were used to interrogate a newly identified white matter degeneration-reactive astrocyte subtype. We define how this reactivity state is induced and its role in governing the molecular and functional specification of local microglia that clear myelin debris from the degenerating white matter to promote repair.
Collapse
|
26
|
Wei H, Withrow J, Rakshit J, Ul Amin F, Nahm J, Mowry FE, Mao Z, Bhattacharjee MB, Zhu JJ, Yang Y, Wu JQ. The identification of a Distinct Astrocyte Subtype that Diminishes in Alzheimer's Disease. Aging Dis 2024; 15:2752-2769. [PMID: 38502590 PMCID: PMC11567244 DOI: 10.14336/ad.2024.0205-1] [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: 12/03/2023] [Accepted: 02/05/2024] [Indexed: 03/21/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by the presence of two hallmark pathologies: the accumulation of Amyloid beta (Aβ) and tau proteins in the brain. There is a growing body of evidence suggesting that astrocytes, a type of glial cell in the brain, play crucial roles in clearing Aβ and binding to tau proteins. However, due to the heterogeneity of astrocytes, the specific roles of different astrocyte subpopulations in response to Aβ and tau remain unclear. To enhance the understanding of astrocyte subpopulations in AD, we investigated astrocyte lineage cells based on single-nuclei transcriptomic data obtained from both human and mouse samples. We characterized the diversity of astrocytes and identified global and subpopulation-specific transcriptomic changes between control and AD samples. Our findings revealed the existence of a specific astrocyte subpopulation marked by low levels of GFAP and the presence of AQP4 and CD63 expression, which showed functional enrichment in Aβ clearance and tau protein binding, and diminished in AD. We verified this type of astrocytes in mouse models and in AD patient brain samples. Furthermore, our research also unveiled significant alterations of the ligand-receptor interactions between astrocytes and other cell types. These changes underscore the complex interplay between astrocytes and neighboring cells in the context of AD. Overall, our work gives insights into astrocyte heterogeneity in the context of AD and reveals a distinct astrocyte subpopulation that holds potential for therapeutic interventions in AD. Targeting specific astrocyte subpopulations may offer new avenues for the development of novel treatments for AD.
Collapse
Affiliation(s)
- Haichao Wei
- The Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
- Center for Stem Cell and Regenerative Medicine, UT Brown Foundation Institute of Molecular Medicine, Houston, TX, USA.
| | - Joseph Withrow
- The Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Jyotirmoy Rakshit
- The Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
- Center for Stem Cell and Regenerative Medicine, UT Brown Foundation Institute of Molecular Medicine, Houston, TX, USA.
| | - Faiz Ul Amin
- The Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
- Center for Stem Cell and Regenerative Medicine, UT Brown Foundation Institute of Molecular Medicine, Houston, TX, USA.
| | - Joshua Nahm
- The Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Francesca E. Mowry
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA.
| | - Zhengmei Mao
- Center for Stem Cell and Regenerative Medicine, UT Brown Foundation Institute of Molecular Medicine, Houston, TX, USA.
| | - Meenakshi B. Bhattacharjee
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Jay-Jiguang Zhu
- The Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Yongjie Yang
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA.
| | - Jia Qian Wu
- The Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
- Center for Stem Cell and Regenerative Medicine, UT Brown Foundation Institute of Molecular Medicine, Houston, TX, USA.
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
| |
Collapse
|
27
|
Yang K, Liu Y, Zhang M. The Diverse Roles of Reactive Astrocytes in the Pathogenesis of Amyotrophic Lateral Sclerosis. Brain Sci 2024; 14:158. [PMID: 38391732 PMCID: PMC10886687 DOI: 10.3390/brainsci14020158] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/17/2024] [Accepted: 01/29/2024] [Indexed: 02/24/2024] Open
Abstract
Astrocytes displaying reactive phenotypes are characterized by their ability to remodel morphologically, molecularly, and functionally in response to pathological stimuli. This process results in the loss of their typical astrocyte functions and the acquisition of neurotoxic or neuroprotective roles. A growing body of research indicates that these reactive astrocytes play a pivotal role in the pathogenesis of amyotrophic lateral sclerosis (ALS), involving calcium homeostasis imbalance, mitochondrial dysfunction, abnormal lipid and lactate metabolism, glutamate excitotoxicity, etc. This review summarizes the characteristics of reactive astrocytes, their role in the pathogenesis of ALS, and recent advancements in astrocyte-targeting strategies.
Collapse
Affiliation(s)
- Kangqin Yang
- Department of Neurology and Psychiatry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yang Liu
- Department of Neurology and Psychiatry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Min Zhang
- Department of Neurology and Psychiatry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
28
|
Mittli D. Inflammatory processes in the prefrontal cortex induced by systemic immune challenge: Focusing on neurons. Brain Behav Immun Health 2023; 34:100703. [PMID: 38033612 PMCID: PMC10682838 DOI: 10.1016/j.bbih.2023.100703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/30/2023] [Accepted: 11/04/2023] [Indexed: 12/02/2023] Open
Abstract
Peripheral immune challenge induces neurobiological alterations in the brain and related neuropsychiatric symptoms both in humans and other mammals. One of the best known physiological effects of systemic inflammation is sickness behavior. However, in addition to this depression-like state, there are other cognitive outcomes of peripherally induced neuroinflammation that can be linked to the dysfunction of higher-order cortical areas, such as the prefrontal cortex (PFC). As the physiological activity of the PFC is largely based on the balanced interplay of excitatory pyramidal cells and inhibitory interneurons, it may be hypothesized that neuroinflammatory processes result in a shift of excitatory/inhibitory balance, which is a common hallmark of several neuropsychiatric conditions. Indeed, many data suggest that peripherally induced neuroinflammation is strongly associated with molecular and functional changes in PFC neurons leading to disturbances in their synaptic networks. Different experimental approaches may cause some incongruence in the reviewed data. However, it is commonly agreed that acute systemic inflammation leads to changes in the excitatory/inhibitory balance in the PFC by proinflammatory signaling at the brain borders and in the brain parenchyma. These cellular changes result in altered local and brain-wide network activity inducing disturbances in the top-down control of goal-directed behavior and cognition regulated by the PFC. Lipopolysaccharide (LPS)-treated rodents are the most widely used experimental models of peripherally induced neuroinflammation, so the majority of the reviewed data come from studies utilizing the LPS model. This may limit their general interpretation regarding the neuronal effects of peripheral immune activation. In addition, several biological variables (e.g., sex, age) can influence the PFC effects of systemic immune challenge, not only the nature and severity of immune activation. Therefore, it would be desirable to investigate inflammation-related neuronal changes in the PFC using other models of systemic inflammation as well, and to focus on the targeted fine-tuning of the affected cell types via common molecular mechanisms of the immune and nervous systems.
Collapse
Affiliation(s)
- Dániel Mittli
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Laboratory of Proteomics, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Department of Physiology and Neurobiology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- InnoScience Ltd., Mátranovák, Hungary
| |
Collapse
|
29
|
Verkhratsky A, Butt A, Li B, Illes P, Zorec R, Semyanov A, Tang Y, Sofroniew MV. Astrocytes in human central nervous system diseases: a frontier for new therapies. Signal Transduct Target Ther 2023; 8:396. [PMID: 37828019 PMCID: PMC10570367 DOI: 10.1038/s41392-023-01628-9] [Citation(s) in RCA: 117] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 08/15/2023] [Accepted: 08/22/2023] [Indexed: 10/14/2023] Open
Abstract
Astroglia are a broad class of neural parenchymal cells primarily dedicated to homoeostasis and defence of the central nervous system (CNS). Astroglia contribute to the pathophysiology of all neurological and neuropsychiatric disorders in ways that can be either beneficial or detrimental to disorder outcome. Pathophysiological changes in astroglia can be primary or secondary and can result in gain or loss of functions. Astroglia respond to external, non-cell autonomous signals associated with any form of CNS pathology by undergoing complex and variable changes in their structure, molecular expression, and function. In addition, internally driven, cell autonomous changes of astroglial innate properties can lead to CNS pathologies. Astroglial pathophysiology is complex, with different pathophysiological cell states and cell phenotypes that are context-specific and vary with disorder, disorder-stage, comorbidities, age, and sex. Here, we classify astroglial pathophysiology into (i) reactive astrogliosis, (ii) astroglial atrophy with loss of function, (iii) astroglial degeneration and death, and (iv) astrocytopathies characterised by aberrant forms that drive disease. We review astroglial pathophysiology across the spectrum of human CNS diseases and disorders, including neurotrauma, stroke, neuroinfection, autoimmune attack and epilepsy, as well as neurodevelopmental, neurodegenerative, metabolic and neuropsychiatric disorders. Characterising cellular and molecular mechanisms of astroglial pathophysiology represents a new frontier to identify novel therapeutic strategies.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- International Joint Research Centre on Purinergic Signalling/School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China.
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
- Achucarro Centre for Neuroscience, IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
- Department of Stem Cell Biology, State Research Institute Centre for Innovative Medicine, LT-01102, Vilnius, Lithuania.
| | - Arthur Butt
- Institute of Biomedical and Biomolecular Sciences, School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Baoman Li
- Department of Forensic Analytical Toxicology, School of Forensic Medicine, China Medical University, Shenyang, China
| | - Peter Illes
- International Joint Research Centre on Purinergic Signalling/School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Rudolf Boehm Institute for Pharmacology and Toxicology, University of Leipzig, 04109, Leipzig, Germany
| | - Robert Zorec
- Celica Biomedical, Lab Cell Engineering, Technology Park, 1000, Ljubljana, Slovenia
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, University of Ljubljana, Faculty of Medicine, Ljubljana, Slovenia
| | - Alexey Semyanov
- Department of Physiology, Jiaxing University College of Medicine, 314033, Jiaxing, China
| | - Yong Tang
- International Joint Research Centre on Purinergic Signalling/School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
- Key Laboratory of Acupuncture for Senile Disease (Chengdu University of TCM), Ministry of Education/Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China.
| | - Michael V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
30
|
Panchenko PE, Hippauf L, Konsman JP, Badaut J. Do astrocytes act as immune cells after pediatric TBI? Neurobiol Dis 2023; 185:106231. [PMID: 37468048 PMCID: PMC10530000 DOI: 10.1016/j.nbd.2023.106231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/28/2023] [Accepted: 07/15/2023] [Indexed: 07/21/2023] Open
Abstract
Astrocytes are in contact with the vasculature, neurons, oligodendrocytes and microglia, forming a local network with various functions critical for brain homeostasis. One of the primary responders to brain injury are astrocytes as they detect neuronal and vascular damage, change their phenotype with morphological, proteomic and transcriptomic transformations for an adaptive response. The role of astrocytic responses in brain dysfunction is not fully elucidated in adult, and even less described in the developing brain. Children are vulnerable to traumatic brain injury (TBI), which represents a leading cause of death and disability in the pediatric population. Pediatric brain trauma, even with mild severity, can lead to long-term health complications, such as cognitive impairments, emotional disorders and social dysfunction later in life. To date, the underlying pathophysiology is still not fully understood. In this review, we focus on the astrocytic response in pediatric TBI and propose a potential immune role of the astrocyte in response to trauma. We discuss the contribution of astrocytes in the local inflammatory cascades and secretion of various immunomodulatory factors involved in the recruitment of local microglial cells and peripheral immune cells through cerebral blood vessels. Taken together, we propose that early changes in the astrocytic phenotype can alter normal development of the brain, with long-term consequences on neurological outcomes, as described in preclinical models and patients.
Collapse
Affiliation(s)
| | - Lea Hippauf
- CNRS UMR 5536 RMSB-University of Bordeaux, Bordeaux, France
| | | | - Jerome Badaut
- CNRS UMR 5536 RMSB-University of Bordeaux, Bordeaux, France; Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| |
Collapse
|
31
|
Makarava N, Mychko O, Molesworth K, Chang JCY, Henry RJ, Tsymbalyuk N, Gerzanich V, Simard JM, Loane DJ, Baskakov IV. Region-Specific Homeostatic Identity of Astrocytes Is Essential for Defining Their Response to Pathological Insults. Cells 2023; 12:2172. [PMID: 37681904 PMCID: PMC10486627 DOI: 10.3390/cells12172172] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/08/2023] [Accepted: 08/28/2023] [Indexed: 09/09/2023] Open
Abstract
The transformation of astrocytes into reactive states constitutes a biological response of the central nervous system under a variety of pathological insults. Astrocytes display diverse homeostatic identities that are developmentally predetermined and regionally specified. Upon transformation into reactive states associated with neurodegenerative diseases and other neurological disorders, astrocytes acquire diverse reactive phenotypes. However, it is not clear whether their reactive phenotypes are dictated by region-specific homeostatic identity or by the nature of an insult. To address this question, region-specific gene expression profiling was performed for four brain regions (cortex, hippocampus, thalamus, and hypothalamus) in mice using a custom NanoString panel consisting of selected sets of genes associated with astrocyte functions and their reactivity for five conditions: prion disease, traumatic brain injury, brain ischemia, 5XFAD Alzheimer's disease model and normal aging. Upon transformation into reactive states, genes that are predominantly associated with astrocytes were found to respond to insults in a region-specific manner. Regardless of the nature of the insult or the insult-specificity of astrocyte response, strong correlations between undirected GSA (gene set analysis) scores reporting on astrocyte reactivity and on their homeostatic functions were observed within each individual brain region. The insult-specific gene expression signatures did not separate well from each other and instead partially overlapped, forming continuums. The current study demonstrates that region-specific homeostatic identities of astrocytes are important for defining their response to pathological insults. Within region-specific populations, reactive astrocytes show continuums of gene expression signatures, partially overlapping between individual insults.
Collapse
Affiliation(s)
- Natallia Makarava
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (N.M.); (J.C.-Y.C.)
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Olga Mychko
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (N.M.); (J.C.-Y.C.)
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Kara Molesworth
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (N.M.); (J.C.-Y.C.)
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jennifer Chen-Yu Chang
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (N.M.); (J.C.-Y.C.)
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Rebecca J. Henry
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA (D.J.L.)
| | - Natalya Tsymbalyuk
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - J. Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - David J. Loane
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA (D.J.L.)
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland
| | - Ilia V. Baskakov
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (N.M.); (J.C.-Y.C.)
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
32
|
Wang J, Deng X, Jiang J, Yao Z, Ju Y, Luo Y. Evaluation of electroacupuncture as a non-pharmacological therapy for astrocytic structural aberrations and behavioral deficits in a post-ischemic depression model in mice. Front Behav Neurosci 2023; 17:1239024. [PMID: 37700911 PMCID: PMC10493307 DOI: 10.3389/fnbeh.2023.1239024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/07/2023] [Indexed: 09/14/2023] Open
Abstract
Background Ascending clinical evidence supports that electroacupuncture (EA) is effective in treating post-ischemic depression (PID), but little is known about how it works at the cellular level. Astrocytes are exquisitely sensitive to their extracellular environment, and under stressful conditions, they may experience aberrant structural remodeling that can potentially cause neuroplastic disturbances and contribute to subsequent changes in mood or behavior. Objectives This study aimed to investigate the effect of EA on behavioral deficits associated with PID in mice and verify the hypothesis that astrocytic morphology may be involved in this impact. Methods We established a PID animal model induced by transient bilateral common carotid artery occlusion (BCCAO, 20 min) and chronic restraint stress (CRS, 21 days). EA treatment (GV20 + ST36) was performed for 3 weeks, from Monday to Friday each week. Depressive- and anxiety-like behaviors and sociability were evaluated using SPT, FST, EPM, and SIT. Immunohistochemistry combined with Sholl and cell morphological analysis was utilized to assess the process morphology of GFAP+ astrocytes in mood-related regions. The potential relationship between morphological changes in astrocytes and behavioral output was detected by correlation analysis. Results Behavioral assays demonstrated that EA treatment induced an overall reduction in behavioral deficits, as measured by the behavioral Z-score. Sholl and morphological analyses revealed that EA prevented the decline in cell complexity of astrocytes in the prefrontal cortex (PFC) and the CA1 region of the hippocampus, where astrocytes displayed evident deramification and atrophy of the branches. Eventually, the correlation analysis showed there was a relationship between behavioral emotionality and morphological changes. Conclusion Our findings imply that EA prevents both behavioral deficits and structural abnormalities in astrocytes in the PID model. The strong correlation between behavioral Z-scores and the observed morphological changes confirms the notion that the weakening of astrocytic processes may play a crucial role in depressive symptoms, and astrocytes could be a potential target of EA in the treatment of PID.
Collapse
Affiliation(s)
- Jingwen Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xin Deng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jin Jiang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhengyu Yao
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yaxin Ju
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Luo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
33
|
Krawczyk MC, Godoy M, Vander P, Zhang AJ, Zhang Y. Loss of Serpin E2 alters antimicrobial gene expression by microglia but not astrocytes. Neurosci Lett 2023; 811:137354. [PMID: 37348749 PMCID: PMC11473033 DOI: 10.1016/j.neulet.2023.137354] [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: 02/14/2023] [Revised: 05/24/2023] [Accepted: 06/15/2023] [Indexed: 06/24/2023]
Abstract
Microglia are the brain-resident immune cells responsible for surveilling and protecting the central nervous system. These cells can express a wide array of immune genes, and that expression can become highly dynamic in response to changes in the environment, such as traumatic injury or neurological disease. Though microglial immune responses are well studied, we still do not know many mechanisms and regulators underlying all the varied microglial responses. Serpin E2 is a serine protease inhibitor that acts on a wide variety of serine proteases, with particularly potent affinity for the blood clotting enzyme thrombin. In the brain, Serpin E2 is highly expressed by many cell types, especially glia, and loss of Serpin E2 leads to behavioral changes as well as deficits in synaptic plasticity. To determine whether Serpin E2 is important for maintaining homeostasis in glia, we performed RNA sequencing of microglia and astrocytes from Serpin E2-deficient mice in a healthy state or under immune activation due to lipopolysaccharide (LPS) injection. We found that microglia in Serpin E2-deficient mice had higher expression of antimicrobial genes, while astrocytes did not display any robust changes in transcription. Furthermore, the lack of Serpin E2 did not affect transcriptional responses to LPS in either microglia or astrocytes. Overall, we find that Serpin E2 is a regulator of antimicrobial genes in microglia.
Collapse
Affiliation(s)
- Mitchell C Krawczyk
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, United States of America
| | - Marlesa Godoy
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, United States of America
| | - Paul Vander
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles (UCLA), CA, United States of America; Molecular, Cellular, and Integrative Physiology Graduate Program, University of California Los Angeles, Los Angeles (UCLA), CA, United States of America
| | - Alice J Zhang
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, United States of America
| | - Ye Zhang
- Department of Psychiatry and Biobehavioral Sciences, Intellectual and Developmental Disabilities Research Center, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, United States of America; Brain Research Institute, University of California Los Angeles (UCLA), Los Angeles, CA, United States of America; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles (UCLA), Los Angeles, CA, United States of America; Molecular Biology Institute, University of California Los Angeles (UCLA), Los Angeles, CA, United States of America.
| |
Collapse
|
34
|
Guebel DV. Human hippocampal astrocytes: Computational dissection of their transcriptome, sexual differences and exosomes across ageing and mild-cognitive impairment. Eur J Neurosci 2023; 58:2677-2707. [PMID: 37427765 DOI: 10.1111/ejn.16081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 02/20/2023] [Accepted: 06/16/2023] [Indexed: 07/11/2023]
Abstract
The role of astrocytes in Alzheimer's disease is often disregarded. Hence, characterization of astrocytes along their early evolution toward Alzheimer would be greatly beneficial. However, due to their exquisite responsiveness, in vivo studies are difficult. So public microarray data of hippocampal homogenates from (healthy) young, (healthy) elder and elder with mild cognitive impairment (MCI) were subjected to re-analysis by a multi-step computational pipeline. Ontologies and pathway analyses were compared after determining the differential genes that, belonging to astrocytes, have splice forms. Likewise, the subset of molecules exportable to exosomes was also determined. The results showed that astrocyte's phenotypes changed significantly. While already 'activated' astrocytes were found in the younger group, major changes occurred during ageing (increased vascular remodelling and response to mechanical stimulus, diminished long-term potentiation and increased long-term depression). MCI's astrocytes showed some 'rejuvenated' features, but their sensitivity to shear stress was markedly lost. Importantly, most of the changes showed to be sex biassed. Men's astrocytes are enriched in a type 'endfeet-astrocytome', whereas women's astrocytes appear close to the 'scar-forming' type (prone to endothelial dysfunction, hypercholesterolemia, loss of glutamatergic synapses, Ca+2 dysregulation, hypoxia, oxidative stress and 'pro-coagulant' phenotype). In conclusion, the computational dissection of the networks based on the hippocampal gene isoforms provides a relevant proxy to in vivo astrocytes, also revealing the occurrence of sexual differences. Analyses of the astrocytic exosomes did not provide an acceptable approximation to the overall functioning of astrocytes in the hippocampus, probably due to the selective cellular mechanisms which charge the cargo molecules.
Collapse
|
35
|
Abstract
Astrocyte endfeet enwrap the entire vascular tree within the central nervous system, where they perform important functions in regulating the blood-brain barrier (BBB), cerebral blood flow, nutrient uptake, and waste clearance. Accordingly, astrocyte endfeet contain specialized organelles and proteins, including local protein translation machinery and highly organized scaffold proteins, which anchor channels, transporters, receptors, and enzymes critical for astrocyte-vascular interactions. Many neurological diseases are characterized by the loss of polarization of specific endfoot proteins, vascular dysregulation, BBB disruption, altered waste clearance, or, in extreme cases, loss of endfoot coverage. A role for astrocyte endfeet has been demonstrated or postulated in many of these conditions. This review provides an overview of the development, composition, function, and pathological changes of astrocyte endfeet and highlights the gaps in our knowledge that future research should address.
Collapse
Affiliation(s)
- Blanca Díaz-Castro
- UK Dementia Research Institute and Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK;
| | - Stefanie Robel
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA;
| | - Anusha Mishra
- Department of Neurology Jungers Center for Neurosciences Research and Knight Cardiovascular Institute, Oregon Health & Science University, Portland, Oregon, USA;
| |
Collapse
|
36
|
Ellen O, Ye S, Nheu D, Dass M, Pagnin M, Ozturk E, Theotokis P, Grigoriadis N, Petratos S. The Heterogeneous Multiple Sclerosis Lesion: How Can We Assess and Modify a Degenerating Lesion? Int J Mol Sci 2023; 24:11112. [PMID: 37446290 DOI: 10.3390/ijms241311112] [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: 05/18/2023] [Revised: 06/21/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Multiple sclerosis (MS) is a heterogeneous disease of the central nervous system that is governed by neural tissue loss and dystrophy during its progressive phase, with complex reactive pathological cellular changes. The immune-mediated mechanisms that promulgate the demyelinating lesions during relapses of acute episodes are not characteristic of chronic lesions during progressive MS. This has limited our capacity to target the disease effectively as it evolves within the central nervous system white and gray matter, thereby leaving neurologists without effective options to manage individuals as they transition to a secondary progressive phase. The current review highlights the molecular and cellular sequelae that have been identified as cooperating with and/or contributing to neurodegeneration that characterizes individuals with progressive forms of MS. We emphasize the need for appropriate monitoring via known and novel molecular and imaging biomarkers that can accurately detect and predict progression for the purposes of newly designed clinical trials that can demonstrate the efficacy of neuroprotection and potentially neurorepair. To achieve neurorepair, we focus on the modifications required in the reactive cellular and extracellular milieu in order to enable endogenous cell growth as well as transplanted cells that can integrate and/or renew the degenerative MS plaque.
Collapse
Affiliation(s)
- Olivia Ellen
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Sining Ye
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Danica Nheu
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Mary Dass
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Maurice Pagnin
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Ezgi Ozturk
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, Department of Neurology, AHEPA University Hospital, Stilponos Kiriakides Str. 1, 54636 Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, Department of Neurology, AHEPA University Hospital, Stilponos Kiriakides Str. 1, 54636 Thessaloniki, Greece
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Melborune, VIC 3004, Australia
| |
Collapse
|
37
|
Patani R, Hardingham GE, Liddelow SA. Functional roles of reactive astrocytes in neuroinflammation and neurodegeneration. Nat Rev Neurol 2023; 19:395-409. [PMID: 37308616 DOI: 10.1038/s41582-023-00822-1] [Citation(s) in RCA: 185] [Impact Index Per Article: 92.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2023] [Indexed: 06/14/2023]
Abstract
Despite advances in uncovering the mechanisms that underlie neuroinflammation and neurodegenerative disease, therapies that prevent neuronal loss remain elusive. Targeting of disease-defining markers in conditions such as Alzheimer disease (amyloid-β and tau) or Parkinson disease (α-synuclein) has been met with limited success, suggesting that these proteins do not act in isolation but form part of a pathological network. This network could involve phenotypic alteration of multiple cell types in the CNS, including astrocytes, which have a major neurosupportive, homeostatic role in the healthy CNS but adopt reactive states under acute or chronic adverse conditions. Transcriptomic studies in human patients and disease models have revealed the co-existence of many putative reactive sub-states of astrocytes. Inter-disease and even intra-disease heterogeneity of reactive astrocytic sub-states are well established, but the extent to which specific sub-states are shared across different diseases is unclear. In this Review, we highlight how single-cell and single-nuclei RNA sequencing and other 'omics' technologies can enable the functional characterization of defined reactive astrocyte states in various pathological scenarios. We provide an integrated perspective, advocating cross-modal validation of key findings to define functionally important sub-states of astrocytes and their triggers as tractable therapeutic targets with cross-disease relevance.
Collapse
Affiliation(s)
- Rickie Patani
- Department of Neuromuscular Disease, UCL Queen Square Institute of Neurology, London, UK
- The Francis Crick Institute, Human Stem Cells and Neurodegeneration Laboratory, London, UK
| | - Giles E Hardingham
- Euan MacDonald Centre for MND, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute at the University of Edinburgh, University of Edinburgh, Edinburgh, UK
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Neuroscience & Physiology, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY, USA.
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
38
|
Pauwels EKJ, Boer GJ. Parkinson's Disease: A Tale of Many Players. Med Princ Pract 2023; 32:155-165. [PMID: 37285828 PMCID: PMC10601631 DOI: 10.1159/000531422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/01/2023] [Indexed: 06/09/2023] Open
Abstract
In 2020, more than 9 million patients suffering from Parkinson's disease (PD) were reported worldwide, and studies predict that the burden of this disease will grow substantially in industrial countries. In the last decade, there has been a better understanding of this neurodegenerative disorder, clinically characterized by motor disturbances, impaired balance, coordination, memory difficulties, and behavioral changes. Various preclinical investigations and studies on human postmortem brains suggest that local oxidative stress and inflammation promote misfolding and aggregation of alpha-synuclein within Lewy bodies and cause nerve cell damage. Parallel to these investigations, the familial contribution to the disease became evident from studies on genome-wide association in which specific genetic defects were linked to neuritic alpha-synuclein pathology. As for treatment, currently available pharmacological and surgical interventions may improve the quality of life but do not stop the progress of neurodegeneration. However, numerous preclinical studies have provided insights into the pathogenesis of PD. Their results provide a solid base for clinical trials and further developments. In this review, we discuss the pathogenesis, the prospects, and challenges of synolytic therapy, CRISPR, gene editing, and gene- and cell-based therapy. We also throw light on the recent observation that targeted physiotherapy may help improve the gait and other motor impairments.
Collapse
Affiliation(s)
| | - Gerard J. Boer
- Netherlands Institute for Brain Research, Royal Academy of Arts and Sciences, Amsterdam, The Netherlands
| |
Collapse
|
39
|
Qian Z, Qin J, Lai Y, Zhang C, Zhang X. Large-Scale Integration of Single-Cell RNA-Seq Data Reveals Astrocyte Diversity and Transcriptomic Modules across Six Central Nervous System Disorders. Biomolecules 2023; 13:692. [PMID: 37189441 PMCID: PMC10135484 DOI: 10.3390/biom13040692] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/30/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
The dysfunction of astrocytes in response to environmental factors contributes to many neurological diseases by impacting neuroinflammation responses, glutamate and ion homeostasis, and cholesterol and sphingolipid metabolism, which calls for comprehensive and high-resolution analysis. However, single-cell transcriptome analyses of astrocytes have been hampered by the sparseness of human brain specimens. Here, we demonstrate how large-scale integration of multi-omics data, including single-cell and spatial transcriptomic and proteomic data, overcomes these limitations. We created a single-cell transcriptomic dataset of human brains by integration, consensus annotation, and analyzing 302 publicly available single-cell RNA-sequencing (scRNA-seq) datasets, highlighting the power to resolve previously unidentifiable astrocyte subpopulations. The resulting dataset includes nearly one million cells that span a wide variety of diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), multiple sclerosis (MS), epilepsy (Epi), and chronic traumatic encephalopathy (CTE). We profiled the astrocytes at three levels, subtype compositions, regulatory modules, and cell-cell communications, and comprehensively depicted the heterogeneity of pathological astrocytes. We constructed seven transcriptomic modules that are involved in the onset and progress of disease development, such as the M2 ECM and M4 stress modules. We validated that the M2 ECM module could furnish potential markers for AD early diagnosis at both the transcriptome and protein levels. In order to accomplish a high-resolution, local identification of astrocyte subtypes, we also carried out a spatial transcriptome analysis of mouse brains using the integrated dataset as a reference. We found that astrocyte subtypes are regionally heterogeneous. We identified dynamic cell-cell interactions in different disorders and found that astrocytes participate in key signaling pathways, such as NRG3-ERBB4, in epilepsy. Our work supports the utility of large-scale integration of single-cell transcriptomic data, which offers new insights into underlying multiple CNS disease mechanisms where astrocytes are involved.
Collapse
Affiliation(s)
- Zhenwei Qian
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Jinglin Qin
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Yiwen Lai
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| | - Chen Zhang
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
- Chinese Institute for Brain Research, Beijing 102206, China
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Nanjing 210000, China
| | - Xiannian Zhang
- School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, China
| |
Collapse
|
40
|
Mussetto V, Moen A, Trofimova L, Sandkühler J, Hogri R. Differential activation of spinal and parabrachial glial cells in a neuropathic pain model. Front Cell Neurosci 2023; 17:1163171. [PMID: 37082205 PMCID: PMC10110840 DOI: 10.3389/fncel.2023.1163171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/17/2023] [Indexed: 04/22/2023] Open
Abstract
The clinical burden faced by chronic pain patients is compounded by affective comorbidities, such as depression and anxiety disorders. Emerging evidence suggests that reactive glial cells in the spinal cord dorsal horn play a key role in the chronification of pain, while supraspinal glia are important for psychological aspects of chronic pain. The lateral parabrachial nucleus (LPBN) in the brainstem is a key node in the ascending pain system, and is crucial for the emotional dimension of pain. Yet, whether astrocytes and microglia in the LPBN are activated during chronic pain is unknown. Here, we evaluated the occurrence of glial activation in the LPBN of male Sprague-Dawley rats 1, 4, and 7 weeks after inducing a chronic constriction injury (CCI) of the sciatic nerve, a prevalent neuropathic pain model. CCI animals developed mechanical and thermal hypersensitivity that persisted for at least 4 weeks, and was mostly reversed after 7 weeks. Using immunohistochemical staining and confocal imaging, we found that CCI caused a strong increase in the expression of the astrocytic marker GFAP and the microglial marker Iba1 in the ipsilateral spinal dorsal horn, with peak expression observed 1 week post-injury. Moreover, morphology analysis revealed changes in microglial phenotype, indicative of microglia activation. In contrast, CCI did not induce any detectable changes in either astrocytes or microglia in the LPBN, at any time point. Thus, our results indicate that while neuropathic pain induces a robust glial reaction in the spinal dorsal horn, it fails to activate glial cells in the LPBN.
Collapse
Affiliation(s)
| | | | | | | | - Roni Hogri
- Department of Neurophysiology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
41
|
Xu Z, Zhang G, Zhang X, Lei Y, Sun Y, He Y, Yang F, Nan W, Xing X, Li Y, Lin J. Menstrual blood-derived endometrial stem cells inhibit neuroinflammation by regulating microglia through the TLR4/MyD88/NLRP3/Casp1 pathway. Int J Biochem Cell Biol 2023; 157:106386. [PMID: 36754162 DOI: 10.1016/j.biocel.2023.106386] [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: 08/22/2022] [Revised: 12/28/2022] [Accepted: 02/03/2023] [Indexed: 02/09/2023]
Abstract
Neuroinflammation is a common response in various neurological disorders. Mesenchymal stem cell-based treatment has become a promising therapy for neuroinflammation-associated diseases. However, the effects of mesenchymal stem cells are controversial, and the underlying mechanism is incompletely understood. In the present study, menstrual blood-derived endometrial stem cells were intravenously transplanted into a mouse model of neuroinflammation established by peripheral injection of lipopolysaccharide. Microglial cells challenged with lipopolysaccharide were cultured with conditioned medium from endometrial stem cells. The levels of cytokines were detected by enzyme-linked immunosorbent assay. Cell proliferation and death were detected by Cell Counting Kit 8 and flow cytometry, respectively. The expression levels of Toll-like receptor 4 (TLR4), myeloid differentiation primary response gene 88 (MyD88), NLR family pyrin domain containing 3 (NLRP3) and caspase 1 (Casp1) were evaluated by western blotting. The results showed that intravenous transplantation of endometrial stem cells downregulated proinflammatory factors and upregulated anti-inflammatory factors in the brain of mice with neuroinflammation. Conditioned medium suppressed the inflammatory reaction and hyperactivation of microglial cells and protected microglial cells from cell death induced by lipopolysaccharide in vitro. The expression of TLR4, MyD88, NLRP3 and Casp1 in the brain of mice with neuroinflammation and in lipopolysaccharide-stimulated microglial cells was downregulated by endometrial stem cells and conditioned medium, respectively. These data suggested that menstrual blood-derived endometrial stem cells may suppress neuroinflammatory reactions partially by regulating microglia through the TLR4/MyD88/NLRP3/Casp1 signalling pathway. Our findings may be very useful for the development of an alternative stem cell-based therapy for neuroinflammation-associated disorders.
Collapse
Affiliation(s)
- Zhihao Xu
- School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, Henan, PR China; Stem Cells and Biotherapy Engineering and Technology Research Center of Henan, Henan Joint International Research Laboratory of Stem Cell Medicine, National Joint Engineering Laboratory of Stem Cells and Biotherapy, Xinxiang 453003, Henan, PR China.
| | - Guoqing Zhang
- School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, Henan, PR China; Stem Cells and Biotherapy Engineering and Technology Research Center of Henan, Henan Joint International Research Laboratory of Stem Cell Medicine, National Joint Engineering Laboratory of Stem Cells and Biotherapy, Xinxiang 453003, Henan, PR China
| | - Xiaoyue Zhang
- School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, Henan, PR China; Stem Cells and Biotherapy Engineering and Technology Research Center of Henan, Henan Joint International Research Laboratory of Stem Cell Medicine, National Joint Engineering Laboratory of Stem Cells and Biotherapy, Xinxiang 453003, Henan, PR China
| | - Yu Lei
- School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, Henan, PR China
| | - Yuliang Sun
- Stem Cells and Biotherapy Engineering and Technology Research Center of Henan, Henan Joint International Research Laboratory of Stem Cell Medicine, National Joint Engineering Laboratory of Stem Cells and Biotherapy, Xinxiang 453003, Henan, PR China; School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, Henan, PR China
| | - Ya'nan He
- Zhongyuan Stem Cell Research Institute, Xinxiang 453003, Henan, PR China
| | - Fen Yang
- Stem Cells and Biotherapy Engineering and Technology Research Center of Henan, Henan Joint International Research Laboratory of Stem Cell Medicine, National Joint Engineering Laboratory of Stem Cells and Biotherapy, Xinxiang 453003, Henan, PR China; School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, Henan, PR China
| | - Wenbin Nan
- School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, Henan, PR China
| | - Xuekun Xing
- College of Public Health, Guilin Medical University, Guilin 541199, Guangxi, PR China
| | - Yonghai Li
- School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, Henan, PR China; Stem Cells and Biotherapy Engineering and Technology Research Center of Henan, Henan Joint International Research Laboratory of Stem Cell Medicine, National Joint Engineering Laboratory of Stem Cells and Biotherapy, Xinxiang 453003, Henan, PR China
| | - Juntang Lin
- School of Life Science and Technology, Xinxiang Medical University, Xinxiang 453003, Henan, PR China; Stem Cells and Biotherapy Engineering and Technology Research Center of Henan, Henan Joint International Research Laboratory of Stem Cell Medicine, National Joint Engineering Laboratory of Stem Cells and Biotherapy, Xinxiang 453003, Henan, PR China; School of Medical Engineering, Xinxiang Medical University, Xinxiang 453003, Henan, PR China.
| |
Collapse
|
42
|
Li JF, Hu WY, Chang HX, Bao JH, Kong XX, Ma H, Li YF. Astrocytes underlie a faster-onset antidepressant effect of hypidone hydrochloride (YL-0919). Front Pharmacol 2023; 14:1175938. [PMID: 37063256 PMCID: PMC10090319 DOI: 10.3389/fphar.2023.1175938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 03/13/2023] [Indexed: 03/31/2023] Open
Abstract
Introduction: Major depression disorder (MDD) is a common and potentially life-threatening mental illness; however, data on its pathogenesis and effective therapeutic measures are lacking. Pathological changes in astrocytes play a pivotal role in MDD. While hypidone hydrochloride (YL-0919), an independently developed antidepressant, has shown rapid action with low side effects, its underlying astrocyte-specific mechanisms remain unclear.Methods: In our study, mice were exposed to chronic restraint stress (CRS) for 14 days or concomitantly administered YL-0919/fluoxetine. Behavioral tests were applied to evaluate the depression model; immunofluorescence and immunohistochemistry staining were used to explore morphological changes in astrocytes; astrocyte-specific RNA sequencing (RNA-Seq) analysis was performed to capture transcriptome wide alterations; and ATP and oxygen consumption rate (OCR) levels of primary astrocytes were measured, followed by YL-0919 incubation to appraise the alteration of energy metabolism and mitochondrial oxidative phosphorylation (OXPHOS).Results: YL-0919 alleviated CRS-induced depressive-like behaviors faster than fluoxetine and attenuated the number and morphologic deficits in the astrocytes of depressed mice. The changes of gene expression profile in astrocytes after CRS were partially reversed by YL-0919. Moreover, YL-0919 improved astrocyte energy metabolism and mitochondrial OXPHOS in astrocytes.Conclusion: Our results provide evidence that YL-0919 exerted a faster-onset antidepressant effect on CRS-mice possibly via astrocyte structural remodeling and mitochondria functional restoration.
Collapse
Affiliation(s)
- Jin-Feng Li
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Wen-Yu Hu
- Beijing Institute of Basic Medical Sciences, Beijing, China
- Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang, China
| | - Hai-Xia Chang
- Beijing Institute of Basic Medical Sciences, Beijing, China
- College of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Jin-Hao Bao
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Xiang-Xi Kong
- Beijing Institute of Basic Medical Sciences, Beijing, China
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Xiang-Xi Kong, ; Hui Ma, ; Yun-Feng Li,
| | - Hui Ma
- Beijing Institute of Basic Medical Sciences, Beijing, China
- *Correspondence: Xiang-Xi Kong, ; Hui Ma, ; Yun-Feng Li,
| | - Yun-Feng Li
- Beijing Institute of Basic Medical Sciences, Beijing, China
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing Institute of Pharmacology and Toxicology, Beijing, China
- *Correspondence: Xiang-Xi Kong, ; Hui Ma, ; Yun-Feng Li,
| |
Collapse
|
43
|
Hasel P, Aisenberg WH, Bennett FC, Liddelow SA. Molecular and metabolic heterogeneity of astrocytes and microglia. Cell Metab 2023; 35:555-570. [PMID: 36958329 DOI: 10.1016/j.cmet.2023.03.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/26/2023] [Accepted: 03/08/2023] [Indexed: 03/25/2023]
Abstract
Astrocytes and microglia are central players in a myriad of processes in the healthy and diseased brain, ranging from metabolism to immunity. The crosstalk between these two cell types contributes to pathology in many if not all neuroinflammatory and neurodegenerative diseases. Recent advancements in integrative multimodal sequencing techniques have begun to highlight how heterogeneous both cell types are and the importance of metabolism to their regulation. We discuss here the transcriptomic, metabolic, and functional heterogeneity of astrocytes and microglia and highlight their interaction in health and disease.
Collapse
Affiliation(s)
- Philip Hasel
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA.
| | - William H Aisenberg
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| | - F Chris Bennett
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Neuroscience and Physiology, NYU Grossman School of Medicine, New York, NY 10016, USA; Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY 10016, USA; Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
44
|
Mittli D, Tukacs V, Ravasz L, Csősz É, Kozma T, Kardos J, Juhász G, Kékesi KA. LPS-induced acute neuroinflammation, involving interleukin-1 beta signaling, leads to proteomic, cellular, and network-level changes in the prefrontal cortex of mice. Brain Behav Immun Health 2023; 28:100594. [PMID: 36713475 PMCID: PMC9880243 DOI: 10.1016/j.bbih.2023.100594] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/12/2022] [Accepted: 01/16/2023] [Indexed: 01/22/2023] Open
Abstract
Neuroinflammation induced by peripheral infections leads to various neuropsychiatric symptoms both in humans and laboratory animals, e.g., to the manifestation of sickness behavior that resembles some features of clinical depression. However, in addition to depression-like behavior, there are other symptoms of acute systemic inflammation that can be associated with the impairment of prefrontal cortex (PFC)-regulated cognitive functions. Thus, we investigated the electrophysiological and proteomic alterations of the PFC using brain slices and the lipopolysaccharide (LPS) model of acute peripheral infection in male mice. Based on the gene expression differences of the coreceptor (Il1rap) of interleukin-1 beta (IL-1β) between neuron types in our previous single-cell sequencing dataset, we first compared the electrophysiological effects of IL-1β on PFC pyramidal cells and interneurons. We found that pyramidal cells are more responsive to IL-1β, as could be presumed from our transcriptomic data. To examine the possible circuit-level correlates of the cellular changes, frontal electroencephalographic (EEG) activity and fronto-occipital functional connectivity were analyzed in LPS-treated mice and significant changes were found in the fronto-occipital EEG correlation and coherence in the delta and high-gamma frequency bands. The upregulation of the prefrontal IL-1 system (IL-1β and its receptor) after LPS treatment was revealed by immunoassays simultaneously with the observed EEG changes. Furthermore, we investigated the LPS-induced alterations of the synaptic proteome in the PFC using 2-D differential gel electrophoresis and mass spectrometry and found 48 altered proteins mainly related to cellular signaling, cytoskeletal organization, and carbohydrate/energy metabolism. Thus, our results indicate remarkable electrophysiological and molecular changes in the PFC related to acute systemic inflammation that may explain some of the concomitant behavioral and physiological symptoms.
Collapse
Affiliation(s)
- Dániel Mittli
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Laboratory of Proteomics, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Department of Physiology and Neurobiology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Vanda Tukacs
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Laboratory of Proteomics, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Lilla Ravasz
- Laboratory of Proteomics, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- CRU Hungary Ltd., Göd, Hungary
| | - Éva Csősz
- Proteomics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | | | - József Kardos
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Gábor Juhász
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Laboratory of Proteomics, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- CRU Hungary Ltd., Göd, Hungary
- InnoScience Ltd., Mátranovák, Hungary
| | - Katalin Adrienna Kékesi
- ELTE NAP Neuroimmunology Research Group, Department of Biochemistry, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Laboratory of Proteomics, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- Department of Physiology and Neurobiology, Institute of Biology, ELTE Eötvös Loránd University, Budapest, Hungary
- InnoScience Ltd., Mátranovák, Hungary
| |
Collapse
|
45
|
Li Y, Guan X, He Y, Jia X, Pan L, Wang Y, Han Y, Zhao R, Yang J, Hou T. ProBDNF signaling is involved in periodontitis-induced depression-like behavior in mouse hippocampus. Int Immunopharmacol 2023; 116:109767. [PMID: 36738676 DOI: 10.1016/j.intimp.2023.109767] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/15/2023] [Accepted: 01/19/2023] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Increasing evidence supports the association between periodontitis and depression. However, the specific mechanisms remain to be further elucidated. The present study aimed to mechanistically investigate the regional roles of proBDNF (the precursor of brain-derived neurotrophic factor) in periodontitis induced depression-like behavior in mice. METHODS Experimental periodontitis model was established by periodontal injection of Porphyromonas gingivalis lipopolysaccharide (Pg-LPS) in 8-week-old male Bdnf-HA/HA mice for 3 weeks. The depression-like behaviors, spontaneous exploratory activity and the level of anxiety were assessed by behavior tests. The activation of microglia and astrocytes, as well as the expression of Interleukin (IL)-1β and Tumor necrosis factor (TNF)-α in the hippocampus, prefrontal cortex, and cortex were further assessed by immunofluorescence and western blots. The levels of IL-1β in blood serum and expression of occludin as well as claudin5 in the hippocampus, prefrontal cortex, and cortex were further determined by enzyme-linked immunosorbent assay and western blot. Finally, the expression of proBDNF, its receptors, and mature BDNF (mBDNF), as well as neuronal activity were measured by western blots and immunofluorescence. RESULTS Pg-LPS successfully induced periodontitis in mice and caused obvious depression-like behavior. Furthermore, we observed an increased activation of astrocytes and microglia, as well as a significant increase in expression of IL-1β and TNF-α in the hippocampus of mice treated with Pg-LPS, with elevated level of IL-1β in serum and decreased expression of occludin and claudin5 in the hippocampus. Importantly, we found that the levels of proBDNF and its receptors, SorCS2 and p75NTR, were increased significantly; however, the level of mBDNF was decreased, therefor leading to greater ratio of proBDNF/mBDNF. In addition, we also detected decreased neuronal activity in the hippocampus of mice treated with Pg-LPS. CONCLUSIONS Our results indicate that Pg-LPS-induced periodontitis could cause depression-like behaviors in mice, and the proBDNF signaling is involved in the process.
Collapse
Affiliation(s)
- Yingxue Li
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Xiaoyue Guan
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yani He
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Xiangbin Jia
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Lifei Pan
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yuting Wang
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yue Han
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Rui Zhao
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Jianmin Yang
- Department of Medicine, Weill Cornell Medical School, Cornell University, New York, NY 10065, USA.
| | - Tiezhou Hou
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| |
Collapse
|
46
|
Myers AJ, Brahimi A, Jenkins IJ, Koob AO. The Synucleins and the Astrocyte. BIOLOGY 2023; 12:biology12020155. [PMID: 36829434 PMCID: PMC9952504 DOI: 10.3390/biology12020155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023]
Abstract
Synucleins consist of three proteins exclusively expressed in vertebrates. α-Synuclein (αS) has been identified as the main proteinaceous aggregate in Lewy bodies, a pathological hallmark of many neurodegenerative diseases. Less is understood about β-synuclein (βS) and γ-synuclein (γS), although it is known βS can interact with αS in vivo to inhibit aggregation. Likewise, both γS and βS can inhibit αS's propensity to aggregate in vitro. In the central nervous system, βS and αS, and to a lesser extent γS, are highly expressed in the neural presynaptic terminal, although they are not strictly located there, and emerging data have shown a more complex expression profile. Synapse loss and astrocyte atrophy are early aspects of degenerative diseases of the brain and correlate with disease progression. Synucleins appear to be involved in synaptic transmission, and astrocytes coordinate and organize synaptic function, with excess αS degraded by astrocytes and microglia adjacent to the synapse. βS and γS have also been observed in the astrocyte and may provide beneficial roles. The astrocytic responsibility for degradation of αS as well as emerging evidence on possible astrocytic functions of βS and γS, warrant closer inspection on astrocyte-synuclein interactions at the synapse.
Collapse
Affiliation(s)
- Abigail J. Myers
- Neuroscience Program, Health Science Research Facility, University of Vermont, 149 Beaumont Ave., Burlington, VT 05405, USA
| | - Ayat Brahimi
- Biology Department, University of Hartford, 200 Bloomfield Ave., West Hartford, CT 06117, USA
| | - Imani J. Jenkins
- Biology Department, University of Hartford, 200 Bloomfield Ave., West Hartford, CT 06117, USA
| | - Andrew O. Koob
- Biology Department, University of Hartford, 200 Bloomfield Ave., West Hartford, CT 06117, USA
- Correspondence: ; Tel.: +1-860-768-5780
| |
Collapse
|
47
|
Li Z, Sun Y, Ding L, Yang J, Huang J, Cheng M, Wu L, Zhuang Z, Chen C, Huang Y, Zhu Z, Jiang S, Huang F, Wang C, Liu S, Liu L, Lei Y. Deciphering the distinct transcriptomic and gene regulatory map in adult macaque basal ganglia cells. Gigascience 2022; 12:giad095. [PMID: 38091510 PMCID: PMC10716911 DOI: 10.1093/gigascience/giad095] [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: 05/06/2023] [Revised: 08/09/2023] [Accepted: 10/10/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND The basal ganglia are a complex of interconnected subcortical structures located beneath the mammalian cerebral cortex. The degeneration of dopaminergic neurons in the basal ganglia is the primary pathological feature of Parkinson's disease. Due to a lack of integrated analysis of multiomics datasets across multiple basal ganglia brain regions, very little is known about the regulatory mechanisms of this area. FINDINGS We utilized high-throughput transcriptomic and epigenomic analysis to profile over 270,000 single-nucleus cells to create a cellular atlas of the basal ganglia, characterizing the cellular composition of 4 regions of basal ganglia in adult macaque brain, including the striatum, substantia nigra (SN), globus pallidum, and amygdala. We found a distinct epigenetic regulation on gene expression of neuronal and nonneuronal cells across regions in basal ganglia. We identified a cluster of SN-specific astrocytes associated with neurodegenerative diseases and further explored the conserved and primate-specific transcriptomics in SN cell types across human, macaque, and mouse. Finally, we integrated our epigenetic landscape of basal ganglia cells with human disease heritability and identified a regulatory module consisting of candidate cis-regulatory elements that are specific to medium spiny neurons and associated with schizophrenia. CONCLUSIONS In general, our macaque basal ganglia atlas provides valuable insights into the comprehensive transcriptome and epigenome of the most important and populous cell populations in the macaque basal ganglia. We have identified 49 cell types based on transcriptomic profiles and 47 cell types based on epigenomic profiles, some of which exhibit region specificity, and characterized the molecular relationships underlying these brain regions.
Collapse
Affiliation(s)
- Zihao Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- BGI Research, Hangzhou 310030, China
| | - Yunong Sun
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- BGI Research, Hangzhou 310030, China
| | | | - Jing Yang
- BGI Research, Hangzhou 310030, China
| | | | | | - Liang Wu
- BGI Research, Shenzhen 518083, China
| | | | - Cheng Chen
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- BGI Research, Hangzhou 310030, China
| | - Yunqi Huang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- BGI Research, Hangzhou 310030, China
| | - Zhiyong Zhu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- BGI Research, Hangzhou 310030, China
| | - Siyuan Jiang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- BGI Research, Hangzhou 310030, China
| | - Fubaoqian Huang
- BGI Research, Hangzhou 310030, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Chunqing Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- BGI Research, Shenzhen 518083, China
| | - Shiping Liu
- BGI Research, Hangzhou 310030, China
- BGI Research, Shenzhen 518083, China
| | - Longqi Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- BGI Research, Hangzhou 310030, China
- BGI Research, Shenzhen 518083, China
| | - Ying Lei
- BGI Research, Shenzhen 518083, China
| |
Collapse
|
48
|
Yang P, Zhang Q, Shen H, Bai X, Liu P, Zhang T. Research progress on the protective effects of aucubin in neurological diseases. PHARMACEUTICAL BIOLOGY 2022; 60:1088-1094. [PMID: 35634723 PMCID: PMC9154787 DOI: 10.1080/13880209.2022.2074057] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/29/2022] [Accepted: 05/01/2022] [Indexed: 06/15/2023]
Abstract
CONTEXT Aucubin (AU), an iridoid glycoside that is one of the active constituents of Eucommia ulmoides Oliv. (EUO) (Eucommiaceae), a traditional Chinese medicine, has been extensively studied in the management of neurological diseases (NDs). However, a comprehensive review of its effects and mechanisms in this regard is currently not available. OBJECTIVE To compile the protective effects and mechanisms of AU in NDs and provide a basis for further research. METHODS We used 'aucubin' as the 'All Fields' or 'MeSH' in PubMed, Web of Science and China National Knowledge Infrastructure without any limitation to search all relevant articles as comprehensively as possible; we selected the articles on AU treatment of NDs for summary. RESULTS Studies reviewed herein reported that AU improved the symptoms or prognosis of Parkinson's disease, Alzheimer's disease, intracerebral haemorrhage, diabetic encephalopathy, epilepsy, anxiety and depression, and traumatic brain injury. The pharmacological mechanisms involved in repairing neuronal loss were postulated to include increasing γ-aminobutyric acid (GABA) content in the synapse, promoting differentiation of neural precursor cells into GABAergic neurons, providing antioxidant and anti-neuroinflammation activities, as well as enhancing autophagy and anti-apoptotic actions. DISCUSSION AND CONCLUSIONS The protective effects of AU on some NDs have been confirmed. According to the pharmacological effects, AU is also highly likely to have protective effects on other NDs, which can be realized by further in vivo and in vitro basic research, and clinical trials. In the future, AU may be used for clinical prevention or treatment of patients with neurological diseases.
Collapse
Affiliation(s)
- Ping Yang
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Qiaoyue Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Hengyan Shen
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Xinyu Bai
- Department of Clinical Pharmacy, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Ping Liu
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Tao Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
49
|
Insulin and IGF-1 elicit robust transcriptional regulation to modulate autophagy in astrocytes. Mol Metab 2022; 66:101647. [PMID: 36503893 PMCID: PMC9731889 DOI: 10.1016/j.molmet.2022.101647] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/08/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE Insulin is a principal metabolic hormone. It regulates a plethora of metabolic pathways in peripheral tissues. The highly homologous insulin-like growth factor 1 (IGF-1), on the other hand, is important for development and growth. Recent studies have shown that insulin and IGF-1 signaling plays fundamental roles in the brain. Loss of insulin or IGF-1 receptors in astrocytes leads to altered glucose handling, mitochondrial metabolism, neurovascular coupling, and behavioral abnormalities in mice. Here, we aim to investigate molecular mechanisms by which insulin and IGF-1 signaling regulates astrocyte functions. METHODS IR-flox and IRKO primary astrocytes were treated with 100 nM insulin or IGF-1 for 6 h, and their transcriptomes were analyzed. Astrocytes with either IR deletion, IGF1R deletion or both were used to examine receptor-dependent transcriptional regulations using qPCR. Additional immunoblotting and confocal imaging studies were performed to functionally validate pathways involved in protein homeostasis. RESULTS Using next-generation RNA sequencing, we show that insulin significantly regulates the expression of over 1,200 genes involved in multiple functional processes in primary astrocytes. Insulin-like growth factor 1 (IGF-1) triggers a similar robust transcriptional regulation in astrocytes. Thus, over 50% of the differentially expressed genes are regulated by both ligands. As expected, these commonly regulated genes are highly enriched in pathways involved in lipid and cholesterol biosynthesis. Additionally, insulin and IGF-1 induce the expression of genes involved in ribosomal biogenesis, while suppressing the expression of genes involved in autophagy, indicating a common role of insulin and IGF-1 on protein homeostasis in astrocytes. Insulin-dependent suppression of autophagy genes, including p62, Ulk1/2, and several Atg genes, is blunted only when both IR and IGF1R are deleted. CONCLUSIONS In summary, insulin and IGF-1 potently suppress autophagy in astrocytes through transcriptional regulation. Both IR and IGF1R can elicit ligand-dependent transcriptional suppression of autophagy. These results demonstrate an important role of astrocytic insulin/IGF-1 signaling on proteostasis. Impairment of this regulation in insulin resistance and diabetes may contribute to neurological complications related to diabetes.
Collapse
|
50
|
Hemati-Gourabi M, Cao T, Romprey MK, Chen M. Capacity of astrocytes to promote axon growth in the injured mammalian central nervous system. Front Neurosci 2022; 16:955598. [PMID: 36203815 PMCID: PMC9530187 DOI: 10.3389/fnins.2022.955598] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/15/2022] [Indexed: 01/02/2023] Open
Abstract
Understanding the regulation of axon growth after injury to the adult central nervous system (CNS) is crucial to improve neural repair. Following acute focal CNS injury, astrocytes are one cellular component of the scar tissue at the primary lesion that is traditionally associated with inhibition of axon regeneration. Advances in genetic models and experimental approaches have broadened knowledge of the capacity of astrocytes to facilitate injury-induced axon growth. This review summarizes findings that support a positive role of astrocytes in axon regeneration and axon sprouting in the mature mammalian CNS, along with potential underlying mechanisms. It is important to recognize that astrocytic functions, including modulation of axon growth, are context-dependent. Evidence suggests that the local injury environment, neuron-intrinsic regenerative potential, and astrocytes’ reactive states determine the astrocytic capacity to support axon growth. An integrated understanding of these factors will optimize therapeutic potential of astrocyte-targeted strategies for neural repair.
Collapse
Affiliation(s)
| | - Tuoxin Cao
- Spinal Cord and Brain Injury Research Center, Lexington, KY, United States
| | - Megan K. Romprey
- Spinal Cord and Brain Injury Research Center, Lexington, KY, United States
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Meifan Chen
- Spinal Cord and Brain Injury Research Center, Lexington, KY, United States
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
- *Correspondence: Meifan Chen,
| |
Collapse
|