1
|
Min E, Ko MY, Kim M, Park H, Kim Y, Kim KK, Lee BS, Hyun SA, Ka M. Perfluorooctanoic acid (PFOA) activates astrogliosis-associated neuroinflammation through ER stress-autophagy axis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 979:179546. [PMID: 40288163 DOI: 10.1016/j.scitotenv.2025.179546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/24/2025] [Accepted: 04/25/2025] [Indexed: 04/29/2025]
Abstract
Perfluorooctanoic acid (PFOA), a perfluoroalkyl acid, induces neuroinflammation. However, present understanding regarding its fundamental role in neuroinflammation remains limited. Therefore, in this study, we aimed to clarify the potential association between PFOA and astrocyte activation via the modulation of the endoplasmic reticulum (ER) stress-autophagy axis. The results obtained revealed that PFOA activated astroglia in A-172 astrocytoma cells and primary astrocytes by upregulating the expression levels of autophagy-related proteins (ATG5, BECN1, SQSTM1, and MAP1LC3B-II). It also activated autophagy in A-172 astrocytoma cells and primary astrocytes via the upstream activation of ER stress-related proteins, such as ATF4, GRP78, and CHOP. Further, the pharmacological inhibition of ER stress as well as autophagy prevented PFOA-induced activation of astrogliosis in PFOA-treated A-172 cells and primary astrocytes. We also observed that PFOA-mediated activation of GFAP upregulated the transcription of pro-inflammatory cytokines, such as IL-1β, TNF-α, and IL-6. These findings confirmed the existence of a relationship between ER stress-induced autophagy and astrogliosis in PFOA-treated astrocytes, suggesting that targeting the ER stress-autophagy axis may be a potential therapeutic strategy for reducing PFOA-induced neuroinflammation.
Collapse
Affiliation(s)
- Euijun Min
- Center for Convergence Toxicology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Department of Biochemistry, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Moon Yi Ko
- Center for Convergence Toxicology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Department of Biochemistry, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Minjeong Kim
- Center for Convergence Toxicology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Heejin Park
- Center for Toxicologic Pathology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Younhee Kim
- Center for Toxicologic Pathology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea
| | - Kee K Kim
- Department of Biochemistry, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Byoung-Seok Lee
- Center for Toxicologic Pathology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea.
| | - Sung-Ae Hyun
- Center for Convergence Toxicology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea.
| | - Minhan Ka
- Center for Convergence Toxicology Research, Division of Next Generation Non-Clinical Research, Korea Institute of Toxicology, Daejeon 34114, Republic of Korea; Human and Environmental Toxicology, University of Science and Technology, Daejeon 34114, Republic of Korea.
| |
Collapse
|
2
|
Wang C, He T, Qin J, Jiao J, Ji F. The roles of immune factors in neurodevelopment. Front Cell Neurosci 2025; 19:1451889. [PMID: 40276707 PMCID: PMC12018394 DOI: 10.3389/fncel.2025.1451889] [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: 03/28/2025] [Indexed: 04/26/2025] Open
Abstract
The development of the nervous system is a highly complex process orchestrated by a multitude of factors, including various immune elements. These immune components play a dual role, not only regulating the immune response but also actively influencing brain development under both physiological and pathological conditions. The brain's immune barrier includes microglia in the brain parenchyma, which act as resident macrophages, astrocytes that support neuronal function and contribute to the inflammatory response, as well as circulating immune cells that reside at the brain's borders, including the choroid plexus, meninges, and perivascular spaces. Cytokines-soluble signaling molecules released by immune cells-play a crucial role in mediating communication between immune cells and the developing nervous system. Cytokines regulate processes such as neurogenesis, synaptic pruning, and inflammation, helping to shape the neural environment. Dysregulation of these immune cells, astrocytes, or cytokine signaling can lead to alterations in neurodevelopment, potentially contributing to neurodevelopmental abnormalities. This article reviews the central role of microglia, astrocytes, cytokines, and other immune factors in neurodevelopment, and explores how neuroinflammation can lead to the onset of neurodevelopmental disorders, shedding new light on their pathogenesis.
Collapse
Affiliation(s)
- Chong Wang
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Tingting He
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jie Qin
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Jianwei Jiao
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Fen Ji
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| |
Collapse
|
3
|
Talifu Z, Xu X, Du H, Li Z, Wang X, Zhang C, Pan Y, Ke H, Liu W, Gao F, Yang D, Jing Y, Yu Y, Du L, Li J. Effect of in vivo reprogramming of astrocytes combined with exercise training on neurorepair in rats with spinal cord injury. Animal Model Exp Med 2025; 8:595-605. [PMID: 39844772 PMCID: PMC12008439 DOI: 10.1002/ame2.12545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 12/15/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND The inability of damaged neurons to regenerate and of axons to establish new functional connections leads to permanent functional deficits after spinal cord injury (SCI). Although astrocyte reprogramming holds promise for neurorepair in various disease models, it is not sufficient on its own to achieve significant functional recovery. METHODS A rat SCI model was established using a spinal cord impactor. Seven days postsurgery, adeno-associated virus were injected to overexpress the transcription factors NeuroD1 and Neurogenin-2 (Ngn2) in the spinal cord. The rats were then trained to walk on a weight-supported treadmill for 4 weeks, starting 14 days after modeling. The effects of these interventions on motor and sensory functions, as well as spinal cord tissue repair, were subsequently evaluated. RESULTS The combination of NeuroD1 and Ngn2 overexpression with weight-supported exercise training significantly improved gait compared to either intervention alone. The group receiving the combined intervention exhibited enhanced sensitivity in sensory assessments. Immunofluorescence analysis revealed increased colocalization of astrocytes and microtubule-associated protein 2-positive neurons in the injury area. These effects were more pronounced than those observed with spinal cord tissue repair alone. Additionally, the combined intervention significantly reduced glial scarring and the size of the injury area. CONCLUSION Exercise intervention enhances the reprogramming effects of astrocytes and restores motor function, yielding better results than either intervention alone.
Collapse
Affiliation(s)
- Zuliyaer Talifu
- School of RehabilitationCapital Medical UniversityBeijingChina
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Chinese Institute of Rehabilitation ScienceBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
- School of Population Medicine and Public HealthChinese Academy of Medical Sciences/Peking Union Medical CollegeBeijingChina
| | - Xin Xu
- School of RehabilitationCapital Medical UniversityBeijingChina
- Department of Neurology, Cheeloo College of MedicineShandong University, Qilu Hospital of Shandong UniversityJinanChina
- School of Health and Life SciencesUniversity of Health and Rehabilitation SciencesQingdaoChina
| | - Huayong Du
- School of RehabilitationCapital Medical UniversityBeijingChina
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Chinese Institute of Rehabilitation ScienceBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
| | - Zehui Li
- School of RehabilitationCapital Medical UniversityBeijingChina
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Chinese Institute of Rehabilitation ScienceBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
| | - Xiaoxin Wang
- School of RehabilitationCapital Medical UniversityBeijingChina
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Chinese Institute of Rehabilitation ScienceBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
| | - Chunjia Zhang
- School of RehabilitationCapital Medical UniversityBeijingChina
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Chinese Institute of Rehabilitation ScienceBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
| | - Yunzhu Pan
- School of RehabilitationCapital Medical UniversityBeijingChina
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Chinese Institute of Rehabilitation ScienceBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
| | - Han Ke
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Department of Neurology, Cheeloo College of MedicineShandong University, Qilu Hospital of Shandong UniversityJinanChina
- School of Health and Life SciencesUniversity of Health and Rehabilitation SciencesQingdaoChina
| | - Wubo Liu
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Department of Neurology, Cheeloo College of MedicineShandong University, Qilu Hospital of Shandong UniversityJinanChina
- School of Health and Life SciencesUniversity of Health and Rehabilitation SciencesQingdaoChina
| | - Feng Gao
- School of RehabilitationCapital Medical UniversityBeijingChina
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
| | - Degang Yang
- School of RehabilitationCapital Medical UniversityBeijingChina
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
| | - Yingli Jing
- School of RehabilitationCapital Medical UniversityBeijingChina
- Chinese Institute of Rehabilitation ScienceBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
| | - Yan Yu
- School of RehabilitationCapital Medical UniversityBeijingChina
- Chinese Institute of Rehabilitation ScienceBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
| | - Liangjie Du
- School of RehabilitationCapital Medical UniversityBeijingChina
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
| | - Jianjun Li
- School of RehabilitationCapital Medical UniversityBeijingChina
- Department of Spinal and Neural Functional Reconstruction, China Rehabilitation Research CenterBeijingChina
- Chinese Institute of Rehabilitation ScienceBeijingChina
- Center of Neural Injury and RepairBeijing Institute for Brain DisordersBeijingChina
- Beijing Key Laboratory of Neural Injury and RehabilitationBeijingChina
- School of Health and Life SciencesUniversity of Health and Rehabilitation SciencesQingdaoChina
| |
Collapse
|
4
|
Santos DE, Silva Lima SA, Moreira LS, Lima Costa S, de Sampaio Schitine C. New perspectives on heterogeneity in astrocyte reactivity in neuroinflammation. Brain Behav Immun Health 2025; 44:100948. [PMID: 40028234 PMCID: PMC11871470 DOI: 10.1016/j.bbih.2025.100948] [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: 11/30/2023] [Revised: 01/03/2025] [Accepted: 01/13/2025] [Indexed: 03/05/2025] Open
Abstract
The inflammatory response is a fundamental aspect of all insults to the central nervous system (CNS), which includes acute trauma, infections, and chronic neurodegenerative conditions. As methods for investigating astrocytes have progressed, recent findings indicate that astrocytes can react to a diverse spectrum of insults affecting the central nervous system. Astrocytes respond to external and internal stimuli from the nervous system in a process called glial reactivity. Astrocyte reactivity, previously considered uniform and functionally inactive, is currently a very diverse event in different inflammatory processes. These differences can occur due to the nature, the intensity of the stimulus, the brain region involved and can range from subtle changes in astrocytic morphology to protein expression alteration, gene transcription profile shifts, and variations in the secretory pattern of molecules. The elucidation of the diverse roles of astrocytes in both normal and pathological conditions has led to increased interest in the notion that various astrocyte subtypes may exist, each contributing with distinct functions. Our study will prioritize the characterization of astrocytic response patterns in the context of the development and progression of neurodegenerative diseases, particularly Alzheimer's and Parkinson's. In addition, we will investigate the astrocyte's response during bacterial and viral infections, given the potential to enhance specific therapeutic interventions based on the reactivity profiles of astrocytes.
Collapse
Affiliation(s)
| | | | - Leticia Santos Moreira
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Health Sciences Institute, Federal University of Bahia, Brazil
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Health Sciences Institute, Federal University of Bahia, Brazil
| | - Clarissa de Sampaio Schitine
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Health Sciences Institute, Federal University of Bahia, Brazil
| |
Collapse
|
5
|
Son M, Nagahawatta DP, Jo HC, Jeon YJ, Ryu B, Kim DY. Diphlorethohydroxycarmalol inhibits Müller cell gliosis by disrupting CXCR4/CXCL12 interaction in violet-blue light-induced retinal phototoxicity. Heliyon 2025; 11:e42475. [PMID: 40034286 PMCID: PMC11874561 DOI: 10.1016/j.heliyon.2025.e42475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 03/05/2025] Open
Abstract
Müller gliosis is a complex process that impairs the ability of retinal Müller glial cells to respond to various forms of retinal injury or disease, leading to retinal damage. Blue light (BL) exposure is a known cause of retinal damage. In this study, we aimed to investigate the potential of DPHC in inhibiting Müller gliosis in models of BL-exposure. We conducted in silico binding analysis to evaluate the binding of DPHC to CXCR4. Then, we developed in vitro and in vivo experimental models to assess the effects of DPHC and BL exposure on Müller gliosis using MIO-M1 cells and zebrafish. Our findings show that DPHC can suppress the Müller gliosis process in BL-exposed MIO-M1 cells in vitro and in BL-exposed zebrafish in vivo. In silico molecular docking, we identified CXCR4 as the target of active site 1 of DPHC. In BL-exposed MIO-M1 cells, DPHC inhibited CXCR4 activity and altered the expression of Müller gliosis markers and NF-κB-related ERK and AKT signaling. In BL-exposed zebrafish, DPHC prevented retinal thickness reduction and inhibited CXCR4 expression and retinal cell apoptosis. This study suggests that DPHC could be a potential therapeutic agent for retinal diseases involving Müller gliosis. By inhibiting CXCR4 activity, DPHC downregulates the ERK/AKT/NF-κB pathway, reducing retinal cell apoptosis and altered expression of Müller gliosis markers. These findings highlight the potential of natural bioactive compounds for treating various diseases, and further research should investigate the therapeutic potential of DPHC and its derivatives.
Collapse
Affiliation(s)
- Myeongjoo Son
- Department of Anatomy & Cell Biology, School of Medicine, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Brain Health Research Laboratory, Institute of Medical Science, Kangwon National University College of Medicine, Chuncheon, 24341, Republic of Korea
| | | | - Hang-Chan Jo
- Center for Sensor Systems, Inha University, Incheon, 22212, Republic of Korea
- Department of Electrical and Computer Engineering, College of Engineering, Inha University, Incheon, 22212, Republic of Korea
| | - You-Jin Jeon
- Department of Marine Life Sciences, Jeju National University, Jeju, 63243, Republic of Korea
- Marine Science Institute, Jeju National University, Jeju, 63333, Republic of Korea
| | - Bomi Ryu
- Major of Food Science and Nutrition, Pukyong National University, Busan, 48513, Republic of Korea
| | - Dae Yu Kim
- Center for Sensor Systems, Inha University, Incheon, 22212, Republic of Korea
- Department of Electrical and Computer Engineering, College of Engineering, Inha University, Incheon, 22212, Republic of Korea
- Inha Research Institute for Aerospace Medicine, Inha University, Incheon, 22212, Republic of Korea
| |
Collapse
|
6
|
Wang J, Zhang Q, Chen L. Microporous annealed particle hydrogels in cell culture, tissue regeneration, and emerging application in cancer immunotherapy. Am J Cancer Res 2025; 15:665-683. [PMID: 40084361 PMCID: PMC11897623 DOI: 10.62347/wrgw4430] [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: 11/07/2024] [Accepted: 02/05/2025] [Indexed: 03/16/2025] Open
Abstract
Microporous annealed particle (MAP) hydrogels consist of densely crosslinked and annealed hydrogel particles. Compared to common hydrogels, the inherent porosity within and among these hydrogel particles offers interconnected channels for substance exchange in addition to sufficient growth space for cells, thereby forming a three-dimensional culture system that highly mimics the in vivo microenvironment. Such characteristics enable MAP hydrogels to adapt to various requirements of biomedical applications, along with their excellent injectability and mechanical properties. This review initially provides a comprehensive summary of the fabrication methods and material types of MAP hydrogels, alongside an assessment of their mechanical properties and porosity. In vitro studies are evaluated based on the impact of MAP hydrogels on cellular behaviors, focusing on cell proliferation, differentiation, migration, activity, and phenotype. In vivo research highlights the promising applications of MAP hydrogels in tissue regeneration, as well as their innovative use in cancer immunotherapy. Current challenges and future research directions are outlined, underscoring the potential of MAP hydrogels to significantly improve clinical outcomes in cancer treatment and regenerative medicine.
Collapse
Affiliation(s)
- Junjie Wang
- Shanghai Institute for Minimally Invasive Therapy, University of Shanghai for Science and TechnologyShanghai 200093, China
| | - Qin Zhang
- Medical Engineering Department of Northern Jiangsu People’s HospitalYangzhou 225009, Jiangsu, China
| | - Liwen Chen
- Shanghai Institute for Minimally Invasive Therapy, University of Shanghai for Science and TechnologyShanghai 200093, China
| |
Collapse
|
7
|
Pu Z, Luo D, Shuai B, Xu Y, Liu M, Zhao J. Focusing on Formyl Peptide Receptors after Traumatic Spinal Cord Injury: from Immune Response to Neurogenesis. Neurochem Res 2025; 50:98. [PMID: 39920516 DOI: 10.1007/s11064-025-04347-5] [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: 11/13/2024] [Revised: 01/01/2025] [Accepted: 01/27/2025] [Indexed: 02/09/2025]
Abstract
The intricate pathophysiological cascades following spinal cord injury (SCI), encompassing cellular demise, axonal degeneration, and the formation of glial scars, pose formidable barriers to neural regeneration and restoration. Notably, neuroinflammation and glial scars emerge as pivotal barrier to post-SCI repair. Formyl peptide receptors (FPRs) emerge as critical regulators of immune responses, exerting significant influence over inflammatory modulation and nerve regeneration subsequent to SCI. Beyond their classical expression in myeloid cells, FPRs demonstrate a pronounced presence within the central nervous system (CNS) with roles in the progression of neurodegenerative disorders and neurological malignancies. Post-SCI, the equilibrium of the inflammatory microenvironment is recalibrated through the strategic modulation of FPRs, including facilitating a balance in microglial polarization, stimulating neural stem cells (NSCs) migration, and promoting neural axon elongation. These observations enlighten the potential of FPRs as innovative targets for neuronal regenerations bolstering SCI repair. This review endeavors to delineate the distribution and function of FPRs in the aftermath of SCI, with a special attention to their roles in inflammatory regulation, NSCs mobilization, and synaptic growth. By elucidating these mechanisms, we aspire to contribute novel insights and strategies for SCI therapy.
Collapse
Affiliation(s)
- Ziheng Pu
- Department of Spine Surgery, Center for Orthopedics, Daping Hospital, Army Medical University, Chongqing, China
| | - Dan Luo
- Yu-Yue Pathology Scientific Research Center, Chongqing, China
| | - Beining Shuai
- Department of Spine Surgery, Center for Orthopedics, Daping Hospital, Army Medical University, Chongqing, China
| | - Yuzhao Xu
- Department of Spine Surgery, Center for Orthopedics, Daping Hospital, Army Medical University, Chongqing, China
| | - Mingyong Liu
- Department of Spine Surgery, Center for Orthopedics, Daping Hospital, Army Medical University, Chongqing, China.
| | - Jianhua Zhao
- Department of Spine Surgery, Center for Orthopedics, Daping Hospital, Army Medical University, Chongqing, China.
| |
Collapse
|
8
|
Sánchez-Sánchez L, Fernández R, Astigarraga E, Barreda-Gómez G, Ganfornina MD. Microarray-Based Methodology for Lipid Profiling, Enzymatic Activity, And Binding Assays in Printed Lipid Raft Membranes from Astrocytes and Neurons. Anal Chem 2025; 97:86-95. [PMID: 39718364 PMCID: PMC11740170 DOI: 10.1021/acs.analchem.4c02421] [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: 05/08/2024] [Revised: 12/07/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024]
Abstract
Lipid rafts are liquid-ordered domains in which specific enzymes and receptors are located. These membrane platforms play crucial roles in a variety of signaling pathways. Alterations in the lipid environment, such as those elicited by oxidative stress, can lead to important functional disruptions in membrane proteins. Cell membrane microarrays have emerged in the past decade as a powerful methodology for the study of both lipids and membrane proteins at large scales. Based on that technology and the importance of liquid-ordered subdomains, we have developed a new printed lipid raft technology with a preserved native protein structure and lipid environment. To validate this technology and evaluate its potential for different aims, raft membrane microarrays (RMMAs) containing two different cell types (astrocytes and neurons) and three different conditions (astrocytes in control situation, metabolic stress, and oxidative stress) were developed. To study differences in lipid profiles between raft domains, the MALDI-MS assay was performed on RMMAs. To evaluate the preservation of native protein activities (enzymatic activity and ligand binding) in the printed raft domains, differences in NADH oxidoreductase, GAPDH, cholinesterase activities, and sigma-1 and sigma-2 binding assays were performed. We demonstrate the performance of this new microarray technology, adapted to membrane subdomains, as valid to explore changes in lipid composition and protein activities in raft domains from brain cell lines under different stress conditions relevant for neuropathology.
Collapse
Affiliation(s)
- Laura Sánchez-Sánchez
- IMG
Pharma Biotech S.L, Zamudio 48170, Spain
- Instituto
de Biomedicina y Genética Molecular, Unidad de Excelencia, University of Valladolid-CSIC, Valladolid 47003, Spain
| | | | | | | | - María Dolores Ganfornina
- Instituto
de Biomedicina y Genética Molecular, Unidad de Excelencia, University of Valladolid-CSIC, Valladolid 47003, Spain
| |
Collapse
|
9
|
Wu N, Zhang X, Li Y, Zhang J, Cui M. Fluorinated Coumarin Derivatives as Selective PET Tracer for MAO-B Imaging. J Med Chem 2025; 68:324-337. [PMID: 39699074 DOI: 10.1021/acs.jmedchem.4c01952] [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: 12/20/2024]
Abstract
Monoamine oxidase-B (MAO-B), predominantly exists on the outer mitochondrial membrane of astrocytes, serves as a crucial biomarker for reactive astrocytes during neuroinflammatory responses and various neurodegenerative diseases. In this study, we synthesized a series of fluorinated coumarin derivatives and evaluated their structure-activity relationship and subtype selectivity for MAO-B. Following this, the preclinical bioevaluation containing in vivo positron emission tomography (PET) imaging and ex vivo autoradiography studies led to the identification of the novel PET tracer, [18F]8, which demonstrated high affinity for MAO-B (IC50 = 0.59 nM) and appreciable brain pharmacokinetics (SUVmax = 2.15 at 2 min, brain2min/60min = 7.67) in rats. Furthermore, the radioactivates from [18F]8 in regions of MAO-B expression could be effectively inhibited by Selegiline. All these positive findings supported that [18F]8 is a promising candidate for MAO-B PET imaging, which merits further evaluation.
Collapse
Affiliation(s)
- Nan Wu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Xiaojun Zhang
- Department of Nuclear Medicine, Chinese PLA General Hospital, Beijing 100853, China
| | - Yuying Li
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Jinming Zhang
- Department of Nuclear Medicine, Chinese PLA General Hospital, Beijing 100853, China
| | - Mengchao Cui
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
- Center for Advanced Materials Research, Beijing Normal University, Zhuhai 519087, China
| |
Collapse
|
10
|
Stadler J, Garmo LG, Doyle D, Cheng CI, Richardson G, Waheed Z, Tofan T, Srinageshwar B, Sharma A, Petersen RB, Dunbar GL, Rossignol J. Curcumin encapsulated in PAMAM dendrimers for the therapeutic treatment of ischemic stroke in rats. Front Cell Dev Biol 2025; 12:1467417. [PMID: 39834388 PMCID: PMC11743639 DOI: 10.3389/fcell.2024.1467417] [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: 07/19/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction Ischemic stroke is a devastating neurovascular condition that occurs when cerebral tissue fails to receive an adequate supply of oxygen. Despite being a leading cause of death and disability worldwide, therapeutic interventions are currently limited. Polyamidoamine (PAMAM) dendrimers are nanomolecules commonly used in biomedical applications due to their ability to encapsulate small-molecules and improve their pharmacokinetic properties. Curcumin is known to have anti-inflammatory and antioxidant effects yet suffers from poor solubility and bioavailability. The purpose of this study is to investigate the efficacy of curcumin encapsulated in PAMAM dendrimers as a potential therapeutic treatment for ischemic stroke by studying post-stroke lesion size, astrocyte reactivity, and functional recovery in a rat model of cerebral ischemia. Methods Forty-eight male and female Sprague-Dawley rats (280-380 g) underwent either a 90-min middle cerebral artery occlusion (MCAo) or sham surgery before receiving one of four treatments: (1) Hanks' balanced salt solution (HBSS) control, (2) empty dendrimer control, (3) curcumin control, or (4) curcumin encapsulated in PAMAM dendrimer. Neurobehavioral outcomes were evaluated at 1-, 3-, 5-, and 7-day post-surgery, after which animals were euthanized on day 8 to assess infarct volume and GFAP immunoreactivity. Results Animals that received formulations containing dendrimers (curcumin encapsulated in dendrimers or empty dendrimers) demonstrated significantly lower levels of GFAP immunoreactivity and improved functional recovery, including weight and neurobehavioral scores, compared to the formulations that did not contain dendrimers (curcumin and HBSS control). Additionally, the dendrimer-curcumin treatment group exhibited a significantly improved paw laterality index over the course of the study compared with the other three treatment groups. Conclusion Although the post-stroke administration of curcumin encapsulated in PAMAM dendrimers modulates the astrocytic response and promotes functional recovery following ischemic stroke in rats, its therapeutic benefits may be driven by PAMAM dendrimers as the empty dendrimer treatment group also showed significant improvements post-stroke. Further investigation regarding PAMAM dendrimers in treating neuroinflammatory conditions remains warranted.
Collapse
Affiliation(s)
- Justin Stadler
- College of Medicine, Central Michigan University, Mount Pleasant, MI, United States
- Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States
| | - Lucas G. Garmo
- College of Medicine, Central Michigan University, Mount Pleasant, MI, United States
- Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States
| | - David Doyle
- College of Medicine, Central Michigan University, Mount Pleasant, MI, United States
- Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States
| | - Chin-I. Cheng
- Department of Statistics, Actuarial and Data Science, Central Michigan University, Mt. Pleasant, MI, United States
| | - Garrett Richardson
- College of Medicine, Central Michigan University, Mount Pleasant, MI, United States
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States
| | - Zain Waheed
- College of Medicine, Central Michigan University, Mount Pleasant, MI, United States
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States
| | - Tim Tofan
- School of Business, Wayne State University, Detroit, MI, United States
| | - Bhairavi Srinageshwar
- College of Medicine, Central Michigan University, Mount Pleasant, MI, United States
- Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States
| | - Ajit Sharma
- Department of Chemistry & Biochemistry, Central Michigan University, Mount Pleasant, MI, United States
| | - Robert B. Petersen
- College of Medicine, Central Michigan University, Mount Pleasant, MI, United States
| | - Gary L. Dunbar
- Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States
- Department of Psychology, Central Michigan University, Mount Pleasant, MI, United States
| | - Julien Rossignol
- College of Medicine, Central Michigan University, Mount Pleasant, MI, United States
- Program in Neuroscience, Central Michigan University, Mount Pleasant, MI, United States
- Field Neurosciences Institute Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, United States
| |
Collapse
|
11
|
Jia Y, Song Y, Xue H, Li X, Zhang Y, Fan S, Yang X, Ding Z, Qiu Y, Wu Z, Zhao P. Sevoflurane postconditioning mitigates neuronal hypoxic-ischemic injury via regulating reactive astrocytic STAT3 protein modification. Chem Biol Interact 2025; 405:111308. [PMID: 39536892 DOI: 10.1016/j.cbi.2024.111308] [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: 09/16/2024] [Revised: 11/01/2024] [Accepted: 11/11/2024] [Indexed: 11/16/2024]
Abstract
Astrocyte activation plays a pivotal role in accelerating the cascade of neuroinflammation associated with the development of hypoxic-ischemic brain injury. This study aimed to investigate the mechanism by which sevoflurane postconditioning mitigates neuronal damage through astrocytes by regulating reactive astrocytic Signal Transducer and Activator of Transcription 3 (STAT3) modifications. A modified Rice‒Vannucci model in rats and a conditioned culture system established by subjecting primary astrocytes to oxygen glucose deprivation, followed by using the conditioned medium to culture the neuron cell line SH-SY5Y were used to simulate HI insult in vivo and in vitro, respectively. These models were followed by 30 min of 2.5 % sevoflurane treatment. Stattic was used to inhibit STAT3 phosphorylation, and (Z)-PUGNAc or OSMI-1 was added to regulate O-linked-β-N-acetylglucosamine modification (O-GlcNAcylation) in primary astrocytes in vitro. Neurobehavioral tests, Nissl staining, CCK8 assay, and flow cytometry for apoptosis were used to assess neuronal function. Immunofluorescence staining was used to detect astrocyte reactivity and the intracellular distribution of STAT3. Immunoprecipitation combined with Western blotting was used to evaluate the O-GlcNAcylation of STAT3. Protein expression and phosphorylation levels were detected by Western blotting. ELISA was conducted to detect the detrimental cytokines IL-6 and IL-1β in astrocyte-conditioned medium. Sevoflurane postconditioning enhanced the O-GlcNAcylation of astrocytic STAT3 following HI insult via the manner of OGT. Crosstalk between O-GlcNAcylation and phosphorylation of STAT3 showed that O-GlcNAcylation inhibited STAT3 phosphorylation. The inhibitory effect on astrocytes suppressed STAT3 nuclear translocation, reduced astrocyte reactivity, decreased the release of the inflammatory cytokines IL6 and IL-1β, attenuated neuronal apoptosis following HI insult, and improved neuron viability. Sevoflurane postconditioning increased astrocytic STAT3 O-GlcNAcylation level to competitively inhibit STAT3 phosphorylation. This deactivated downstream inflammation pathways and reduced astrocyte reactivity, thereby mitigating HI insult in neurons both in vivo and in vitro.
Collapse
Affiliation(s)
- Yufei Jia
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yanhong Song
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Hang Xue
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xingyue Li
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yinong Zhang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Shiyue Fan
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xu Yang
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Zixuan Ding
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yue Qiu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Ziyi Wu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Ping Zhao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
12
|
Dzyubenko E, Hermann DM. Neuroglia and extracellular matrix molecules. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:197-211. [PMID: 40122625 DOI: 10.1016/b978-0-443-19104-6.00010-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
This chapter provides a comprehensive overview of the roles of astrocytes, microglia, and the extracellular matrix (ECM) in regulating neuroplasticity and maintaining brain homeostasis. Astrocytes provide essential metabolic support to neurons, regulate synapse development, support neuroplasticity mechanisms, and modulate neurotransmission. Microglia, the resident immune cells of the brain, play a critical role in neuroinflammatory responses and homeostatic brain regulation by modulating synapse formation and pruning. The extracellular space (ECS) mediates intercellular interactions, provides a highly regulated environment for intercellular communication, and is filled with ECM molecules. Proteoglycans and polysaccharides of the ECM play a vital role not only in brain development but also in brain function throughout life. In the injured brain, neuroplasticity and regeneration can be bidirectionally regulated as a result of the interplay between ECM, astrocytes, and microglia. The modulation of synaptic strength, structural remodeling, and modification of intrinsic neuronal properties are among the central mechanisms that contribute to neuronal plasticity in health and disease. We believe that the understanding of ECM-glia interactions and their role in neuroplasticity regulation is key to the development of novel therapeutic strategies in neurologic disorders.
Collapse
Affiliation(s)
- Egor Dzyubenko
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| | - Dirk M Hermann
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
13
|
Da Silva K, Kumar P, Choonara YE. The paradigm of stem cell secretome in tissue repair and regeneration: Present and future perspectives. Wound Repair Regen 2025; 33:e13251. [PMID: 39780313 PMCID: PMC11711308 DOI: 10.1111/wrr.13251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025]
Abstract
As the number of patients requiring organ transplants continues to rise exponentially, there is a dire need for therapeutics, with repair and regenerative properties, to assist in alleviating this medical crisis. Over the past decade, there has been a shift from conventional stem cell treatments towards the use of the secretome, the protein and factor secretions from cells. These components may possess novel druggable targets and hold the key to profoundly altering the field of regenerative medicine. Despite the progress in this field, clinical translation of secretome-containing products is limited by several challenges including but not limited to ensuring batch-to-batch consistency, the prevention of further heterogeneity, production of sufficient secretome quantities, product registration, good manufacturing practice protocols and the pharmacokinetic/pharmacodynamic profiles of all the components. Despite this, the secretome may hold the key to unlocking the regenerative blockage scientists have encountered for years. This review critically analyses the secretome derived from different cell sources and used in several tissues for tissue regeneration. Furthermore, it provides an overview of the current delivery strategies and the future perspectives for the secretome as a potential therapeutic. The success and possible shortcomings of the secretome are evaluated.
Collapse
Affiliation(s)
- Kate Da Silva
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Pradeep Kumar
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Yahya E. Choonara
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| |
Collapse
|
14
|
Li N, Wang H, Hu C, Qie S, Liu Z. Regulatory T Cells for Stroke Recovery: A Promising Immune Therapeutic Strategy. CNS Neurosci Ther 2025; 31:e70248. [PMID: 39878387 PMCID: PMC11775944 DOI: 10.1111/cns.70248] [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: 11/28/2024] [Revised: 01/07/2025] [Accepted: 01/19/2025] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Stroke remains a leading cause of mortality and disability among adults. Given the restricted therapeutic window for intravascular interventions and neuroprotection during the acute phase, there has been a growing focus on tissue repair and functional recovery in the subacute and chronic phases after stroke. The pro-inflammatory microglial polarization occurs in subacute and chronic phases after stroke and may represent therapeutic targets for stroke recovery. CD4+ regulatory T cells (Tregs), a subtype of T cells with immunosuppressive effects, have been shown to be important in stroke. Tregs infiltrate into the brain primarily during the subacute and chronic phases following a stroke. Infiltrating Tregs play a critical role in mitigating pro-inflammatory microglial responses, modulating the immune microenvironment, and promoting the functional restoration of the damaged brain following a stroke. METHODS A systematic literature search was conducted in PubMed, Scopus, and Web of Science and then conduct a comprehensive analysis of the searched literature. RESULTS This review provides a comprehensive summary of recent preclinical research advances on the role of Tregs in stroke, with a particular focus on their reparative functions during the subacute and chronic phases. It discusses changes in peripheral and brain infiltrating Tregs post-stroke, their functions and underlying mechanisms, and therapeutic strategies involving Tregs. Additionally, this review explores the potential and challenges associated with the clinical application of Tregs in ischemic stroke. CONCLUSION Treg cell-related therapy represents a promising immune-therapeutic strategy for stroke recovery. However, there are several critical issues that must be resolved before its advancement to clinical application.
Collapse
Affiliation(s)
- Ning Li
- Department of Rehabilitation, Beijing Rehabilitation HospitalCapital Medical UniversityBeijingChina
| | - Hujun Wang
- Department of Rehabilitation, Beijing Rehabilitation HospitalCapital Medical UniversityBeijingChina
| | - Changbin Hu
- Department of Rehabilitation, Beijing Rehabilitation HospitalCapital Medical UniversityBeijingChina
| | - Shuyan Qie
- Department of Rehabilitation, Beijing Rehabilitation HospitalCapital Medical UniversityBeijingChina
| | - Zongjian Liu
- Department of Research, Beijing Rehabilitation HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
15
|
Tichauer JE, Rovegno M. Role of astrocytes connexins - pannexins in acute brain injury. Neurotherapeutics 2025; 22:e00523. [PMID: 39848901 PMCID: PMC11840357 DOI: 10.1016/j.neurot.2025.e00523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/25/2025] Open
Abstract
Acute brain injuries (ABIs) encompass a broad spectrum of primary injuries such as ischemia, hypoxia, trauma, and hemorrhage that converge into secondary injury where some mechanisms show common determinants. In this regard, astroglial connexin and pannexin channels have been shown to play an important role. These channels are transmembrane proteins sharing similar topology and form gateways between adjacent cells named gap junctions (GJs) and pores into unopposed membranes named hemichannels (HCs). In astrocytes, GJs and HCs enable intercellular communication and have active participation in normal brain physiological processes, such as calcium waves, synapsis modulation, regional blood flow regulation, and homeostatic control of the extracellular environment, among others. However, after acute brain injury, astrocytes can change their phenotype and modify the activity of both channels and hemichannels, which can result in the amplification of danger signals, increased mediators of inflammation, and neuronal death, contributing to the expansion of brain damage and neurological deterioration. This is known as secondary brain damage. In this review, we discussed the main biological mechanism of secondary brain damage with a particular focus on astroglial connexin and pannexin participation during acute brain injuries.
Collapse
Affiliation(s)
- Juan E Tichauer
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile.
| | - Maximiliano Rovegno
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile.
| |
Collapse
|
16
|
Gobbo D, Kirchhoff F. Animal-based approaches to understanding neuroglia physiology in vitro and in vivo. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:229-263. [PMID: 40122627 DOI: 10.1016/b978-0-443-19104-6.00012-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
This chapter describes the pivotal role of animal models for unraveling the physiology of neuroglial cells in the central nervous system (CNS). The two rodent species Mus musculus (mice) and Rattus norvegicus (rats) have been indispensable in scientific research due to their remarkable resemblance to humans anatomically, physiologically, and genetically. Their ease of maintenance, short gestation times, and rapid development make them ideal candidates for studying the physiology of astrocytes, oligodendrocyte-lineage cells, and microglia. Moreover, their genetic similarity to humans facilitates the investigation of molecular mechanisms governing neural physiology. Mice are largely the predominant model of neuroglial research, owing to advanced genetic manipulation techniques, whereas rats remain invaluable for applications requiring larger CNS structures for surgical manipulations. Next to rodents, other animal models, namely, Danio rerio (zebrafish) and Drosophila melanogaster (fruit fly), will be discussed to emphasize their critical role in advancing our understanding of glial physiology. Each animal model provides distinct advantages and disadvantages. By combining the strengths of each of them, researchers can gain comprehensive insights into glial function across species, ultimately promoting the understanding of glial physiology in the human CNS and driving the development of novel therapeutic interventions for CNS disorders.
Collapse
Affiliation(s)
- Davide Gobbo
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany.
| | - Frank Kirchhoff
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany; Center for Gender-specific Biology and Medicine (CGBM), University of Saarland, Homburg, Germany.
| |
Collapse
|
17
|
Hayes CA, Wilson D, De Leon MA, Mustapha MJ, Morales S, Odden MC, Ashpole NM. Insulin-like growth factor-1 and cognitive health: Exploring cellular, preclinical, and clinical dimensions. Front Neuroendocrinol 2025; 76:101161. [PMID: 39536910 DOI: 10.1016/j.yfrne.2024.101161] [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: 07/31/2024] [Revised: 11/08/2024] [Accepted: 11/10/2024] [Indexed: 11/16/2024]
Abstract
Age and insulin-like growth factor-1 (IGF-1) have an inverse association with cognitive decline and dementia. IGF-1 is known to have important pleiotropic functions beginning in neurodevelopment and extending into adulthood such as neurogenesis. At the cellular level, IGF-1 has pleiotropic signaling mechanisms through the IGF-1 receptor on neurons and neuroglia to attenuate inflammation, promote myelination, maintain astrocytic functions for homeostatic balances, and neuronal synaptogenesis. In preclinical rodent models of aging and transgenic models of IGF-1, increased IGF-1 improves cognition in a variety of behavioral paradigms along with reducing IGF-1 via knockout models being able to induce cognitive impairment. At the clinical levels, most studies highlight that increased levels of IGF-1 are associated with better cognition. This review provides a comprehensive and up-to-date evaluation of the association between IGF-1 and cognition at the cellular signaling levels, preclinical, and clinical levels.
Collapse
Affiliation(s)
- Cellas A Hayes
- Department of Epidemiology and Population Health, School of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA.
| | - Destiny Wilson
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA
| | - Miguel A De Leon
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA
| | | | - Sharon Morales
- Department of Biomedical Science, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Michelle C Odden
- Department of Epidemiology and Population Health, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Nicole M Ashpole
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, University, MS 38677, USA
| |
Collapse
|
18
|
McKeever PM, Sababi AM, Sharma R, Xu Z, Xiao S, McGoldrick P, Ketela T, Sato C, Moreno D, Visanji N, Kovacs GG, Keith J, Zinman L, Rogaeva E, Goodarzi H, Bader GD, Robertson J. Single-nucleus transcriptome atlas of orbitofrontal cortex in amyotrophic lateral sclerosis with a deep learning-based decoding of alternative polyadenylation mechanisms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.22.573083. [PMID: 38187588 PMCID: PMC10769403 DOI: 10.1101/2023.12.22.573083] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) are two age-related and fatal neurodegenerative disorders that lie on a shared disease spectrum. While both disorders involve complex interactions between neuronal and glial cells, the specific cell-type alterations and their contributions to disease pathophysiology remain incompletely understood. Here, we applied single-nucleus RNA sequencing of the orbitofrontal cortex, a region affected in ALS-FTLD, to map cell-type specific transcriptional signatures in C9orf72-related ALS (with and without FTLD) and sporadic ALS cases. Our findings reveal disease- and cell-type-specific transcriptional changes, with neurons exhibiting the most pronounced alterations, primarily affecting mitochondrial function, protein homeostasis, and chromatin remodeling. A comparison with independent datasets from different cortical regions of C9orf72 and sporadic ALS cases showed concordance in several pathways, with neuronal STMN2 and NEFL showing consistent up-regulation between brain regions and disease subtypes. We also interrogated alternative polyadenylation (APA) as an additional layer of transcriptional regulation, demonstrating that APA events are not correlated with identified gene expression changes. To interpret these events, we developed APA-Net, a deep learning model that integrates transcript sequences with RNA-binding protein expression profiles, revealing cell type-specific patterns of APA regulation. Our atlas illuminates cell type-specific pathomechanisms of ALS/FTLD, providing a valuable resource for further investigation.
Collapse
|
19
|
Meng F, Cui J, Wang P, Wang J, Sun J, Li L. The Phenotype Changes of Astrocyte During Different Ischemia Conditions. Brain Sci 2024; 14:1256. [PMID: 39766455 PMCID: PMC11674399 DOI: 10.3390/brainsci14121256] [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: 10/23/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVES Dementia is becoming a major health problem in the world, and chronic brain ischemia is an established important risk factor in predisposing this disease. Astrocytes, as one major part of the blood-brain barrier (BBB), are activated during chronic cerebral blood flow hypoperfusion. Reactive astrocytes have been classified into phenotype pro-inflammatory type A1 or neuroprotective type A2. However, the specific subtype change of astrocyte and the mechanisms of chronic brain ischemia are still unknown. METHODS In order to depict the phenotype changes and their possible roles during this process, a rat bilateral common carotid artery occlusion model (BCAO) was employed in the present study. Meanwhile, the signaling pathways that possibly regulate these changes were investigated as well. RESULTS After four-week occlusion, astrocytes in the cortex of BCAO rats were shown to be the A2 phenotype, identified by the significant up-regulation of S100a10 accompanied by the down-regulation of Connexin 43 (CX43) protein. Next, we established in vitro hypoxia models, which were set up by stimulating primary astrocyte cultures from rat cortex with cobalt chloride, low glucose, or/and fibrinogen. Consistent with in vivo data, the cultured astrocytes also transformed into the A2 phenotype with the up-regulation of S100a10 and the down-regulation of CX43. In order to explore the mechanism of CX43 protein changes, C6 astrocyte cells were handled in both hypoxia and low-glucose stimulus, in which decreased pERK and pJNK expression were found. CONCLUSIONS In conclusion, our data suggest that in chronic cerebral ischemia conditions, the gradual ischemic insults could promote the transformation of astrocytes into A2 type instead of A1 type, and the phosphorylation of CX43 was negatively regulated by the phosphorylation of ERK and JNK. Also, our data could provide some new evidence of how to leverage the endogenous astrocytes phenotype changes during CNS injury by promoting them to be "protector" and not "culprit".
Collapse
Affiliation(s)
- Fei Meng
- Cardiac Valve Center, Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 101100, China;
| | - Jing Cui
- Department of Pathology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (J.C.); (J.S.)
| | - Peng Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Department of Cardiology, Qilu Hospital, Shandong University, Jinan 250012, China;
| | - Junhui Wang
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada;
| | - Jing Sun
- Department of Pathology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (J.C.); (J.S.)
| | - Liang Li
- Department of Pathology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (J.C.); (J.S.)
| |
Collapse
|
20
|
Sámano C, Mazzone GL. The role of astrocytes response triggered by hyperglycaemia during spinal cord injury. Arch Physiol Biochem 2024; 130:724-741. [PMID: 37798949 DOI: 10.1080/13813455.2023.2264538] [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: 03/24/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023]
Abstract
OBJECTIVE This manuscript aimed to provide a comprehensive overview of the physiological, molecular, and cellular mechanisms triggered by reactive astrocytes (RA) in the context of spinal cord injury (SCI), with a particular focus on cases involving hyperglycaemia. METHODS The compilation of articles related to astrocyte responses in neuropathological conditions, with a specific emphasis on those related to SCI and hyperglycaemia, was conducted by searching through databases including Science Direct, Web of Science, and PubMed. RESULTS AND CONCLUSIONS This article explores the dual role of astrocytes in both neurophysiological and neurodegenerative conditions within the central nervous system (CNS). In the aftermath of SCI and hyperglycaemia, astrocytes undergo a transformation into RA, adopting a distinct phenotype. While there are currently no approved therapies for SCI, various therapeutic strategies have been proposed to alleviate the detrimental effects of RAs following SCI and hyperglycemia. These strategies show promising potential in the treatment of SCI and its likely comorbidities.
Collapse
Affiliation(s)
- C Sámano
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana, Unidad Cuajimalpa (UAM-C), Ciudad de México, México
| | - G L Mazzone
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Pilar, Buenos Aires, Argentina
- Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Buenos Aires, Argentina
| |
Collapse
|
21
|
Kong J, Zhang Q, Zheng H, Tang D, Fang L, An S, Li J, Fan Z. TGN-020 ameliorates motor dysfunction post-spinal cord injury via enhancing astrocyte autophagy and mitigating inflammation by activating AQP4/PPAR-γ/mTOR pathway. Exp Neurol 2024; 382:114975. [PMID: 39326822 DOI: 10.1016/j.expneurol.2024.114975] [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: 08/15/2024] [Revised: 09/17/2024] [Accepted: 09/22/2024] [Indexed: 09/28/2024]
Abstract
Spinal Cord Injury (SCI) is a severe condition that often leads to substantial neurological impairments. This study aimed to explore the role of Aquaporin-4 (AQP4) in regulating astrocyte autophagy and neuroinflammation post-SCI, as well as to evaluate the therapeutic potential of AQP4 inhibition using the specific inhibitor TGN-020. Using Western blot, CCK8 assays, immunofluorescence staining, histopathological assessments, and behavioral analyses, we investigated the effects of TGN-020 on SCI-induced alterations in autophagy, neuroinflammation, astrocyte proliferation, neuronal damage, and motor function recovery in both rat and astrocyte models. Our findings indicate that TGN-020 significantly enhances astrocyte autophagy, reduces neuroinflammation, thereby leading to mitigated astrocyte activation by suppressing AQP4 expression. These beneficial effects are associated with the activation of the peroxisome proliferator-activated receptor-γ/mammalian target of rapamycin (PPAR-γ/mTOR) signaling pathway. Notably, the introduction of the PPAR-γ specific inhibitor GW9662 abrogated the positive regulatory effects of TGN-020 on SCI-induced autophagy and neuroinflammation. Collectively, our in vivo and in vitro experiments demonstrate that TGN-020, by down-regulating AQP4, activates the PPAR-γ/mTOR pathway, ameliorates astrocyte autophagy, diminishes neuroinflammation, and ultimately enhances motor function recovery.
Collapse
Affiliation(s)
- Jundong Kong
- Department of Orthopedics, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China
| | - Qiangqiang Zhang
- Department of Orthopedics, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China
| | - Haohong Zheng
- Department of Orthopedics, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China
| | - Diandong Tang
- Department of Orthopedics, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China
| | - Li Fang
- Department of Orthopedics, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China
| | - Shuaihao An
- Department of Orthopedics, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China
| | - Jian Li
- Department of Orthopedics, The First Affiliated Hospital, Jinzhou Medical University, Jinzhou 121000, China.
| | - Zhongkai Fan
- Jinzhou Medical University, Jinzhou 121000, China.
| |
Collapse
|
22
|
Rigo YR, Benvenutti R, Portela LV, Strogulski NR. Neurogenic potential of NG2 in neurotrauma: a systematic review. Neural Regen Res 2024; 19:2673-2683. [PMID: 38595286 PMCID: PMC11168526 DOI: 10.4103/nrr.nrr-d-23-01031] [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: 06/21/2023] [Revised: 12/20/2023] [Accepted: 02/07/2024] [Indexed: 04/11/2024] Open
Abstract
Regenerative approaches towards neuronal loss following traumatic brain or spinal cord injury have long been considered a dogma in neuroscience and remain a cutting-edge area of research. This is reflected in a large disparity between the number of studies investigating primary and secondary injury as therapeutic targets in spinal cord and traumatic brain injuries. Significant advances in biotechnology may have the potential to reshape the current state-of-the-art and bring focus to primary injury neurotrauma research. Recent studies using neural-glial factor/antigen 2 (NG2) cells indicate that they may differentiate into neurons even in the developed brain. As these cells show great potential to play a regenerative role, studies have been conducted to test various manipulations in neurotrauma models aimed at eliciting a neurogenic response from them. In the present study, we systematically reviewed the experimental protocols and findings described in the scientific literature, which were peer-reviewed original research articles (1) describing preclinical experimental studies, (2) investigating NG2 cells, (3) associated with neurogenesis and neurotrauma, and (4) in vitro and/or in vivo, available in PubMed/MEDLINE, Web of Science or SCOPUS, from 1998 to 2022. Here, we have reviewed a total of 1504 papers, and summarized findings that ultimately suggest that NG2 cells possess an inducible neurogenic potential in animal models and in vitro. We also discriminate findings of NG2 neurogenesis promoted by different pharmacological and genetic approaches over functional and biochemical outcomes of traumatic brain injury and spinal cord injury models, and provide mounting evidence for the potential benefits of manipulated NG2 cell ex vivo transplantation in primary injury treatment. These findings indicate the feasibility of NG2 cell neurogenesis strategies and add new players in the development of therapeutic alternatives for neurotrauma.
Collapse
Affiliation(s)
- Yuri R. Rigo
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Radharani Benvenutti
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Luis V. Portela
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Nathan R. Strogulski
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, University of Dublin, Dublin, Ireland
| |
Collapse
|
23
|
Hatakeyama T, Kawai N, Maruo T, Norikane T, Yamamoto Y, Miyake K. Reactive Astrocytes Promote Axonal Remodeling of the Corticospinal Tract During Neuronal Recovery Revealed by 18F-THK5351 PET. Clin Nucl Med 2024; 49:1145-1147. [PMID: 39485873 DOI: 10.1097/rlu.0000000000005509] [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: 11/03/2024]
Abstract
ABSTRACT A teenager who suffered from left hemiparesis after traumatic brain injury underwent 18F-THK5351 PET 48, 286, and 810 days after the injury. The first scan showed slight uptake in the right corticospinal tract (CST), and the second scan showed intense uptake along the CST, which was significantly reduced in the third scan. The hemiparesis has improved between the first and second scans. 18F-THK5351 binds to monoamine oxidase B, which is expressed in reactive astrocytes (RAs). Recently, the beneficial role of RAs in plasticity and reconstruction after traumatic brain injury has been reported. 18F-THK5351 uptake may represent axonal remodeling accompanied with RAs in the CST.
Collapse
Affiliation(s)
- Tetsuhiro Hatakeyama
- From the Department of Neurological Surgery, Faculty of Medicine, Kagawa University
| | - Nobuyuki Kawai
- Department of Neurological Surgery, Kagawa Rehabilitation Hospital
| | - Tomoko Maruo
- Department of Neurological Surgery, Kagawa Rehabilitation Hospital
| | - Takashi Norikane
- Department of Radiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Yuka Yamamoto
- Department of Radiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Keisuke Miyake
- From the Department of Neurological Surgery, Faculty of Medicine, Kagawa University
| |
Collapse
|
24
|
Maguire AS, Ta L, Gross AL, Osterhoudt DE, Cannon JS, Hall PI, Sandey M, Seyfried TN, Gray-Edwards HL, Sena-Esteves M, Martin DR. Intravenous gene therapy improves lifespan and clinical outcomes in feline Sandhoff Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.15.623838. [PMID: 39605340 PMCID: PMC11601349 DOI: 10.1101/2024.11.15.623838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Sandhoff Disease (SD), a fatal neurodegenerative disorder, is caused by the absence of ß-hexosaminidase (Hex) and subsequent accumulation of GM2 ganglioside in lysosomes. Previous studies have led to adeno-associated virus (AAV) gene therapy for children with GM2 gangliosidosis in both expanded access and Phase I/II clinical trials via intracranial and/or cerebrospinal fluid-based delivery. The current study investigated intravenous (IV) gene therapy of SD cats, treated at one month of age with a bicistronic AAV vector. While untreated SD cats lived to 4.3±0.2 months, cats treated with low and high doses lived to 8.3±1.2 and 12.4±2.7 months, respectively. In-life assessments revealed clear clinical benefit of AAV treatment, with the most dramatic improvement seen in the reduction of overt full-body tremors. Cerebrospinal fluid levels of aspartate aminotransferase (AST) and lactate dehydrogenase (LDH) were decreased, indicating a reduction of cell damage within the central nervous system. Magnetic resonance imaging (MRI) and spectroscopy (MRS) acquired on a 7 Tesla scanner indicated that structural pathology and metabolite abnormalities are partially normalized by AAV treatment. Dose-dependent reduction of GM2 ganglioside storage and increases in Hex activity were most substantial in the caudal regions of the brain and in the spinal cord. Immunohistochemistry revealed reduction in neuroinflammatory cell populations and partial correction of myelin deficits. These results support the dose-dependent efficacy of AAV delivered IV for significant restoration of clinical metrics and Hex function in a feline model of SD.
Collapse
Affiliation(s)
- Anne S. Maguire
- Scott Ritchey Research Center, Auburn University College of Veterinary Medicine; Auburn, AL, USA
- Department of Anatomy, Physiology, & Pharmacology, Auburn University College of Veterinary Medicine; Auburn, AL, USA
| | - Linh Ta
- Biology Department, Boston College, Chestnut Hill, MA, USA
| | - Amanda L. Gross
- Scott Ritchey Research Center, Auburn University College of Veterinary Medicine; Auburn, AL, USA
| | - Devin E. Osterhoudt
- Scott Ritchey Research Center, Auburn University College of Veterinary Medicine; Auburn, AL, USA
| | - Jessica S. Cannon
- Scott Ritchey Research Center, Auburn University College of Veterinary Medicine; Auburn, AL, USA
| | - Paige I. Hall
- Scott Ritchey Research Center, Auburn University College of Veterinary Medicine; Auburn, AL, USA
| | - Maninder Sandey
- Department of Pathobiology, Auburn University College of Veterinary Medicine; Auburn, AL, USA
| | | | - Heather L. Gray-Edwards
- Department of Radiology, University of Massachusetts Medical School, Worcester, MA, USA
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Miguel Sena-Esteves
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Douglas R. Martin
- Scott Ritchey Research Center, Auburn University College of Veterinary Medicine; Auburn, AL, USA
- Department of Anatomy, Physiology, & Pharmacology, Auburn University College of Veterinary Medicine; Auburn, AL, USA
| |
Collapse
|
25
|
Zheng X, Zhang H, Zhang Y, Ding Z, Huang Z, Li H, Yao M, Song W, Liu J. Salidroside ameliorates cerebral ischemic injury and regulates the glutamate metabolism pathway in astrocytes. Front Pharmacol 2024; 15:1472100. [PMID: 39600364 PMCID: PMC11588439 DOI: 10.3389/fphar.2024.1472100] [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: 08/02/2024] [Accepted: 10/17/2024] [Indexed: 11/29/2024] Open
Abstract
Background and Aim Salidroside (SA) is the main active component of Rhodiola rosea L., with potential in treating cardiovascular and cerebrovascular diseases and cerebral ischemia. However, its efficacy and mechanism in cerebral ischemia remain unclear, particularly regarding its effect on glutamate (Glu) metabolism. In this paper, we aimed to investigate the efficacy of SA in treating cerebral ischemia and its pharmacological mechanism. Experimental procedure We studied the effects of SA on SD rats with cerebral ischemia, evaluating neurobehavior, cerebral water content, infarct size, and brain microstructure. We also assessed its impact on glial fibrillary acidic protein (GFAP), glutamine synthetase (GS), and glutamate transporter 1 (GLT-1) proteins using immunohistochemistry and Western blot. Additionally, we used SVGp12 cells to simulate cerebral ischemia and measured Glu levels and used Western blot to observe the level of GS and GLT-1. Results SA improved neural function, reduced infarct size, and regulated GSH and Glu levels in rats. In cell experiments, SA increased cell viability and decreased Glu concentration after ischemia induction. It also regulated the expression of GFAP, GS, and GLT-1. Conclusion SA alleviates cerebral ischemia-induced injury by acting on astrocytes, possibly through regulating the glutamate metabolic pathway.
Collapse
Affiliation(s)
- Xiaoyu Zheng
- Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing, China
| | - Hongwei Zhang
- Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, China
| | - Yehao Zhang
- Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing, China
| | - Zhao Ding
- Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing, China
| | - Zishan Huang
- Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing, China
- Research Institute of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Haoran Li
- Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing, China
- Research Institute of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Mingjiang Yao
- Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing, China
| | - Wenting Song
- Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing, China
| | - Jianxun Liu
- Beijing Key Laboratory of Traditional Chinese Medicine Pharmacology, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, National Research Center for Clinical Medicine of Cardiovascular Diseases of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
26
|
Soltani Khaboushan A, Zafari R, Sabahi M, Khorasanizadeh M, Dabbagh Ohadi MA, Flouty O, Ranjan M, Slavin KV. Focused ultrasound for treatment of epilepsy: a systematic review and meta-analysis of preclinical and clinical studies. Neurosurg Rev 2024; 47:839. [PMID: 39521750 DOI: 10.1007/s10143-024-03078-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 09/28/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024]
Abstract
Various preclinical and clinical studies have demonstrated the neuromodulatory and ablative effects of focused ultrasound (FUS). However, the safety and efficacy of FUS in clinical settings for treating epilepsy have not been well established. This study aims to provide a systematic review of all preclinical and clinical studies that have used FUS for the treatment of epilepsy. A systematic search was conducted using Scopus, Web of Science, PubMed, and Embase databases. All preclinical and clinical studies reporting outcomes of FUS in the treatment of epilepsy were included in the systematic review. Random-effect meta-analysis was performed to determine safety in clinical studies and seizure activity reduction in preclinical studies. A total of 24 articles were included in the study. Meta-analysis demonstrated that adverse events occurred in 13% (95% CI = 2-57%) of patients with epilepsy who underwent FUS. The frequency of adverse events was higher with the use of FUS for lesioning (36%, 95% CI = 4-88%) in comparison to neuromodulation (5%, 95% CI = 0-71%), although this difference was not significant (P = 0.31). Three-level meta-analysis in preclinical studies demonstrated a reduced spike rate in neuromodulating FUS compared to the control group (P = 0.02). According to this systematic review and meta-analysis, FUS can be considered a safe and feasible approach for treating epileptic seizures, especially in drug-resistant patients. While the efficacy of FUS has been demonstrated in several preclinical studies, further research is necessary to confirm its effectiveness in clinical practice and to determine the adverse events.
Collapse
Affiliation(s)
- Alireza Soltani Khaboushan
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Neurosurgery, Tehran University of Medical Sciences, Tehran, Iran
| | - Rasa Zafari
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadmahdi Sabahi
- Department of Neurological Surgery, Pauline Braathen Neurological Centre, Cleveland Clinic Florida, Weston, FL, USA
| | - MirHojjat Khorasanizadeh
- Department of Neurosurgery, Mount Sinai Hospital, Icahn School of Medicine, New York City, NY, USA
| | - Mohammad Amin Dabbagh Ohadi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Neurosurgery, Tehran University of Medical Sciences, Tehran, Iran
| | - Oliver Flouty
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Manish Ranjan
- Department of Neurosurgery, Rockefeller Neuroscience Institute, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Konstantin V Slavin
- Department of Neurosurgery, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
27
|
Jeon SY, Yang HW, Son BR, Baek J, Kim JL. Caregiving-Related Depression Increases Neuroinflammation in Spousal Caregivers to Individuals With Cognitive Impairment: A Longitudinal Study. J Gerontol A Biol Sci Med Sci 2024; 79:glae235. [PMID: 39297507 PMCID: PMC11638088 DOI: 10.1093/gerona/glae235] [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: 04/24/2024] [Indexed: 12/14/2024] Open
Abstract
BACKGROUND The caregiving burden of the spousal caregivers (SCGs) to individuals with cognitive impairment poses public health challenges with adverse psychosocial and physiological effects. However, few studies have investigated the neurobiological impact of caregiving, particularly through the investigation of neuroinflammation and neurodegeneration. METHODS Using data from a longitudinal cohort at Chungnam National University Hospital, the relationship between caregiving burden, neuroinflammation, and neurodegeneration was examined in 38 older adult couples over a 16-month period. Caregiving burden was assessed through a multifaceted approach. For factors related to the care recipient, we assessed cognitive function and neuropsychiatric symptoms. Factors regarding the SCGs included the measurement of perceived depression. Glial fibrillary acidic protein (GFAP) was used as a plasma biomarker for neuroinflammation and neurofilament light chain (NfL) for neurodegeneration. Regression analyses were adjusted for age, sex, apolipoprotein E status, follow-up interval, vascular risk factors, and physical activity. RESULTS Changes in depression among SCGs were significantly correlated with increased GFAP levels (p = .003), indicating that greater depressive symptoms during caregiving are associated with increased neuroinflammation. In contrast, no significant correlations were found between changes in cognitive function or neuropsychiatric symptoms in care recipients and the plasma biomarker levels of SCGs. Additionally, there was no significant association between changes in depression and NfL levels in SCGs. CONCLUSIONS The psychological stress experienced by SCGs while caring for partners with cognitive impairment actively contributes to neuroinflammation, a well-known risk factor for various diseases. This study emphasizes the need to address psychological stress experienced by older adult caregivers.
Collapse
Affiliation(s)
- So Yeon Jeon
- Department of Psychiatry, Chungnam National University Hospital, Daejeon, Republic of Korea
- Department of Psychiatry, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hee Won Yang
- Department of Psychiatry, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Bo Ran Son
- Brain Research Institute, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jimin Baek
- Department of Psychiatry, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Jeong Lan Kim
- Department of Psychiatry, Chungnam National University Hospital, Daejeon, Republic of Korea
- Department of Psychiatry, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
28
|
Saito A, Omura I, Imaizumi K. CREB3L1/OASIS: cell cycle regulator and tumor suppressor. FEBS J 2024; 291:4853-4866. [PMID: 38215153 DOI: 10.1111/febs.17052] [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: 09/11/2023] [Revised: 11/09/2023] [Accepted: 01/05/2024] [Indexed: 01/14/2024]
Abstract
Cell cycle checkpoints detect DNA errors, eventually arresting the cell cycle to promote DNA repair. Failure of such cell cycle arrest causes aberrant cell proliferation, promoting the pathogenesis of multiple diseases, including cancer. Endoplasmic reticulum (ER) stress transducers activate the unfolded protein response, which not only deals with unfolded proteins in ER lumen but also orchestrates diverse physiological phenomena such as cell differentiation and lipid metabolism. Among ER stress transducers, cyclic AMP-responsive element-binding protein 3-like protein 1 (CREB3L1) [also known as old astrocyte specifically induced substance (OASIS)] is an ER-resident transmembrane transcription factor. This molecule is cleaved by regulated intramembrane proteolysis, followed by activation as a transcription factor. OASIS is preferentially expressed in specific cells, including astrocytes and osteoblasts, to regulate their differentiation. In accordance with its name, OASIS was originally identified as being upregulated in long-term-cultured astrocytes undergoing cell cycle arrest because of replicative stress. In the context of cell cycle regulation, previously unknown physiological roles of OASIS have been discovered. OASIS is activated as a transcription factor in response to DNA damage to induce p21-mediated cell cycle arrest. Although p21 is directly induced by the master regulator of the cell cycle, p53, no crosstalk occurs between p21 induction by OASIS or p53. Here, we summarize previously unknown cell cycle regulation by ER-resident transcription factor OASIS, particularly focusing on commonalities and differences in cell cycle arrest between OASIS and p53. This review also mentions tumorigenesis caused by OASIS dysfunctions, and OASIS's potential as a tumor suppressor and therapeutic target.
Collapse
Affiliation(s)
- Atsushi Saito
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Issei Omura
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Kazunori Imaizumi
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, Japan
| |
Collapse
|
29
|
Moon CE, Lee JK, Kim H, Kwon JM, Kang Y, Han J, Ji YW, Seo Y. Proteomic analysis of CD29+ Müller cells reveals metabolic reprogramming in rabbit myopia model. Sci Rep 2024; 14:24072. [PMID: 39402218 PMCID: PMC11473955 DOI: 10.1038/s41598-024-75637-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
The prevalence of myopia is rapidly increasing, significantly impacting the quality of life of affected individuals. Prior research by our group revealed reactive gliosis in Müller cells within myopic retina, prompting further investigation of their role in myopia, which remains unclear. In this study, we analyzed protein expression changes in CD29+ Müller cells isolated from a form deprivation-induced rabbit model of myopia using magnetic activated cell sorting to investigate the role of these cells in myopia. As the principal glial cells in the retina, Müller cells exhibited significant alterations in the components of metabolic pathways, particularly glycolysis and angiogenesis, including the upregulation of glycolytic enzymes, such as lactate dehydrogenase A and pyruvate kinase, implicated in the adaptation to increased metabolic demands under myopic stress. Additionally, a decrease in the expression of proteins associated with oxygen transport suggested enhanced vulnerability to oxidative stress. These findings highlight the proactive role of CD29+ Müller cells in modifying the retinal environment in response to myopic stress and provide valuable insights into mechanisms that could help mitigate myopia progression.
Collapse
Affiliation(s)
- Chae-Eun Moon
- Department of Ophthalmology, Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, 50 Yonsei‑ro, Seodaemun‑gu, Seoul, 03722, Republic of Korea
| | - Jun-Ki Lee
- Department of Ophthalmology, Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, 50 Yonsei‑ro, Seodaemun‑gu, Seoul, 03722, Republic of Korea
| | - Hyunjin Kim
- Department of Ophthalmology, Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, 50 Yonsei‑ro, Seodaemun‑gu, Seoul, 03722, Republic of Korea
| | - Ji-Min Kwon
- Department of Ophthalmology, Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, 50 Yonsei‑ro, Seodaemun‑gu, Seoul, 03722, Republic of Korea
| | - Yujin Kang
- Department of Ophthalmology, Institute of Vision Research, Severance Hospital, Yonsei University College of Medicine, 50 Yonsei‑ro, Seodaemun‑gu, Seoul, 03722, Republic of Korea
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jinu Han
- Department of Ophthalmology, Gangnam Severance Hospital, Yonsei University College of Medicine, 211, Eounju-ro, Gangnam-gu, Seoul, 03722, Republic of Korea
| | - Yong Woo Ji
- Department of Ophthalmology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin-si, 16995, Gyeonggi-do, Republic of Korea.
| | - Yuri Seo
- Department of Ophthalmology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin-si, 16995, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
30
|
Zhang AY, Elias E, Manners MT. Sex-dependent astrocyte reactivity: Unveiling chronic stress-induced morphological changes across multiple brain regions. Neurobiol Dis 2024; 200:106610. [PMID: 39032799 PMCID: PMC11500746 DOI: 10.1016/j.nbd.2024.106610] [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: 05/28/2024] [Revised: 07/08/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024] Open
Abstract
Chronic stress is a major precursor to various neuropsychiatric disorders and is linked with increased inflammation in the brain. However, the bidirectional association between inflammation and chronic stress has yet to be fully understood. Astrocytes are one of the key inflammatory regulators in the brain, and the morphological change in reactive astrocytes serves as an important indicator of inflammation. In this study, we evaluated the sex-specific astrocyte response to chronic stress or systemic inflammation in key brain regions associated with mood disorders. We conducted the unpredictable chronic mild stress (UCMS) paradigm to model chronic stress, or lipopolysaccharide (LPS) injection to model systemic inflammation. To evaluate stress-induced morphological changes in astrocyte complexity, we measured GFAP fluorescent intensity for astrocyte expression, branch bifurcation by quantifying branch points and terminal points, branch arborization by conducting Sholl analysis, and calculated the ramification index. Our analysis indicated that chronic stress-induced morphological changes in astrocytes in all brain regions investigated. The effects of chronic stress were region and sex specific. Notably, females had greater stress or inflammation-induced astrocyte activation in the hypothalamus (HYPO), CA1, CA3, and amygdala (AMY) than males. These findings indicate that chronic stress induces astrocyte activation that may drive sex and region-specific effects in females, potentially contributing to sex-dependent mechanisms of disease.
Collapse
Affiliation(s)
- Ariel Y Zhang
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA.
| | - Elias Elias
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA.
| | - Melissa T Manners
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA.
| |
Collapse
|
31
|
Mishra RR, Nielsen BE, Trudrung MA, Lee S, Bolstad LJ, Hellenbrand DJ, Hanna AS. The Effect of Tissue Inhibitor of Metalloproteinases on Scar Formation after Spinal Cord Injury. Cells 2024; 13:1547. [PMID: 39329731 PMCID: PMC11430430 DOI: 10.3390/cells13181547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
Spinal cord injury (SCI) often results in permanent loss of motor and sensory function. After SCI, the blood-spinal cord barrier (BSCB) is disrupted, causing the infiltration of neutrophils and macrophages, which secrete several kinds of cytokines, as well as matrix metalloproteinases (MMPs). MMPs are proteases capable of degrading various extracellular matrix (ECM) proteins, as well as many non-matrix substrates. The tissue inhibitor of MMPs (TIMP)-1 is significantly upregulated post-SCI and operates via MMP-dependent and MMP-independent pathways. Through the MMP-dependent pathway, TIMP-1 directly reduces inflammation and destruction of the ECM by binding and blocking the catalytic domains of MMPs. Thus, TIMP-1 helps preserve the BSCB and reduces immune cell infiltration. The MMP-independent pathway involves TIMP-1's cytokine-like functions, in which it binds specific TIMP surface receptors. Through receptor binding, TIMP-1 can stimulate the proliferation of several types of cells, including keratinocytes, aortic smooth muscle cells, skin epithelial cells, corneal epithelial cells, and astrocytes. TIMP-1 induces astrocyte proliferation, modulates microglia activation, and increases myelination and neurite extension in the central nervous system (CNS). In addition, TIMP-1 also regulates apoptosis and promotes cell survival through direct signaling. This review provides a comprehensive assessment of TIMP-1, specifically regarding its contribution to inflammation, ECM remodeling, and scar formation after SCI.
Collapse
Affiliation(s)
- Raveena R. Mishra
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.R.M.); (B.E.N.); (M.A.T.); (S.L.); (L.J.B.)
| | - Brooke E. Nielsen
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.R.M.); (B.E.N.); (M.A.T.); (S.L.); (L.J.B.)
| | - Melissa A. Trudrung
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.R.M.); (B.E.N.); (M.A.T.); (S.L.); (L.J.B.)
| | - Samuel Lee
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.R.M.); (B.E.N.); (M.A.T.); (S.L.); (L.J.B.)
| | - Luke J. Bolstad
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.R.M.); (B.E.N.); (M.A.T.); (S.L.); (L.J.B.)
| | - Daniel J. Hellenbrand
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.R.M.); (B.E.N.); (M.A.T.); (S.L.); (L.J.B.)
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Amgad S. Hanna
- Department of Neurosurgery, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.R.M.); (B.E.N.); (M.A.T.); (S.L.); (L.J.B.)
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
32
|
Pérez LA, Palacios E, González MF, Leyton-Rivera I, Martínez-Meza S, Pérez-Núñez R, Jeldes E, Avalos AM, Díaz J, Leyton L. A Pro-Inflammatory Stimulus versus Extensive Passaging of DITNC1 Astrocyte Cultures as Models to Study Astrogliosis. Int J Mol Sci 2024; 25:9454. [PMID: 39273404 PMCID: PMC11394751 DOI: 10.3390/ijms25179454] [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: 07/12/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Astrogliosis is a process by which astrocytes, when exposed to inflammation, exhibit hypertrophy, motility, and elevated expression of reactivity markers such as Glial Fibrillar Acidic Protein, Vimentin, and Connexin43. Since 1999, our laboratory in Chile has been studying molecular signaling pathways associated with "gliosis" and has reported that reactive astrocytes upregulate Syndecan 4 and αVβ3 Integrin, which are receptors for the neuronal glycoprotein Thy-1. Thy-1 engagement stimulates adhesion and migration of reactive astrocytes and induces neurons to retract neurites, thus hindering neuronal network repair. Reportedly, we have used DITNC1 astrocytes and neuron-like CAD cells to study signaling mechanisms activated by the Syndecan 4-αVβ3 Integrin/Thy-1 interaction. Importantly, the sole overexpression of β3 Integrin in non-reactive astrocytes turns them into reactive cells. In vitro, extensive passaging is a simile for "aging", and aged fibroblasts have shown β3 Integrin upregulation. However, it is not known if astrocytes upregulate β3 Integrin after successive cell passages. Here, we hypothesized that astrocytes undergoing long-term passaging increase β3 Integrin expression levels and behave as reactive astrocytes without needing pro-inflammatory stimuli. We used DITNC1 cells with different passage numbers to study reactivity markers using immunoblots, immunofluorescence, and astrocyte adhesion/migration assays. We also evaluated β3 Integrin levels by immunoblot and flow cytometry, as well as the neurotoxic effects of reactive astrocytes. Serial cell passaging mimicked the effects of inflammatory stimuli, inducing astrocyte reactivity. Indeed, in response to Thy-1, β3 Integrin levels, as well as cell adhesion and migration, gradually increased with multiple passages. Importantly, these long-lived astrocytes expressed and secreted factors that inhibited neurite outgrowth and caused neuronal death, just like reactive astrocytes in culture. Therefore, we describe two DITNC1 cell types: a non-reactive type that can be activated with Tumor Necrosis Factor (TNF) and another one that exhibits reactive astrocyte features even in the absence of TNF treatment. Our results emphasize the importance of passage numbers in cell behavior. Likewise, we compare the pro-inflammatory stimulus versus long-term in-plate passaging of cell cultures and introduce them as astrocyte models to study the reactivity process.
Collapse
Affiliation(s)
- Leonardo A Pérez
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
| | - Esteban Palacios
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
- Laboratorio de Microbiología Celular, Instituto de Investigación y Postgrado, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago 833-0546, Chile
| | - María Fernanda González
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
| | - Ignacio Leyton-Rivera
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
| | - Samuel Martínez-Meza
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
| | - Ramón Pérez-Núñez
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
| | - Emanuel Jeldes
- Andes Biotechnologies SpA, Santiago 7750000, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia y Vida, Santiago 7750000, Chile
| | - Ana María Avalos
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 7500912, Chile
| | - Jorge Díaz
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 838-0453, Chile
| |
Collapse
|
33
|
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
|
34
|
Bubik A, Frangež R, Žužek MC, Gutiérrez-Aguirre I, Lah TT, Sedmak B. Cyanobacterial Cyclic Peptides Can Disrupt Cytoskeleton Organization in Human Astrocytes-A Contribution to the Understanding of the Systemic Toxicity of Cyanotoxins. Toxins (Basel) 2024; 16:374. [PMID: 39330832 PMCID: PMC11436104 DOI: 10.3390/toxins16090374] [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: 07/27/2024] [Revised: 08/13/2024] [Accepted: 08/21/2024] [Indexed: 09/28/2024] Open
Abstract
The systemic toxicity of cyclic peptides produced by cyanobacteria (CCPs) is not yet completely understood. Apart from the most known damages to the liver and kidneys, symptoms of their neurotoxicity have also been reported. Hepatotoxic CCPs, like microcystins, as well as non-hepatotoxic anabaenopeptins and planktopeptins, all exhibit cytotoxic and cytostatic effects on mammalian cells. However, responses of different cell types to CCPs depend on their specific modes of interaction with cell membranes. This study demonstrates that non-hepatotoxic planktopeptin BL1125 and anabaenopeptins B and F, at concentrations up to 10 µM, affect normal and tumor human astrocytes (NHA and U87-GM) in vitro by their almost immediate insertion into the lipid monolayer. Like microcystin-LR (up to 1 µM), they inhibit Ser/Thr phosphatases and reorganize cytoskeletal elements, with modest effects on their gene expression. Based on the observed effects on intermediate filaments and intermediate filament linkage elements, their direct or indirect influence on tubulin cytoskeletons via post-translational modifications, we conclude that the basic mechanism of CCP toxicities is the induction of inter- and intracellular communication failure. The assessed inhibitory activity on Ser/Thr phosphatases is also crucial since the signal transduction cascades are modulated by phosphorylation/dephosphorylation processes.
Collapse
Affiliation(s)
- Anja Bubik
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 121, SI-1000 Ljubljana, Slovenia; (T.T.L.); (B.S.)
- Faculty of Environmental Protection, Trg mladosti 7, SI-3320 Velenje, Slovenia
| | - Robert Frangež
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, SI-1000 Ljubljana, Slovenia; (R.F.); (M.C.Ž.)
| | - Monika C. Žužek
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, SI-1000 Ljubljana, Slovenia; (R.F.); (M.C.Ž.)
| | - Ion Gutiérrez-Aguirre
- Department of Biotechnology and Systems Biology, National Institute of Biology, Večna pot 121, SI-1000 Ljubljana, Slovenia
| | - Tamara T. Lah
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 121, SI-1000 Ljubljana, Slovenia; (T.T.L.); (B.S.)
| | - Bojan Sedmak
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 121, SI-1000 Ljubljana, Slovenia; (T.T.L.); (B.S.)
- Faculty of Environmental Protection, Trg mladosti 7, SI-3320 Velenje, Slovenia
| |
Collapse
|
35
|
Xue X, Wu X, Fan Y, Han S, Zhang H, Sun Y, Yin Y, Yin M, Chen B, Sun Z, Zhao S, Zhang Q, Liu W, Zhang J, Li J, Shi Y, Xiao Z, Dai J, Zhao Y. Heterogeneous fibroblasts contribute to fibrotic scar formation after spinal cord injury in mice and monkeys. Nat Commun 2024; 15:6321. [PMID: 39060269 PMCID: PMC11282111 DOI: 10.1038/s41467-024-50564-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
Spinal cord injury (SCI) leads to fibrotic scar formation at the lesion site, yet the heterogeneity of fibrotic scar remains elusive. Here we show the heterogeneity in distribution, origin, and function of fibroblasts within fibrotic scars after SCI in mice and female monkeys. Utilizing lineage tracing and single-cell RNA sequencing (scRNA-seq), we found that perivascular fibroblasts (PFs), and meningeal fibroblasts (MFs), rather than pericytes/vascular smooth cells (vSMCs), primarily contribute to fibrotic scar in both transection and crush SCI. Crabp2 + /Emb+ fibroblasts (CE-F) derived from meninges primarily localize in the central region of fibrotic scars, demonstrating enhanced cholesterol synthesis and secretion of type I collagen and fibronectin. In contrast, perivascular/pial Lama1 + /Lama2+ fibroblasts (LA-F) are predominantly found at the periphery of the lesion, expressing laminin and type IV collagen and functionally involved in angiogenesis and lipid transport. These findings may provide a comprehensive understanding for remodeling heterogeneous fibrotic scars after SCI.
Collapse
Affiliation(s)
- Xiaoyu Xue
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xianming Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yongheng Fan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Shuyu Han
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Haipeng Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Yuting Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Yanyun Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Man Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zheng Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Shuaijing Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Qi Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Weiyuan Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Jiaojiao Zhang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jiayin Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ya Shi
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100101, China.
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China.
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
36
|
Kim M, Oh S, Kim S, Kim IS, Kim J, Han J, Ahn JW, Chung S, Jang JH, Shin JE, Park KI. In vivo neural regeneration via AAV-NeuroD1 gene delivery to astrocytes in neonatal hypoxic-ischemic brain injury. Inflamm Regen 2024; 44:33. [PMID: 39014391 PMCID: PMC11253351 DOI: 10.1186/s41232-024-00349-y] [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: 03/26/2024] [Accepted: 07/06/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Neonatal hypoxic-ischemic brain injury (HIBI) is a significant contributor to neonatal mortality and long-term neurodevelopmental disability, characterized by massive neuronal loss and reactive astrogliosis. Current therapeutic approaches for neonatal HIBI have been limited to general supportive therapy because of the lack of methods to compensate for irreversible neuronal loss. This study aimed to establish a feasible regenerative therapy for neonatal HIBI utilizing in vivo direct neuronal reprogramming technology. METHODS Neonatal HIBI was induced in ICR mice at postnatal day 7 by permanent right common carotid artery occlusion and exposure to hypoxia with 8% oxygen and 92% nitrogen for 90 min. Three days after the injury, NeuroD1 was delivered to reactive astrocytes of the injury site using the astrocyte-tropic adeno-associated viral (AAV) vector AAVShH19. AAVShH19 was engineered with the Cre-FLEX system for long-term tracking of infected cells. RESULTS AAVShH19-mediated ectopic NeuroD1 expression effectively converted astrocytes into GABAergic neurons, and the converted cells exhibited electrophysiological properties and synaptic transmitters. Additionally, we found that NeuroD1-mediated in vivo direct neuronal reprogramming protected injured host neurons and altered the host environment, i.e., decreased the numbers of activated microglia, reactive astrocytes, and toxic A1-type astrocytes, and decreased the expression of pro-inflammatory factors. Furthermore, NeuroD1-treated mice exhibited significantly improved motor functions. CONCLUSIONS This study demonstrates that NeuroD1-mediated in vivo direct neuronal reprogramming technology through AAV gene delivery can be a novel regenerative therapy for neonatal HIBI.
Collapse
Affiliation(s)
- Miri Kim
- Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Seokmin Oh
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Songyeon Kim
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Il-Sun Kim
- Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Joowon Kim
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jungho Han
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Ji Woong Ahn
- BnH Research. Co., Ltd. Goyang-Si, Gyeonggi-Do, Republic of Korea
| | - Seungsoo Chung
- Department of Physiology, Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jae-Hyung Jang
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- GluGene Therapeutics Inc., Seoul, Republic of Korea
| | - Jeong Eun Shin
- Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea.
| | - Kook In Park
- Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
- Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| |
Collapse
|
37
|
Wei J, Wang M, Li S, Han R, Xu W, Zhao A, Yu Q, Li H, Li M, Chi G. Reprogramming of astrocytes and glioma cells into neurons for central nervous system repair and glioblastoma therapy. Biomed Pharmacother 2024; 176:116806. [PMID: 38796971 DOI: 10.1016/j.biopha.2024.116806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/18/2024] [Accepted: 05/20/2024] [Indexed: 05/29/2024] Open
Abstract
Central nervous system (CNS) damage is usually irreversible owing to the limited regenerative capability of neurons. Following CNS injury, astrocytes are reactively activated and are the key cells involved in post-injury repair mechanisms. Consequently, research on the reprogramming of reactive astrocytes into neurons could provide new directions for the restoration of neural function after CNS injury and in the promotion of recovery in various neurodegenerative diseases. This review aims to provide an overview of the means through which reactive astrocytes around lesions can be reprogrammed into neurons, to elucidate the intrinsic connection between the two cell types from a neurogenesis perspective, and to summarize what is known about the neurotranscription factors, small-molecule compounds and MicroRNA that play major roles in astrocyte reprogramming. As the malignant proliferation of astrocytes promotes the development of glioblastoma multiforme (GBM), this review also examines the research advances on and the theoretical basis for the reprogramming of GBM cells into neurons and discusses the advantages of such approaches over traditional treatment modalities. This comprehensive review provides new insights into the field of GBM therapy and theoretical insights into the mechanisms of neurological recovery following neurological injury and in GBM treatment.
Collapse
Affiliation(s)
- Junyuan Wei
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Miaomiao Wang
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Shilin Li
- School of Public Health, Jilin University, Changchun 130021, China.
| | - Rui Han
- Department of Neurovascular Surgery, First Hospital of Jilin University, 1xinmin Avenue, Changchun, Jilin Province 130021, China.
| | - Wenhong Xu
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Anqi Zhao
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Qi Yu
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Haokun Li
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Meiying Li
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Guangfan Chi
- The Key Laboratory of Pathobiology, Ministry of Education, and College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
38
|
Sriram S, Carstens K, Dewing W, Fiacco TA. Astrocyte regulation of extracellular space parameters across the sleep-wake cycle. Front Cell Neurosci 2024; 18:1401698. [PMID: 38988660 PMCID: PMC11233815 DOI: 10.3389/fncel.2024.1401698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/07/2024] [Indexed: 07/12/2024] Open
Abstract
Multiple subfields of neuroscience research are beginning to incorporate astrocytes into current frameworks of understanding overall brain physiology, neuronal circuitry, and disease etiology that underlie sleep and sleep-related disorders. Astrocytes have emerged as a dynamic regulator of neuronal activity through control of extracellular space (ECS) volume and composition, both of which can vary dramatically during different levels of sleep and arousal. Astrocytes are also an attractive target of sleep research due to their prominent role in the glymphatic system, a method by which toxic metabolites generated during wakefulness are cleared away. In this review we assess the literature surrounding glial influences on fluctuations in ECS volume and composition across the sleep-wake cycle. We also examine mechanisms of astrocyte volume regulation in glymphatic solute clearance and their role in sleep and wake states. Overall, findings highlight the importance of astrocytes in sleep and sleep research.
Collapse
Affiliation(s)
- Sandhya Sriram
- Interdepartmental Graduate Program in Neuroscience, University of California, Riverside, Riverside, CA, United States
- Department of Biochemistry and Molecular Biology, University of California, Riverside, Riverside, CA, United States
| | - Kaira Carstens
- Department of Biochemistry and Molecular Biology, University of California, Riverside, Riverside, CA, United States
| | - Wayne Dewing
- Undergraduate Major in Neuroscience, University of California, Riverside, Riverside, CA, United States
| | - Todd A Fiacco
- Interdepartmental Graduate Program in Neuroscience, University of California, Riverside, Riverside, CA, United States
- Department of Biochemistry and Molecular Biology, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
39
|
Risby-Jones G, Lee JD, Woodruff TM, Fung JN. Sex differences in Huntington's disease from a neuroinflammation perspective. Front Neurol 2024; 15:1384480. [PMID: 38915800 PMCID: PMC11194371 DOI: 10.3389/fneur.2024.1384480] [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: 02/09/2024] [Accepted: 05/28/2024] [Indexed: 06/26/2024] Open
Abstract
Huntington's disease (HD) is a debilitating neurodegenerative condition characterized by motor, cognitive and psychiatric abnormalities. Immune dysregulation, prominently featuring increased immune activity, plays a significant role in HD pathogenesis. In addition to the central nervous system (CNS), systemic innate immune activation and inflammation are observed in HD patients, exacerbating the effects of the Huntingtin (HTT) gene mutation. Recent attention to sex differences in HD symptom severity underscores the need to consider gender as a biological variable in neurodegenerative disease research. Understanding sex-specific immune responses holds promise for elucidating HD pathophysiology and informing targeted treatment strategies to mitigate cognitive and functional decline. This perspective will highlight the importance of investigating gender influence in HD, particularly focusing on sex-specific immune responses predisposing individuals to disease.
Collapse
Affiliation(s)
- Grace Risby-Jones
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| | - John D. Lee
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| | - Trent M. Woodruff
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Jenny N. Fung
- School of Biomedical Sciences, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
40
|
Lombardi AM, Wong H, Bower ME, Milstead R, Borski C, Schmitt E, Griffioen M, LaPlante L, Ehringer MA, Stitzel J, Hoeffer CA. AKT2 modulates astrocytic nicotine responses in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.31.596856. [PMID: 38854016 PMCID: PMC11160815 DOI: 10.1101/2024.05.31.596856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
A better understanding of nicotine neurobiology is needed to reduce or prevent chronic addiction, ameliorate the detrimental effects of nicotine withdrawal, and increase successful cessation of use. Nicotine binds and activates two astrocyte-expressed nicotinic acetylcholine receptors (nAChRs), α4β2 and α7. We recently found that Protein kinase B-β (Pkb-β or Akt2) expression is restricted to astrocytes in mice and humans. To determine if AKT2 plays a role in astrocytic nicotinic responses, we generated astrocyte-specific Akt2 conditional knockout (cKO) and full Akt2 KO mice for in vivo and in vitro experiments. For in vivo studies, we examined mice exposed to chronic nicotine for two weeks in drinking water (200 μg/mL) and following acute nicotine challenge (0.09, 0.2 mg/kg) after 24 hrs. Our in vitro studies used cultured mouse astrocytes to measure nicotine-dependent astrocytic responses. We validated our approaches using lipopolysaccharide (LPS) exposure inducing astrogliosis. Sholl analysis was used to measure glial fibrillary acidic protein responses in astrocytes. Our data show that wild-type (WT) mice exhibit increased astrocyte morphological complexity during acute nicotine exposure, with decreasing complexity during chronic nicotine use, whereas Akt2 cKO mice showed increased astrocyte morphology complexity. In culture, we found that 100μM nicotine was sufficient for morphological changes and blocking α7 or α4β2 nAChRs prevented observed morphologic changes. Finally, we performed conditioned place preference (CPP) in Akt2 cKO mice and found that astrocytic AKT2 deficiency reduced nicotine preference compared to controls. These findings show the importance of nAChRs and Akt2 signaling in the astrocytic response to nicotine.
Collapse
Affiliation(s)
- Andrew M. Lombardi
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
| | - Helen Wong
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Myra E. Bower
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Ryan Milstead
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Curtis Borski
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Emily Schmitt
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
| | - Mina Griffioen
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Lauren LaPlante
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Marissa A. Ehringer
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Jerry Stitzel
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
| | - Charles A. Hoeffer
- Department of Integrative Physiology, University of Colorado, Boulder, CO 80303
- Institute for Behavioral Genetics, University of Colorado, Boulder, CO 80309
- Linda Crnic Institute, Anschutz Medical Center, Aurora, CO 80045
| |
Collapse
|
41
|
Moreira R, Nóbrega C, de Almeida LP, Mendonça L. Brain-targeted drug delivery - nanovesicles directed to specific brain cells by brain-targeting ligands. J Nanobiotechnology 2024; 22:260. [PMID: 38760847 PMCID: PMC11100082 DOI: 10.1186/s12951-024-02511-7] [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: 02/15/2024] [Accepted: 04/29/2024] [Indexed: 05/19/2024] Open
Abstract
Neurodegenerative diseases are characterized by extensive loss of function or death of brain cells, hampering the life quality of patients. Brain-targeted drug delivery is challenging, with a low success rate this far. Therefore, the application of targeting ligands in drug vehicles, such as lipid-based and polymeric nanoparticles, holds the promise to overcome the blood-brain barrier (BBB) and direct therapies to the brain, in addition to protect their cargo from degradation and metabolization. In this review, we discuss the barriers to brain delivery and the different types of brain-targeting ligands currently in use in brain-targeted nanoparticles, such as peptides, proteins, aptamers, small molecules, and antibodies. Moreover, we present a detailed review of the different targeting ligands used to direct nanoparticles to specific brain cells, like neurons (C4-3 aptamer, neurotensin, Tet-1, RVG, and IKRG peptides), astrocytes (Aquaporin-4, D4, and Bradykinin B2 antibodies), oligodendrocytes (NG-2 antibody and the biotinylated DNA aptamer conjugated to a streptavidin core Myaptavin-3064), microglia (CD11b antibody), neural stem cells (QTRFLLH, VPTQSSG, and NFL-TBS.40-63 peptides), and to endothelial cells of the BBB (transferrin and insulin proteins, and choline). Reports demonstrated enhanced brain-targeted delivery with improved transport to the specific cell type targeted with the conjugation of these ligands to nanoparticles. Hence, this strategy allows the implementation of high-precision medicine, with reduced side effects or unwanted therapy clearance from the body. Nevertheless, the accumulation of some of these nanoparticles in peripheral organs has been reported indicating that there are still factors to be improved to achieve higher levels of brain targeting. This review is a collection of studies exploring targeting ligands for the delivery of nanoparticles to the brain and we highlight the advantages and limitations of this type of approach in precision therapies.
Collapse
Grants
- under BrainHealth2020 projects (CENTRO-01-0145-FEDER-000008), through the COMPETE 2020 - Operational Programme for Competitiveness and Internationalization and Portuguese national funds via FCT - Fundação para a Ciência e a Tecnologia, under projects - UIDB/04539/2020 and UIDP/04539/2020, POCI-01-0145-FEDER-030737 (NeuroStemForMJD, PTDC/BTM-ORG/30737/2017), CEECIND/04242/2017, and PhD Scholarship European Regional Development Fund (ERDF) through the Centro 2020 Regional Operational Programme
- under BrainHealth2020 projects (CENTRO-01-0145-FEDER-000008), through the COMPETE 2020 - Operational Programme for Competitiveness and Internationalization and Portuguese national funds via FCT - Fundação para a Ciência e a Tecnologia, under projects - UIDB/04539/2020 and UIDP/04539/2020, POCI-01-0145-FEDER-030737 (NeuroStemForMJD, PTDC/BTM-ORG/30737/2017), CEECIND/04242/2017, and PhD Scholarship European Regional Development Fund (ERDF) through the Centro 2020 Regional Operational Programme
Collapse
Affiliation(s)
- Ricardo Moreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, polo 1, Coimbra, FMUC, 3004-504, Portugal
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, 3004-504, Portugal
- Faculty of Pharmacy, University of Coimbra, Coimbra, 3000-548, Portugal
| | - Clévio Nóbrega
- Algarve Biomedical Center Research Institute (ABC-RI), University of Algarve, Faro, 8005-139, Portugal
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro, 8005-139, Portugal
| | - Luís Pereira de Almeida
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, polo 1, Coimbra, FMUC, 3004-504, Portugal
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, 3004-504, Portugal
- Faculty of Pharmacy, University of Coimbra, Coimbra, 3000-548, Portugal
- Institute of Interdisciplinary Research, University of Coimbra, Coimbra, 3030-789, Portugal
| | - Liliana Mendonça
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, polo 1, Coimbra, FMUC, 3004-504, Portugal.
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, 3004-504, Portugal.
- Institute of Interdisciplinary Research, University of Coimbra, Coimbra, 3030-789, Portugal.
| |
Collapse
|
42
|
Sintakova K, Romanyuk N. The role of small extracellular vesicles and microRNA as their cargo in the spinal cord injury pathophysiology and therapy. Front Neurosci 2024; 18:1400413. [PMID: 38774785 PMCID: PMC11106386 DOI: 10.3389/fnins.2024.1400413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 04/16/2024] [Indexed: 05/24/2024] Open
Abstract
Spinal cord injury (SCI) is a devastating condition with a complex pathology that affects a significant portion of the population and causes long-term consequences. After primary injury, an inflammatory cascade of secondary injury occurs, followed by neuronal cell death and glial scar formation. Together with the limited regenerative capacity of the central nervous system, these are the main reasons for the poor prognosis after SCI. Despite recent advances, there is still no effective treatment. Promising therapeutic approaches include stem cells transplantation, which has demonstrated neuroprotective and immunomodulatory effects in SCI. This positive effect is thought to be mediated by small extracellular vesicles (sEVs); membrane-bound nanovesicles involved in intercellular communication through transport of functional proteins and RNA molecules. In this review, we summarize the current knowledge about sEVs and microRNA as their cargo as one of the most promising therapeutic approaches for the treatment of SCI. We provide a comprehensive overview of their role in SCI pathophysiology, neuroprotective potential and therapeutic effect.
Collapse
Affiliation(s)
- Kristyna Sintakova
- Department of Neuroregeneration, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
- Department of Neuroscience, 2nd Faculty of Medicine, Charles University, Prague, Czechia
| | - Nataliya Romanyuk
- Department of Neuroregeneration, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
43
|
He Q, Qi Q, Ibeanu GC, Li PA. B355252 Suppresses LPS-Induced Neuroinflammation in the Mouse Brain. Brain Sci 2024; 14:467. [PMID: 38790446 PMCID: PMC11119117 DOI: 10.3390/brainsci14050467] [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: 03/28/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
B355252 is a small molecular compound known for potentiating neural growth factor and protecting against neuronal cell death induced by glutamate in vitro and cerebral ischemia in vivo. However, its other biological functions remain unclear. This study aims to investigate whether B355252 suppresses neuroinflammatory responses and cell death in the brain. C57BL/6j mice were intraperitoneally injected with a single dosage of lipopolysaccharide (LPS, 1 mg/kg) to induce inflammation. B355252 (1 mg/kg) intervention was started two days prior to the LPS injection. The animal behavioral changes were assessed pre- and post-LPS injections. The animal brains were harvested at 4 and 24 h post-LPS injection, and histological, biochemical, and cytokine array outcomes were examined. Results showed that B355252 improved LPS-induced behavioral deterioration, mitigated brain tissue damage, and suppressed the activation of microglial and astrocytes. Furthermore, B355252 reduced the protein levels of key pyroptotic markers TLR4, NLRP3, and caspase-1 and inhibited the LPS-induced increases in IL-1β, IL-18, and cytokines. In conclusion, B355252 demonstrates a potent anti-neuroinflammatory effect in vivo, suggesting that its potential therapeutic value warrants further investigation.
Collapse
Affiliation(s)
- Qingping He
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise (BRITE), College of Health and Sciences, North Carolina Central University, Durham, NC 27707, USA; (Q.H.); (G.C.I.)
| | - Qi Qi
- Human Vaccine Institute, Department of Surgery, Duke University Medical Center, Durham, NC 27707, USA;
| | - Gordon C. Ibeanu
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise (BRITE), College of Health and Sciences, North Carolina Central University, Durham, NC 27707, USA; (Q.H.); (G.C.I.)
| | - P. Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise (BRITE), College of Health and Sciences, North Carolina Central University, Durham, NC 27707, USA; (Q.H.); (G.C.I.)
| |
Collapse
|
44
|
Kim D, Ko HY, Chung JI, Park YM, Lee S, Kim SY, Kim J, Chun JH, Han KS, Lee M, Ju YH, Park SJ, Park KD, Nam MH, Kim SH, Shim JK, Park Y, Lim H, Park J, Lee GH, Kim H, Kim S, Park U, Ryu H, Lee SY, Park S, Kang SG, Chang JH, Lee CJ, Yun M. Visualizing cancer-originating acetate uptake through monocarboxylate transporter 1 in reactive astrocytes in the glioblastoma tumor microenvironment. Neuro Oncol 2024; 26:843-857. [PMID: 38085571 PMCID: PMC11066945 DOI: 10.1093/neuonc/noad243] [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] [Indexed: 05/04/2024] Open
Abstract
BACKGROUND Reactive astrogliosis is a hallmark of various brain pathologies, including neurodegenerative diseases and glioblastomas. However, the specific intermediate metabolites contributing to reactive astrogliosis remain unknown. This study investigated how glioblastomas induce reactive astrogliosis in the neighboring microenvironment and explore 11C-acetate PET as an imaging technique for detecting reactive astrogliosis. METHODS Through in vitro, mouse models, and human tissue experiments, we examined the association between elevated 11C-acetate uptake and reactive astrogliosis in gliomas. We explored acetate from glioblastoma cells, which triggers reactive astrogliosis in neighboring astrocytes by upregulating MAO-B and monocarboxylate transporter 1 (MCT1) expression. We evaluated the presence of cancer stem cells in the reactive astrogliosis region of glioblastomas and assessed the correlation between the volume of 11C-acetate uptake beyond MRI and prognosis. RESULTS Elevated 11C-acetate uptake is associated with reactive astrogliosis and astrocytic MCT1 in the periphery of glioblastomas in human tissues and mouse models. Glioblastoma cells exhibit increased acetate production as a result of glucose metabolism, with subsequent secretion of acetate. Acetate derived from glioblastoma cells induces reactive astrogliosis in neighboring astrocytes by increasing the expression of MAO-B and MCT1. We found cancer stem cells within the reactive astrogliosis at the tumor periphery. Consequently, a larger volume of 11C-acetate uptake beyond contrast-enhanced MRI was associated with a worse prognosis. CONCLUSIONS Our results highlight the role of acetate derived from glioblastoma cells in inducing reactive astrogliosis and underscore the potential value of 11C-acetate PET as an imaging technique for detecting reactive astrogliosis, offering important implications for the diagnosis and treatment of glioblastomas.
Collapse
Affiliation(s)
- Dongwoo Kim
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hae Young Ko
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jee-In Chung
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yongmin Mason Park
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- IBS School, University of Science and Technology, Daejeon, Republic of Korea
| | - Sangwon Lee
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seon Yoo Kim
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jisu Kim
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Joong-Hyun Chun
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyung-Seok Han
- Department of Biological Sciences, Chungnam National University, Daejeon, Republic of Korea
| | - Misu Lee
- Division of Life Science, College of Life Science and Bioengineering, Incheon National University, Incheon, Republic of Korea
| | - Yeon Ha Ju
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, Republic of Korea
- IBS School, University of Science and Technology, Daejeon, Republic of Korea
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Sun Jun Park
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Med Science & Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
| | - Ki Duk Park
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Med Science & Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Med Science & Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
- Department of KHU-KIST Convergence Science and Technology, Kyung Hee University, Seoul, Republic of Korea
| | - Se Hoon Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin-Kyoung Shim
- Department of Neurosurgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Youngjoo Park
- Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyunkeong Lim
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jaekyung Park
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Gwan-Ho Lee
- Research Resources Division, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Hyunjin Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Suhyun Kim
- K-Laboratory, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Uiyeol Park
- K-Laboratory, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Hoon Ryu
- K-Laboratory, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - So Yun Lee
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Sunghyouk Park
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Seok-Gu Kang
- Department of Neurosurgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - C Justin Lee
- IBS School, University of Science and Technology, Daejeon, Republic of Korea
| | - Mijin Yun
- Department of Nuclear Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
45
|
Wang J, Yuan Y, Zhang S, Lu S, Han G, Bian M, huang L, Meng D, Su D, Xiao L, Xiao Y, Zhang J, Gong N, Jiang L. Remodeling of the Intra-Conduit Inflammatory Microenvironment to Improve Peripheral Nerve Regeneration with a Neuromechanical Matching Protein-Based Conduit. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302988. [PMID: 38430538 PMCID: PMC11077661 DOI: 10.1002/advs.202302988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 12/22/2023] [Indexed: 03/04/2024]
Abstract
Peripheral nerve injury (PNI) remains a challenging area in regenerative medicine. Nerve guide conduit (NGC) transplantation is a common treatment for PNI, but the prognosis of NGC treatment is unsatisfactory due to 1) neuromechanical unmatching and 2) the intra-conduit inflammatory microenvironment (IME) resulting from Schwann cell pyroptosis and inflammatory-polarized macrophages. A neuromechanically matched NGC composed of regenerated silk fibroin (RSF) loaded with poly(3,4-ethylenedioxythiophene): poly(styrene sulfonate) (P:P) and dimethyl fumarate (DMF) are designed, which exhibits a matched elastic modulus (25.1 ± 3.5 MPa) for the peripheral nerve and the highest 80% elongation at break, better than most protein-based conduits. Moreover, the NGC can gradually regulate the intra-conduit IME by releasing DMF and monitoring sciatic nerve movements via piezoresistive sensing. The combination of NGC and electrical stimulation modulates the IME to support PNI regeneration by synergistically inhibiting Schwann cell pyroptosis and reducing inflammatory factor release, shifting macrophage polarization from the inflammatory M1 phenotype to the tissue regenerative M2 phenotype and resulting in functional recovery of neurons. In a rat sciatic nerve crush model, NGC promoted remyelination and functional and structural regeneration. Generally, the DMF/RSF/P:P conduit provides a new potential therapeutic approach to promote nerve repair in future clinical treatments.
Collapse
Affiliation(s)
- Jia‐Yi Wang
- Department of Orthopaedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Ya Yuan
- Department of Orthopaedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
- Department of RehabilitationZhongshan HospitalFudan UniversityShanghai200032China
| | - Shu‐Yan Zhang
- The Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Centre for Biomedical Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistrySchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Shun‐Yi Lu
- Department of Orthopaedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Guan‐Jie Han
- Department of Orthopaedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Meng‐Xuan Bian
- Department of Orthopaedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Lei huang
- Department of Orthopaedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - De‐Hua Meng
- Department of Orthopaedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Di‐Han Su
- Department of Orthopaedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Lan Xiao
- School of MechanicalMedical and Process EngineeringCentre for Biomedical TechnologiesQueensland University of TechnologyBrisbane4059Australia
- Australia‐China Centre for Tissue Engineering and Regenerative MedicineQueensland University of TechnologyBrisbane4059Australia
| | - Yin Xiao
- School of MechanicalMedical and Process EngineeringCentre for Biomedical TechnologiesQueensland University of TechnologyBrisbane4059Australia
- Australia‐China Centre for Tissue Engineering and Regenerative MedicineQueensland University of TechnologyBrisbane4059Australia
- School of Medicine and Dentistry & Menzies Health Institute QueenslandGriffith UniversityGold Coast4222Australia
| | - Jian Zhang
- Department of Orthopaedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Ning‐Ji Gong
- Department of EmergencyDepartment of OrthopedicsThe Second HospitalCheeloo College of MedicineShandong UniversityJinanShandong250033China
| | - Li‐Bo Jiang
- Department of Orthopaedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| |
Collapse
|
46
|
Nguyen HS, Kang SJ, Kim S, Cha BH, Park KS, Jeong SW. Changes in the expression of satellite glial cell-specific markers during postnatal development of rat sympathetic ganglia. Brain Res 2024; 1829:148809. [PMID: 38354998 DOI: 10.1016/j.brainres.2024.148809] [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: 11/16/2023] [Revised: 01/31/2024] [Accepted: 02/08/2024] [Indexed: 02/16/2024]
Abstract
The sympathetic ganglia represent a final motor pathway that mediates homeostatic "fight and flight" responses in the visceral organs. Satellite glial cells (SGCs) form a thin envelope close to the neuronal cell body and synapses in the sympathetic ganglia. This unique morphological feature suggests that neurons and SGCs form functional units for regulation of sympathetic output. In the present study, we addressed whether SGC-specific markers undergo age-dependent changes in the postnatal development of rat sympathetic ganglia. We found that fatty acid-binding protein 7 (FABP7) is an early SGC marker, whereas the S100B calcium-binding protein, inwardly rectifying potassium channel, Kir4.1 and small conductance calcium-activated potassium channel, SK3 are late SGC markers in the postnatal development of sympathetic ganglia. Unlike in sensory ganglia, FABP7 + SGC was barely detectable in adult sympathetic ganglia. The expression of connexin 43, a gap junction channel gradually increased with age, although it was detected in both SGCs and neurons in sympathetic ganglia. Glutamine synthetase was expressed in sensory, but not sympathetic SGCs. Unexpectedly, the sympathetic SGCs expressed a water-selective channel, aquaporin 1 instead of aquaporin 4, a pan-glial marker. However, aquaporin 1 was not detected in the SGCs encircling large neurons. Nerve injury and inflammation induced the upregulation of glial fibrillary acidic protein, suggesting that this protein is a hall marker of glial activation in the sympathetic ganglia. In conclusion, our findings provide basic information on the in vivo profiles of specific markers for identifying sympathetic SGCs at different stages of postnatal development in both healthy and diseased states.
Collapse
Affiliation(s)
- Huu Son Nguyen
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea; Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Seong Jun Kang
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Sohyun Kim
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Byung Ho Cha
- Department of Pediatrics, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Kyu-Sang Park
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea; Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea
| | - Seong-Woo Jeong
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, Republic of Korea.
| |
Collapse
|
47
|
Wang S, Xie S, Zheng Q, Zhang Z, Wang T, Zhang G. Biofluid biomarkers for Alzheimer's disease. Front Aging Neurosci 2024; 16:1380237. [PMID: 38659704 PMCID: PMC11039951 DOI: 10.3389/fnagi.2024.1380237] [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: 02/01/2024] [Accepted: 03/27/2024] [Indexed: 04/26/2024] Open
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disease, with a complex pathogenesis and an irreversible course. Therefore, the early diagnosis of AD is particularly important for the intervention, prevention, and treatment of the disease. Based on the different pathophysiological mechanisms of AD, the research progress of biofluid biomarkers are classified and reviewed. In the end, the challenges and perspectives of future research are proposed.
Collapse
Affiliation(s)
- Sensen Wang
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China
| | - Sitan Xie
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
| | - Qinpin Zheng
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China
| | - Zhihui Zhang
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
| | - Tian Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China
| | - Guirong Zhang
- Shandong Yinfeng Academy of Life Science, Jinan, Shandong, China
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, Shandong, China
| |
Collapse
|
48
|
Gentenaar M, Meulmeester FL, van der Burg XR, Hoekstra AT, Hunt H, Kroon J, van Roon-Mom WMC, Meijer OC. Glucocorticoid receptor antagonist CORT113176 attenuates motor and neuropathological symptoms of Huntington's disease in R6/2 mice. Exp Neurol 2024; 374:114675. [PMID: 38216109 DOI: 10.1016/j.expneurol.2024.114675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 11/17/2023] [Accepted: 01/02/2024] [Indexed: 01/14/2024]
Abstract
Huntington's Disease (HD) is a progressive neurodegenerative disease caused by a mutation in the huntingtin gene. The mutation leads to a toxic gain of function of the mutant huntingtin (mHtt) protein resulting in cellular malfunction, aberrant huntingtin aggregation and eventually neuronal cell death. Patients with HD show impaired motor functions and cognitive decline. Elevated levels of glucocorticoids have been found in HD patients and in HD mouse models, and there is a positive correlation between increased glucocorticoid levels and the progression of HD. Therefore, antagonism of the glucocorticoid receptor (GR) may be an interesting strategy for the treatment of HD. In this study, we evaluated the efficacy of the selective GR antagonist CORT113176 in the commonly used R6/2 mouse model. In male mice, CORT113176 treatment significantly delayed the loss of grip strength, the development of hindlimb clasping, gait abnormalities, and the occurrence of epileptic seizures. CORT113176 treatment delayed loss of DARPP-32 immunoreactivity in the dorsolateral striatum. It also restored HD-related parameters including astrocyte markers in both the dorsolateral striatum and the hippocampus, and microglia markers in the hippocampus. This suggests that CORT113176 has both cell-type and brain region-specific effects. CORT113176 delayed the formation of mHtt aggregates in the striatum and the hippocampus. In female mice, we did not observe major effects of CORT113176 treatment on HD-related symptoms, with the exception of the anti-epileptic effects. We conclude that CORT113176 effectively delays several key symptoms related to the HD phenotype in male R6/2 mice and believe that GR antagonism may be a possible treatment option.
Collapse
Affiliation(s)
- Max Gentenaar
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands.
| | - Fleur L Meulmeester
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ximaine R van der Burg
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
| | - Anna T Hoekstra
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands; Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Hazel Hunt
- Corcept Therapeutics, Menlo Park, CA, USA
| | - Jan Kroon
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands; Corcept Therapeutics, Menlo Park, CA, USA
| | | | - Onno C Meijer
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
49
|
Hamani C, Davidson B, Lipsman N, Abrahao A, Nestor SM, Rabin JS, Giacobbe P, Pagano RL, Campos ACP. Insertional effect following electrode implantation: an underreported but important phenomenon. Brain Commun 2024; 6:fcae093. [PMID: 38707711 PMCID: PMC11069120 DOI: 10.1093/braincomms/fcae093] [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: 09/11/2023] [Revised: 12/08/2023] [Accepted: 03/26/2024] [Indexed: 05/07/2024] Open
Abstract
Deep brain stimulation has revolutionized the treatment of movement disorders and is gaining momentum in the treatment of several other neuropsychiatric disorders. In almost all applications of this therapy, the insertion of electrodes into the target has been shown to induce some degree of clinical improvement prior to stimulation onset. Disregarding this phenomenon, commonly referred to as 'insertional effect', can lead to biased results in clinical trials, as patients receiving sham stimulation may still experience some degree of symptom amelioration. Similar to the clinical scenario, an improvement in behavioural performance following electrode implantation has also been reported in preclinical models. From a neurohistopathologic perspective, the insertion of electrodes into the brain causes an initial trauma and inflammatory response, the activation of astrocytes, a focal release of gliotransmitters, the hyperexcitability of neurons in the vicinity of the implants, as well as neuroplastic and circuitry changes at a distance from the target. Taken together, it would appear that electrode insertion is not an inert process, but rather triggers a cascade of biological processes, and, as such, should be considered alongside the active delivery of stimulation as an active part of the deep brain stimulation therapy.
Collapse
Affiliation(s)
- Clement Hamani
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Benjamin Davidson
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Nir Lipsman
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Division of Neurosurgery, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Agessandro Abrahao
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Sean M Nestor
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Psychiatry, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Jennifer S Rabin
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
- Rehabilitation Sciences Institute, University of Toronto, Toronto M5G 1V7, Canada
| | - Peter Giacobbe
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Psychiatry, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Rosana L Pagano
- Laboratory of Neuroscience, Hospital Sírio-Libanês, São Paulo, SP CEP 01308-060, Brazil
| | - Ana Carolina P Campos
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Laboratory of Neuroscience, Hospital Sírio-Libanês, São Paulo, SP CEP 01308-060, Brazil
| |
Collapse
|
50
|
Koukalova L, Chmelova M, Amlerova Z, Vargova L. Out of the core: the impact of focal ischemia in regions beyond the penumbra. Front Cell Neurosci 2024; 18:1336886. [PMID: 38504666 PMCID: PMC10948541 DOI: 10.3389/fncel.2024.1336886] [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: 11/11/2023] [Accepted: 02/08/2024] [Indexed: 03/21/2024] Open
Abstract
The changes in the necrotic core and the penumbra following induction of focal ischemia have been the focus of attention for some time. However, evidence shows, that ischemic injury is not confined to the primarily affected structures and may influence the remote areas as well. Yet many studies fail to probe into the structures beyond the penumbra, and possibly do not even find any significant results due to their short-term design, as secondary damage occurs later. This slower reaction can be perceived as a therapeutic opportunity, in contrast to the ischemic core defined as irreversibly damaged tissue, where the window for salvation is comparatively short. The pathologies in remote structures occur relatively frequently and are clearly linked to the post-stroke neurological outcome. In order to develop efficient therapies, a deeper understanding of what exactly happens in the exo-focal regions is necessary. The mechanisms of glia contribution to the ischemic damage in core/penumbra are relatively well described and include impaired ion homeostasis, excessive cell swelling, glutamate excitotoxic mechanism, release of pro-inflammatory cytokines and phagocytosis or damage propagation via astrocytic syncytia. However, little is known about glia involvement in post-ischemic processes in remote areas. In this literature review, we discuss the definitions of the terms "ischemic core", "penumbra" and "remote areas." Furthermore, we present evidence showing the array of structural and functional changes in the more remote regions from the primary site of focal ischemia, with a special focus on glia and the extracellular matrix. The collected information is compared with the processes commonly occurring in the ischemic core or in the penumbra. Moreover, the possible causes of this phenomenon and the approaches for investigation are described, and finally, we evaluate the efficacy of therapies, which have been studied for their anti-ischemic effect in remote areas in recent years.
Collapse
Affiliation(s)
- Ludmila Koukalova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Martina Chmelova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Zuzana Amlerova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Lydia Vargova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|