1
|
Castillo-González J, González-Rey E. Beyond wrecking a wall: revisiting the concept of blood-brain barrier breakdown in ischemic stroke. Neural Regen Res 2025; 20:1944-1956. [PMID: 39254550 PMCID: PMC11691464 DOI: 10.4103/nrr.nrr-d-24-00392] [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: 04/10/2024] [Revised: 06/17/2024] [Accepted: 07/04/2024] [Indexed: 09/11/2024] Open
Abstract
The blood-brain barrier constitutes a dynamic and interactive boundary separating the central nervous system and the peripheral circulation. It tightly modulates the ion transport and nutrient influx, while restricting the entry of harmful factors, and selectively limiting the migration of immune cells, thereby maintaining brain homeostasis. Despite the well-established association between blood-brain barrier disruption and most neurodegenerative/neuroinflammatory diseases, much remains unknown about the factors influencing its physiology and the mechanisms underlying its breakdown. Moreover, the role of blood-brain barrier breakdown in the translational failure underlying therapies for brain disorders is just starting to be understood. This review aims to revisit this concept of "blood-brain barrier breakdown," delving into the most controversial aspects, prevalent challenges, and knowledge gaps concerning the lack of blood-brain barrier integrity. By moving beyond the oversimplistic dichotomy of an "open"/"bad" or a "closed"/"good" barrier, our objective is to provide a more comprehensive insight into blood-brain barrier dynamics, to identify novel targets and/or therapeutic approaches aimed at mitigating blood-brain barrier dysfunction. Furthermore, in this review, we advocate for considering the diverse time- and location-dependent alterations in the blood-brain barrier, which go beyond tight-junction disruption or brain endothelial cell breakdown, illustrated through the dynamics of ischemic stroke as a case study. Through this exploration, we seek to underscore the complexity of blood-brain barrier dysfunction and its implications for the pathogenesis and therapy of brain diseases.
Collapse
Affiliation(s)
- Julia Castillo-González
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, Granada, Spain
| | - Elena González-Rey
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, Granada, Spain
| |
Collapse
|
2
|
Qian M, Wang Z, Liu H, Zhang X, Xu J, Zhang Y, Chen L, Zhou Z, Yu Y, Dong W. Reactive astrocytes in spinal cord injury: An analysis of heterogeneity based on temporality and spatiality, potential therapies, and limitations. J Neuropathol Exp Neurol 2025:nlaf042. [PMID: 40314931 DOI: 10.1093/jnen/nlaf042] [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] [Indexed: 05/03/2025] Open
Abstract
Spinal cord injury (SCI) constitutes a profound central nervous system disorder characterized by significant neurological dysfunction and sensory loss below the injury site. SCI elicits a multifaceted cellular response in which the proliferation of reactive astrocytes and the ensuing diversity in their functions and phenotypes play pivotal roles within the injury microenvironment, especially during the secondary phases of the condition. This review explores the activation and heterogeneity of astrocytes following SCI. It underscores the necessity of delineating the heterogeneity among reactive astrocyte subpopulations throughout the secondary injury phase of SCI. Developing therapeutic strategies that capitalize on the beneficial properties of certain reactive astrocyte subpopulations while mitigating the adverse effects of others could have profound implications for future clinical management of SCI.
Collapse
Affiliation(s)
- Mengting Qian
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Zheng Wang
- Department of Oncology, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Hang Liu
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Xinyu Zhang
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Jingyi Xu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Yiwen Zhang
- Department of Neurosurgery, The Affiliated Hospital of Panzhihua University, Panzhihua, Sichuan, People's Republic of China
| | - Ligang Chen
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Zhengjun Zhou
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Yang Yu
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
- Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China
| | - Wei Dong
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| |
Collapse
|
3
|
Lu M, Li J, Huang Q, Mao D, Yang G, Lan Y, Zeng J, Pan M, Shi S, Zou D. Single-Nucleus Landscape of Glial Cells and Neurons in Alzheimer's Disease. Mol Neurobiol 2025; 62:2695-2709. [PMID: 39153159 DOI: 10.1007/s12035-024-04428-6] [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: 02/21/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with a projected significant increase in incidence. Therefore, this study analyzed single-nucleus AD data to provide a theoretical basis for the clinical development and treatment of AD. We downloaded AD-related monocyte data from the Gene Expression Omnibus database, annotated cells, compared cell abundance between groups, and investigated glial and neuronal cell biological processes and pathways through functional enrichment analysis. Furthermore, we constructed a global regulatory network for AD based on cell communication and ecological analyses. Our findings revealed increased abundance of Capping Protein Regulator And Myosin 1 linker 1 (CARMIL1)+ astrocytes (AST), Immunoglobulin Superfamily Member 21 (IGSF21)+ microglia (MIC), SRY-Box Transcription Factor 6 (SOX6)+ inhibitory neurons (InNeu), and laminin alpha-2 chain (LAMA2)+ oligodendrocytes (OLI) cell subgroups in tissues of patients with AD, while prostaglandin D2 synthase (PTGDS)+ AST, Src Family Tyrosine Kinase (FYN)+ MIC, and Proteolipid Protein 1 (PLP1)+ InNeu subgroups specifically decreased. We found that the cell phenotype of patients with AD shifted from a simpler to a more complex state compared to the control group. Cell communication analysis revealed strong communication between MIC and NEU. Furthermore, AST, MIC, NEU, and OLI were involved in oxidative stress- and inflammation-related pathways, potentially contributing to disease development. This study provides a theoretical basis for further exploring the specific mechanisms underlying AD.
Collapse
Affiliation(s)
- Mengru Lu
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, No 166 Daxuedong Road, Nanning, Guangxi, 530007, China
| | - Jiaxin Li
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Qi Huang
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, No 166 Daxuedong Road, Nanning, Guangxi, 530007, China
| | - Daniel Mao
- Department of Biology, Pennsylvania State University, University Park, PA, 16802, USA
| | - Grace Yang
- State College Area High School, State College, PA, 16801, USA
| | - Yating Lan
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, No 166 Daxuedong Road, Nanning, Guangxi, 530007, China
| | - Jingyi Zeng
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, No 166 Daxuedong Road, Nanning, Guangxi, 530007, China
| | - Mika Pan
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, No 166 Daxuedong Road, Nanning, Guangxi, 530007, China
| | - Shengliang Shi
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, No 166 Daxuedong Road, Nanning, Guangxi, 530007, China.
| | - Donghua Zou
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, No 166 Daxuedong Road, Nanning, Guangxi, 530007, China.
| |
Collapse
|
4
|
Liu Y, Peng H, Liu Q, Hao J, Tang C, Yan H. Differential Expression of GABA Receptor-Related Genes in Alzheimer's Disease and the Positive Regulatory Role of Aerobic Exercise-From Genetic Screening to D-gal-induced AD-like Pathology Model. Neuromolecular Med 2024; 27:1. [PMID: 39752101 DOI: 10.1007/s12017-024-08821-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 11/25/2024] [Indexed: 01/04/2025]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder. The neuropathology of AD appears in the hippocampus. The purpose of this work was to reveal key differentially expressed genes (DEGs) in the hippocampus of AD patients and healthy individuals. Furthermore, we established an in vivo AD-like model to validate and explore the effects of exercise on these risky genes. The datasets GSE36980 and GSE48350 were downloaded from the GEO database and visualized using R packages to obtain DEGs. Subsequently, the potential biological functions of these DEGs were predicted, PPI network interactions were screened for core genes, and Pearson correlation analysis was performed. Additionally, we determined the diagnostic value of core DEGs using ROC curves. Single-cell analysis was used to verify the cell type specificity of hub genes. Finally, we used RT-qPCR, immunohistochemistry, and immunofluorescence to validate the expression of core DEGs in model mice and to explore the beneficial mechanisms of exercise. A total of 13 differentially expressed genes (DEGs) associated with the development of AD were identified, comprising 11 down-regulated genes and 2 up-regulated genes. PPI network visualization acquired four down-regulated core DEGs with good diagnostic value. The findings from the in vivo study indicated that the mRNA expression of GABRA1, GABRG2, and SVOP decreased, and the astrocyte marker GFAP notably increased in AD mice. Surprisingly, exercise increased hippocampal GABRA1 and GABRG2 expression and decreased GFAP-positive intensity of GABRG1 localization, reducing expression of inflammatory markers TNF-α and IL-1β. In addition, exercise improved the spatial exploration ability but had little effect on the preference index in AD mice. Our data highlighted the mechanism by which exercise improves memory performance in AD patients by reducing astrocyte neurotoxicity inducing decreased hippocampal GABA receptor expression.
Collapse
Affiliation(s)
- Yang Liu
- College of Physical Education, Nanchang Institute of Technology, Nanchang, 330044, China.
- Department of Neurology, People's Hospital of Henan University, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China.
| | - Haoran Peng
- Department of Neurology, People's Hospital of Henan University, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
| | - Qi Liu
- College of Physical Education, Nanchang Institute of Technology, Nanchang, 330044, China
| | - Jianying Hao
- College of Physical Education, Nanchang Institute of Technology, Nanchang, 330044, China
| | - Chao Tang
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, 410012, China
| | - Hanhui Yan
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha, 410012, China
- School of Sports Science Division of Sport Physiology, Beijing Sport University, Beijing, 100084, China
| |
Collapse
|
5
|
Chen H, Deng C, Meng Z, Zhu M, Yang R, Yuan J, Meng S. Combined Catalpol and Tetramethylpyrazine Promote Axonal Plasticity in Alzheimer's Disease by Inducing Astrocytes to Secrete Exosomes Carrying CDK5 mRNA and Regulating STAT3 Phosphorylation. Mol Neurobiol 2024; 61:10770-10791. [PMID: 38789892 DOI: 10.1007/s12035-024-04251-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
Alzheimer's disease (AD) is a common progressive degenerative disease of the central nervous system in aging populations. This study aimed to investigate the effects of combined catalpol and tetramethylpyrazine (CT) in promoting axonal plasticity in AD and the potential underlying mechanism. Astrocytes were treated with different concentrations of compatible CT. Exosomes were collected and subjected to sequencing analysis, which was followed by the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of differentially expressed genes. Amyloid precursor protein/presenilin 1 (APP/PS1) double-transfected male mice were used as the in vivo AD models. Astrocyte-derived exosomes that were transfected with cyclin-dependent kinase 5 (CDK5) or CT treatment were injected into the tail vein of mice. The levels of CDK5, synaptic plasticity marker protein neurofilament 200 (NF200), and growth-associated protein 43 (GAP-43) in the hippocampus of mice were compared in each group. Immunofluorescence staining was used to detect the localization of STAT3 and to visualize synaptic morphology via β-tubulin-III (TUBB3). Astrocyte-derived exosomes transfected with siCDK5 or treated with CT were co-cultured with HT-22 cells, which were untransfected or silenced for signal transducer and activator of transcription 3 (STAT3). Amyloid β-protein (Aβ)1-42 was induced in the in vitro AD models. The viability, apoptosis, and expression levels of NF200 and GAP-43 proteins in the hippocampal neurons of each group were compared. In total, 166 differentially expressed genes in CT-induced astrocyte-derived exosomes were included in the KEGG analysis, and they were found to be enriched in 12 pathways, mainly in axon guidance. CT treatment significantly increased the level of CDK5 mRNA in astrocyte-derived exosomes-these exosomes restored CDK5 mRNA and protein levels in the hippocampus of the in vivo AD model mice and the in vitro AD model; promoted p-STAT3 (Ser727), NF200 and GAP-43 proteins; and promoted the regeneration and extension of neuronal synapses. Silencing of CDK5 blocked both neuronal protection as well as induction of axonal plasticity in AD by CT-treated exosomes in vitro and in vivo. Moreover, silencing of STAT3 blocked both neuronal protection as well as induction of axonal plasticity in AD caused by CDK5 overexpression or CT-treated astrocyte-induced exosomes. CT promotes axonal plasticity in AD by inducing astrocytes to secrete exosomes carrying CDK5 mRNA and regulating STAT3 (Ser727) phosphorylation.
Collapse
Affiliation(s)
- Huize Chen
- Department of Traditional Chinese Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine,Xuhui District, 600 Yishan Road, Shanghai, 200233, China
| | - Chujun Deng
- Department of Traditional Chinese Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine,Xuhui District, 600 Yishan Road, Shanghai, 200233, China
| | - Zeyu Meng
- Second Clinical Medicine College of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Mengting Zhu
- Graduate School of Jiangxi, University of Traditional Chinese Medicine, Nanchang, China
| | - Ruoyu Yang
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Yuan
- Department of Traditional Chinese Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine,Xuhui District, 600 Yishan Road, Shanghai, 200233, China
| | - Shengxi Meng
- Department of Traditional Chinese Medicine, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine,Xuhui District, 600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
6
|
Yan M, Sun Z, Zhang S, Yang G, Jiang X, Wang G, Li R, Wang Q, Tian X. SOCS modulates JAK-STAT pathway as a novel target to mediate the occurrence of neuroinflammation: Molecular details and treatment options. Brain Res Bull 2024; 213:110988. [PMID: 38805766 DOI: 10.1016/j.brainresbull.2024.110988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/28/2024] [Accepted: 05/26/2024] [Indexed: 05/30/2024]
Abstract
SOCS (Suppressor of Cytokine Signalling) proteins are intracellular negative regulators that primarily modulate and inhibit cytokine-mediated signal transduction, playing a crucial role in immune homeostasis and related inflammatory diseases. SOCS act as inhibitors by regulating the Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling pathway, thereby intervening in the pathogenesis of inflammation and autoimmune diseases. Recent studies have also demonstrated their involvement in central immunity and neuroinflammation, showing a dual functionality. However, the specific mechanisms of SOCS in the central nervous system remain unclear. This review thoroughly elucidates the specific mechanisms linking the SOCS-JAK-STAT pathway with the inflammatory manifestations of neurodegenerative diseases. Based on this, it proposes the theory that SOCS proteins can regulate the JAK-STAT pathway and inhibit the occurrence of neuroinflammation. Additionally, this review explores in detail the current therapeutic landscape and potential of targeting SOCS in the brain via the JAK-STAT pathway for neuroinflammation, offering insights into potential targets for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Min Yan
- College of Graduate Education, Shandong Sport University, Jinan 255300, China
| | - Zhiyuan Sun
- College of Graduate Education, Shandong Sport University, Jinan 255300, China
| | - Sen Zhang
- College of Graduate Education, Shandong Sport University, Jinan 255300, China
| | - Guangxin Yang
- College of Graduate Education, Shandong Sport University, Jinan 255300, China
| | - Xing Jiang
- College of Graduate Education, Shandong Sport University, Jinan 255300, China
| | - Guilong Wang
- College of Graduate Education, Shandong Sport University, Jinan 255300, China
| | - Ran Li
- College of Graduate Education, Shandong Sport University, Jinan 255300, China.
| | - Qinglu Wang
- College of Graduate Education, Shandong Sport University, Jinan 255300, China.
| | - Xuewen Tian
- College of Graduate Education, Shandong Sport University, Jinan 255300, China.
| |
Collapse
|
7
|
Mateos-Martínez P, Coronel R, Sachse M, González-Sastre R, Maeso L, Rodriguez MJ, Terrón MC, López-Alonso V, Liste I. Human cerebral organoids: cellular composition and subcellular morphological features. Front Cell Neurosci 2024; 18:1406839. [PMID: 38933177 PMCID: PMC11199856 DOI: 10.3389/fncel.2024.1406839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Introduction Human cerebral organoids (hCOs) derived from pluripotent stem cells are very promising for the study of neurodevelopment and the investigation of the healthy or diseased brain. To help establish hCOs as a powerful research model, it is essential to perform the morphological characterization of their cellular components in depth. Methods In this study, we analyzed the cell types consisting of hCOs after culturing for 45 days using immunofluorescence and reverse transcriptase qualitative polymerase chain reaction (RT-qPCR) assays. We also analyzed their subcellular morphological characteristics by transmission electron microscopy (TEM). Results Our results show the development of proliferative zones to be remarkably similar to those found in human brain development with cells having a polarized structure surrounding a central cavity with tight junctions and cilia. In addition, we describe the presence of immature and mature migrating neurons, astrocytes, oligodendrocyte precursor cells, and microglia-like cells. Discussion The ultrastructural characterization presented in this study provides valuable information on the structural development and morphology of the hCO, and this information is of general interest for future research on the mechanisms that alter the cell structure or function of hCOs.
Collapse
Affiliation(s)
- Patricia Mateos-Martínez
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Unidad de Biología Computacional, Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Escuela Internacional de Doctorado de la Universidad Nacional de Educación a Distancia (UNED), Programa en Ciencias Biomédicas y Salud Pública, Madrid, Spain
| | - Raquel Coronel
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Martin Sachse
- Unidad de Microscopía Electrónica, Unidades Centrales Científico Técnicas, Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa González-Sastre
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Unidad de Biología Computacional, Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Escuela Internacional de Doctorado de la Universidad Nacional de Educación a Distancia (UNED), Programa en Ciencias Biomédicas y Salud Pública, Madrid, Spain
| | - Laura Maeso
- Unidad de Biología Computacional, Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Maria Josefa Rodriguez
- Unidad de Microscopía Electrónica, Unidades Centrales Científico Técnicas, Instituto de Salud Carlos III, Madrid, Spain
| | - María C. Terrón
- Unidad de Microscopía Electrónica, Unidades Centrales Científico Técnicas, Instituto de Salud Carlos III, Madrid, Spain
| | - Victoria López-Alonso
- Unidad de Biología Computacional, Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Isabel Liste
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| |
Collapse
|
8
|
Ma T, Wang Y, Yu L, Liu J, Wang T, Sun P, Feng Y, Zhang D, Shi L, He K, Zhao L, Xu Z. Mea6/cTAGE5 cooperates with TRAPPC12 to regulate PTN secretion and white matter development. iScience 2024; 27:109180. [PMID: 38439956 PMCID: PMC10909747 DOI: 10.1016/j.isci.2024.109180] [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: 07/31/2023] [Revised: 11/11/2023] [Accepted: 02/06/2024] [Indexed: 03/06/2024] Open
Abstract
Mutations of TRAPPC12 are associated with progressive childhood encephalopathy including abnormal white matter. However, the underlying pathogenesis is still unclear. Here, we found that Trappc12 deficiency in CG4 and oligodendrocyte progenitor cells (OPCs) affects their differentiation and maturation. In addition, TRAPPC12 interacts with Mea6/cTAGE5, and Mea6/cTAGE5 ablation in OPCs affects their proliferation and differentiation, leading to marked hypomyelination, compromised synaptic functionality, and aberrant behaviors in mice. We reveal that TRAPPC12 is associated with COPII components at ER exit site, and Mea6/cTAGE5 cKO disrupts the trafficking pathway by affecting the distribution and/or expression of TRAPPC12, SEC13, SEC31A, and SAR1. Moreover, we observed marked disturbances in the secretion of pleiotrophin (PTN) in Mea6-deficient OPCs. Notably, exogenous PTN supplementation ameliorated the differentiation deficits of these OPCs. Collectively, our findings indicate that the association between TRAPPC12 and MEA6 is important for cargo trafficking and white matter development.
Collapse
Affiliation(s)
- Tiantian Ma
- 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 100083, China
| | - Yaqing Wang
- 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 100083, China
| | - Laikang Yu
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, Haidian District, China
| | - Jinghua 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 100083, China
| | - Tao Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Pengyu 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 100083, China
| | - Yinghang Feng
- 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 100083, China
| | - Dan 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 100083, China
| | - Lei Shi
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Kangmin He
- 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 100083, China
| | - Li Zhao
- Key Laboratory of Physical Fitness and Exercise, Ministry of Education, Beijing Sport University, Beijing, Haidian District, China
| | - Zhiheng Xu
- 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 100083, China
| |
Collapse
|
9
|
da Cunha Nones DC, Novais CO, Rojas VCT, de Paula Franco P, da Silva Estevam E, Silva MS, Giusti-Paiva A, Dos Anjos-Garcia T, Vilela FC. Litter reduction-induced obesity promotes early depressive-like behavior and elevated prefrontal cortex GFAP expression in male offspring. Behav Brain Res 2024; 461:114839. [PMID: 38154508 DOI: 10.1016/j.bbr.2023.114839] [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: 09/15/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 12/30/2023]
Abstract
AIMS The present study was developed to investigate how litter reduction-induced obesity promotes early depressive-related behaviors in rodent offspring. MAIN METHODS We employed a standardized litter size reduction protocol, dividing litters into groups: normal litters (NL), consisting of six males and six females pups and small litters (SL), comprising two males and two females pups. Maternal behavior was monitored during the initial week of lactation. Subsequently, we assessed the pups for weight gain, locomotor activity, social play behavior, and performance in forced swimming test. We further evaluated the weights of retroperitoneal and perigonadal fat tissues, along with the expression of glial fibrillary acidic pprotein (GFAP) in the hippocampus and prefrontal cortex of the offspring. KEY FINDINGS Our results indicated that litter size reduction led to an increased the maternal behavior. In contrast, offspring from the SL group displayed greater weight gain and increased, retroperitoneal and perigonadal fat. Both male and female rodents in the SL group exhibited decreased social play behavior, and male offspring spent more time immobile during the forced swimming test, suggesting a depressive-like phenotype. Notably, we observed an increase in the GFAP expression in the prefrontal cortex of male rodents, with a trend toward increased expression in the hippocampus. SIGNIFICANCE Obesity may facilitate the development of early depressive-like behaviors, potentially associated with elevated GFAP expression in the prefrontal cortex.
Collapse
Affiliation(s)
- Débora Cristina da Cunha Nones
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde (PPGB), Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil; Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil
| | - Cíntia Onofra Novais
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas (PPGMCF), Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil; Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil
| | - Viviana Carolina Trujillo Rojas
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas (PPGMCF), Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil; Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil
| | - Priscila de Paula Franco
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas (PPGMCF), Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil; Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil
| | - Elisa da Silva Estevam
- Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil
| | - Mariana Santos Silva
- Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil
| | - Alexandre Giusti-Paiva
- Departamento de Ciências Fisiológicas, Centro de Ciências Biológicas da Universidade Federal de Santa Catarina (UFSC), Florianópolis, Santa Catarina, Brazil
| | - Tayllon Dos Anjos-Garcia
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas (PPGMCF), Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil; Departamento de Ciências Fisiológicas, Instituto de Ciências Biomédicas, Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil.
| | - Fabiana Cardoso Vilela
- Programa de Pós-Graduação em Biociências Aplicadas à Saúde (PPGB), Universidade Federal de Alfenas (Unifal-MG), Alfenas, Minas Gerais, Brazil; Centro de Inovação e Ensaios Pré-Clínicos (CIEnP), Florianópolis, Santa Catarina, Brazil
| |
Collapse
|
10
|
Liu J, Xin X, Sun J, Fan Y, Zhou X, Gong W, Yang M, Li Z, Wang Y, Yang Y, Gao C. Dual-targeting AAV9P1-mediated neuronal reprogramming in a mouse model of traumatic brain injury. Neural Regen Res 2024; 19:629-635. [PMID: 37721294 PMCID: PMC10581548 DOI: 10.4103/1673-5374.380907] [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: 11/16/2022] [Revised: 03/09/2023] [Accepted: 06/06/2023] [Indexed: 09/19/2023] Open
Abstract
Traumatic brain injury results in neuronal loss and glial scar formation. Replenishing neurons and eliminating the consequences of glial scar formation are essential for treating traumatic brain injury. Neuronal reprogramming is a promising strategy to convert glial scars to neural tissue. However, previous studies have reported inconsistent results. In this study, an AAV9P1 vector incorporating an astrocyte-targeting P1 peptide and glial fibrillary acidic protein promoter was used to achieve dual-targeting of astrocytes and the glial scar while minimizing off-target effects. The results demonstrate that AAV9P1 provides high selectivity of astrocytes and reactive astrocytes. Moreover, neuronal reprogramming was induced by downregulating the polypyrimidine tract-binding protein 1 gene via systemic administration of AAV9P1 in a mouse model of traumatic brain injury. In summary, this approach provides an improved gene delivery vehicle to study neuronal programming and evidence of its applications for traumatic brain injury.
Collapse
Affiliation(s)
- Jingzhou Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Xin Xin
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Jiejie Sun
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yueyue Fan
- Academy of Medical Engineering and Translational Medicine, Medical College, Tianjin University, Tianjin, China
| | - Xun Zhou
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Wei Gong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Meiyan Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Zhiping Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yuli Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Yang Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Chunsheng Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
11
|
Rodríguez JJ, Gardenal E, Zallo F, Arrue A, Cabot J, Busquets X. Astrocyte S100β expression and selective differentiation to GFAP and GS in the entorhinal cortex during ageing in the 3xTg-Alzheimer's disease mouse model. Acta Histochem 2024; 126:152131. [PMID: 38159478 DOI: 10.1016/j.acthis.2023.152131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
The study of astrocytes and its role in the development and evolution of neurodegenerative diseases, including Alzheimer's disease (AD) is essential to fully understand their aetiology. The aim if this study is to deepen into the concept of the heterogeneity of astrocyte subpopulations in the EC and in particular the identification of differentially functioning astrocyte subpopulations that respond differently to AD progression. S100β protein belongs to group of small calcium regulators of cell membrane channels and pumps that are expressed by astrocytes and is hypothesised to play and have a relevant role in AD development. We analysed the selective differentiation of S100β-positive astrocytes into Glutamine synthetase (GS) and Glial fibrillary acidic protein (GFAP)-positive sub-groups in the entorhinal cortex (EC) of AD triple transgenic animal model (3xTg-AD). EC is the brain region earliest affected in humans AD but also in this closest animal model regarding their pathology and time course. We observed no changes in the number of S100β-positive astrocytes between 1 and 18 months of age in the EC of 3xTg-AD mice. However, we identified relevant morphological changes in S100β/GFAP positive astrocytes showing a significant reduction in their surface and volume whilst an increase in number and percentage. Furthermore, the percentage of S100β/GS positive astrocyte population was also increased in 18 months old 3xTg-AD mice compared to the non-Tg mice. Our findings reveal the presence of differentially controlled astrocyte populations that respond differently to AD progression in the EC of 3xTg-AD mice. These results highpoints the major astrocytic role together with its early and marked affection in AD and arguing in favour of its importance in neurogenerative diseases and potential target for new therapeutic approaches.
Collapse
Affiliation(s)
- J J Rodríguez
- Functional Neuroanatomy Group, IKERBASQUE, Basque Foundation for Science, Dept. of Neurosciences, Medical Faculty, University of the Basque Country (UPV/EHU), 48009 Bilbao, 48940 Leioa, Bizkaia, Spain
| | - E Gardenal
- Functional Neuroanatomy Group, IKERBASQUE, Basque Foundation for Science, Dept. of Neurosciences, Medical Faculty, University of the Basque Country (UPV/EHU), 48009 Bilbao, 48940 Leioa, Bizkaia, Spain
| | - F Zallo
- Functional Neuroanatomy Group, IKERBASQUE, Basque Foundation for Science, Dept. of Neurosciences, Medical Faculty, University of the Basque Country (UPV/EHU), 48009 Bilbao, 48940 Leioa, Bizkaia, Spain
| | - A Arrue
- Neurochemical Research Unit, Bizkaia Mental Health Network, Osakidetza-Basque Health Service, Barakaldo 48903, Spain
| | - Joan Cabot
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, 07122 Palma, Spain
| | - X Busquets
- Laboratory of Molecular Cell Biomedicine, Department of Biology, University of the Balearic Islands, 07122 Palma, Spain.
| |
Collapse
|
12
|
Gopar-Cuevas Y, Saucedo-Cardenas O, Loera-Arias MJ, Montes-de-Oca-Luna R, Rodriguez-Rocha H, Garcia-Garcia A. Metformin and Trehalose-Modulated Autophagy Exerts a Neurotherapeutic Effect on Parkinson's Disease. Mol Neurobiol 2023; 60:7253-7273. [PMID: 37542649 DOI: 10.1007/s12035-023-03530-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/20/2023] [Indexed: 08/07/2023]
Abstract
Since the number of aged people will increase in the next years, neurodegenerative diseases, including Parkinson's Disease (PD), will also rise. Recently, we demonstrated that autophagy stimulation with rapamycin decreases dopaminergic neuronal death mediated by oxidative stress in the paraquat (PQ)-induced PD model. Assessing the neurotherapeutic efficacy of autophagy-inducing molecules is critical for preventing or delaying neurodegeneration. Therefore, we evaluated the autophagy inducers metformin and trehalose effect in a PD model. Autophagy induced by both molecules was confirmed in the SH-SY5Y dopaminergic cells by detecting increased LC3-II marker and autophagosome number compared to the control by western blot and transmission electron microscopy. Both autophagy inducers showed an antioxidant effect, improved mitochondrial activity, and decreased dopaminergic cell death induced by PQ. Next, we evaluated the effect of both inducers in vivo. C57BL6 mice were pretreated with metformin or trehalose before PQ administration. Cognitive and motor deteriorated functions in the PD model were evaluated through the nest building and the gait tests and were prevented by metformin and trehalose. Both autophagy inducers significantly reduced the dopaminergic neuronal loss, astrocytosis, and microgliosis induced by PQ. Also, cell death mediated by PQ was prevented by metformin and trehalose, assessed by TUNEL assay. Metformin and trehalose induced autophagy through AMPK phosphorylation and decreased α-synuclein accumulation. Therefore, metformin and trehalose are promising neurotherapeutic autophagy inducers with great potential for treating neurodegenerative diseases such as PD.
Collapse
Affiliation(s)
- Yareth Gopar-Cuevas
- Departamento de Histologia, Facultad de Medicina, Universidad Autonoma de Nuevo Leon, Francisco I. Madero S/N, 64460, Monterrey, Nuevo Leon, Mexico
| | - Odila Saucedo-Cardenas
- Departamento de Histologia, Facultad de Medicina, Universidad Autonoma de Nuevo Leon, Francisco I. Madero S/N, 64460, Monterrey, Nuevo Leon, Mexico
| | - Maria J Loera-Arias
- Departamento de Histologia, Facultad de Medicina, Universidad Autonoma de Nuevo Leon, Francisco I. Madero S/N, 64460, Monterrey, Nuevo Leon, Mexico
| | - Roberto Montes-de-Oca-Luna
- Departamento de Histologia, Facultad de Medicina, Universidad Autonoma de Nuevo Leon, Francisco I. Madero S/N, 64460, Monterrey, Nuevo Leon, Mexico
| | - Humberto Rodriguez-Rocha
- Departamento de Histologia, Facultad de Medicina, Universidad Autonoma de Nuevo Leon, Francisco I. Madero S/N, 64460, Monterrey, Nuevo Leon, Mexico.
| | - Aracely Garcia-Garcia
- Departamento de Histologia, Facultad de Medicina, Universidad Autonoma de Nuevo Leon, Francisco I. Madero S/N, 64460, Monterrey, Nuevo Leon, Mexico.
| |
Collapse
|
13
|
Gao MY, Wang JQ, He J, Gao R, Zhang Y, Li X. Single-Cell RNA-Sequencing in Astrocyte Development, Heterogeneity, and Disease. Cell Mol Neurobiol 2023; 43:3449-3464. [PMID: 37552355 PMCID: PMC11409980 DOI: 10.1007/s10571-023-01397-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 07/29/2023] [Indexed: 08/09/2023]
Abstract
Astrocytes are the most plentiful cell type in the central nervous system (CNS) and perform complicated functions in health and disease. It is obvious that different astrocyte subpopulations, or activation states, are relevant with specific genomic programs and functions. In recent years, the emergence of new technologies such as single-cell RNA sequencing (scRNA-seq) has made substantial advance in the characterization of astrocyte heterogeneity, astrocyte developmental trajectory, and its role in CNS diseases which has had a significant impact on neuroscience. In this review, we present an overview of astrocyte development, heterogeneity, and its essential role in the physiological and pathological environments of the CNS. We focused on the critical role of single-cell sequencing in revealing astrocyte development, heterogeneity, and its role in different CNS diseases.
Collapse
Affiliation(s)
- Meng-Yuan Gao
- A National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Jia-Qi Wang
- A National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Jin He
- A National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Rui Gao
- A National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Yuan Zhang
- A National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Xing Li
- A National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China.
| |
Collapse
|
14
|
Yao ZM, Sun XR, Huang J, Chen L, Dong SY. Astrocyte-Neuronal Communication and Its Role in Stroke. Neurochem Res 2023; 48:2996-3006. [PMID: 37329448 DOI: 10.1007/s11064-023-03966-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/03/2023] [Accepted: 06/06/2023] [Indexed: 06/19/2023]
Abstract
Astrocytes are the most abundant glial cells in the central nervous system. These cells are an important hub for intercellular communication. They participate in various pathophysiological processes, including synaptogenesis, metabolic transformation, scar production, and blood-brain barrier repair. The mechanisms and functional consequences of astrocyte-neuron signaling are more complex than previously thought. Stroke is a disease associated with neurons in which astrocytes also play an important role. Astrocytes respond to the alterations in the brain microenvironment after stroke, providing required substances to neurons. However, they can also have harmful effects. In this review, we have summarized the function of astrocytes, their association with neurons, and two paradigms of the inflammatory response, which suggest that targeting astrocytes may be an effective strategy for treating stroke.
Collapse
Affiliation(s)
- Zi-Meng Yao
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Xiao-Rong Sun
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Jie Huang
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Lei Chen
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Shu-Ying Dong
- Department of Pharmacology, School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China.
- Bengbu Medical College Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Bengbu, Anhui, China.
- Anhui Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, Anhui, China.
| |
Collapse
|
15
|
Huang B, Chen K, Li Y. Aerobic exercise, an effective prevention and treatment for mild cognitive impairment. Front Aging Neurosci 2023; 15:1194559. [PMID: 37614470 PMCID: PMC10442561 DOI: 10.3389/fnagi.2023.1194559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/28/2023] [Indexed: 08/25/2023] Open
Abstract
Aerobic exercise has emerged as a promising intervention for mild cognitive impairment (MCI), a precursor to dementia. The therapeutic benefits of aerobic exercise are multifaceted, encompassing both clinical and molecular domains. Clinically, aerobic exercise has been shown to mitigate hypertension and type 2 diabetes mellitus, conditions that significantly elevate the risk of MCI. Moreover, it stimulates the release of nitric oxide, enhancing arterial elasticity and reducing blood pressure. At a molecular level, it is hypothesized that aerobic exercise modulates the activation of microglia and astrocytes, cells crucial to brain inflammation and neurogenesis, respectively. It has also been suggested that aerobic exercise promotes the release of exercise factors such as irisin, cathepsin B, CLU, and GPLD1, which could enhance synaptic plasticity and neuroprotection. Consequently, regular aerobic exercise could potentially prevent or reduce the likelihood of MCI development in elderly individuals. These molecular mechanisms, however, are hypotheses that require further validation. The mechanisms of action are intricate, and further research is needed to elucidate the precise molecular underpinnings and to develop targeted therapeutics for MCI.
Collapse
Affiliation(s)
- Baiqing Huang
- Sports Institute, Yunnan Minzu University, Kunming, China
| | - Kang Chen
- Tianjin Key Lab of Exercise Physiology and Sports Medicine, Tianjin University of Sport, Tianjin, China
| | - Ying Li
- Sports Institute, Yunnan Minzu University, Kunming, China
| |
Collapse
|
16
|
Non-genomic Effect of Estradiol on the Neurovascular Unit and Possible Involvement in the Cerebral Vascular Accident. Mol Neurobiol 2023; 60:1964-1985. [PMID: 36596967 DOI: 10.1007/s12035-022-03178-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 12/16/2022] [Indexed: 01/05/2023]
Abstract
Cerebrovascular diseases, such as ischemic cerebral vascular accident (CVA), are responsible for causing high rates of morbidity, mortality, and disability in the population. The neurovascular unit (NVU) during and after ischemic CVA plays crucial roles in cell regulation and preservation, the immune and inflammatory response, and cell and/or tissue survival and repair. Cellular responses to 17β-estradiol (E2) can be triggered by two mechanisms: one called classical or genomic, which is due to the activation of the "classical" nuclear estrogen receptors α (ERα) and β (ERβ), and the non-genomic or rapid mechanism, which is due to the activation of the G protein-coupled estrogen receptor 1 (GPER) that is located in the plasma membrane and some in intracellular membranes, such as in the Golgi apparatus and endoplasmic reticulum. Nuclear receptors can regulate gene expression and cellular functions. On the contrary, activating the GPER by E2 and/or its G-1 agonist triggers several rapid cell signaling pathways. Therefore, E2 or its G-1 agonist, by mediating GPER activation and/or expression, can influence several NVU cell types. Most studies argue that the activation of the GPER may be used as a potential therapeutic target in various pathologies, such as CVA. Thus, with this review, we aimed to summarize the existing literature on the role of GPER mediated by E2 and/or its agonist G-1 in the physiology and pathophysiology of NVU.
Collapse
|
17
|
Mhatre-Winters I, Eid A, Han Y, Tieu K, Richardson JR. Sex and APOE Genotype Alter the Basal and Induced Inflammatory States of Primary Astrocytes from Humanized Targeted Replacement Mice. ASN Neuro 2023; 15:17590914221144549. [PMID: 36604975 PMCID: PMC9982390 DOI: 10.1177/17590914221144549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Apolipoprotein E4 (APOE4) genotype and sex are significant risk factors for Alzheimer's disease (AD), with females demonstrating increased risk modulated by APOE genotype. APOE is predominantly expressed in astrocytes, however, there is a lack of comprehensive assessments of sex differences in astrocytes stratified by APOE genotype. Here, we examined the response of mixed-sex and sex-specific neonatal APOE3 and APOE4 primary mouse astrocytes (PMA) to a cytokine mix of IL1b, TNFa, and IFNg. Pro-inflammatory and anti-inflammatory cytokine profiles were assessed by qRT-PCR and Meso Scale Discovery multiplex assay. Mixed-sex APOE4 PMA were found to have higher basal messenger RNA expression of several pro-inflammatory cytokines including Il6, Tnfa, Il1b, Mcp1, Mip1a, and Nos2 compared to APOE3 PMA, which was accompanied by increased levels of these secreted cytokines. In sex-specific cultures, basal expression of Il1b, Il6, and Nos2 was 1.5 to 2.5 fold higher in APOE4 female PMA compared to APOE4 males, with both being higher than APOE3 PMA. Similar results were found for secreted levels of these cytokines. Together, these findings indicate that APOE4 genotype and female sex, contribute to a greater inflammatory response in primary astrocytes and these data may provide a framework for investigating the mechanisms contributing to genotype and sex differences in AD-related neuroinflammation.
Collapse
Affiliation(s)
- Isha Mhatre-Winters
- Department of Environmental Health Sciences, Robert Stempel College
of Public Health and Social Work, Florida International
University, Miami, FL, USA,Department of Neurosciences, School of Biomedical Sciences, Kent
State University, Kent, OH, USA
| | - Aseel Eid
- Department of Environmental Health Sciences, Robert Stempel College
of Public Health and Social Work, Florida International
University, Miami, FL, USA
| | - Yoonhee Han
- Department of Environmental Health Sciences, Robert Stempel College
of Public Health and Social Work, Florida International
University, Miami, FL, USA
| | - Kim Tieu
- Department of Environmental Health Sciences, Robert Stempel College
of Public Health and Social Work, Florida International
University, Miami, FL, USA
| | - Jason R. Richardson
- Department of Environmental Health Sciences, Robert Stempel College
of Public Health and Social Work, Florida International
University, Miami, FL, USA,Jason R. Richardson, Department of
Environmental Health Sciences, Robert Stempel College of Public Health and
Social Work, Florida International University, Miami, FL 33199-2156, USA.
| |
Collapse
|
18
|
Naranjo O, Osborne O, Torices S, Toborek M. In Vivo Targeting of the Neurovascular Unit: Challenges and Advancements. Cell Mol Neurobiol 2022; 42:2131-2146. [PMID: 34086179 PMCID: PMC9056891 DOI: 10.1007/s10571-021-01113-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/28/2021] [Indexed: 12/26/2022]
Abstract
The blood-brain barrier (BBB) is essential for the homeostasis of the central nervous system (CNS). Functions of the BBB are performed by the neurovascular unit (NVU), which consists of endothelial cells, pericytes, astrocytes, microglia, basement membrane, and neurons. NVU cells interact closely and together are responsible for neurovascular coupling, BBB integrity, and transendothelial fluid transport. Studies have shown that NVU dysfunction is implicated in several acute and chronic neurological diseases, including Alzheimer's disease, multiple sclerosis, and stroke. The mechanisms of NVU disruption remain poorly understood, partially due to difficulties in selective targeting of NVU cells. In this review, we discuss the relative merits of available protein markers and drivers of the NVU along with recent advancements that have been made in the field to increase efficiency and specificity of NVU research.
Collapse
Affiliation(s)
- Oandy Naranjo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Olivia Osborne
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Silvia Torices
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Institute of Physiotherapy and Health Sciences, The Jerzy Kukuczka Academy of Physical Education, Katowice, Poland.
- Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, Gautier Bldg., Room 528, 1011 NW 15th Street, Miami, FL, 33136, USA.
| |
Collapse
|
19
|
Treviño S, Pulido G, Fuentes E, Handal-Silva A, Moreno-Rodríguez A, Venegas B, Flores G, Guevara J, Díaz A. Effect of cadmium administration on the antioxidant system and neuronal death in the hippocampus of rats. Synapse 2022; 76:1-16. [PMID: 35709361 DOI: 10.1002/syn.22242] [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: 12/09/2021] [Revised: 05/12/2022] [Accepted: 05/30/2022] [Indexed: 11/12/2022]
Abstract
Cadmium (Cd) is a heavy metal classified as a carcinogen whose exposure could affect the function of the central nervous system. Studies suggest that Cd modifies neuronal morphology in the hippocampus and affects cognitive tasks. The oxidative stress pathway is proposed as a mechanism of toxicity. However, this mechanism is not precise yet. This study aimed to evaluate the effect of Cd administration on oxidative stress markers in the male rat's hippocampus. Male Wistar rats were divided into (1) control (drinking water) and (2) treatment with Cd (32.5 ppm of cadmium chloride (CdCl2 ) in water). The Cd was administered for 2, 3, and 4 months. The results show that the oral administration of CdCl2 increased the concentration of Cd in plasma and hippocampus, and this response is time-dependent on its administration. Likewise, it caused an increase in lipid peroxidation and nitrosative stress markers. Moreover, it increased reactive astrogliosis and antioxidant enzyme activity. Consequently, the progression of the oxidative response exacerbated neurodegeneration in hippocampal cells. Our results suggest that Cd exposure induces a severe oxidative response that contributes critically to hippocampal neurodegeneration. It is suggested that exposure to Cd increases the risk of developing neurological diseases, which contributes to a decrease in the quality of life of the human and the environment in which it lives.
Collapse
Affiliation(s)
- Samuel Treviño
- Faculty of Chemical Sciences, Department of Pharmacy, Benemerita Autonomous University of Puebla, Puebla, Mexico
| | - Guadalupe Pulido
- Faculty of Chemical Sciences, Department of Pharmacy, Benemerita Autonomous University of Puebla, Puebla, Mexico
| | - Estefania Fuentes
- Faculty of Chemical Sciences, Department of Pharmacy, Benemerita Autonomous University of Puebla, Puebla, Mexico
| | - Anabella Handal-Silva
- Department of Biology and Reproductive Toxicology, Science Institute, Benemerita Autonomous University of Puebla, Puebla, Mexico
| | - Albino Moreno-Rodríguez
- Faculty of Chemical Sciences, Department of Pharmacy, Benemerita Autonomous University of Puebla, Puebla, Mexico
| | - Berenice Venegas
- Biological Sciences Faculty, Benemerita Autonomous University of Puebla, Puebla, Mexico
| | - Gonzalo Flores
- Neuropsychiatry Laboratory, Institute of Physiology, Benemerita Autonomous University of Puebla, Puebla, Mexico
| | - Jorge Guevara
- Faculty of Medicine, Department of Biochemistry, National Autonomous University of Mexico, Mexico City, Mexico
| | - Alfonso Díaz
- Faculty of Chemical Sciences, Department of Pharmacy, Benemerita Autonomous University of Puebla, Puebla, Mexico
| |
Collapse
|
20
|
Castellanos DB, Martín-Jiménez CA, Pinzón A, Barreto GE, Padilla-González GF, Aristizábal A, Zuluaga M, González Santos J. Metabolomic Analysis of Human Astrocytes in Lipotoxic Condition: Potential Biomarker Identification by Machine Learning Modeling. Biomolecules 2022; 12:biom12070986. [PMID: 35883542 PMCID: PMC9313230 DOI: 10.3390/biom12070986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/15/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023] Open
Abstract
The association between neurodegenerative diseases (NDs) and obesity has been well studied in recent years. Obesity is a syndrome of multifactorial etiology characterized by an excessive accumulation and release of fatty acids (FA) in adipose and non-adipose tissue. An excess of FA generates a metabolic condition known as lipotoxicity, which triggers pathological cellular and molecular responses, causing dysregulation of homeostasis and a decrease in cell viability. This condition is a hallmark of NDs, and astrocytes are particularly sensitive to it, given their crucial role in energy production and oxidative stress management in the brain. However, analyzing cellular mechanisms associated with these conditions represents a challenge. In this regard, metabolomics is an approach that allows biochemical analysis from the comprehensive perspective of cell physiology. This technique allows cellular metabolic profiles to be determined in different biological contexts, such as those of NDs and specific metabolic insults, including lipotoxicity. Since data provided by metabolomics can be complex and difficult to interpret, alternative data analysis techniques such as machine learning (ML) have grown exponentially in areas related to omics data. Here, we developed an ML model yielding a 93% area under the receiving operating characteristic (ROC) curve, with sensibility and specificity values of 80% and 93%, respectively. This study aimed to analyze the metabolomic profiles of human astrocytes under lipotoxic conditions to provide powerful insights, such as potential biomarkers for scenarios of lipotoxicity induced by palmitic acid (PA). In this work, we propose that dysregulation in seleno-amino acid metabolism, urea cycle, and glutamate metabolism pathways are major triggers in astrocyte lipotoxic scenarios, while increased metabolites such as alanine, adenosine, and glutamate are suggested as potential biomarkers, which, to our knowledge, have not been identified in human astrocytes and are proposed as candidates for further research and validation.
Collapse
Affiliation(s)
- Daniel Báez Castellanos
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110311, Colombia; (D.B.C.); (A.A.)
| | - Cynthia A. Martín-Jiménez
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center, Atlanta, GA 30329-4208, USA;
| | - Andrés Pinzón
- Laboratorio de Bioinformática y Biología de Sistemas, Universidad Nacional de Colombia, Bogotá 111321, Colombia;
| | - George E. Barreto
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland;
| | | | - Andrés Aristizábal
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110311, Colombia; (D.B.C.); (A.A.)
| | - Martha Zuluaga
- Escuela de Ciencias Básicas Tecnologías e Ingenierías, Universidad Nacional Abierta y a Distancia, Dosquebradas 661001, Colombia;
| | - Janneth González Santos
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110311, Colombia; (D.B.C.); (A.A.)
- Correspondence: ; Tel.: +57-60-1-3208320
| |
Collapse
|
21
|
Kou JJ, Shi JZ, He YY, Hao JJ, Zhang HY, Luo DM, Song JK, Yan Y, Xie XM, Du GH, Pang XB. Luteolin alleviates cognitive impairment in Alzheimer's disease mouse model via inhibiting endoplasmic reticulum stress-dependent neuroinflammation. Acta Pharmacol Sin 2022; 43:840-849. [PMID: 34267346 DOI: 10.1038/s41401-021-00702-8] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 05/17/2021] [Indexed: 12/14/2022]
Abstract
Luteolin is a flavonoid in a variety of fruits, vegetables, and herbs, which has shown anti-inflammatory, antioxidant, and anti-cancer neuroprotective activities. In this study, we investigated the potential beneficial effects of luteolin on memory deficits and neuroinflammation in a triple-transgenic mouse model of Alzheimer's disease (AD) (3 × Tg-AD). The mice were treated with luteolin (20, 40 mg · kg-1 · d-1, ip) for 3 weeks. We showed that luteolin treatment dose-dependently improved spatial learning, ameliorated memory deficits in 3 × Tg-AD mice, accompanied by inhibiting astrocyte overactivation (GFAP) and neuroinflammation (TNF-α, IL-1β, IL-6, NO, COX-2, and iNOS protein), and decreasing the expression of endoplasmic reticulum (ER) stress markers GRP78 and IRE1α in brain tissues. In rat C6 glioma cells, treatment with luteolin (1, 10 µM) dose-dependently inhibited LPS-induced cell proliferation, excessive release of inflammatory cytokines, and increase of ER stress marker GRP78. In conclusion, luteolin is an effective agent in the treatment of learning and memory deficits in 3 × Tg-AD mice, which may be attributable to the inhibition of ER stress in astrocytes and subsequent neuroinflammation. These results provide the experimental basis for further research and development of luteolin as a therapeutic agent for AD.
Collapse
|
22
|
Mekhaeil M, Dev KK, Conroy MJ. Existing Evidence for the Repurposing of PARP-1 Inhibitors in Rare Demyelinating Diseases. Cancers (Basel) 2022; 14:cancers14030687. [PMID: 35158955 PMCID: PMC8833351 DOI: 10.3390/cancers14030687] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/23/2022] [Accepted: 01/27/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Poly (ADP-ribose) polymerase-1 (PARP-1) inhibitors are successful cancer therapeutics that impair DNA repair machinery, leading to an accumulation of DNA damage and consequently cell death. The shared underlying mechanisms driving malignancy and demyelinating disease, together with the success of anticancer drugs as repurposed therapeutics, makes the repurposing of PARP-1 inhibitors for demyelinating diseases a worthy concept to consider. In addition, PARP-1 inhibitors demonstrate notable neuroprotective effects in demyelinating disorders, including multiple sclerosis which is considered the archetypical demyelinating disease. Abstract Over the past decade, Poly (ADP-ribose) polymerase-1 (PARP-1) inhibitors have arisen as a novel and promising targeted therapy for breast cancer gene (BRCA)-mutated ovarian and breast cancer patients. Therapies targeting the enzyme, PARP-1, have since established their place as maintenance drugs for cancer. Here, we present existing evidence that implicates PARP-1 as a player in the development and progression of both malignancy and demyelinating disease. These findings, together with the proven clinical efficacy and marketed success of PARP-1 inhibitors in cancer, present the repurposing of these drugs for demyelinating diseases as a desirable therapeutic concept. Indeed, PARP-1 inhibitors are noted to demonstrate neuroprotective effects in demyelinating disorders such as multiple sclerosis and Parkinson’s disease, further supporting the use of these drugs in demyelinating, neuroinflammatory, and neurodegenerative diseases. In this review, we discuss the potential for repurposing PARP-1 inhibitors, with a focus on rare demyelinating diseases. In particular, we address the possible use of PARP-1 inhibitors in examples of rare leukodystrophies, for which there are a paucity of treatment options and an urgent need for novel therapeutic approaches.
Collapse
Affiliation(s)
- Marianna Mekhaeil
- Drug Development Research Group, Department of Physiology, School of Medicine, Trinity College Dublin, D18 DH50 Dublin, Ireland; (M.M.); (K.K.D.)
- Cancer Immunology Research Group, Department of Physiology, School of Medicine, Trinity College Dublin, D18 DH50 Dublin, Ireland
| | - Kumlesh Kumar Dev
- Drug Development Research Group, Department of Physiology, School of Medicine, Trinity College Dublin, D18 DH50 Dublin, Ireland; (M.M.); (K.K.D.)
| | - Melissa Jane Conroy
- Cancer Immunology Research Group, Department of Physiology, School of Medicine, Trinity College Dublin, D18 DH50 Dublin, Ireland
- Correspondence:
| |
Collapse
|
23
|
Contribution of senescent and reactive astrocytes on central nervous system inflammaging. Biogerontology 2022; 23:21-33. [PMID: 35084630 DOI: 10.1007/s10522-022-09952-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/19/2022] [Indexed: 01/10/2023]
Abstract
Astrocytes, the most predominant cells in the central nervous system (CNS), have well-recognized neuroprotective functions. However, during the CNS aging, astrocytes can become neurotoxic and contribute to chronic inflammation in age-associated brain deterioration and disease. Astrocytes are known to become senescent or reactive due to the exposure to stressful stimuli, in both cases they contribute to an impaired cognitive function through the production of pro-inflammatory mediators. Although both scenarios (senescence and reactive gliosis) have been studied independently, there are no direct studies comparing their secretomes simultaneously in the aging-brain. In this review we discuss the most recent studies in that respect, in order to analyze their simultaneous participation in brain aging.
Collapse
|
24
|
Zhao Z, Hu X, Wang J, Wang J, Hou Y, Chen S. Zinc finger E-Box binding homeobox 2 (ZEB2)-induced astrogliosis protected neuron from pyroptosis in cerebral ischemia and reperfusion injury. Bioengineered 2021; 12:12917-12930. [PMID: 34852714 PMCID: PMC8809936 DOI: 10.1080/21655979.2021.2012551] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/24/2021] [Accepted: 11/25/2021] [Indexed: 11/15/2022] Open
Abstract
Ischemia injury can cause cell death or impairment of neuron and astrocytes, and thus induce loss of nerve function. central nervous systems injury induces an aberrant activation of astrocytes called astrogliosis. Pyroptosis, which is a kind of programmed cell death, was proved play an important role in ischemia injury. Zinc Finger E-Box Binding Homeobox 2 (ZEB2) promoted neuron astrogliosis, which play a protected role in neuron regeneration. However, its precise mechanism remains unclear. This study investigated the mechanism of ZEB2 on astrogliosis and neuron regeneration after cerebral ischemia reperfusion condition. To confirm our hypothesis, Neurons and astrocytes were isolated from fetal Sprague Dawley rats, in vivo Middle Cerebral Artery Occlusion/reperfusion (MCAO/R) rat model and in vitro oxygen-glucose deprivation/reperfusion (OGD/R)-treated astrocytes and neurocytes model were constructed. Our results showed that ZEB2 was expressed in nucleus of astrocyte and upregulated after OGD/R induction, ZEB2 promoted astrogliosis. Further upregulation of ZEB2 promoted the astrogliosis, which promoted neuron proliferation and regeneration by decreased pyroptosis. Moreover, this finding was further confirmed in vivo MCAO/R rat model. Overexpression of ZEB2 promoted astrogliosis, which decreased infarct volume and accumulated recovery of neurological function by alleviated pyroptosis. This finding revealed that ZEB2 was a regulator of the astrogliosis after ischemia/reperfusion injury, and then astrogliosis promoted neuron regeneration via decreased neuron pyroptosis. Therefore, ZEB2 may be a potential therapeutic target for ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Zhixin Zhao
- Department of Neurosurgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Zhejiang Province, China
| | - Xiaoming Hu
- Department of Neurosurgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Zhejiang Province, China
| | - Jie Wang
- Department of Neurosurgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Zhejiang Province, China
| | - Jianfeng Wang
- Department of Neurosurgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Zhejiang Province, China
| | - Yong Hou
- Department of Neurosurgery, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Zhejiang Province, China
| | - Suyun Chen
- Department of Clinical Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Zhejiang Province, China
| |
Collapse
|
25
|
Xu J, Wang C, Xu P. Effects of hydroxyapatite extract on rats with transient ischemia: Long-term potentiation and axon regeneration. Exp Ther Med 2021; 22:1486. [PMID: 34765027 DOI: 10.3892/etm.2021.10921] [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: 04/03/2020] [Accepted: 08/10/2021] [Indexed: 11/05/2022] Open
Abstract
Hydroxyapatite (HA) has been extensively used as a reconstructive and prosthetic material for osseous tissue. The present study aimed to determine whether HA extract exerted effects on central nervous system injury following transient cerebral ischemia/reperfusion in rats. Male Wistar rats were treated with HA following bilateral common carotid artery clamping (two-vessel occlusion). The results demonstrated that treatment with HA extract attenuated the inhibition of long-term potential in a rat model of transient cerebral ischemia/reperfusion. Furthermore, HA extract improved axon regeneration, which was confirmed via the immunohistochemical analysis of growth associated protein 43 and glial fibrillary acidic protein. Taken together, the results of the present study provided preliminary evidence of the protective effect of HA on neuronal damage.
Collapse
Affiliation(s)
- Jing Xu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Chunyang Wang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Pengjuan Xu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, P.R. China
| |
Collapse
|
26
|
Salcman B, Affleck K, Bulfone-Paus S. P2X Receptor-Dependent Modulation of Mast Cell and Glial Cell Activities in Neuroinflammation. Cells 2021; 10:cells10092282. [PMID: 34571930 PMCID: PMC8471135 DOI: 10.3390/cells10092282] [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: 07/13/2021] [Revised: 08/23/2021] [Accepted: 08/30/2021] [Indexed: 11/27/2022] Open
Abstract
Localisation of mast cells (MCs) at the abluminal side of blood vessels in the brain favours their interaction with glial cells, neurons, and endothelial cells, resulting in the activation of these cells and the release of pro-inflammatory mediators. In turn, stimulation of glial cells, such as microglia, astrocytes, and oligodendrocytes may result in the modulation of MC activities. MCs, microglia, astrocytes, and oligodendrocytes all express P2X receptors (P2XRs) family members that are selectively engaged by ATP. As increased concentrations of extracellular adenosine 5′-triphosphate (ATP) are present in the brain in neuropathological conditions, P2XR activation in MCs and glial cells contributes to the control of their communication and amplification of the inflammatory response. In this review we discuss P2XR-mediated MC activation, its bi-directional effect on microglia, astrocytes and oligodendrocytes and role in neuroinflammation.
Collapse
Affiliation(s)
- Barbora Salcman
- Lydia Becker Institute of Immunology and Inflammation, Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester M13 9NT, UK;
| | - Karen Affleck
- GlaxoSmithKline, Immunology Research Unit, Stevenage SG1 2NY, UK;
| | - Silvia Bulfone-Paus
- Lydia Becker Institute of Immunology and Inflammation, Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester M13 9NT, UK;
- Correspondence:
| |
Collapse
|
27
|
Gao L, Zheng WG, Wu XK, Du GH, Qin XM. Baicalein Delays H 2O 2-Induced Astrocytic Senescence through Inhibition of Senescence-Associated Secretory Phenotype (SASP), Suppression of JAK2/STAT1/NF-κB Pathway, and Regulation of Leucine Metabolism. ACS Chem Neurosci 2021; 12:2320-2335. [PMID: 34152720 DOI: 10.1021/acschemneuro.1c00024] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Baicalein is an active ingredient extracted from the dried roots of the Scutellaria baicalensis Georgi. It has been demonstrated to improve memory impairment in multiple animal models; however, the underlying mechanisms remain ambiguous. The accumulation of senescent astrocytes and senescence-associated secretory phenotype (SASP) secreted by senescent astrocytes has been deemed as potential contributors to neurodegenerative diseases. Therefore, this study explored the protective effects of baicalein against astrocyte senescence and investigated the molecular mechanisms and metabolic mechanisms of baicalein against astrocyte senescence. Our results demonstrated that treatment with baicalein protects T98G cells from H2O2-induced damage, delays cell senescence, inhibits the secretion of SASP (IL-6, IL-8, TNF-α, CXCL1, and MMP-1), and inhibits SASP-related pathways NF-κB and JAK2/STAT1. 1H NMR metabolomics analysis and correlation analysis revealed that leucine was significantly correlated with SASP factors. Further study demonstrated that supplement with leucine could restrain SASP secretion, and baicalein could significantly increase leucine level through down-regulation of BCAT1 and up-regulation of SLC7A5 expression. The above results revealed that baicalein exerted protective and antisenescence effects in H2O2-induced T98G cells possibly through inhibition of SASP, suppression of JAK2/STAT1/NF-κB pathway, and regulation of leucine metabolism. Consistent results were obtained in primary astrocytes of newborn SD rats, which suggests that baicalein significantly increases viabilities, delays senescence, inhibits IL-6 secretion, and increases leucine level in H2O2-induced primary astrocytes.
Collapse
Affiliation(s)
- Li Gao
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan 030006, China
- Key Laboratory of Chemical Biology and Molecular Engineering of Minstry of Education, Taiyuan 030006, China
| | - Wen-ge Zheng
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan 030006, China
- Key Laboratory of Chemical Biology and Molecular Engineering of Minstry of Education, Taiyuan 030006, China
| | - Xing-kang Wu
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan 030006, China
- Key Laboratory of Chemical Biology and Molecular Engineering of Minstry of Education, Taiyuan 030006, China
| | - Guan-hua Du
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xue-mei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China
- Key Laboratory of Effective Substances Research and Utilization in TCM of Shanxi Province, Taiyuan 030006, China
- Key Laboratory of Chemical Biology and Molecular Engineering of Minstry of Education, Taiyuan 030006, China
| |
Collapse
|
28
|
Liu Y, Chu S, Hu Y, Yang S, Li X, Zheng Q, Ai Q, Ren S, Wang H, Gong L, Xu X, Chen NH. Exogenous Adenosine Antagonizes Excitatory Amino Acid Toxicity in Primary Astrocytes. Cell Mol Neurobiol 2021; 41:687-704. [PMID: 32632892 PMCID: PMC11448567 DOI: 10.1007/s10571-020-00876-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 05/12/2020] [Indexed: 12/29/2022]
Abstract
Excitatory toxicity is still a hot topic in the study of ischemic stroke, and related research has focused mainly on neurons. Adenosine is an important neuromodulator that is known as a "biosignature" in the central nervous system (CNS). The protective effect of exogenous adenosine on neurons has been confirmed, but its mechanism remains elusive. In this study, astrocytes were pretreated with adenosine, and the effects of an A2a receptor (A2aR) inhibitor (SCH58261) and A2b receptor (A2bR) inhibitor (PSB1115) on excitatory glutamate were investigated. An oxygen glucose deprivation/reoxygenation (OGD/R) and glutamate model was generated in vitro. Post-model assessment included expression levels of glutamate transporters (glt-1), gap junction protein (Cx43) and glutamate receptor (AMPAR), Na+-K+-ATPase activity, and diffusion distance of dyes. Glutamate and glutamine contents were determined at different time points. The results showed that (1) adenosine could improve the function of Na+-K+-ATPase, upregulate the expression of glt-1, and enhance the synthesis of glutamine in astrocytes. This effect was associated with A2aR activation but not with A2bR activation. (2) Adenosine could inhibit the expression of gap junction protein (Cx43) and reduce glutamate diffusion. Inhibition of A2aR attenuated adenosine inhibition of gap junction intercellular communication (GJIC) in the OGD/R model, while it enhanced adenosine inhibition of GJIC in the glutamate model, depending on the glutamate concentration. (3) Adenosine could cause AMPAR gradually entered the nucleus from the cytoplasm, thereby reducing the expression of AMPAR on the cell membrane. Taken together, the results indicate that adenosine plays a role of anti-excitatory toxicity effect in protection against neuronal death and the functional recovery of ischemic stroke mainly by targeting astrocytes, which are closely related to A2aR. The present study provided a scientific basis for adenosine prevention and ischemic stroke treatment, thereby providing a new approach for alleviating ischemic stroke.
Collapse
Affiliation(s)
- Yingjiao Liu
- College of Pharmacy, Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Shifeng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yaomei Hu
- College of Pharmacy, Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, China
| | - Songwei Yang
- College of Pharmacy, Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, China
| | - Xun Li
- College of Pharmacy, Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Qinglian Zheng
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
| | - Qidi Ai
- College of Pharmacy, Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, China
| | - Siyu Ren
- College of Pharmacy, Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, China
| | - Huiqin Wang
- College of Pharmacy, Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, China
| | - Limin Gong
- College of Pharmacy, Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, China
| | - Xin Xu
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China
| | - Nai-Hong Chen
- College of Pharmacy, Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha, 410208, China.
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, 510631, China.
| |
Collapse
|
29
|
Low-Dose Copper Exposure Exacerbates Depression-Like Behavior in ApoE4 Transgenic Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6634181. [PMID: 33833851 PMCID: PMC8018851 DOI: 10.1155/2021/6634181] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/20/2021] [Accepted: 03/04/2021] [Indexed: 11/18/2022]
Abstract
Depression is one of the most common neuropsychiatric disorders. Although the pathogenesis of depression is still unknown, environmental risk factors and genetics are implicated. Copper (Cu), a cofactor of multiple enzymes, is involved in regulating depression-related processes. Depressed patients carrying the apolipoprotein ε4 allele display more severe depressive symptoms, indicating that ApoE4 is closely associated with an increased risk of depression. The study explored the effect of low-dose Cu exposure and ApoE4 on depression-like behavior of mice and further investigates the possible mechanisms. The ApoE4 mice and wild-type (WT) mice were treated with 0.13 ppm CuCl2 for 4 months. After the treatment, ApoE4 mice displayed obvious depression-like behavior compared with the WT mice, and Cu exposure further exacerbated the depression-like behavior of ApoE4 mice. There was no significant difference in anxiety behavior and memory behavior. Proteomic analysis revealed that the differentially expressed proteins between Cu-exposed and nonexposed ApoE4 mice were mainly involved in the Ras signaling pathway, protein export, axon guidance, serotonergic synapse, GABAergic synapse, and dopaminergic synapse. Among these differentially expressed proteins, immune response and synaptic function are highly correlated. Representative protein expression changes are quantified by western blot, showing consistent results as determined by proteomic analysis. Hippocampal astrocytes and microglia were increased in Cu-exposed ApoE4 mice, suggesting that neuroglial cells played an important role in the pathogenesis of depression. Taken together, our study demonstrated that Cu exposure exacerbates depression-like behavior of ApoE4 mice and the mechanisms may involve the dysregulation of synaptic function and immune response and overactivation of neuroinflammation.
Collapse
|
30
|
Ding ZB, Song LJ, Wang Q, Kumar G, Yan YQ, Ma CG. Astrocytes: a double-edged sword in neurodegenerative diseases. Neural Regen Res 2021; 16:1702-1710. [PMID: 33510058 PMCID: PMC8328766 DOI: 10.4103/1673-5374.306064] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Astrocytes play multifaceted and vital roles in maintaining neurophysiological function of the central nervous system by regulating homeostasis, increasing synaptic plasticity, and sustaining neuroprotective effects. Astrocytes become activated as a result of inflammatory responses during the progression of pathological changes associated with neurodegenerative disorders. Reactive astrocytes (neurotoxic A1 and neuroprotective A2) are triggered during disease progression and pathogenesis due to neuroinflammation and ischemia. However, only a limited body of literature describes morphological and functional changes of astrocytes during the progression of neurodegenerative diseases. The present review investigated the detrimental and beneficial roles of astrocytes in neurodegenerative diseases reported in recent studies, as these cells have promising therapeutic potential and offer new approaches for treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Zhi-Bin Ding
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine; Department of Neurology, Affiliated Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Li-Juan Song
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine; Department of Neurology, Affiliated Shanxi Bethune Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Qing Wang
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan, Shanxi Province, China
| | - Gajendra Kumar
- Department of Neuroscience, City University of Hong Kong, Tat Chee Avenue, Hong Kong Special Administrative Region, China
| | - Yu-Qing Yan
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan; Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, Shanxi Province, China
| | - Cun-Gen Ma
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine, Research Center of Neurobiology, Shanxi University of Chinese Medicine, Taiyuan; Institute of Brain Science, Shanxi Key Laboratory of Inflammatory Neurodegenerative Diseases, Medical School of Shanxi Datong University, Datong, Shanxi Province, China
| |
Collapse
|
31
|
Yu M, Zheng N, Jiang D, Wang L, Zhan Q, Zhao J. Chemokine C-C motif ligand 2 suppressed the growth of human brain astrocytes under Ischemic/hypoxic conditions via regulating ERK1/2 pathway. Brain Inj 2020; 34:1277-1282. [PMID: 32749897 DOI: 10.1080/02699052.2020.1797167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PRIMARY OBJECTIVE Chemokine C-C motif ligand 2 (CCL2) plays a critical role in inflammation-related diseases in the central nervous system (CNS). However, the role of CCL2 in ischemic stroke remains unclear. RESEARCH DESIGN To investigate the role of CCL2 in ischemic stroke, we performed oxygen-glucose deprivation (OGD) on human brain astrocytes. METHODS AND PROCEDURES To assess cell proliferation, the CCK-8 assay was performed. Cell apoptosis was determined using flow cytometry. qRT-PCR and western blotting were utilized to measure gene expression. MAIN OUTCOMES AND RESULTS Our results suggest that CCL2 and its receptor CCR2 are upregulated in OGD cells. Moreover, a CCL2 antibody significantly alleviated the ischemic/hypoxic-induced suppression of growth in human brain astrocytes. Human recombinant protein, CCL2, inhibited the growth of human brain astrocytes under normoxia conditions. These results demonstrate that CCL2 upregulation suppresses the recovery of human brain astrocytes under ischemic/hypoxic conditions. This effect was abolished by the ERK inhibitor PD98059. Therefore, CCL2/CCR2 activation may suppress the growth of human brain astrocytes through enhancing the activity of ERK1/2. CONCLUSIONS Our results not only developed a deeper understanding of the role of CCL2 in human brain astrocytes but also provided novel insight into potential treatments for ischemic stroke.
Collapse
Affiliation(s)
- Min Yu
- Department of Neurology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine , Shanghai, China
| | - Ni Zheng
- Department of Nuclear Medicine, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine , Shanghai, China
| | - Dudu Jiang
- Department of Neurology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine , Shanghai, China
| | - Lijing Wang
- Department of Neurology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine , Shanghai, China
| | - Qing Zhan
- Department of Neurology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine , Shanghai, China
| | - Jiangmin Zhao
- Department of Radiology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai, China
| |
Collapse
|
32
|
Zhu Y, Fan Z, Wang R, Xie R, Guo H, Zhang M, Guo B, Sun T, Zhang H, Zhuo L, Li Y, Wu S. Single-Cell Analysis for Glycogen Localization and Metabolism in Cultured Astrocytes. Cell Mol Neurobiol 2020; 40:801-812. [PMID: 31863221 PMCID: PMC7261284 DOI: 10.1007/s10571-019-00775-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/08/2019] [Indexed: 12/22/2022]
Abstract
Cerebral glycogen is principally localized in astrocytes rather than in neurons. Glycogen metabolism has been implicated in higher brain functions, including learning and memory, yet the distribution patterns of glycogen in different types of astrocytes have not been fully described. Here, we applied a method based on the incorporation of 2-NBDG, a D-glucose fluorescent derivative that can trace glycogen, to investigate glycogen's distribution in the brain. We identified two types of astrocytes, namely, 2-NBDGI (glycogen-deficient) and 2-NBDGII (glycogen-rich) cells. Whole-cell patch-clamp and fluorescence-activated cell sorting (FACS) were used to separate 2-NBDGII astrocytes from 2-NBDGI astrocytes. The expression levels of glycogen metabolic enzymes were analyzed in 2-NBDGI and 2-NBDGII astrocytes. We found unique glycogen metabolic patterns between 2-NBDGI and 2-NBDGII astrocytes. We also observed that 2-NBDGII astrocytes were mainly identified as fibrous astrocytes but not protoplasmic astrocytes. Our data reveal cell type-dependent glycogen distribution and metabolism patterns, suggesting diverse functions of these different astrocytes.
Collapse
Affiliation(s)
- Yuanyuan Zhu
- Department of Neurobiology, The School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Ze Fan
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital of the Fourth Military Medical University, Xi'an, China
| | - Rui Wang
- Department of Neurobiology, The School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Rougang Xie
- Department of Neurobiology, The School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Haiyun Guo
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital of the Fourth Military Medical University, Xi'an, China
| | - Ming Zhang
- Department of Neurobiology, The School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Baolin Guo
- Department of Neurobiology, The School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Tangna Sun
- Department of Neurobiology, The School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Haifeng Zhang
- Department of Neurobiology, The School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Lixia Zhuo
- Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Li
- Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| | - Shengxi Wu
- Department of Neurobiology, The School of Basic Medicine, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
33
|
Zhang Y, Shi Q, Li X, Xia C. Fasciculation and Elongation Protein Zeta-1 Expression in Reactive Astrocytes in a Rat Model of Frontal Lobe Injury. J Neuropathol Exp Neurol 2020; 79:194-208. [PMID: 31774489 DOI: 10.1093/jnen/nlz113] [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: 03/11/2019] [Revised: 06/05/2019] [Indexed: 11/12/2022] Open
Abstract
There are reports that depression induced by frontal lobe injury (FLI) has a devastating effect on human mental health. We previously reported that fasciculation and elongation protein zeta-1 (FEZ1) was essential for astrocytic protection of dopamine neurons. Studies of glutamate-glutamine cycle in mental illness have been reported, whereas not from the perspective of astrocytes. This study was designed to investigate the roles of astrocytic FEZ1 and glutamate-glutamine cycle after FLI. A model of FLI was established by inserting a blade into the right frontal lobe of rats. Behavioral tests were used to observe the behavioral changes of FLI rats. Neuropathologic examinations, including immunohistochemistry, were conducted. Behavioral tests showed that FLI decreased exploratory activity. Western blot analysis revealed that the expression of astroglial proteins overall decreased in the initial injury stage, as well as FEZ1. Immunohistochemistry showed a shift of FEZ1 localization from neurons in sham-lesioned rats to astrocytes in FLI rats, and showed the expression profile of glutamate transporter 1 and glutamine synthetase (GS) was consistent with Western blot observation. Our results indicate that astrocytic FEZ1 and glutamate-glutamine cycle dysfunction may be involved in the pathogenesis of depression after FLI.
Collapse
Affiliation(s)
- Ye Zhang
- From the Cytoneurobiology Unit, Department of Anatomy, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Qing Shi
- From the Cytoneurobiology Unit, Department of Anatomy, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Xiwen Li
- From the Cytoneurobiology Unit, Department of Anatomy, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| | - Chunlin Xia
- From the Cytoneurobiology Unit, Department of Anatomy, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
34
|
Morris G, Maes M, Berk M, Carvalho AF, Puri BK. Nutritional ketosis as an intervention to relieve astrogliosis: Possible therapeutic applications in the treatment of neurodegenerative and neuroprogressive disorders. Eur Psychiatry 2020; 63:e8. [PMID: 32093791 PMCID: PMC8057392 DOI: 10.1192/j.eurpsy.2019.13] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Nutritional ketosis, induced via either the classical ketogenic diet or the use of emulsified medium-chain triglycerides, is an established treatment for pharmaceutical resistant epilepsy in children and more recently in adults. In addition, the use of oral ketogenic compounds, fractionated coconut oil, very low carbohydrate intake, or ketone monoester supplementation has been reported to be potentially helpful in mild cognitive impairment, Parkinson’s disease, schizophrenia, bipolar disorder, and autistic spectrum disorder. In these and other neurodegenerative and neuroprogressive disorders, there are detrimental effects of oxidative stress, mitochondrial dysfunction, and neuroinflammation on neuronal function. However, they also adversely impact on neurone–glia interactions, disrupting the role of microglia and astrocytes in central nervous system (CNS) homeostasis. Astrocytes are the main site of CNS fatty acid oxidation; the resulting ketone bodies constitute an important source of oxidative fuel for neurones in an environment of glucose restriction. Importantly, the lactate shuttle between astrocytes and neurones is dependent on glycogenolysis and glycolysis, resulting from the fact that the astrocytic filopodia responsible for lactate release are too narrow to accommodate mitochondria. The entry into the CNS of ketone bodies and fatty acids, as a result of nutritional ketosis, has effects on the astrocytic glutamate–glutamine cycle, glutamate synthase activity, and on the function of vesicular glutamate transporters, EAAT, Na+, K+-ATPase, Kir4.1, aquaporin-4, Cx34 and KATP channels, as well as on astrogliosis. These mechanisms are detailed and it is suggested that they would tend to mitigate the changes seen in many neurodegenerative and neuroprogressive disorders. Hence, it is hypothesized that nutritional ketosis may have therapeutic applications in such disorders.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia
| | - Michael Maes
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia.,Department of Psychiatry, Chulalongkorn University, Faculty of Medicine, Bangkok, Thailand
| | - Michael Berk
- Deakin University, IMPACT Strategic Research Centre, Barwon Health, School of Medicine, Geelong, Victoria, Australia.,Deakin University, CMMR Strategic Research Centre, School of Medicine, Geelong, Victoria, Australia.,Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and the Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - André F Carvalho
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada.,Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
| | | |
Collapse
|
35
|
Hindeya Gebreyesus H, Gebrehiwot Gebremichael T. The Potential Role of Astrocytes in Parkinson's Disease (PD). Med Sci (Basel) 2020; 8:E7. [PMID: 32012713 PMCID: PMC7151567 DOI: 10.3390/medsci8010007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 08/08/2019] [Accepted: 08/09/2019] [Indexed: 12/11/2022] Open
Abstract
Astrocytes are multi-functional cells, now recognized as critical participants in many brain functions. They play a critical physiological role in the clearance of neurotransmitters, such as glutamate and gamma-aminobutyric acid (GABA), and in the regulation of K+ from the space of synaptic clefts. Astrocytes also express the excitatory amino acid transporters (EAATs) and aquaporin-4 (AQP4) water channel, which are involved in both physiological functions and neurodegenerative diseases (ND). Some of the ND are the Alzheimer's (AD), Huntington's (HD), Parkinson's diseases (PD), Cerebral edema, amyotrophic lateral sclerosis (ALS), and epilepsy pathological conditions in specific regions of the CNS. Parkinson's disease is the second most common age-related neurodegenerative disorder, characterized by degeneration of dopaminergic neurons of the substantia nigra pars compacta (SNpc). These project to the striatum, forming an important pathway within the basal ganglia. Mostly, PD has no clear etiology, and the mechanism of dopaminergic (DA) neuron loss is not well illustrated. The results of various studies suggest that astrocytes are involved in the pathophysiology of PD. Evidence has shown that the down-regulation of EAAT-2/GLT-1 and AQP4 expression is associated with PD pathogenesis. However, controversial results were reported in different experimental studies about the expression and function of EAAT-2/GLT-1 and AQP4, as well as their colocalization in different brain regions, and their involvement in PD development. Therefore, under neurological disorders, Parkinson's disease is related to the genetic and phenotypic change of astrocytes' biology. In this review, the authors summarized recent their research findings, which revealed the involvement of EAAT-2/GLT-1 and AQP4 expression, the physical interaction between EAAT-2/GLT-1 and AQP4 in astrocyte function, and their potential role in the development of PD in SNpc and Subthalamic nucleus (STN) of the basal ganglia nuclei.
Collapse
Affiliation(s)
- Hiluf Hindeya Gebreyesus
- School of Medicine, Biomedical Sciences, College of Health Sciences, Mekelle University, P.O. Box: 1871 Mekelle, Tigray, Ethiopia
| | | |
Collapse
|
36
|
Ginsenosides Rb1 and Rg1 Protect Primary Cultured Astrocytes against Oxygen-Glucose Deprivation/Reoxygenation-Induced Injury via Improving Mitochondrial Function. Int J Mol Sci 2019; 20:ijms20236086. [PMID: 31816825 PMCID: PMC6929005 DOI: 10.3390/ijms20236086] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 11/28/2019] [Accepted: 11/29/2019] [Indexed: 11/23/2022] Open
Abstract
This study aimed to evaluate whether ginsenosides Rb1 (20-S-protopanaxadiol aglycon) and Rg1 (20-S-protopanaxatriol aglycon) have mitochondrial protective effects against oxygen-glucose deprivation/reoxygenation (OGD/R)-induced injury in primary mouse astrocytes and to explore the mechanisms involved. The OGD/R model was used to mimic the pathological process of cerebral ischemia-reperfusion in vitro. Astrocytes were treated with normal conditions, OGD/R, OGD/R plus Rb1, or OGD/R plus Rg1. Cell viability was measured to evaluate the cytotoxicity of Rb1 and Rg1. Intracellular reactive oxygen species (ROS) and catalase (CAT) were detected to evaluate oxidative stress. The mitochondrial DNA (mtDNA) copy number and mitochondrial membrane potential (MMP) were measured to evaluate mitochondrial function. The activities of the mitochondrial respiratory chain (MRC) complexes I–V and the level of cellular adenosine triphosphate (ATP) were measured to evaluate oxidative phosphorylation (OXPHOS) levels. Cell viability was significantly decreased in the OGD/R group compared to the control group. Rb1 or Rg1 administration significantly increased cell viability. Moreover, OGD/R caused a significant increase in ROS formation and, subsequently, it decreased the activity of CAT and the mtDNA copy number. At the same time, treatment with OGD/R depolarized the MMP in the astrocytes. Rb1 or Rg1 administration reduced ROS production, increased CAT activity, elevated the mtDNA content, and attenuated the MMP depolarization. In addition, Rb1 or Rg1 administration increased the activities of complexes I, II, III, and V and elevated the level of ATP, compared to those in the OGD/R groups. Rb1 and Rg1 have different chemical structures, but exert similar protective effects against astrocyte damage induced by OGD/R. The mechanism may be related to improved efficiency of mitochondrial oxidative phosphorylation and the reduction in ROS production in cultured astrocytes.
Collapse
|
37
|
Cruz-Rivera YE, Perez-Morales J, Santiago YM, Gonzalez VM, Morales L, Cabrera-Rios M, Isaza CE. A Selection of Important Genes and Their Correlated Behavior in Alzheimer's Disease. J Alzheimers Dis 2019; 65:193-205. [PMID: 30040709 PMCID: PMC6087431 DOI: 10.3233/jad-170799] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
In 2017, approximately 5 million Americans were living with Alzheimer’s disease (AD), and it is estimated that by 2050 this number could increase to 16 million. In this study, we apply mathematical optimization to approach microarray analysis to detect differentially expressed genes and determine the most correlated structure among their expression changes. The analysis of GSE4757 microarray dataset, which compares expression between AD neurons without neurofibrillary tangles (controls) and with neurofibrillary tangles (cases), was casted as a multiple criteria optimization (MCO) problem. Through the analysis it was possible to determine a series of Pareto efficient frontiers to find the most differentially expressed genes, which are here proposed as potential AD biomarkers. The Traveling Sales Problem (TSP) model was used to find the cyclical path of maximal correlation between the expression changes among the genes deemed important from the previous stage. This leads to a structure capable of guiding biological exploration with enhanced precision and repeatability. Ten genes were selected (FTL, GFAP, HNRNPA3, COX1, ND2, ND3, ND4, NUCKS1, RPL41, and RPS10) and their most correlated cyclic structure was found in our analyses. The biological functions of their products were found to be linked to inflammation and neurodegenerative diseases and some of them had not been reported for AD before. The TSP path connects genes coding for mitochondrial electron transfer proteins. Some of these proteins are closely related to other electron transport proteins already reported as important for AD.
Collapse
Affiliation(s)
- Yazeli E Cruz-Rivera
- The Applied Optimization Group/Department of Industrial Engineering, University of Puerto Rico, Mayagüez Campus, Mayagüez, Puerto Rico
| | - Jaileene Perez-Morales
- Department of Basic Science-Biochemistry Division, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Yaritza M Santiago
- The Applied Optimization Group/Department of Industrial Engineering, University of Puerto Rico, Mayagüez Campus, Mayagüez, Puerto Rico
| | - Valerie M Gonzalez
- The Applied Optimization Group/Department of Industrial Engineering, University of Puerto Rico, Mayagüez Campus, Mayagüez, Puerto Rico
| | - Luisa Morales
- Public Health Program, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Mauricio Cabrera-Rios
- The Applied Optimization Group/Department of Industrial Engineering, University of Puerto Rico, Mayagüez Campus, Mayagüez, Puerto Rico
| | - Clara E Isaza
- The Applied Optimization Group/Department of Industrial Engineering, University of Puerto Rico, Mayagüez Campus, Mayagüez, Puerto Rico.,Public Health Program, Ponce Health Sciences University, Ponce, Puerto Rico
| |
Collapse
|
38
|
Infection by Trypanosoma cruzi in the central nervous system in non-human mammals: a systematic review. Parasitology 2019; 146:983-1005. [PMID: 30873928 DOI: 10.1017/s0031182019000210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Currently, the types and distribution of the lesions induced in the central nervous system (CNS) by Trypanosoma cruzi remain unclear as the available evidence is based on fragmented data. Therefore, we developed a systematic review to analyse the main characteristics of the CNS lesions in non-human hosts infected. From a structured search on the PubMed/Medline and Scopus platforms, 32 studies were retrieved, subjected to data extraction and methodological bias analysis. Our results show that the most frequent alterations in the CNS are the presence of different forms of T. cruzi and intense lymphocytes infiltrates. The encephalon is the main target of T. cruzi, and inflammatory changes in the CNS are more frequent and severe in the acute phase of infection. The parasite's genotype and phenotype are associated with the tropism and severity of the CNS lesions. The methodological limitations found in the studies were divergences in inoculation pathways, under-reporting of animal age and weight, sample calculation strategies and histopathological characterization. Since the changes were dependent on the pathogenicity and virulence of the T. cruzi strains, the genotype and phenotype characterization of the parasite are extremely relevant to predict changes in the CNS and the neurological manifestations associated with Chagas' disease.
Collapse
|
39
|
Mechanisms of Toxicity of Industrially Relevant Silicomanganese Dust on Human 1321N1 Astrocytoma Cells: An In Vitro Study. Int J Mol Sci 2019; 20:ijms20030740. [PMID: 30744184 PMCID: PMC6386893 DOI: 10.3390/ijms20030740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/27/2019] [Accepted: 02/02/2019] [Indexed: 12/19/2022] Open
Abstract
Tremendous efforts are applied in the ferroalloy industry to control and reduce exposure to dust generated during the production process, as inhalable Mn-containing particulate matter has been linked to neurodegenerative diseases. This study aimed to investigate the toxicity and biological effects of dust particles from laboratory-scale processes where molten silicomanganese (SiMn) was exposed to air, using a human astrocytoma cell line, 1321N1, as model system. Characterization of the dust indicated presence of both nano-sized and larger particles averaging between 100 and 300 nm. The dust consisted mainly of Si, Mn and O. Investigation of cellular mechanisms showed a dose- and time-dependent effect on cell viability, with only minor changes in the expression of proteins involved in apoptosis. Moreover, gene expression of the neurotoxic biomarker amyloid precursor protein (APP) increased, whereas APP protein expression decreased. Finally, induction of gap junctional intercellular communication (GJIC) increased with higher doses and correlated with the other endpoints. Thus, the effects of SiMn dust on 1321N1 cells are highly dependent on the dose of exposure and involves changes in APP, apoptosis-related proteins and intercellular communication.
Collapse
|
40
|
Toro-Urrego N, Vesga-Jiménez DJ, Herrera MI, Luaces JP, Capani F. Neuroprotective Role of Hypothermia in Hypoxic-ischemic Brain Injury: Combined Therapies using Estrogen. Curr Neuropharmacol 2019; 17:874-890. [PMID: 30520375 PMCID: PMC7052835 DOI: 10.2174/1570159x17666181206101314] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 10/26/2018] [Accepted: 11/28/2018] [Indexed: 12/15/2022] Open
Abstract
Hypoxic-ischemic brain injury is a complex network of factors, which is mainly characterized by a decrease in levels of oxygen concentration and blood flow, which lead to an inefficient supply of nutrients to the brain. Hypoxic-ischemic brain injury can be found in perinatal asphyxia and ischemic-stroke, which represent one of the main causes of mortality and morbidity in children and adults worldwide. Therefore, knowledge of underlying mechanisms triggering these insults may help establish neuroprotective treatments. Selective Estrogen Receptor Modulators and Selective Tissue Estrogenic Activity Regulators exert several neuroprotective effects, including a decrease of reactive oxygen species, maintenance of cell viability, mitochondrial survival, among others. However, these strategies represent a traditional approach of targeting a single factor of pathology without satisfactory results. Hence, combined therapies, such as the administration of therapeutic hypothermia with a complementary neuroprotective agent, constitute a promising alternative. In this sense, the present review summarizes the underlying mechanisms of hypoxic-ischemic brain injury and compiles several neuroprotective strategies, including Selective Estrogen Receptor Modulators and Selective Tissue Estrogenic Activity Regulators, which represent putative agents for combined therapies with therapeutic hypothermia.
Collapse
Affiliation(s)
- Nicolás Toro-Urrego
- Address correspondence to this author at the Laboratorio de Citoarquitectura y Plasticidad Neuronal, Instituto de Investigaciones Cardiológicas, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina; E-mail:
| | | | | | | | | |
Collapse
|
41
|
Gómez-Pinedo U, Duran-Moreno M, Sirerol-Piquer S, Matias-Guiu J. Myelin changes in Alexander disease. NEUROLOGÍA (ENGLISH EDITION) 2018. [DOI: 10.1016/j.nrleng.2017.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
42
|
Ng YP, Yip TF, Peiris JSM, Ip NY, Lee SMY. Avian influenza A H7N9 virus infects human astrocytes and neuronal cells and induces inflammatory immune responses. J Neurovirol 2018; 24:752-760. [PMID: 29987581 PMCID: PMC7094989 DOI: 10.1007/s13365-018-0659-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 06/05/2018] [Accepted: 06/25/2018] [Indexed: 11/05/2022]
Abstract
Seasonal, pandemic, and avian influenza virus infections may be associated with central nervous system pathology, albeit with varying frequency and different mechanisms. Here, we demonstrate that differentiated human astrocytic (T98G) and neuronal (SH-SY5Y) cells can be infected by avian H7N9 and pandemic H1N1 viruses. However, infectious progeny viruses can only be detected in H7N9 virus infected human neuronal cells. Neither of these viral strains can generate infectious progeny virus in human astrocytes despite replication of viral genome was observed. Furthermore, H7N9 virus triggered high pro-inflammatory cytokine expression, while pandemic H1N1 virus induced only low cytokine expression in either brain cell type. The experimental finding here is the first data to demonstrate that avian H7N9 virus can infect, transcribe, and replicate its viral genome; induce cytokine upregulation; and cause cytopathic effects in human brain cells, which may potentially lead to profound central nervous system injury. Observation for neurological problems due to H7N9 virus infection deserves further attention when managing these patients.
Collapse
Affiliation(s)
- Y P Ng
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - T F Yip
- HKU-Pasteur Research Pole and Centre of Influenza Research, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - J S Malik Peiris
- HKU-Pasteur Research Pole and Centre of Influenza Research, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Nancy Y Ip
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Suki M Y Lee
- HKU-Pasteur Research Pole and Centre of Influenza Research, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
43
|
Wang Y, Qin Y, Guo T, Tang C, Liu L, Gao D. High Concentration of Glial Cell Line-Derived Neurotrophic Factor Protects Primary Astrocytes from Apoptosis. Dev Neurosci 2018; 40:134-144. [DOI: 10.1159/000487853] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 02/20/2018] [Indexed: 12/18/2022] Open
Abstract
Background: Studies have shown that astrocytes play an important role in a variety of biological processes, so damage to astrocytes can cause a series of related diseases. Glial cell line-derived neurotrophic factor (GDNF) has always been considered a protective factor for dopamine neurons. However, it remains unclear whether GDNF has a protective effect on glial cells, especially astrocytes. In this study, we put forward the hypothesis that a high concentration of GDNF in the microenvironment of astrocytes exerts an inhibitory effect on the apoptosis of astrocytes by DNA-damaging reagents. Methods: We isolated, purified, and identified primary astrocytes from neonate rats. Astrocytes were exposed to mitoxantrone (MTN, a DNA-damaging compound) for 24 h. The effects of MTN on astrocytes were tested by Hoechst 33342 staining, CCK-8 assay, and flow cytometry assay. One of the concentrations of MTN was applied to construct an apoptotic model of astrocytes. The astrocytes were then treated with GDNF together with a selected concentration of MTN for 24 h. The cell viability, cell nucleus morphology, and apoptosis ratio of the cells was assessed by Hoechst 33342 staining, CCK-8 assay, and flow cytometry assay, respectively. RNA sequencing (RNA-Seq), quantitative PCR analysis, and KEGG pathway mapping were performed to examine the genes involved in the procedure. Finally, Western blot analysis was applied to confirm the expression levels of the proteins of interest. Results: Hoechst 33342 staining revealed a one-tenth change in the percentage of Hoechst-positive cells after the addition of 500 ng/mL GDNF combined with 1,000 nM MTN for 24 h. The viability of the cells treated the same as described above was 1.4-fold that of the control group. Flow cytometry assays indicated that the apoptotic rates were 17.67, 8.67, and 4.34% for 0, 200, and 500 ng/mL GDNF, respectively. Birc2, Birc3, and Gadd45b were linked to the antiapoptotic process induced by GDNF in astrocytes. Western blot analysis confirmed the elevated expression of Birc2 and Gadd45b. Conclusions: Our studies revealed that GDNF has a noticeable antiapoptotic effect on gene-injured astrocytes. This may provide critical clues for the treatment of a series of diseases in which damaged astrocytes are involved.
Collapse
|
44
|
Fernández-Moriano C, Divakar PK, Crespo A, Gómez-Serranillos MP. Protective effects of lichen metabolites evernic and usnic acids against redox impairment-mediated cytotoxicity in central nervous system-like cells. Food Chem Toxicol 2017; 105:262-277. [PMID: 28450128 DOI: 10.1016/j.fct.2017.04.030] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 04/10/2017] [Accepted: 04/23/2017] [Indexed: 01/08/2023]
Abstract
Lichens species produce unique secondary metabolites that attract increasing pharmacological interest, including their redox modulatory activities. Current work evaluated for the first time the in vitro cytoprotective properties, based on the antioxidant activities, of the Parmeliaceae lichens Evernia prunastri and Usnea ghattensis and the mechanism of action of their major phenolic constituents: the evernic and usnic acids, respectively. In two models of central nervous system-like cells (U373-MG and SH-SY5Y cell lines), exogenous H2O2 induced oxidative stress-mediated cytotoxicity. We first assessed their radical scavenging capacities (ORAC and DPPH tests) and the phenolic content of the extracts. At the optimal concentrations, pretreatments with evernic acid displayed significant protection against H2O2-induced cytotoxic damage in both models. It reversed the alterations in oxidative stress markers (including ROS generation, glutathione system and lipid peroxidation levels) and cellular apoptosis (caspase-3 activity). Such effects were in part mediated by a notable enhancement of the expression of intracellular phase-II antioxidant enzymes; a plausible involvement of the Nrf2 cytoprotective pathway is suggested. Usnic acid exerted similar effects, to some extent more moderate. Results suggest that lichen polyketides evernic and usnic acids merit further research as promising antioxidant candidates in the therapy of oxidative stress-related diseases, including the neurodegenerative disorders.
Collapse
Affiliation(s)
- Carlos Fernández-Moriano
- Department of Pharmacology, Faculty of Pharmacy, University Complutense of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain
| | - Pradeep Kumar Divakar
- Department of Plant Biology II, Faculty of Pharmacy, University Complutense of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain
| | - Ana Crespo
- Department of Plant Biology II, Faculty of Pharmacy, University Complutense of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain
| | - M Pilar Gómez-Serranillos
- Department of Pharmacology, Faculty of Pharmacy, University Complutense of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain.
| |
Collapse
|
45
|
Gómez-Pinedo U, Duran-Moreno M, Sirerol-Piquer S, Matias-Guiu J. Myelin changes in Alexander disease. Neurologia 2017; 33:526-533. [PMID: 28342553 DOI: 10.1016/j.nrl.2017.01.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 01/26/2017] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION Alexander disease (AxD) is a type of leukodystrophy. Its pathological basis, along with myelin loss, is the appearance of Rosenthal bodies, which are cytoplasmic inclusions in astrocytes. Mutations in the gene coding for GFAP have been identified as a genetic basis for AxD. However, the mechanism by which these variants produce the disease is not understood. DEVELOPMENT The most widespread hypothesis is that AxD develops when a gain of function mutation causes an increase in GFAP. However, this mechanism does not explain myelin loss, given that experimental models in which GFAP expression is normal or mutated do not exhibit myelin disorders. This review analyses other possibilities that may explain this alteration, such as epigenetic or inflammatory alterations, presence of NG2 (+) - GFAP (+) cells, or post-translational modifications in GFAP that are unrelated to increased expression. CONCLUSIONS The different hypotheses analysed here may explain the myelin alteration affecting these patients, and multiple mechanisms may coexist. These theories raise the possibility of designing therapies based on these mechanisms.
Collapse
Affiliation(s)
- U Gómez-Pinedo
- Laboratorio de Neurobiología, Servicio de Neurología, Instituto de Neurociencias, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, España.
| | - M Duran-Moreno
- Laboratorio de Neurobiología Comparada, Instituto Cavanilles de Biodiversidad y Biología Evolutiva, Universidad de Valencia, Valencia, España
| | - S Sirerol-Piquer
- Laboratorio de Neurobiología Comparada, Instituto Cavanilles de Biodiversidad y Biología Evolutiva, Universidad de Valencia, Valencia, España
| | - J Matias-Guiu
- Laboratorio de Neurobiología, Servicio de Neurología, Instituto de Neurociencias, IdISSC, Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, España
| |
Collapse
|
46
|
Martín-Jiménez CA, Salazar-Barreto D, Barreto GE, González J. Genome-Scale Reconstruction of the Human Astrocyte Metabolic Network. Front Aging Neurosci 2017; 9:23. [PMID: 28243200 PMCID: PMC5303712 DOI: 10.3389/fnagi.2017.00023] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 01/27/2017] [Indexed: 12/22/2022] Open
Abstract
Astrocytes are the most abundant cells of the central nervous system; they have a predominant role in maintaining brain metabolism. In this sense, abnormal metabolic states have been found in different neuropathological diseases. Determination of metabolic states of astrocytes is difficult to model using current experimental approaches given the high number of reactions and metabolites present. Thus, genome-scale metabolic networks derived from transcriptomic data can be used as a framework to elucidate how astrocytes modulate human brain metabolic states during normal conditions and in neurodegenerative diseases. We performed a Genome-Scale Reconstruction of the Human Astrocyte Metabolic Network with the purpose of elucidating a significant portion of the metabolic map of the astrocyte. This is the first global high-quality, manually curated metabolic reconstruction network of a human astrocyte. It includes 5,007 metabolites and 5,659 reactions distributed among 8 cell compartments, (extracellular, cytoplasm, mitochondria, endoplasmic reticle, Golgi apparatus, lysosome, peroxisome and nucleus). Using the reconstructed network, the metabolic capabilities of human astrocytes were calculated and compared both in normal and ischemic conditions. We identified reactions activated in these two states, which can be useful for understanding the astrocytic pathways that are affected during brain disease. Additionally, we also showed that the obtained flux distributions in the model, are in accordance with literature-based findings. Up to date, this is the most complete representation of the human astrocyte in terms of inclusion of genes, proteins, reactions and metabolic pathways, being a useful guide for in-silico analysis of several metabolic behaviors of the astrocyte during normal and pathologic states.
Collapse
Affiliation(s)
- Cynthia A Martín-Jiménez
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana Bogotá, Colombia
| | - Diego Salazar-Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana Bogotá, Colombia
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad JaverianaBogotá, Colombia; Instituto de Ciencias Biomédicas, Universidad Autónoma de ChileSantiago, Chile
| | - Janneth González
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana Bogotá, Colombia
| |
Collapse
|
47
|
Relationship Between Obesity, Alzheimer’s Disease, and Parkinson’s Disease: an Astrocentric View. Mol Neurobiol 2016; 54:7096-7115. [PMID: 27796748 DOI: 10.1007/s12035-016-0193-8] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 10/03/2016] [Indexed: 12/13/2022]
|
48
|
Abstract
Astrocytes are the most explored non-neuronal cells in the brain under neurophysiological and neurodegenerative conditions. Extensive research has been done to understand their specific role during neuropathological conditions but still the existing findings could not conclude their mechanism of action and their specific role in neurodegenerative conditions. This review discusses their physiological and pathological roles, their activation, morphological alterations and their probable use in search of new therapeutic targets for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sarika Singh
- a 1 Toxicology Division, CSIR-CDRI , Lucknow , India.,b 2 Department of Biochemistry and Biophysics , University of California , San Francisco, San Francisco , CA , USA
| | - Neeraj Joshi
- a 1 Toxicology Division, CSIR-CDRI , Lucknow , India.,b 2 Department of Biochemistry and Biophysics , University of California , San Francisco, San Francisco , CA , USA
| |
Collapse
|
49
|
Vartak-Sharma N, Nooka S, Ghorpade A. Astrocyte elevated gene-1 (AEG-1) and the A(E)Ging HIV/AIDS-HAND. Prog Neurobiol 2016; 157:133-157. [PMID: 27090750 DOI: 10.1016/j.pneurobio.2016.03.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 03/11/2016] [Accepted: 03/19/2016] [Indexed: 12/23/2022]
Abstract
Recent attempts to analyze human immunodeficiency virus (HIV)-1-induced gene expression changes in astrocytes uncovered a multifunctional oncogene, astrocyte elevated gene-1 (AEG-1). Our previous studies revealed that AEG-1 regulates reactive astrocytes proliferation, migration and inflammation, hallmarks of aging and CNS injury. Moreover, the involvement of AEG-1 in neurodegenerative disorders, such as Huntington's disease and migraine, and its induction in the aged brain suggest a plausible role in regulating overall CNS homeostasis and aging. Therefore, it is important to investigate AEG-1 specifically in aging-associated cognitive decline. In this study, we decipher the common mechanistic links in cancer, aging and HIV-1-associated neurocognitive disorders that likely contribute to AEG-1-based regulation of astrocyte responses and function. Despite AEG-1 incorporation into HIV-1 virions and its induction by HIV-1, tumor necrosis factor-α and interleukin-1β, the specific role(s) of AEG-1 in astrocyte-driven HIV-1 neuropathogenesis are incompletely defined. We propose that AEG-1 plays a central role in a multitude of cellular stress responses involving mitochondria, endoplasmic reticulum and the nucleolus. It is thus important to further investigate AEG-1-based cellular and molecular regulation in order to successfully develop better therapeutic approaches that target AEG-1 to combat cancer, HIV-1 and aging.
Collapse
Affiliation(s)
- Neha Vartak-Sharma
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, TX, 76107-2699, USA; Institute for Integrated Cell-Material Sciences, Kyoto University, Japan; Institute for Stem Cell Research and Regenerative Medicine, National Center for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - Shruthi Nooka
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, TX, 76107-2699, USA
| | - Anuja Ghorpade
- Department of Cell Biology and Immunology, University of North Texas Health Science Center, Fort Worth, TX, 76107-2699, USA.
| |
Collapse
|
50
|
Pearce WJ. In cerebrovascular circadian rhythms, EETs keep the beat. Focus on "Rhythmic expression of cytochrome P450 epoxygenases CYP4x1 and CYP2c11 in the rat brain and vasculature". Am J Physiol Cell Physiol 2014; 307:C986-8. [PMID: 25273881 DOI: 10.1152/ajpcell.00327.2014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- William J Pearce
- Loma Linda University School of Medicine, Loma Linda, California
| |
Collapse
|