1
|
Sapkota A, Halder SK, Milner R. Evaluating Matrix Metalloproteinase Expression in Primary Mouse Glial Cultures Using Gel Zymography. Methods Mol Biol 2025; 2918:73-88. [PMID: 40261615 DOI: 10.1007/978-1-0716-4482-9_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Microglia and astrocytes are major glial cell types in the central nervous system (CNS). Under normal conditions, they play a variety of supportive and protective functions, but when inappropriately stimulated by excessive or chronic stimuli, they can become detrimental to CNS health, both in their own effector functions and by orchestrating inflammatory processes. Microglia and astrocytes both produce matrix metalloproteinases (MMPs), which play important roles in physiological and pathological processes. To characterize MMP expression in glial cells, and to gain insights into the roles they play in regulating tissue repair and remodeling, we employ the method of gel zymography to quantify levels of MMP-2 and MMP-9, as well as their inactive pro-forms. This technique is relatively easy and simple to perform and provides quick and quantifiable data. In this chapter, we describe this approach to analyze MMP expression in supernatants from primary mouse cultures of microglia and astrocytes.
Collapse
Affiliation(s)
- Arjun Sapkota
- San Diego Biomedical Research Institute, San Diego, CA, USA
| | - Sebok K Halder
- San Diego Biomedical Research Institute, San Diego, CA, USA
| | - Richard Milner
- San Diego Biomedical Research Institute, San Diego, CA, USA.
| |
Collapse
|
2
|
Radosinska D, Radosinska J. The Link Between Matrix Metalloproteinases and Alzheimer's Disease Pathophysiology. Mol Neurobiol 2025; 62:885-899. [PMID: 38935232 PMCID: PMC11711632 DOI: 10.1007/s12035-024-04315-0] [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: 01/10/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024]
Abstract
Alzheimer's disease (AD) is a major contributor to dementia and the most common neurodegenerative disorder. In AD pathophysiology, matrix metalloproteinases (MMPs)-proteolytic enzymes, best known to be responsible for remodeling and degradation of the extracellular matrix-were suggested to play an important role. Due to the diverse nature of the published data and frequent inconsistent results presented in available papers, it was considered essential to analyze all aspects of MMP literature with respect to AD pathophysiology and attempt to outline a unifying concept for understanding their role in AD. Thus, the main contribution of this review article is to summarize the most recent research on the participation of MMP in AD pathophysiology obtained using the cell cultures to understand the molecular principles of their action. Furthermore, an updated comprehensive view regarding this topic based exclusively on papers from human studies is provided as well. It can be concluded that determining the exact role of any particular MMPs in the AD pathophysiology holds promise for establishing their role as potential biomarkers reflecting the severity or progression of this disease or for developing new therapeutic agents targeting the processes that lead to AD.
Collapse
Affiliation(s)
- Dominika Radosinska
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University in Bratislava, Bratislava, Slovak Republic
| | - Jana Radosinska
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, Sasinkova 2, 81372, Bratislava, Slovak Republic.
| |
Collapse
|
3
|
Amontree M, Nelson M, Stefansson L, Pak D, Maguire-Zeiss K, Turner RS, Conant K. Resveratrol differentially affects MMP-9 release from neurons and glia; implications for therapeutic efficacy. J Neurochem 2024; 168:1895-1908. [PMID: 38163875 DOI: 10.1111/jnc.16031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/03/2023] [Accepted: 11/21/2023] [Indexed: 01/03/2024]
Abstract
Resveratrol, a naturally occurring polyphenol that activates sirtuin 1 (SIRT1), has been shown to reduce overall levels of matrix metalloprotease-9 (MMP-9) in cerebrospinal fluid (CSF) samples from patients with Alzheimer's dementia (AD). Depending on the site of release, however, MMP-9 has the potential to improve or impair cognition. In particular, its release from microglia or pericytes proximal to the blood brain barrier can damage the basement membrane, while neuronal activity-dependent release of this protease from glutamatergic neurons can instead promote dendritic spine expansion and long-term potentiation of synaptic plasticity. In the present study, we test the hypothesis that resveratrol reduces overall MMP-9 levels in CSF samples from patients with APOE4, an allele associated with increased glial inflammation. We also examine the possibility that resveratrol reduces inflammation-associated MMP release from cultured glia but spares neuronal activity-dependent release from cultured cortical neurons. We observe that resveratrol decreases overall levels of MMP-2 and MMP-9 in CSF samples from AD patients. Resveratrol also reduces CSF levels of tissue inhibitor of metalloproteinases-1 (TIMP-1), glial-derived protein that restricts long-term potentiation of synaptic transmission, in individuals homozygous for APOE4. Consistent with these results, we observe that resveratrol reduces basal and lipopolysaccharide (LPS)-stimulated MMP and TIMP-1 release from cultured microglia and astrocytes. In contrast, however, resveratrol does not inhibit release of MMP-9 from cortical neurons. Overall, these results are consistent with the possibility that while resveratrol reduces potentially maladaptive MMP and TIMP-1 release from activated glia, neuroplasticity-promoting MMP release from neurons is spared. In contrast, resveratrol reduces release of neurocan and brevican, extracellular matrix components that restrict neuroplasticity, from both neurons and glia. These data underscore the diversity of resveratrol's actions with respect to affected cell types and molecular targets and also suggest that further studies may be warranted to determine if its effects on glial MMP release could make it a useful adjunct for AD- and/or anti-amyloid therapy-related damage to the blood brain barrier.
Collapse
Affiliation(s)
- Matthew Amontree
- Department of Neuroscience, Georgetown University Medical Center, Washington, District of Columbia, USA
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, District of Columbia, USA
| | - Matthew Nelson
- Department of Neuroscience, Georgetown University Medical Center, Washington, District of Columbia, USA
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, District of Columbia, USA
| | - Lara Stefansson
- Department of Neuroscience, Georgetown University Medical Center, Washington, District of Columbia, USA
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, District of Columbia, USA
| | - Daniel Pak
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, District of Columbia, USA
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Kathleen Maguire-Zeiss
- Department of Neuroscience, Georgetown University Medical Center, Washington, District of Columbia, USA
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, District of Columbia, USA
- Department of Biology, Georgetown University, Washington, District of Columbia, USA
| | - R Scott Turner
- Department of Neurology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Katherine Conant
- Department of Neuroscience, Georgetown University Medical Center, Washington, District of Columbia, USA
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, District of Columbia, USA
| |
Collapse
|
4
|
Jiang F, Xu C, Fan X, Yang S, Fan W, Li M, Song J, Wei W, Chen H, Zhong D, Li G. MyD88 Inhibition Attenuates Cerebral Ischemia-reperfusion Injury by Regulating the Inflammatory Response and Reducing Blood-brain Barrier Damage. Neuroscience 2024; 549:121-137. [PMID: 38754722 DOI: 10.1016/j.neuroscience.2024.05.010] [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: 12/30/2023] [Revised: 05/01/2024] [Accepted: 05/09/2024] [Indexed: 05/18/2024]
Abstract
Myeloid differentiation primary response gene 88 (MyD88), a downstream molecule directly linked to Toll-like receptor (TLRs) and IL1 receptor, has been implicated in ischemia-reperfusion injury across various organs. However, its role in cerebral ischemia-reperfusion injury (CIRI) remains unclear. Five transient middle cerebral artery occlusion (tMCAO) microarray datasets were obtained from the Gene Expression Omnibus (GEO) database. We screened these datasets for differentially expressed genes (DEGs) using the GSE35338 and GSE58720 datasets and performed weighted gene co-expression network analysis (WGCNA) using the GSE30655, GSE28731, and GSE32529 datasets to identify the core module related to tMCAO. A protein-protein interaction (PPI) network was constructed using the intersecting DEGs and genes in the core module. Finally, we identified Myd88 was the core gene. In addition, Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Set Enrichment Analysis (GSEA) validated that TNFα, IL17, and MyD88 signaling pathways were significantly enriched in tMCAO. Subsequently, we investigated the mechanistic role of MyD88 in the tMCAO model using male C57BL/6 mice. MyD88 expression increased significantly 24 h after reperfusion. After intraperitoneal administration of TJ-M2010-5, a MyD88-specific inhibitor, during reperfusion, the infarction volumes in the mice were ameliorated. TJ-M2010-5 inhibits the activation of microglia and astrocytes. Moreover, it attenuates the upregulation of inflammatory cytokines TNFα, IL17, and MMP9 while preserving the expression level of ZO1 after tMCAO, thereby safeguarding against blood-brain barrier (BBB) disruption. Finally, our findings suggest that MyD88 regulates the IRAK4/IRF5 signaling pathway associated with microglial activation. MyD88 participates in CIRI by regulating the inflammatory response and BBB damage following tMCAO.
Collapse
Affiliation(s)
- Fangchao Jiang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chen Xu
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xuehui Fan
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuai Yang
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wei Fan
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Meng Li
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jihe Song
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wan Wei
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongping Chen
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Di Zhong
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Guozhong Li
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Department of Neurology, Heilongjiang Provincial Hospital, Harbin, China.
| |
Collapse
|
5
|
Zhao Y, Yang Y, Yang R, Sun C, Gao X, Gu X, Yuan Y, Nie Y, Xu S, Han R, Zhang L, Li J, Hu P, Wang Y, Chen H, Cao X, Wu J, Wang Z, Gu Y, Ye J. IDH1 mutation inhibits differentiation of astrocytes and glioma cells with low oxoglutarate dehydrogenase expression by disturbing α-ketoglutarate-related metabolism and epigenetic modification. LIFE METABOLISM 2024; 3:loae002. [PMID: 39872214 PMCID: PMC11749698 DOI: 10.1093/lifemeta/loae002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 01/03/2024] [Accepted: 01/13/2024] [Indexed: 01/30/2025]
Abstract
Isocitrate dehydrogenase (IDH) mutations frequently occur in lower-grade gliomas and secondary glioblastomas. Mutant IDHs exhibit a gain-of-function activity, leading to the production of D-2-hydroxyglutarate (D-2HG) by reducing α-ketoglutarate (α-KG), a central player in metabolism and epigenetic modifications. However, the role of α-KG homeostasis in IDH-mutated gliomagenesis remains elusive. In this study, we found that low expression of oxoglutarate dehydrogenase (OGDH) was a common feature in IDH-mutated gliomas, as well as in astrocytes. This low expression of OGDH resulted in the accumulation of α-KG and promoted astrocyte maturation. However, IDH1 mutation significantly reduced α-KG levels and increased glutaminolysis and DNA/histone methylation in astrocytes. These metabolic and epigenetic alterations inhibited astrocyte maturation and led to cortical dysplasia in mice. Moreover, our results also indicated that reduced OGDH expression can promote the differentiation of glioma cells, while IDH1 mutations impeded the differentiation of glioma cells with low OGDH by reducing the accumulation of α-KG and increasing glutaminolysis. Finally, we found that l-glutamine increased α-KG levels and augmented the differentiation-promoting effects of AGI5198, an IDH1-mutant inhibitor, in IDH1-mutant glioma cells. Collectively, this study reveals that low OGDH expression is a crucial metabolic characteristic of IDH-mutant gliomas, providing a potential strategy for the treatment of IDH-mutant gliomas by targeting α-KG homeostasis.
Collapse
Affiliation(s)
- Yuanlin Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Ying Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Risheng Yang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
- Department of Pathology, Air Force Hospital of Southern Theater Command, Guangzhou, Guangdong 510000, China
| | - Chao Sun
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi 710038, China
| | - Xing Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Xiwen Gu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Yuan Yuan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Yating Nie
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Shenhui Xu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Ruili Han
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Lijun Zhang
- Department of Clinical Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi’an, Shaanxi 710038, China
| | - Jing Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Peizhen Hu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Yingmei Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Huangtao Chen
- Department of Neurosurgery, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710004, China
| | - Xiangmei Cao
- Department of Pathology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, China
| | - Jing Wu
- Institute of Analytical Chemistry and Instrument for Life Science, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, Shaanxi 710049, China
| | - Zhe Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Yu Gu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Jing Ye
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| |
Collapse
|
6
|
Yonesi M, Ramos M, Ramirez-Castillejo C, Fernández-Serra R, Panetsos F, Belarra A, Chevalier M, Rojo FJ, Pérez-Rigueiro J, Guinea GV, González-Nieto D. Resistance to Degradation of Silk Fibroin Hydrogels Exposed to Neuroinflammatory Environments. Polymers (Basel) 2023; 15:polym15112491. [PMID: 37299290 DOI: 10.3390/polym15112491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/18/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
Central nervous system (CNS) diseases represent an extreme burden with significant social and economic costs. A common link in most brain pathologies is the appearance of inflammatory components that can jeopardize the stability of the implanted biomaterials and the effectiveness of therapies. Different silk fibroin scaffolds have been used in applications related to CNS disorders. Although some studies have analyzed the degradability of silk fibroin in non-cerebral tissues (almost exclusively upon non-inflammatory conditions), the stability of silk hydrogel scaffolds in the inflammatory nervous system has not been studied in depth. In this study, the stability of silk fibroin hydrogels exposed to different neuroinflammatory contexts has been explored using an in vitro microglial cell culture and two in vivo pathological models of cerebral stroke and Alzheimer's disease. This biomaterial was relatively stable and did not show signs of extensive degradation across time after implantation and during two weeks of in vivo analysis. This finding contrasted with the rapid degradation observed under the same in vivo conditions for other natural materials such as collagen. Our results support the suitability of silk fibroin hydrogels for intracerebral applications and highlight the potentiality of this vehicle for the release of molecules and cells for acute and chronic treatments in cerebral pathologies.
Collapse
Affiliation(s)
- Mahdi Yonesi
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Spain
| | - Milagros Ramos
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Spain
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Carmen Ramirez-Castillejo
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Spain
| | - Rocío Fernández-Serra
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Spain
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Silk Biomed SL, Calle Navacerrada 18, Urb. Puerto Galapagar, 28260 Madrid, Spain
| | - Fivos Panetsos
- Silk Biomed SL, Calle Navacerrada 18, Urb. Puerto Galapagar, 28260 Madrid, Spain
- Bioactive Surfaces SL, Puerto de Navacerrada 18. Galapagar, 28260 Madrid, Spain
- Neurocomputing and Neurorobotics Research Group, Faculty of Biology and Faculty of Optics, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Biomaterials and Regenerative Medicine Group, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Calle Prof. Martín Lagos s/n, 28040 Madrid, Spain
| | - Adrián Belarra
- Laboratorio Micro-CT UCM, Departamento de Radiología, Rehabilitación y Fisioterapia, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Margarita Chevalier
- Laboratorio Micro-CT UCM, Departamento de Radiología, Rehabilitación y Fisioterapia, Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Francisco J Rojo
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Silk Biomed SL, Calle Navacerrada 18, Urb. Puerto Galapagar, 28260 Madrid, Spain
- Bioactive Surfaces SL, Puerto de Navacerrada 18. Galapagar, 28260 Madrid, Spain
- Biomaterials and Regenerative Medicine Group, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Calle Prof. Martín Lagos s/n, 28040 Madrid, Spain
| | - José Pérez-Rigueiro
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Silk Biomed SL, Calle Navacerrada 18, Urb. Puerto Galapagar, 28260 Madrid, Spain
- Bioactive Surfaces SL, Puerto de Navacerrada 18. Galapagar, 28260 Madrid, Spain
- Biomaterials and Regenerative Medicine Group, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Calle Prof. Martín Lagos s/n, 28040 Madrid, Spain
| | - Gustavo V Guinea
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Silk Biomed SL, Calle Navacerrada 18, Urb. Puerto Galapagar, 28260 Madrid, Spain
- Bioactive Surfaces SL, Puerto de Navacerrada 18. Galapagar, 28260 Madrid, Spain
- Biomaterials and Regenerative Medicine Group, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Calle Prof. Martín Lagos s/n, 28040 Madrid, Spain
| | - Daniel González-Nieto
- Center for Biomedical Technology, Universidad Politécnica de Madrid, 28223 Pozuelo de Alarcón, Spain
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Silk Biomed SL, Calle Navacerrada 18, Urb. Puerto Galapagar, 28260 Madrid, Spain
- Bioactive Surfaces SL, Puerto de Navacerrada 18. Galapagar, 28260 Madrid, Spain
| |
Collapse
|
7
|
Baričević Z, Ayar Z, Leitao SM, Mladinic M, Fantner GE, Ban J. Label-Free Long-Term Methods for Live Cell Imaging of Neurons: New Opportunities. BIOSENSORS 2023; 13:404. [PMID: 36979616 PMCID: PMC10046152 DOI: 10.3390/bios13030404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 06/18/2023]
Abstract
Time-lapse light microscopy combined with in vitro neuronal cultures has provided a significant contribution to the field of Developmental Neuroscience. The establishment of the neuronal polarity, i.e., formation of axons and dendrites, key structures responsible for inter-neuronal signaling, was described in 1988 by Dotti, Sullivan and Banker in a milestone paper that continues to be cited 30 years later. In the following decades, numerous fluorescently labeled tags and dyes were developed for live cell imaging, providing tremendous advancements in terms of resolution, acquisition speed and the ability to track specific cell structures. However, long-term recordings with fluorescence-based approaches remain challenging because of light-induced phototoxicity and/or interference of tags with cell physiology (e.g., perturbed cytoskeletal dynamics) resulting in compromised cell viability leading to cell death. Therefore, a label-free approach remains the most desirable method in long-term imaging of living neurons. In this paper we will focus on label-free high-resolution methods that can be successfully used over a prolonged period. We propose novel tools such as scanning ion conductance microscopy (SICM) or digital holography microscopy (DHM) that could provide new insights into live cell dynamics during neuronal development and regeneration after injury.
Collapse
Affiliation(s)
- Zrinko Baričević
- Department of Biotechnology, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia; (Z.B.); (M.M.)
| | - Zahra Ayar
- Laboratory for Bio- and Nano-Instrumentation, Institute of Bioengineering, School of Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), 1015 Lausanne, Switzerland; (Z.A.); (S.M.L.)
| | - Samuel M. Leitao
- Laboratory for Bio- and Nano-Instrumentation, Institute of Bioengineering, School of Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), 1015 Lausanne, Switzerland; (Z.A.); (S.M.L.)
| | - Miranda Mladinic
- Department of Biotechnology, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia; (Z.B.); (M.M.)
| | - Georg E. Fantner
- Laboratory for Bio- and Nano-Instrumentation, Institute of Bioengineering, School of Engineering, Swiss Federal Institute of Technology Lausanne (EPFL), 1015 Lausanne, Switzerland; (Z.A.); (S.M.L.)
| | - Jelena Ban
- Department of Biotechnology, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia; (Z.B.); (M.M.)
| |
Collapse
|
8
|
Zelenka L, Pägelow D, Krüger C, Seele J, Ebner F, Rausch S, Rohde M, Lehnardt S, van Vorst K, Fulde M. Novel protocol for the isolation of highly purified neonatal murine microglia and astrocytes. J Neurosci Methods 2022; 366:109420. [PMID: 34808220 DOI: 10.1016/j.jneumeth.2021.109420] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/02/2021] [Accepted: 11/11/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND The crosstalk and reactivity of the cell type glia, especially microglia and astrocytes, have progressively gathered research attention in understanding proper brain function regulated by the innate immune response. Therefore, methods to isolate highly viable and pure glia for the analysis on a cell-specific level are indispensable. NEW METHOD We modified previously established techniques: Animal numbers were reduced by multiple microglial harvests from the same mixed glial culture, thereby maximizing microglial yields following the principles of the 3Rs (replacement, reduction, and refinement). We optimized Magnetic-activated cell sorting (MACS®) of microglia and astrocytes by applying cultivated primary glial cell suspensions instead of directly sorting dissociated single cell suspension. RESULTS We generated highly viable and pure microglia and astrocytes derived from a single mixed culture with a purity of ~99%, as confirmed by FACS analysis. Field emission scanning electron microscopy (FESEM) demonstrated integrity of the MACS-purified glial cells. Tumor necrosis factor (TNF) and Interleukin-10 (IL-10) ELISA confirmed pro- and anti-inflammatory responses to be functional in purified glia, but significantly weakened compared to non-purified cells, further highlighting the importance of cellular crosstalk for proper immune activation. COMPARISON WITH EXISTING METHOD(S) Unlike previous studies that either isolated a single type of glia or displayed a substantial proportion of contamination with other cell types, we achieved isolation of both microglia and astrocytes at an increased purity (99-100%). CONCLUSIONS We have created an optimized protocol for the efficient purification of both primary microglia and astrocytes. Our results clearly demonstrate the importance of purity in glial cell cultivation in order to examine immune responses, which particularly holds true for astrocytes. We propose the novel protocol as a tool to investigate the cell type-specific crosstalk between microglia and astrocytes in the frame of CNS diseases.
Collapse
Affiliation(s)
- Laura Zelenka
- Institute of Microbiology and Epizootics, Centre of Infection Medicine, Freie Universität Berlin, Robert-von-Ostertag-Straße 7-13, 14163 Berlin, Germany
| | - Dennis Pägelow
- Institute of Microbiology and Epizootics, Centre of Infection Medicine, Freie Universität Berlin, Robert-von-Ostertag-Straße 7-13, 14163 Berlin, Germany
| | - Christina Krüger
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jana Seele
- University Medical Center Göttingen, Institute of Neuropathology, Göttingen, Germany
| | - Friederike Ebner
- Freie Universität Berlin, Institute of Immunology, Berlin, Germany
| | - Sebastian Rausch
- Freie Universität Berlin, Institute of Immunology, Berlin, Germany
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Seija Lehnardt
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Department of Neurology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Kira van Vorst
- Institute of Microbiology and Epizootics, Centre of Infection Medicine, Freie Universität Berlin, Robert-von-Ostertag-Straße 7-13, 14163 Berlin, Germany
| | - Marcus Fulde
- Institute of Microbiology and Epizootics, Centre of Infection Medicine, Freie Universität Berlin, Robert-von-Ostertag-Straße 7-13, 14163 Berlin, Germany.
| |
Collapse
|
9
|
Cameron T, Bennet T, Rowe EM, Anwer M, Wellington CL, Cheung KC. Review of Design Considerations for Brain-on-a-Chip Models. MICROMACHINES 2021; 12:441. [PMID: 33921018 PMCID: PMC8071412 DOI: 10.3390/mi12040441] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/12/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023]
Abstract
In recent years, the need for sophisticated human in vitro models for integrative biology has motivated the development of organ-on-a-chip platforms. Organ-on-a-chip devices are engineered to mimic the mechanical, biochemical and physiological properties of human organs; however, there are many important considerations when selecting or designing an appropriate device for investigating a specific scientific question. Building microfluidic Brain-on-a-Chip (BoC) models from the ground-up will allow for research questions to be answered more thoroughly in the brain research field, but the design of these devices requires several choices to be made throughout the design development phase. These considerations include the cell types, extracellular matrix (ECM) material(s), and perfusion/flow considerations. Choices made early in the design cycle will dictate the limitations of the device and influence the end-point results such as the permeability of the endothelial cell monolayer, and the expression of cell type-specific markers. To better understand why the engineering aspects of a microfluidic BoC need to be influenced by the desired biological environment, recent progress in microfluidic BoC technology is compared. This review focuses on perfusable blood-brain barrier (BBB) and neurovascular unit (NVU) models with discussions about the chip architecture, the ECM used, and how they relate to the in vivo human brain. With increased knowledge on how to make informed choices when selecting or designing BoC models, the scientific community will benefit from shorter development phases and platforms curated for their application.
Collapse
Affiliation(s)
- Tiffany Cameron
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.C.); (T.B.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Tanya Bennet
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.C.); (T.B.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Elyn M. Rowe
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (E.M.R.); (M.A.); (C.L.W.)
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Mehwish Anwer
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (E.M.R.); (M.A.); (C.L.W.)
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Cheryl L. Wellington
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (E.M.R.); (M.A.); (C.L.W.)
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Karen C. Cheung
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; (T.C.); (T.B.)
- Centre for Blood Research, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Electrical & Computer Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
10
|
Birck C, Ginolhac A, Pavlou MAS, Michelucci A, Heuschling P, Grandbarbe L. NF-κB and TNF Affect the Astrocytic Differentiation from Neural Stem Cells. Cells 2021; 10:840. [PMID: 33917855 PMCID: PMC8068246 DOI: 10.3390/cells10040840] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 04/02/2021] [Accepted: 04/06/2021] [Indexed: 01/26/2023] Open
Abstract
The NF-κB signaling pathway is crucial during development and inflammatory processes. We have previously shown that NF-κB activation induces dedifferentiation of astrocytes into neural progenitor cells (NPCs). Here, we provide evidence that the NF-κB pathway plays also a fundamental role during the differentiation of NPCs into astrocytes. First, we show that the NF-κB pathway is essential to initiate astrocytic differentiation as its early inhibition induces NPC apoptosis and impedes their differentiation. Second, we demonstrate that persistent NF-κB activation affects NPC-derived astrocyte differentiation. Tumor necrosis factor (TNF)-treated NPCs show NF-κB activation, maintain their multipotential and proliferation properties, display persistent expression of immature markers and inhibit astrocyte markers. Third, we analyze the effect of NF-κB activation on the main known astrocytic differentiation pathways, such as NOTCH and JAK-STAT. Our findings suggest that the NF-κB pathway plays a dual fundamental role during NPC differentiation into astrocytes: it promotes astrocyte specification, but its persistent activation impedes their differentiation.
Collapse
Affiliation(s)
- Cindy Birck
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Communication, University of Luxembourg, L-1511 Luxembourg, Luxembourg; (C.B.); (A.G.); (M.A.S.P.); (P.H.)
| | - Aurélien Ginolhac
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Communication, University of Luxembourg, L-1511 Luxembourg, Luxembourg; (C.B.); (A.G.); (M.A.S.P.); (P.H.)
| | - Maria Angeliki S. Pavlou
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Communication, University of Luxembourg, L-1511 Luxembourg, Luxembourg; (C.B.); (A.G.); (M.A.S.P.); (P.H.)
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg;
| | - Alessandro Michelucci
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg;
- Neuro-Immunology Group, Department of Oncology, Luxembourg Institute of Health, L-1526 Luxembourg, Luxembourg
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Paul Heuschling
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Communication, University of Luxembourg, L-1511 Luxembourg, Luxembourg; (C.B.); (A.G.); (M.A.S.P.); (P.H.)
| | - Luc Grandbarbe
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Communication, University of Luxembourg, L-1511 Luxembourg, Luxembourg; (C.B.); (A.G.); (M.A.S.P.); (P.H.)
| |
Collapse
|
11
|
Wen SJ, Zheng XM, Liu LF, Li NN, Mao HA, Huang L, Yuan QL. Effects of primary microglia and astrocytes on neural stem cells in in vitro and in vivo models of ischemic stroke. Neural Regen Res 2021; 16:1677-1685. [PMID: 33510055 PMCID: PMC8328755 DOI: 10.4103/1673-5374.306093] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Transplantation of neural stem cells (NSCs) can protect neurons in animal stroke models; however, their low rates of survival and neuronal differentiation limit their clinical application. Glial niches, an important location of neural stem cells, regulate survival, proliferation and differentiation of neural stem cells. However, the effects of activated glial cells on neural stem cells remain unclear. In the present study, we explored the effects of activated astrocytes and microglia on neural stem cells in vitro stroke models. We also investigated the effects of combined transplantation of neural stem cells and glial cells after stroke in rats. In a Transwell co-culture system, primary cultured astrocytes, microglia or mixed glial cells were exposed to glutamate or H2O2 and then seeded in the upper inserts, while primary neural stem cells were seeded in the lower uncoated wells and cultured for 7 days. Our results showed that microglia were conducive to neurosphere formation and had no effects on apoptosis within neurospheres, while astrocytes and mixed glial cells were conducive to neurosphere differentiation and reduced apoptosis within neurospheres, regardless of their pretreatment. In contrast, microglia and astrocytes induced neuronal differentiation of neural stem cells in differentiation medium, regardless of their pretreatment, with an exception of astrocytes pretreated with H2O2. Rat models of ischemic stroke were established by occlusion of the middle cerebral artery. Three days later, 5 × 105 neural stem cells with microglia or astrocytes were injected into the right lateral ventricle. Neural stem cell/astrocyte-treated rats displayed better improvement of neurological deficits than neural stem cell only-treated rats at 4 days after cell transplantation. Moreover, neural stem cell/microglia-, and neural stem cell/astrocyte-treated rats showed a significant decrease in ischemic volume compared with neural stem cell-treated rats. These findings indicate that microglia and astrocytes exert different effects on neural stem cells, and that co-transplantation of neural stem cells and astrocytes is more conducive to the recovery of neurological impairment in rats with ischemic stroke. The study was approved by the Animal Ethics Committee of Tongji University School of Medicine, China (approval No. 2010-TJAA08220401) in 2010.
Collapse
Affiliation(s)
- Sheng-Jun Wen
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xi-Min Zheng
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Li-Fen Liu
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Na-Na Li
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hai-An Mao
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liang Huang
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qiong-Lan Yuan
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
12
|
Kant R, Halder SK, Fernández JA, Griffin JH, Milner R. Activated Protein C Attenuates Experimental Autoimmune Encephalomyelitis Progression by Enhancing Vascular Integrity and Suppressing Microglial Activation. Front Neurosci 2020; 14:333. [PMID: 32351356 PMCID: PMC7174764 DOI: 10.3389/fnins.2020.00333] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 03/20/2020] [Indexed: 12/16/2022] Open
Abstract
Background Activated protein C (APC), a serine protease with antithrombotic effects, protects in animal models of ischemic stroke by suppressing inflammation and enhancing vascular integrity, angiogenesis, neurogenesis and neuroprotection. A small number of animal studies suggest it might also have therapeutic potential in multiple sclerosis (MS), though results have been mixed. Based on these conflicting data, the goals of this study were to clarify the therapeutic potential of APC in the experimental autoimmune encephalomyelitis (EAE) model of MS and to determine mechanistically how APC mediates this protective effect. Methods The protective potential of APC was examined in a chronic progressive model of EAE. Vascular breakdown, tight junction protein expression and vascular expression of fibronectin and α5β1 integrin as well as vascularity and glial activation were analyzed using immunofluorescence (IF) of spinal cord sections taken from mice with established EAE. The direct influence of APC on microglial activation was evaluated in vitro by a combination of morphology and MMP-9 expression. Results APC attenuated the progression of EAE, and this was strongly associated at the histopathological level with reduced levels of leukocyte infiltration and concomitant demyelination. Further analysis revealed that APC reduced vascular breakdown which was associated with maintained endothelial expression of the tight junction protein zonula occludens-1 (ZO-1). In addition, APC suppressed microglial activation in this EAE model and in vitro studies revealed that APC strongly inhibited microglial activation at both the morphological level and by the expression of the pro-inflammatory protease MMP-9. Conclusion These findings build on the work of others in demonstrating strong therapeutic potential for APC in the treatment of inflammatory demyelinating disease and suggest that enhancement of vascular integrity and suppression of microglial activation may be important mediators of this protection.
Collapse
Affiliation(s)
- Ravi Kant
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Sebok K Halder
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Jose A Fernández
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - John H Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Richard Milner
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| |
Collapse
|
13
|
Astrocyte Support for Oligodendrocyte Differentiation can be Conveyed via Extracellular Vesicles but Diminishes with Age. Sci Rep 2020; 10:828. [PMID: 31964978 PMCID: PMC6972737 DOI: 10.1038/s41598-020-57663-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 01/06/2020] [Indexed: 01/06/2023] Open
Abstract
The aging brain is associated with significant changes in physiology that alter the tissue microenvironment of the central nervous system (CNS). In the aged CNS, increased demyelination has been associated with astrocyte hypertrophy and aging has been implicated as a basis for these pathological changes. Aging tissues accumulate chronic cellular stress, which can lead to the development of a pro-inflammatory phenotype that can be associated with cellular senescence. Herein, we provide evidence that astrocytes aged in culture develop a spontaneous pro-inflammatory and senescence-like phenotype. We found that extracellular vesicles (EVs) from young astrocyte were sufficient to convey support for oligodendrocyte differentiation while this support was lost by EVs from aged astrocytes. Importantly, the negative influence of culture age on astrocytes, and their cognate EVs, could be countered by treatment with rapamycin. Comparative proteomic analysis of EVs from young and aged astrocytes revealed peptide repertoires unique to each age. Taken together, these findings provide new information on the contribution of EVs as potent mediators by which astrocytes can extert changing influence in either the disease or aged brain.
Collapse
|
14
|
Tajalli-Nezhad S, Karimian M, Beyer C, Atlasi MA, Azami Tameh A. The regulatory role of Toll-like receptors after ischemic stroke: neurosteroids as TLR modulators with the focus on TLR2/4. Cell Mol Life Sci 2019; 76:523-537. [PMID: 30377701 PMCID: PMC11105485 DOI: 10.1007/s00018-018-2953-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 10/19/2018] [Indexed: 02/07/2023]
Abstract
Ischemic stroke is the most common cerebrovascular disease and considered as a worldwide leading cause of death. After cerebral ischemia, different pathophysiological processes including neuroinflammation, invasion and aggregation of inflammatory cells and up-regulation of cytokines occur simultaneously. In this respect, Toll-like receptors (TLRs) are the first identified important mediators for the activation of the innate immune system and are widely expressed in glial cells and neurons following brain trauma. TLRs are also able to interact with endogenous and exogenous molecules released during ischemia and can increase tissue damage. Particularly, TLR2 and TLR4 activate different downstream inflammatory signaling pathways. In addition, TLR signaling can alternatively play a role for endogenous neuroprotection. In this review, the gene and protein structures, common genetic polymorphisms of TLR2 and TLR4, TLR-related molecular pathways and their putative role after ischemic stroke are delineated. Furthermore, the relationship between neurosteroids and TLRs as neuroprotective mechanism is highlighted in the context of brain ischemia.
Collapse
Affiliation(s)
- Saeedeh Tajalli-Nezhad
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Karimian
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Cordian Beyer
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Mohammad Ali Atlasi
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Abolfazl Azami Tameh
- Anatomical Sciences Research Center, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
15
|
Barca Mayo O, Berdondini L, De Pietri Tonelli D. Astrocytes and Circadian Rhythms: An Emerging Astrocyte-Neuron Synergy in the Timekeeping System. Methods Mol Biol 2019; 1938:131-154. [PMID: 30617978 DOI: 10.1007/978-1-4939-9068-9_10] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Animals have an internal timekeeping system to anticipate daily changes associated with the transition of day to night, which is deeply involved in the regulation and maintenance of behavioral and physiological processes. Prevailing knowledge associated the control of circadian clocks to a network of neurons in the central pacemaker, the suprachiasmatic nucleus (SCN), but astrocytes are rapidly emerging as key cellular contributors to the timekeeping system. However, how these glial cells impact the neuronal clock to modulate rhythmic neurobehavioral outputs just begin to be investigated. Astrocyte-neuron cocultures are an excellent exploratory method to further characterize the critical role of circadian communication between nerve cells, as well as to address the role of astrocytes as modulators and targets of neuronal rhythmic behaviors. Here, we describe a robust method to study astrocyte rhythmic interactions with neurons by coculturing them with primary neurons in physically separated layers. This simple coculture system provides hints on in vivo signaling processes. Moreover, it allows investigating cell-type specific effects separately as well as the identification of extracellular astrocytic or neuronal factors involved in rhythm generation in both cell types.
Collapse
Affiliation(s)
- Olga Barca Mayo
- Neurobiology of miRNAs Laboratory, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy.
| | - Luca Berdondini
- Microtechnology for Neuroelectronics (Nets3) Laboratory, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Davide De Pietri Tonelli
- Neurobiology of miRNAs Laboratory, Neuroscience and Brain Technologies Department, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| |
Collapse
|
16
|
Facci L, Barbierato M, Zusso M, Skaper SD, Giusti P. Serum amyloid A primes microglia for ATP-dependent interleukin-1β release. J Neuroinflammation 2018; 15:164. [PMID: 29803222 PMCID: PMC5970445 DOI: 10.1186/s12974-018-1205-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/15/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Acute-phase response is a systemic reaction to environmental/inflammatory insults and involves production of acute-phase proteins, including serum amyloid A (SAA). Interleukin-1β (IL-1β), a master regulator of neuroinflammation produced by activated inflammatory cells of the myeloid lineage, in particular microglia, plays a key role in the pathogenesis of acute and chronic diseases of the peripheral nervous system and CNS. IL-1β release is promoted by ATP acting at the purinergic P2X7 receptor (P2X7R) in cells primed with toll-like receptor (TLR) ligands. METHODS Purified (> 99%) microglia cultured from neonatal rat cortex and cerebellum were first primed with the putative TLR4/TLR2 agonist SAA (recombinant human Apo-SAA) or the established TLR4 agonist lipopolysaccharide (LPS) followed by addition of ATP. Expression of genes for the NLRP3 inflammasome, IL-1β, tumor necrosis factor-α (TNF-α), and SAA1 was measured by quantitative real-time polymerase chain reaction (q-PCR). Intracellular and extracellular amounts of IL-1β were determined by ELISA. RESULTS Apo-SAA stimulated, in a time-dependent manner, the expression of NLRP3, IL-1β, and TNF-α in cortical microglia, and produced a concentration-dependent increase in the intracellular content of IL-1β in these cells. A 2-h 'priming' of the microglia with Apo-SAA followed by addition of ATP for 1 h, resulting in a robust release of IL-1β into the culture medium, with a concomitant reduction in its intracellular content. The selective P2X7R antagonist A740003 blocked ATP-dependent release of IL-1β. Microglia prepared from rat cerebellum displayed similar behaviors. As with LPS, Apo-SAA upregulated SAA1 and TLR2 mRNA, and downregulated that of TLR4. LPS was less efficacious than Apo-SAA, perhaps reflecting an action of the latter at TLR4 and TLR2. The TLR4 antagonist CLI-095 fully blocked the action of LPS, but only partially that of Apo-SAA. Although the TLR2 antagonist CU-CPT22 was inactive against Apo-SAA, it also failed to block the TLR2 agonist Pam3CSK4. CONCLUSIONS Microglia are central to the inflammatory process and a major source of IL-1β when activated. P2X7R-triggered IL-1β maturation and export is thus likely to represent an important contributor to this cytokine pool. Given that SAA is detected in Alzheimer disease and multiple sclerosis brain, together with IL-1β-immunopositive microglia, these findings propose a link between P2X7R, SAA, and IL-1β in CNS pathophysiology.
Collapse
Affiliation(s)
- Laura Facci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "E. Meneghetti" 2, 35131, Padua, Italy
| | - Massimo Barbierato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "E. Meneghetti" 2, 35131, Padua, Italy
| | - Morena Zusso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "E. Meneghetti" 2, 35131, Padua, Italy
| | - Stephen D Skaper
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "E. Meneghetti" 2, 35131, Padua, Italy.
| | - Pietro Giusti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "E. Meneghetti" 2, 35131, Padua, Italy
| |
Collapse
|
17
|
O'Sullivan SA, O'Sullivan C, Healy LM, Dev KK, Sheridan GK. Sphingosine 1-phosphate receptors regulate TLR4-induced CXCL5 release from astrocytes and microglia. J Neurochem 2018; 144:736-747. [PMID: 29377126 DOI: 10.1111/jnc.14313] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/06/2018] [Accepted: 01/18/2018] [Indexed: 12/13/2022]
Abstract
Sphingosine 1-phosphate receptors (S1PR) are G protein-coupled and compose a family with five subtypes, S1P1R-S1P5R. The drug Gilenya® (Novartis, Basel, Switzerland) (Fingolimod; FTY720) targets S1PRs and was the first oral therapy for patients with relapsing-remitting multiple sclerosis (MS). The phosphorylated form of FTY720 (pFTY720) binds S1PRs causing initial agonism, then subsequent receptor internalization and functional antagonism. Internalization of S1P1R attenuates sphingosine 1-phosphate (S1P)-mediated egress of lymphocytes from lymph nodes, limiting aberrant immune function in MS. pFTY720 also exerts direct actions on neurons and glial cells which express S1PRs. In this study, we investigated the regulation of pro-inflammatory chemokine release by S1PRs in enriched astrocytes and microglial cultures. Astrocytes and microglia were stimulated with lipopolysaccharide (LPS) and increases in C-X-C motif chemokine 5 (CXCL5), also known as LIX (lipopolysaccharide-induced CXC chemokine) expression were quantified. Results showed that pFTY720 attenuated LPS-induced CXCL5 (LIX) protein release from astrocytes, as did the S1P1R selective agonist, SEW2871. In addition, pFTY720 blocked messenger ribonucleic acid (mRNA) transcription of the chemokines, (i) CXCL5/LIX, (ii) C-X-C motif chemokine 10 (CXCL10) also known as interferon gamma-induced protein 10 (IP10) and (iii) chemokine (C-C motif) ligand 2 (CCL2) also known as monocyte chemoattractant protein 1 (MCP1). Interestingly, inhibition of sphingosine kinase attenuated LPS-induced increases in mRNA levels of all three chemokines, suggesting that LPS-TLR4 (Toll-like receptor 4) signalling may enhance chemokine expression via S1P-S1PR transactivation. Lastly, these observations were not limited to astrocytes since we also found that pFTY720 attenuated LPS-induced release of CXCL5 from microglia. These data highlight a role for S1PR signalling in regulating the levels of chemokines in glial cells and support the notion that pFTY720 efficacy in multiple sclerosis may involve the direct modulation of astrocytes and microglia.
Collapse
Affiliation(s)
- Sinead A O'Sullivan
- Drug Development, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Department of Neurology, University of Chicago, Chicago, IL, USA
| | | | - Luke M Healy
- Drug Development, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Kumlesh K Dev
- Drug Development, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Graham K Sheridan
- Drug Development, School of Medicine, Trinity College Dublin, Dublin, Ireland.,School of Pharmacy & Biomolecular Sciences, University of Brighton, Brighton, UK
| |
Collapse
|
18
|
Smith JA, Braga A, Verheyen J, Basilico S, Bandiera S, Alfaro-Cervello C, Peruzzotti-Jametti L, Shu D, Haque F, Guo P, Pluchino S. RNA Nanotherapeutics for the Amelioration of Astroglial Reactivity. MOLECULAR THERAPY. NUCLEIC ACIDS 2017; 10:103-121. [PMID: 29499926 PMCID: PMC5738063 DOI: 10.1016/j.omtn.2017.11.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 11/20/2017] [Accepted: 11/20/2017] [Indexed: 12/22/2022]
Abstract
In response to injuries to the CNS, astrocytes enter a reactive state known as astrogliosis, which is believed to be deleterious in some contexts. Activated astrocytes overexpress intermediate filaments including glial fibrillary acidic protein (GFAP) and vimentin (Vim), resulting in entangled cells that inhibit neurite growth and functional recovery. Reactive astrocytes also secrete inflammatory molecules such as Lipocalin 2 (Lcn2), which perpetuate reactivity and adversely affect other cells of the CNS. Herein, we report proof-of-concept use of the packaging RNA (pRNA)-derived three-way junction (3WJ) motif as a platform for the delivery of siRNAs to downregulate such reactivity-associated genes. In vitro, siRNA-3WJs induced a significant knockdown of Gfap, Vim, and Lcn2 in a model of astroglial activation, with a concomitant reduction in protein expression. Knockdown of Lcn2 also led to reduced protein secretion from reactive astroglial cells, significantly impeding the perpetuation of inflammation in otherwise quiescent astrocytes. Intralesional injection of anti-Lcn2-3WJs in mice with contusion spinal cord injury led to knockdown of Lcn2 at mRNA and protein levels in vivo. Our results provide evidence for siRNA-3WJs as a promising platform for ameliorating astroglial reactivity, with significant potential for further functionalization and adaptation for therapeutic applications in the CNS.
Collapse
Affiliation(s)
- Jayden A Smith
- Department of Clinical Neurosciences, Division of Stem Cell Neurobiology, Wellcome Trust-Medical Research Council Stem Cell Institute and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK.
| | - Alice Braga
- Department of Clinical Neurosciences, Division of Stem Cell Neurobiology, Wellcome Trust-Medical Research Council Stem Cell Institute and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK; Department of Diagnostics and Public Health, University of Verona, Verona 37134, Italy
| | - Jeroen Verheyen
- Department of Clinical Neurosciences, Division of Stem Cell Neurobiology, Wellcome Trust-Medical Research Council Stem Cell Institute and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Silvia Basilico
- Department of Clinical Neurosciences, Division of Stem Cell Neurobiology, Wellcome Trust-Medical Research Council Stem Cell Institute and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Sara Bandiera
- Department of Clinical Neurosciences, Division of Stem Cell Neurobiology, Wellcome Trust-Medical Research Council Stem Cell Institute and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK; Department of Life Sciences, University of Trieste, Trieste 34127, Italy
| | - Clara Alfaro-Cervello
- Department of Clinical Neurosciences, Division of Stem Cell Neurobiology, Wellcome Trust-Medical Research Council Stem Cell Institute and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Luca Peruzzotti-Jametti
- Department of Clinical Neurosciences, Division of Stem Cell Neurobiology, Wellcome Trust-Medical Research Council Stem Cell Institute and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Dan Shu
- College of Pharmacy, Division of Pharmaceutics and Pharmaceutical Chemistry, The Ohio State University, Columbus, OH, USA; College of Medicine, Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; NCI Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA; Center for RNA Nanobiotechnology and Nanomedicine, The Ohio State University, Columbus, OH, USA
| | - Farzin Haque
- College of Pharmacy, Division of Pharmaceutics and Pharmaceutical Chemistry, The Ohio State University, Columbus, OH, USA; College of Medicine, Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; NCI Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA; Center for RNA Nanobiotechnology and Nanomedicine, The Ohio State University, Columbus, OH, USA
| | - Peixuan Guo
- College of Pharmacy, Division of Pharmaceutics and Pharmaceutical Chemistry, The Ohio State University, Columbus, OH, USA; College of Medicine, Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH, USA; NCI Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA; Center for RNA Nanobiotechnology and Nanomedicine, The Ohio State University, Columbus, OH, USA.
| | - Stefano Pluchino
- Department of Clinical Neurosciences, Division of Stem Cell Neurobiology, Wellcome Trust-Medical Research Council Stem Cell Institute and NIHR Biomedical Research Centre, University of Cambridge, Cambridge, UK.
| |
Collapse
|
19
|
Expression and Differential Responsiveness of Central Nervous System Glial Cell Populations to the Acute Phase Protein Serum Amyloid A. Sci Rep 2017; 7:12158. [PMID: 28939905 PMCID: PMC5610307 DOI: 10.1038/s41598-017-12529-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 09/12/2017] [Indexed: 02/07/2023] Open
Abstract
Acute-phase response is a systemic reaction to environmental/inflammatory insults and involves hepatic production of acute-phase proteins, including serum amyloid A (SAA). Extrahepatically, SAA immunoreactivity is found in axonal myelin sheaths of cortex in Alzheimer's disease and multiple sclerosis (MS), although its cellular origin is unclear. We examined the responses of cultured rat cortical astrocytes, microglia and oligodendrocyte precursor cells (OPCs) to master pro-inflammatory cytokine tumour necrosis factor (TNF)-α and lipopolysaccaride (LPS). TNF-α time-dependently increased Saa1 (but not Saa3) mRNA expression in purified microglia, enriched astrocytes, and OPCs (as did LPS for microglia and astrocytes). Astrocytes depleted of microglia were markedly less responsive to TNF-α and LPS, even after re-addition of microglia. Microglia and enriched astrocytes showed complementary Saa1 expression profiles following TNF-α or LPS challenge, being higher in microglia with TNF-α and higher in astrocytes with LPS. Recombinant human apo-SAA stimulated production of both inflammatory mediators and its own mRNA in microglia and enriched, but not microglia-depleted astrocytes. Co-ultramicronized palmitoylethanolamide/luteolin, an established anti-inflammatory/ neuroprotective agent, reduced Saa1 expression in OPCs subjected to TNF-α treatment. These last data, together with past findings suggest that co-ultramicronized palmitoylethanolamide/luteolin may be a novel approach in the treatment of inflammatory demyelinating disorders like MS.
Collapse
|
20
|
Białecka M, Kurzawski M, Vlaykova T, Tacheva T, Dziedziejko V, Pierzchlińska A, Droździk M. Effects of common functional MMP12 gene polymorphisms on PD in a Polish population. Neurol Neurochir Pol 2017; 51:347-353. [DOI: 10.1016/j.pjnns.2017.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/08/2017] [Indexed: 10/19/2022]
|
21
|
Willis CM, Ménoret A, Jellison ER, Nicaise AM, Vella AT, Crocker SJ. A Refined Bead-Free Method to Identify Astrocytic Exosomes in Primary Glial Cultures and Blood Plasma. Front Neurosci 2017; 11:335. [PMID: 28663721 PMCID: PMC5471332 DOI: 10.3389/fnins.2017.00335] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 05/29/2017] [Indexed: 01/05/2023] Open
Abstract
Astrocytes are the most abundant glial cell type in the central nervous system (CNS) and are known to fulfill critical homeostatic functions. Dysfunction of activated astrocytes is also known to participate in the development of several neurological diseases. Astrocytes can be uniquely identified by expression of the intermediate filament protein glial acidic fibrillary protein (GFAP). Herein, we report on the development of a rigorous and sensitive methodology to identify GFAP+ exosomes in primary culture using flow cytometry. We then demonstrate that activated astrocytes release increased amounts of exosomes in response to treatment with interleukin-1β. Using this methodology, we report the identification of GFAP+ exosomes in blood and then use a mouse model of inflammatory demyelination, experimental autoimmune encephalomyelitis (EAE), to examine whether the abundance of GFAP+ exosomes in blood circulation changes during clinical illness. We find a detectable increase in the presence of GFAP+ exosomes in EAE mice when compared with non-EAE, control mice. Our data provide a novel perspective on the presence of GFAP in blood as it identifies exosomes as potential astrocyte-derived signals within blood. These data are complementary to previous clinical studies that reported elevated GFAP protein in blood samples from multiple sclerosis (MS) patients during a clinical relapse. These data also reveal the existence of a potential systemic role for astrocyte-derived exosomes in CNS conditions involving inflammation such as multiple sclerosis.
Collapse
Affiliation(s)
- Cory M Willis
- Departments of Neuroscience, University of Connecticut School of MedicineFarmington, CT, United States
| | - Antoine Ménoret
- Departments of Immunology, University of Connecticut School of MedicineFarmington, CT, United States
| | - Evan R Jellison
- Departments of Immunology, University of Connecticut School of MedicineFarmington, CT, United States
| | - Alexandra M Nicaise
- Departments of Neuroscience, University of Connecticut School of MedicineFarmington, CT, United States
| | - Anthony T Vella
- Departments of Immunology, University of Connecticut School of MedicineFarmington, CT, United States
| | - Stephen J Crocker
- Departments of Neuroscience, University of Connecticut School of MedicineFarmington, CT, United States
| |
Collapse
|
22
|
Matsye P, Zheng L, Si Y, Kim S, Luo W, Crossman DK, Bratcher PE, King PH. HuR promotes the molecular signature and phenotype of activated microglia: Implications for amyotrophic lateral sclerosis and other neurodegenerative diseases. Glia 2017; 65:945-963. [PMID: 28300326 DOI: 10.1002/glia.23137] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 02/16/2017] [Accepted: 02/16/2017] [Indexed: 12/25/2022]
Abstract
In neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS), chronic activation of microglia contributes to disease progression. Activated microglia produce cytokines, chemokines, and other factors that normally serve to clear infection or damaged tissue either directly or through the recruitment of other immune cells. The molecular program driving this phenotype is classically linked to the transcription factor NF-κB and characterized by the upregulation of proinflammatory factors such as IL-1β, TNF-α, and IL-6. Here, we investigated the role of HuR, an RNA-binding protein that regulates gene expression through posttranscriptional pathways, on the molecular and cellular phenotypes of activated microglia. We performed RNA sequencing of HuR-silenced microglia and found significant attenuation of lipopolysaccharide-induced IL-1β and TNF-α inflammatory pathways and other factors that promote microglial migration and invasion. RNA kinetics and luciferase reporter studies suggested that the attenuation was related to altered promoter activity rather than a change in RNA stability. HuR-silenced microglia showed reduced migration, invasion, and chemotactic properties but maintained viability. MMP-12, a target exquisitely sensitive to HuR knockdown, participates in the migration/invasion phenotype. HuR is abundantly detected in the cytoplasmic compartment of activated microglia from ALS spinal cords consistent with its increased activity. Microglia from ALS-associated mutant SOD1 mice demonstrated higher migration/invasion properties which can be blocked with HuR inhibition. These findings underscore an important role for HuR in sculpting the molecular signature and phenotype of activated microglia, and as a possible therapeutic target in ALS and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Prachi Matsye
- Department of Neurology, University of Alabama, Birmingham, Alabama.,Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | - Lei Zheng
- Department of Neurology, University of Alabama, Birmingham, Alabama.,Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | - Ying Si
- Department of Neurology, University of Alabama, Birmingham, Alabama.,Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | - Soojin Kim
- Department of Neurology, University of Alabama, Birmingham, Alabama
| | - Wenyi Luo
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - David K Crossman
- Department of Genetics, University of Alabama, Birmingham, Alabama
| | - Preston E Bratcher
- Department of Pediatrics, Division of Pediatric Pulmonary Medicine, National Jewish Health, Denver, Colorado
| | - Peter H King
- Department of Neurology, University of Alabama, Birmingham, Alabama.,Department of Genetics, University of Alabama, Birmingham, Alabama.,Department of Pediatrics, Division of Pediatric Pulmonary Medicine, National Jewish Health, Denver, Colorado.,Department of Cell, Developmental and Integrative Biology, University of Alabama, Birmingham, Alabama
| |
Collapse
|
23
|
Peña-Ortega F. Pharmacological Tools to Activate Microglia and their Possible use to Study Neural Network Patho-physiology. Curr Neuropharmacol 2017; 15:595-619. [PMID: 27697040 PMCID: PMC5543677 DOI: 10.2174/1570159x14666160928151546] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 08/05/2016] [Accepted: 09/26/2016] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Microglia are the resident immunocompetent cells of the CNS and also constitute a unique cell type that contributes to neural network homeostasis and function. Understanding microglia cell-signaling not only will reveal their diverse functions but also will help to identify pharmacological and non-pharmacological tools to modulate the activity of these cells. METHODS We undertook a search of bibliographic databases for peer-reviewed research literature to identify microglial activators and their cell-specificity. We also looked for their effects on neural network function and dysfunction. RESULTS We identified several pharmacological targets to modulate microglial function, which are more or less specific (with the proper control experiments). We also identified pharmacological targets that would require the development of new potent and specific modulators. We identified a wealth of evidence about the participation of microglia in neural network function and their alterations in pathological conditions. CONCLUSION The identification of specific microglia-activating signals provides experimental tools to modulate the activity of this heterogeneous cell type in order to evaluate its impact on other components of the nervous system, and it also helps to identify therapeutic approaches to ease some pathological conditions related to microglial dysfunction.
Collapse
Affiliation(s)
- Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, UNAM-Campus Juriquilla, México
| |
Collapse
|
24
|
Marinelli C, Di Liddo R, Facci L, Bertalot T, Conconi MT, Zusso M, Skaper SD, Giusti P. Ligand engagement of Toll-like receptors regulates their expression in cortical microglia and astrocytes. J Neuroinflammation 2015; 12:244. [PMID: 26714634 PMCID: PMC4696218 DOI: 10.1186/s12974-015-0458-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 12/15/2015] [Indexed: 12/13/2022] Open
Abstract
Background Toll-like receptor (TLR) activation on microglia and astrocytes are key elements in neuroinflammation which accompanies a number of neurological disorders. While TLR activation on glia is well-established to up-regulate pro-inflammatory mediator expression, much less is known about how ligand engagement of one TLR may affect expression of other TLRs on microglia and astrocytes. Methods In the present study, we evaluated the effects of agonists for TLR2 (zymosan), TLR3 (polyinosinic-polycytidylic acid (poly(I:C)), a synthetic analogue of double-stranded RNA) and TLR4 (lipopolysaccaride (LPS)) in influencing expression of their cognate receptor as well as that of the other TLRs in cultures of rat cortical purified microglia (>99.5 %) and nominally microglia-free astrocytes. Elimination of residual microglia (a common contaminant of astrocyte cultures) was achieved by incubation with the lysosomotropic agent l-leucyl-l-leucine methyl ester (L-LME). Results Flow cytometric analysis confirmed the purity (essentially 100 %) of the obtained microglia, and up to 5 % microglia contamination of astrocytes. L-LME treatment effectively removed microglia from the latter (real-time polymerase chain reaction). The three TLR ligands robustly up-regulated gene expression for pro-inflammatory markers (interleukin-1 and interleukin-6, tumor necrosis factor) in microglia and enriched, but not purified, astrocytes, confirming cellular functionality. LPS, zymosan and poly(I:C) all down-regulated TLR4 messenger RNA (mRNA) and up-regulated TLR2 mRNA at 6 and 24 h. In spite of their inability to elaborate pro-inflammatory mediator output, the nominally microglia-free astrocytes (>99 % purity) also showed similar behaviours to those of microglia, as well as changes in TLR3 gene expression. LPS interaction with TLR4 activates downstream mitogen-activated protein kinase and nuclear factor-κB signalling pathways and subsequently causes inflammatory mediator production. The effects of LPS on TLR2 mRNA in both cell populations were antagonized by a nuclear factor-κB inhibitor. Conclusions TLR2 and TLR4 activation in particular, in concert with microglia and astrocytes, comprise key elements in the initiation and maintenance of neuropathic pain. The finding that both homologous (zymosan) and heterologous (LPS, poly(I:C)) TLR ligands are capable of regulating TLR2 gene expression, in particular, may have important implications in understanding the relative contributions of different TLRs in neurological disorders associated with neuroinflammation. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0458-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Carla Marinelli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "E. Meneghetti" 2, 35131, Padua, Italy.
| | - Rosa Di Liddo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "E. Meneghetti" 2, 35131, Padua, Italy.
| | - Laura Facci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "E. Meneghetti" 2, 35131, Padua, Italy.
| | - Thomas Bertalot
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "E. Meneghetti" 2, 35131, Padua, Italy.
| | - Maria Teresa Conconi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "E. Meneghetti" 2, 35131, Padua, Italy.
| | - Morena Zusso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "E. Meneghetti" 2, 35131, Padua, Italy.
| | - Stephen D Skaper
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "E. Meneghetti" 2, 35131, Padua, Italy.
| | - Pietro Giusti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Largo "E. Meneghetti" 2, 35131, Padua, Italy.
| |
Collapse
|
25
|
Kleiderman S, Sá JV, Teixeira AP, Brito C, Gutbier S, Evje LG, Hadera MG, Glaab E, Henry M, Sachinidis A, Alves PM, Sonnewald U, Leist M. Functional and phenotypic differences of pure populations of stem cell-derived astrocytes and neuronal precursor cells. Glia 2015; 64:695-715. [DOI: 10.1002/glia.22954] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 11/19/2015] [Accepted: 11/23/2015] [Indexed: 12/19/2022]
Affiliation(s)
- Susanne Kleiderman
- The Doerenkamp-Zbinden Chair of in-Vitro Toxicology and Biomedicine/Alternatives to Animal Experimentation; University of Konstanz; Konstanz Germany
| | - João V. Sá
- Instituto de Tecnologia Química e Biológica António Xavier; Universidade Nova de Lisboa; Av. da República 2780-157 Oeiras Portugal
- IBET; Instituto de Biologia Experimental e Tecnológica; Apartado 12 2780-901 Oeiras Portugal
| | - Ana P. Teixeira
- Instituto de Tecnologia Química e Biológica António Xavier; Universidade Nova de Lisboa; Av. da República 2780-157 Oeiras Portugal
- IBET; Instituto de Biologia Experimental e Tecnológica; Apartado 12 2780-901 Oeiras Portugal
| | - Catarina Brito
- Instituto de Tecnologia Química e Biológica António Xavier; Universidade Nova de Lisboa; Av. da República 2780-157 Oeiras Portugal
- IBET; Instituto de Biologia Experimental e Tecnológica; Apartado 12 2780-901 Oeiras Portugal
| | - Simon Gutbier
- The Doerenkamp-Zbinden Chair of in-Vitro Toxicology and Biomedicine/Alternatives to Animal Experimentation; University of Konstanz; Konstanz Germany
| | - Lars G. Evje
- Department of Earth Science, University of Bergen; Allégaten 41 5007 Bergen Norway
| | - Mussie G. Hadera
- Department of Pharmacy; College of Health Sciences; Mekelle University, Tigray Ethiopia
| | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg; Belvaux L-4366 Luxembourg
| | - Margit Henry
- Institute of Neurophysiology and Center for Molecular Medicine, Cologne (CMMC), University of Cologne; Cologne Germany
| | - Agapios Sachinidis
- Institute of Neurophysiology and Center for Molecular Medicine, Cologne (CMMC), University of Cologne; Cologne Germany
| | - Paula M. Alves
- Instituto de Tecnologia Química e Biológica António Xavier; Universidade Nova de Lisboa; Av. da República 2780-157 Oeiras Portugal
- IBET; Instituto de Biologia Experimental e Tecnológica; Apartado 12 2780-901 Oeiras Portugal
| | - Ursula Sonnewald
- Department of Drug Design and Pharmacology; Faculty of Health and Medical Sciences; Copenhagen Denmark
- Department of Neuroscience; Norwegian University of Science and Technology; Faculty of Medicine; Trondheim Norway
| | - Marcel Leist
- The Doerenkamp-Zbinden Chair of in-Vitro Toxicology and Biomedicine/Alternatives to Animal Experimentation; University of Konstanz; Konstanz Germany
| |
Collapse
|
26
|
Lorea-Hernández JJ, Morales T, Rivera-Angulo AJ, Alcantara-Gonzalez D, Peña-Ortega F. Microglia modulate respiratory rhythm generation and autoresuscitation. Glia 2015; 64:603-19. [DOI: 10.1002/glia.22951] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 11/18/2015] [Accepted: 11/18/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Jonathan-Julio Lorea-Hernández
- Departamento de Neurobiología del Desarrollo y Neurofisiología; Instituto De Neurobiología, UNAM Campus Juriquilla; Querétaro México
| | - Teresa Morales
- Departamento de Neurobiología del Desarrollo y Neurofisiología; Instituto De Neurobiología, UNAM Campus Juriquilla; Querétaro México
| | - Ana-Julia Rivera-Angulo
- Departamento de Neurobiología del Desarrollo y Neurofisiología; Instituto De Neurobiología, UNAM Campus Juriquilla; Querétaro México
| | - David Alcantara-Gonzalez
- Departamento de Neurobiología del Desarrollo y Neurofisiología; Instituto De Neurobiología, UNAM Campus Juriquilla; Querétaro México
| | - Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología; Instituto De Neurobiología, UNAM Campus Juriquilla; Querétaro México
| |
Collapse
|
27
|
Astrocytic CCAAT/Enhancer-Binding Protein Delta Contributes to Glial Scar Formation and Impairs Functional Recovery After Spinal Cord Injury. Mol Neurobiol 2015; 53:5912-5927. [PMID: 26510742 PMCID: PMC5085997 DOI: 10.1007/s12035-015-9486-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 10/12/2015] [Indexed: 02/06/2023]
Abstract
After spinal cord injury, inflammatory reaction induces the aggregation of astrocytes to form a glial scar that eventually blocks axonal regeneration. Transcription factor CCAAT/enhancer-binding protein delta (C/EBPδ) is a regulatory protein of genes responsive to inflammatory factors, but its role in glial scar formation after spinal cord injury remains unknown. By using a model of moderate spinal cord contusion injury at the mid-thoracic level, we found that C/EBPδ was expressed mostly in the reactive astrocytes bordering the lesion in wild-type mice from 7 days after the injury. C/EBPδ-deficient mice showed reduced glial scar formation, more residual white matter, and better motor function recovery compared with wild-type mice 28 days after the injury. Upon interleukin (IL)-1β stimulation in vitro, the increased expression of C/EBPδ in reactive astrocytes inhibited RhoA expression and, subsequently, the ability of astrocyte migration. However, these reactive astrocytes also produced an increased amount of matrix metalloproteinase-3, which promoted the migration of non-IL-1β-treated, inactive astrocytes. Although the involvement of other non-astroglial C/EBPδ cannot be entirely excluded, our studies suggest that astrocytic C/EBPδ is integral to the inflammatory cascades leading to glial scar formation after spinal cord injury.
Collapse
|
28
|
Johnson KM, Crocker SJ. TIMP-1 couples RhoK activation to IL-1β-induced astrocyte responses. Neurosci Lett 2015; 609:165-70. [PMID: 26484505 DOI: 10.1016/j.neulet.2015.10.038] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 09/21/2015] [Accepted: 10/13/2015] [Indexed: 12/27/2022]
Abstract
Interleukin-1β (IL-1β) is a pleotropic cytokine known to influence the central nervous system (CNS) responses to injury or infection. IL-1β also directly induces astrocytic expression of tissue inhibitor of metalloproteinases (TIMP)-1, a potent trophic factor and regulator of matrix metalloproteinase activity. In this study, we examined the functional relationship between IL-1β and TIMP-1 and determined that the behavior of astrocytes in response to IL-1β is determined by TIMP-1 expression. Using primary astrocytes from C57Bl/6 mice, we found astrocytes from wildtype (Wt) mice exhibited a robust wound healing response to a scratch wound that was arrested in response to IL-1β. In contrast, TIMP-1 knockout (TIMP-1KO) astrocytes, exhibited minimal response to the scratch wound but an accelerated response following IL-1β-treatment. We also determined that the scratch wound effect in Wt cultures was attenuated by inhibition of Rho kinase but amplified in the TIMP-1KO cultures. We propose that the specific induction of TIMP-1 from astrocytes in response to IL-1β reflects a previously unrecognized physiological relationship where the directionality of astrocytic behavior is determined by the actions of TIMP‑1. These findings may provide additional insight into glial responses in the context of neuropathology where expression of TIMP-1 may vary and astrocytic responses may be impacted by the inflammatory milieu of the CNS.
Collapse
Affiliation(s)
- Kasey M Johnson
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Stephen J Crocker
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, United States.
| |
Collapse
|
29
|
Inflammation Promotes a Conversion of Astrocytes into Neural Progenitor Cells via NF-κB Activation. Mol Neurobiol 2015; 53:5041-55. [PMID: 26381429 PMCID: PMC5012156 DOI: 10.1007/s12035-015-9428-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Accepted: 09/07/2015] [Indexed: 11/23/2022]
Abstract
Brain inflammation, a common feature in neurodegenerative diseases, is a complex series of events, which can be detrimental and even lead to neuronal death. Nonetheless, several studies suggest that inflammatory signals are also positively influencing neural cell proliferation, survival, migration, and differentiation. Recently, correlative studies suggested that astrocytes are able to dedifferentiate upon injury and may thereby re-acquire neural stem cell (NSC) potential. However, the mechanism underlying this dedifferentiation process upon injury remains unclear. Here, we report that during the early response of reactive gliosis, inflammation induces a conversion of mature astrocytes into neural progenitors. A TNF treatment induces the decrease of specific astrocyte markers, such as glial fibrillary acidic protein (GFAP) or genes related to glycogen metabolism, while a subset of these cells re-expresses immaturity markers, such as CD44, Musashi-1, and Oct4. Thus, TNF treatment results in the appearance of cells that exhibit a neural progenitor phenotype and are able to proliferate and differentiate into neurons and/or astrocytes. This dedifferentiation process is maintained as long as TNF is present in the culture medium. In addition, we highlight a role for Oct4 in this process, since the TNF-induced dedifferentiation can be prevented by inhibiting Oct4 expression. Our results show that activation of the NF-κB pathway through TNF plays an important role in the dedifferentiation of astrocytes via the re-expression of Oct4. These findings indicate that the first step of reactive gliosis is in fact a dedifferentiation process of resident astrocytes mediated by the NF-κB pathway.
Collapse
|
30
|
Xing L, McDonald H, Da Fonte DF, Gutierrez-Villagomez JM, Trudeau VL. Dopamine D1 receptor activation regulates the expression of the estrogen synthesis gene aromatase B in radial glial cells. Front Neurosci 2015; 9:310. [PMID: 26388722 PMCID: PMC4557113 DOI: 10.3389/fnins.2015.00310] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 08/18/2015] [Indexed: 11/13/2022] Open
Abstract
Radial glial cells (RGCs) are abundant stem-like non-neuronal progenitors that are important for adult neurogenesis and brain repair, yet little is known about their regulation by neurotransmitters. Here we provide evidence for neuronal-glial interactions via a novel role for dopamine to stimulate RGC function. Goldfish were chosen as the model organism due to the abundance of RGCs and regenerative abilities of the adult central nervous system. A close anatomical relationship was observed between tyrosine hydroxylase-positive catecholaminergic cell bodies and axons and dopamine-D1 receptor expressing RGCs along the ventricular surface of telencephalon, a site of active neurogenesis. A primary cell culture model was established and immunofluorescence analysis indicates that in vitro RGCs from female goldfish retain their major characteristics in vivo, including expression of glial fibrillary acidic protein and brain lipid binding protein. The estrogen synthesis enzyme aromatase B is exclusively found in RGCs, but this is lost as cells differentiate to neurons and other glial types in adult teleost brain. Pharmacological experiments using the cultured RGCs established that specific activation of dopamine D1 receptors up-regulates aromatase B mRNA through a cyclic adenosine monophosphate-dependent molecular mechanism. These data indicate that dopamine enhances the steroidogenic function of this neuronal progenitor cell.
Collapse
Affiliation(s)
- Lei Xing
- Department of Biology, Centre for Advanced Research in Environmental Genomics, University of Ottawa Ottawa, ON, Canada
| | - Heather McDonald
- Department of Biology, Centre for Advanced Research in Environmental Genomics, University of Ottawa Ottawa, ON, Canada
| | - Dillon F Da Fonte
- Department of Biology, Centre for Advanced Research in Environmental Genomics, University of Ottawa Ottawa, ON, Canada
| | - Juan M Gutierrez-Villagomez
- Department of Biology, Centre for Advanced Research in Environmental Genomics, University of Ottawa Ottawa, ON, Canada
| | - Vance L Trudeau
- Department of Biology, Centre for Advanced Research in Environmental Genomics, University of Ottawa Ottawa, ON, Canada
| |
Collapse
|
31
|
Jebelli J, Piers T, Pocock J. Selective Depletion of Microglia from Cerebellar Granule Cell Cultures Using L-leucine Methyl Ester. J Vis Exp 2015:e52983. [PMID: 26275019 DOI: 10.3791/52983] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Microglia, the resident immunocompetent cells of the CNS, play multifaceted roles in modulating and controlling neuronal function, as well as mediating innate immunity. Primary rodent cell culture models have greatly advanced our understanding of neuronal-glial interactions, but only recently have methods to specifically eliminate microglia from mixed cultures been utilized. One such technique - described here - is the use of L-leucine methyl ester, a lysomotropic agent that is internalized by macrophages and microglia, wherein it causes lysosomal disruption and subsequent apoptosis(13,14). Experiments using L-leucine methyl ester have the power to identify the contribution of microglia to the surrounding cellular environment under diverse culture conditions. Using a protocol optimized in our laboratory, we describe how to eliminate microglia from P5 rodent cerebellar granule cell culture. This approach allows one to assess the relative impact of microglia on experimental data, as well as determine whether microglia are playing a neuroprotective or neurotoxic role in culture models of neurological conditions, such as stroke, Alzheimer's or Parkinson's disease.
Collapse
Affiliation(s)
| | - Thomas Piers
- Therapeutic Innovation Group, School of Life and Medical Sciences, University College London
| | | |
Collapse
|
32
|
Gustin A, Kirchmeyer M, Koncina E, Felten P, Losciuto S, Heurtaux T, Tardivel A, Heuschling P, Dostert C. NLRP3 Inflammasome Is Expressed and Functional in Mouse Brain Microglia but Not in Astrocytes. PLoS One 2015; 10:e0130624. [PMID: 26091541 PMCID: PMC4474809 DOI: 10.1371/journal.pone.0130624] [Citation(s) in RCA: 287] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 05/21/2015] [Indexed: 11/19/2022] Open
Abstract
Neuroinflammation is the local reaction of the brain to infection, trauma, toxic molecules or protein aggregates. The brain resident macrophages, microglia, are able to trigger an appropriate response involving secretion of cytokines and chemokines, resulting in the activation of astrocytes and recruitment of peripheral immune cells. IL-1β plays an important role in this response; yet its production and mode of action in the brain are not fully understood and its precise implication in neurodegenerative diseases needs further characterization. Our results indicate that the capacity to form a functional NLRP3 inflammasome and secretion of IL-1β is limited to the microglial compartment in the mouse brain. We were not able to observe IL-1β secretion from astrocytes, nor do they express all NLRP3 inflammasome components. Microglia were able to produce IL-1β in response to different classical inflammasome activators, such as ATP, Nigericin or Alum. Similarly, microglia secreted IL-18 and IL-1α, two other inflammasome-linked pro-inflammatory factors. Cell stimulation with α-synuclein, a neurodegenerative disease-related peptide, did not result in the release of active IL-1β by microglia, despite a weak pro-inflammatory effect. Amyloid-β peptides were able to activate the NLRP3 inflammasome in microglia and IL-1β secretion occurred in a P2X7 receptor-independent manner. Thus microglia-dependent inflammasome activation can play an important role in the brain and especially in neuroinflammatory conditions.
Collapse
Affiliation(s)
- Audrey Gustin
- Life Sciences Research Unit, Faculty of Science, Technology and Communication, University of Luxembourg, Luxembourg, Luxembourg
| | - Mélanie Kirchmeyer
- Life Sciences Research Unit, Faculty of Science, Technology and Communication, University of Luxembourg, Luxembourg, Luxembourg
| | - Eric Koncina
- Life Sciences Research Unit, Faculty of Science, Technology and Communication, University of Luxembourg, Luxembourg, Luxembourg
| | - Paul Felten
- Life Sciences Research Unit, Faculty of Science, Technology and Communication, University of Luxembourg, Luxembourg, Luxembourg
| | - Sophie Losciuto
- Life Sciences Research Unit, Faculty of Science, Technology and Communication, University of Luxembourg, Luxembourg, Luxembourg
| | - Tony Heurtaux
- Life Sciences Research Unit, Faculty of Science, Technology and Communication, University of Luxembourg, Luxembourg, Luxembourg
| | - Aubry Tardivel
- Biochemistry Institute, University of Lausanne, Epalinges, Switzerland
| | - Paul Heuschling
- Life Sciences Research Unit, Faculty of Science, Technology and Communication, University of Luxembourg, Luxembourg, Luxembourg
| | - Catherine Dostert
- Life Sciences Research Unit, Faculty of Science, Technology and Communication, University of Luxembourg, Luxembourg, Luxembourg
- * E-mail:
| |
Collapse
|
33
|
Johnson KM, Milner R, Crocker SJ. Extracellular matrix composition determines astrocyte responses to mechanical and inflammatory stimuli. Neurosci Lett 2015; 600:104-9. [PMID: 26067407 DOI: 10.1016/j.neulet.2015.06.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/03/2015] [Accepted: 06/05/2015] [Indexed: 01/19/2023]
Abstract
Astrocytes perform critical homeostatic physiological functions in the central nervous system (CNS) and are robustly responsive to injury, inflammation, or infection. We hypothesized that the components of the extracellular matrix (ECM), which are known to vary during development and in response to disease, determine astrocytic responses to injury and inflammation. We examined the response of primary astrocyte cultures grown on different ECM proteins to a mechanical wound (i.e., scratch). ECM substrates selected were laminin (Ln), vitronectin (Vn), fibronectin (Fn) or Tenascin C (TnC). We found that regrowth of the scratch wound was ECM dependent: recovery was arrested on fibronectin (Fn), almost complete on either Vn, Ln, or TnC. To determine whether ECM responses were also influenced by inflammation, we treated ECM plated cultures with interleukin-1β (IL-1β). We found that IL-1β arrested astrocyte growth on Ln, accelerated astrocyte growth on Fn and had no significant effect on astrocyte growth on TnC or Vn. We also determined that blocking β1integrins, the major class of receptors for all ECM proteins tested, prevented the robust response of astrocytes exposed to TnC, Ln and Vn, and also inhibited the robust effect of IL-1β to stimulate astrocyte growth on Fn. In addition, we evaluated downstream targets of integrin signaling, specifically the mammalian target of rapamycin (mTOR), and determined that activation of this pathway contributed to the response of astrocytes grown on TnC, but not on Ln, Vn or Fn. These findings provide new insights into the role of ECM as a source of heterogeneity of glial responses that may have important implications for neuropathological sequelae.
Collapse
Affiliation(s)
- Kasey M Johnson
- Department of Neuroscience,University of Connecticut School of Medicine, Farmington, CT, USA
| | - Richard Milner
- Department of Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Stephen J Crocker
- Department of Neuroscience,University of Connecticut School of Medicine, Farmington, CT, USA.
| |
Collapse
|
34
|
Cui X, Chopp M, Zacharek A, Karasinska JM, Cui Y, Ning R, Zhang Y, Wang Y, Chen J. Deficiency of brain ATP-binding cassette transporter A-1 exacerbates blood-brain barrier and white matter damage after stroke. Stroke 2015; 46:827-34. [PMID: 25593138 DOI: 10.1161/strokeaha.114.007145] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE The ATP-binding cassette transporter A-1 (ABCA1) gene is a key target of the transcription factors liver X receptors. Liver X receptor activation has anti-inflammatory and neuroprotective effects in animal ischemic stroke models. Here, we tested the hypothesis that brain ABCA1 reduces blood-brain barrier (BBB) and white matter (WM) impairment in the ischemic brain after stroke. METHODS Adult brain-specific ABCA1-deficient (ABCA1(-B/-B)) and floxed-control (ABCA1(fl/fl)) mice were subjected to permanent distal middle cerebral artery occlusion and were euthanized 7 days after distal middle cerebral artery occlusion. Functional outcome, infarct volume, BBB leakage, and WM damage were analyzed. RESULTS Compared with ABCA1(fl/fl) mice, ABCA1(-B/-B) mice showed marginally (P=0.052) increased lesion volume but significantly increased BBB leakage and WM damage in the ischemic brain and more severe neurological deficits. Brain ABCA1-deficient mice exhibited increased the level of matrix metalloproteinase-9 and reduced the level of insulin-like growth factor 1 in the ischemic brain. BBB leakage was inversely correlated (r=-0.073; P<0.05) with aquaporin-4 expression. Reduction of insulin-like growth factor 1 and aquaporin-4, but upregulation of matrix metalloproteinase-9 expression were also found in the primary astrocyte cultures derived from ABCA1(-B/-B) mice. Cultured primary cortical neurons derived from C57BL/6 wild-type mice with ABCA1(-B/-B) astrocyte-conditioned medium exhibited decreased neurite outgrowth compared with culture with ABCA1(fl/fl) astrocyte-conditioned medium. ABCA1(-B/-B) primary cortical neurons show significantly decreased neurite outgrowth, which was attenuated by insulin-like growth factor 1 treatment. CONCLUSIONS We demonstrate that brain ABCA1 deficiency increases BBB leakage, WM/axonal damage, and functional deficits after stroke. Concomitant reduction of insulin-like growth factor 1 and upregulation of matrix metalloproteinase-9 may contribute to brain ABCA1 deficiency-induced BBB and WM/axonal damage in the ischemic brain.
Collapse
Affiliation(s)
- Xu Cui
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (X.C., M.C., A.Z., Y.C., R.N., Y.Z., J.C.); Department of Physics, Oakland University, Rochester, MI (M.C.); Neural Protection and Regeneration section, Center for Neuropsychiatric Research, National Institute on Drug Abuse, NIH, Baltimore, MD (Y.W.); and Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada (J.M.K.).
| | - Michael Chopp
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (X.C., M.C., A.Z., Y.C., R.N., Y.Z., J.C.); Department of Physics, Oakland University, Rochester, MI (M.C.); Neural Protection and Regeneration section, Center for Neuropsychiatric Research, National Institute on Drug Abuse, NIH, Baltimore, MD (Y.W.); and Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada (J.M.K.)
| | - Alex Zacharek
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (X.C., M.C., A.Z., Y.C., R.N., Y.Z., J.C.); Department of Physics, Oakland University, Rochester, MI (M.C.); Neural Protection and Regeneration section, Center for Neuropsychiatric Research, National Institute on Drug Abuse, NIH, Baltimore, MD (Y.W.); and Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada (J.M.K.)
| | - Joanna M Karasinska
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (X.C., M.C., A.Z., Y.C., R.N., Y.Z., J.C.); Department of Physics, Oakland University, Rochester, MI (M.C.); Neural Protection and Regeneration section, Center for Neuropsychiatric Research, National Institute on Drug Abuse, NIH, Baltimore, MD (Y.W.); and Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada (J.M.K.)
| | - Yisheng Cui
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (X.C., M.C., A.Z., Y.C., R.N., Y.Z., J.C.); Department of Physics, Oakland University, Rochester, MI (M.C.); Neural Protection and Regeneration section, Center for Neuropsychiatric Research, National Institute on Drug Abuse, NIH, Baltimore, MD (Y.W.); and Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada (J.M.K.)
| | - Ruizhuo Ning
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (X.C., M.C., A.Z., Y.C., R.N., Y.Z., J.C.); Department of Physics, Oakland University, Rochester, MI (M.C.); Neural Protection and Regeneration section, Center for Neuropsychiatric Research, National Institute on Drug Abuse, NIH, Baltimore, MD (Y.W.); and Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada (J.M.K.)
| | - Yi Zhang
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (X.C., M.C., A.Z., Y.C., R.N., Y.Z., J.C.); Department of Physics, Oakland University, Rochester, MI (M.C.); Neural Protection and Regeneration section, Center for Neuropsychiatric Research, National Institute on Drug Abuse, NIH, Baltimore, MD (Y.W.); and Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada (J.M.K.)
| | - Yun Wang
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (X.C., M.C., A.Z., Y.C., R.N., Y.Z., J.C.); Department of Physics, Oakland University, Rochester, MI (M.C.); Neural Protection and Regeneration section, Center for Neuropsychiatric Research, National Institute on Drug Abuse, NIH, Baltimore, MD (Y.W.); and Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada (J.M.K.)
| | - Jieli Chen
- From the Department of Neurology, Henry Ford Hospital, Detroit, MI (X.C., M.C., A.Z., Y.C., R.N., Y.Z., J.C.); Department of Physics, Oakland University, Rochester, MI (M.C.); Neural Protection and Regeneration section, Center for Neuropsychiatric Research, National Institute on Drug Abuse, NIH, Baltimore, MD (Y.W.); and Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada (J.M.K.).
| |
Collapse
|
35
|
Aberrant production of tenascin-C in globoid cell leukodystrophy alters psychosine-induced microglial functions. J Neuropathol Exp Neurol 2014; 73:964-74. [PMID: 25192051 DOI: 10.1097/nen.0000000000000117] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Globoid cell leukodystrophy (GLD), or Krabbe disease, is a rare and often fatal demyelinating disease caused by mutations in the galactocerebrosidase (galc) gene that result in accumulation of galactosylsphingosine (psychosine). We recently reported that the extracellular matrix (ECM) protease, matrix metalloproteinase-3, is elevated in GLD and that it regulates psychosine-induced microglial activation. Here, we examined central nervous system ECM component expression in human GLD patients and in the twitcher mouse model of GLD using immunohistochemistry. The influence of ECM proteins on primary murine microglial responses to psychosine was evaluated using ECM proteins as substrates and analyzed by quantitative real-time polymerase chain reaction, immunocytochemistry, and ELISA. Functional analysis of microglial cytotoxicity was performed on oligodendrocytes in coculture, and cell death was measured by lactose dehydrogenase assay. Tenascin-C (TnC) was expressed at higher levels in human GLD and in twitcher mice versus controls. Microglial responses to psychosine were enhanced by TnC, as determined by an increase in globoid-like cell formation, matrix metalloproteinase-3 mRNA expression, and higher toxicity toward oligodendrocytes in culture. These findings were consistent with a shift toward the M1 microglial phenotype in TnC-grown microglia. Thus, elevated TnC expression in GLD modified microglial responses to psychosine. These data offer a novel perspective and enhance understanding of the microglial contribution to GLD pathogenesis.
Collapse
|
36
|
Claycomb KI, Johnson KM, Bongarzone ER, Crocker SJ. An in vitro model for the study of cellular pathophysiology in globoid cell leukodystrophy. J Vis Exp 2014:e51903. [PMID: 25350151 DOI: 10.3791/51903] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The precise function of multi-nucleated microglia, called globoid cells, that are uniquely abundant in the central nervous system of globoid cell leukodystrophy (GLD) is unclear. This gap in knowledge has been hindered by the lack of an appropriate in vitro model for study. Herein, we describe a primary murine glial culture system in which treatment with psychosine results in multinucleation of microglia resembling the characteristic globoid cells found in GLD. Using this novel system, we defined the conditions and modes of analysis for study of globoid cells. The potential use of this model system was validated in our previous study, which identified a potential role for matrix metalloproteinase (MMP)-3 in GLD. This novel in vitro system may be a useful model in which to study the formation and function, but also the potential therapeutic manipulation, of these unique cells.
Collapse
Affiliation(s)
| | - Kasey M Johnson
- Department of Neuroscience, University of Connecticut Health Center
| | | | - Stephen J Crocker
- Department of Neuroscience, University of Connecticut Health Center;
| |
Collapse
|
37
|
Chen SH, Oyarzabal EA, Sung YF, Chu CH, Wang Q, Chen SL, Lu RB, Hong JS. Microglial regulation of immunological and neuroprotective functions of astroglia. Glia 2014; 63:118-31. [PMID: 25130274 DOI: 10.1002/glia.22738] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 07/23/2014] [Indexed: 01/06/2023]
Abstract
Microglia and astroglia play critical roles in the development, function, and survival of neurons in the CNS. However, under inflammatory conditions the role of astrogliosis in the inflammatory process and its effects on neurons remains unclear. Here, we used several types of cell cultures treated with the bacterial inflammogen LPS to address these questions. We found that the presence of astroglia reduced inflammation-driven neurotoxicity, suggesting that astrogliosis is principally neuroprotective. Neutralization of supernatant glial cell line-derived neurotrophic factor (GDNF) released from astroglia significantly reduced this neuroprotective effect during inflammation. To determine the immunological role of astroglia, we optimized a highly-enriched astroglial culture protocol and demonstrated that LPS failed to induce the synthesis and release of TNF-α and iNOS/NO. Instead we found significant enhancement of TNF-α and iNOS expression in highly-enriched astroglial cultures required the presence of 0.5-1% microglia, respectively. Thus suggesting that microglial-astroglial interactions are required for LPS to induce the expression of pro-inflammatory factors and GDNF from astroglia. Specifically, we found that microglia-derived TNF-α plays a pivotal role as a paracrine signal to regulate the neuroprotective functions of astrogliosis. Taken together, these findings suggest that astroglia may not possess the ability to directly recognize the innate immune stimuli LPS, but rather depend on crosstalk with microglia to elicit release of neurotrophic factors as a counterbalance to support neuronal survival from the collateral damage generated by activated microglia during neuroinflammation.
Collapse
Affiliation(s)
- Shih-Heng Chen
- Laboratory of Toxicology and Pharmacology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | | | | | | | | | | | | | | |
Collapse
|
38
|
NADPH oxidase mediates TNF-α-evoked in vitro brain barrier dysfunction: roles of apoptosis and time. Mol Cell Neurosci 2014; 61:72-84. [PMID: 24907586 DOI: 10.1016/j.mcn.2014.06.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 05/20/2014] [Accepted: 06/02/2014] [Indexed: 11/20/2022] Open
Abstract
The pro-inflammatory cytokine TNF-α severely perturbs the integrity of the blood-brain barrier (BBB). This study explored the specific roles of NADPH oxidase and associated downstream effectors by using human brain microvascular endothelial cells (HBMECs) and human astrocytes (HAs), the key components of BBB, alone or in co-cultures to mimic human BBB. Exposure to TNF-α (6h) impaired BBB integrity as evidenced by marked decreases in transendothelial electrical resistance and concurrent increases in paracellular flux which appeared to subside with time (24h). Increased barrier dysfunction concurred with increases in endothelial NADPH oxidase activity, O2(-) production, actin stress fibre formation, MMP-2/9 activities and concomitant decreases in antioxidant (CuZn-SOD and catalase) and tight junction (claudin-5 and occludin) protein expressions. Conversely, TNF-α did not affect astrocytic MMP activities and antioxidant enzyme expressions. Unlike BBB damage, rates of HBMEC and HA apoptosis increased by time. Suppression of NADPH oxidase by apocynin or diphenyleneiodonium prevented TNF-α-evoked morphological changes and apoptosis, attenuated endothelial MMP activity and helped retain usual tight junction protein expression and barrier function. In conclusion, HBMECs constitute the main source of oxidative stress and basement-membrane degrading endopeptidases in inflammatory conditions associated with excessive release of TNF-α where targeting NADPH oxidase may prove extremely beneficial in maintaining proper barrier activity through prevention of cytoskeletal and tight junction reorganisations.
Collapse
|
39
|
Caesar M, Felk S, Zach S, Brønstad G, Aasly JO, Gasser T, Gillardon F. Changes in matrix metalloprotease activity and progranulin levels may contribute to the pathophysiological function of mutant leucine-rich repeat kinase 2. Glia 2014; 62:1075-92. [DOI: 10.1002/glia.22663] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 03/04/2014] [Accepted: 03/07/2014] [Indexed: 12/28/2022]
Affiliation(s)
- Mareike Caesar
- Boehringer Ingelheim Pharma GmbH & Co KG; CNS Diseases Research; Biberach an der Riss Germany
| | - Sandra Felk
- Boehringer Ingelheim Pharma GmbH & Co KG; CNS Diseases Research; Biberach an der Riss Germany
| | - Susanne Zach
- Boehringer Ingelheim Pharma GmbH & Co KG; CNS Diseases Research; Biberach an der Riss Germany
| | - Gunnar Brønstad
- St. Olav's University Hospital; Department of Neurology; Trondheim Norway
| | - Jan O. Aasly
- St. Olav's University Hospital; Department of Neurology; Trondheim Norway
| | - Thomas Gasser
- Hertie Institut fuer klinische Hirnforschung; Tuebingen Germany
| | - Frank Gillardon
- Boehringer Ingelheim Pharma GmbH & Co KG; CNS Diseases Research; Biberach an der Riss Germany
| |
Collapse
|
40
|
Liang H, Guan D, Gao A, Yin Y, Jing M, Yang L, Ma W, Hu E, Zhang X. Human amniotic epithelial stem cells inhibit microglia activation through downregulation of tumor necrosis factor-α, interleukin-1β and matrix metalloproteinase-12 in vitro and in a rat model of intracerebral hemorrhage. Cytotherapy 2014; 16:523-34. [PMID: 24424266 DOI: 10.1016/j.jcyt.2013.11.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 10/20/2013] [Accepted: 11/08/2013] [Indexed: 01/01/2023]
Abstract
BACKGROUND AIMS The molecular mechanisms by which stem cell transplantation improves functional recovery after intracerebral hemorrhage (ICH) are not well understood. Accumulating evidence suggests that microglia cells are activated shortly after ICH and that this activation contributes to secondary ICH-induced brain injury. We studied the effect of human amniotic epithelial stem cells (HAESCs) on microglia activation. METHODS To study the effect of HAESCs in vitro, we used thrombin to activate the microglia cells. Twenty-four hours after thrombin treatment, the levels of tumor necrosis factor-α and interleukin-1β were measured by enzyme-linked immunosorbent assay. In vivo, the HAESCs were transplanted into the rat striatum 1 day after collagenase-induced ICH. The expression levels of matrix metalloproteinase (MMP)-12 and microglia infiltration in the peri-hematoma tissues were determined 7 days after ICH through the use of reverse transcriptase-polymerase chain reaction and immunohistochemical analysis, respectively. RESULTS Thrombin-activated microglia expression of tumor necrosis factor-α, interleukin-1β and MMP-12 was significantly reduced through contact-dependent and paracrine mechanisms when the HAESCs were co-cultured with microglia cells. After transplantation of HAESCs in rat brains, the expression levels of MMP-12 and microglia infiltration in the peri-hematoma tissues were significantly reduced. CONCLUSIONS Our observations suggest that microglia activation could be inhibited by HAESCs both in vitro and in vivo, which may be an important mechanism by which the transplantation of HAESCs reduces brain edema and ameliorates the neurologic deficits after ICH. Therefore, we hypothesize that methods for suppressing the activation of microglia and reducing the inflammatory response can be used for designing effective treatment strategies for ICH.
Collapse
Affiliation(s)
- Hongsheng Liang
- Key Laboratory of Neurosurgery, College of Heilongjiang Province, First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Dong Guan
- Department of Neurosurgery, Qingdao Hiser Medical Group, Qingdao, People's Republic of China
| | - Aili Gao
- School of Life Science, Northeast Agricultural University, Harbin, People's Republic of China
| | - Yibo Yin
- Key Laboratory of Neurosurgery, College of Heilongjiang Province, First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Meng Jing
- Key Laboratory of Neurosurgery, College of Heilongjiang Province, First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Lin Yang
- Key Laboratory of Neurosurgery, College of Heilongjiang Province, First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Wei Ma
- Key Laboratory of Neurosurgery, College of Heilongjiang Province, First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Enxi Hu
- Key Laboratory of Neurosurgery, College of Heilongjiang Province, First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Xiangtong Zhang
- Key Laboratory of Neurosurgery, College of Heilongjiang Province, First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China.
| |
Collapse
|
41
|
Langenfurth A, Rinnenthal JL, Vinnakota K, Prinz V, Carlo AS, Stadelmann C, Siffrin V, Peaschke S, Endres M, Heppner F, Glass R, Wolf SA, Kettenmann H. Membrane-type 1 metalloproteinase is upregulated in microglia/brain macrophages in neurodegenerative and neuroinflammatory diseases. J Neurosci Res 2013; 92:275-86. [DOI: 10.1002/jnr.23288] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 06/14/2013] [Accepted: 07/25/2013] [Indexed: 01/09/2023]
Affiliation(s)
- Anika Langenfurth
- Cellular Neurosciences; Max Delbrück Centre for Molecular Medicine; Berlin Germany
- Department of Neurology; Charité, Universitätsmedizin Berlin; Charité Campus Virchow Berlin Germany
| | - Jan Leo Rinnenthal
- Institute for Neuropathology; Charité, Universitätsmedizin Berlin; Charité Campus Mitte Berlin Germany
| | - Katyayni Vinnakota
- Cellular Neurosciences; Max Delbrück Centre for Molecular Medicine; Berlin Germany
| | - Vincent Prinz
- Department of Neurology and Center for Stroke Research Berlin; Charité, Universitätsmedizin Berlin; Charité Campus Mitte Berlin Germany
- Department of Neurosurgery; Charité, Universitätsmedizin Berlin; Charité Campus Virchow Berlin Germany
| | - Anne-Sophie Carlo
- Molecular Cardiovascular Research; Max Delbrück Centre for Molecular Medicine; Berlin Germany
- Max Planck Institute for Infection Biology; Berlin Germany
| | | | - Volker Siffrin
- Department of Neurology; University Medical Center Mainz; Johannes Gutenberg University Mainz; Mainz Germany
| | - Susann Peaschke
- Cellular Neurosciences; Max Delbrück Centre for Molecular Medicine; Berlin Germany
| | - Matthias Endres
- Department of Neurology; Charité, Universitätsmedizin Berlin; Charité Campus Virchow Berlin Germany
- Department of Neurology and Center for Stroke Research Berlin; Charité, Universitätsmedizin Berlin; Charité Campus Mitte Berlin Germany
| | - Frank Heppner
- Institute for Neuropathology; Charité, Universitätsmedizin Berlin; Charité Campus Mitte Berlin Germany
| | - Rainer Glass
- Neurosurgical Research; University Clinics Munich (LMU); Munich Germany
| | - Susanne A. Wolf
- Cellular Neurosciences; Max Delbrück Centre for Molecular Medicine; Berlin Germany
| | - Helmut Kettenmann
- Cellular Neurosciences; Max Delbrück Centre for Molecular Medicine; Berlin Germany
| |
Collapse
|
42
|
Song K, Na JY, Oh MH, Kim S, Kim YH, Park BY, Shin GW, Kim B, You M, Kwon J. Synthetic prion Peptide 106-126 resulted in an increase matrix metalloproteinases and inflammatory cytokines from rat astrocytes and microglial cells. Toxicol Res 2013; 28:5-9. [PMID: 24278583 PMCID: PMC3834397 DOI: 10.5487/tr.2012.28.1.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 03/12/2012] [Accepted: 03/21/2012] [Indexed: 12/22/2022] Open
Abstract
It has been shown that the accumulation of prion in the cytoplasm can result in neurodegenerative disorders. Synthetic prion peptide 106-126 (PrP) is a glycoprotein that is expressed predominantly by neurons and other cells, including glial cells. Prion-induced chronic neurodegeneration has a substantial inflammatory component, and an increase in the levels of matrix metalloproteinases (MMPs) may play an important role in neurodegenerative development and progression. However, the expression of MMPs in PrP induced rat astrocytes and microglia has not yet been compared. Thus, in this study, we examined the fluorescence intensity of CD11b positive microglia and Glial Fibrillary Acidic Protein (GFAP) positive astrocytes and found that the fluorescent intensity was increased following incubation with PrP at 24 hours in a dose-dependent manner. We also observed an increase in interleukin-1 beta (IL-1β) and tumor necrosis factor alpha (TNF-α) protein expression, which are initial inflammatory cytokines, in both PrP induced astrocytes and microglia. Furthermore, an increase MMP-1, 3 and 11 expressions in PrP induced astrocytes and microglia was observed by real time PCR. Our results demonstrated PrP induced activation of astrocytes and microglia respectively, which resulted in an increase in inflammatory cytokines and MMPs expression. These results provide the insight into the different sensitivities of glial cells to PrP.
Collapse
Affiliation(s)
- Kibbeum Song
- Bio-Safety Research Institute, College of Veterinary Medicine, Chonbuk National University, Jeonju 561-156, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Petters C, Dringen R. Comparison of primary and secondary rat astrocyte cultures regarding glucose and glutathione metabolism and the accumulation of iron oxide nanoparticles. Neurochem Res 2013; 39:46-58. [PMID: 24190598 DOI: 10.1007/s11064-013-1189-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Revised: 10/24/2013] [Accepted: 10/25/2013] [Indexed: 12/31/2022]
Abstract
Astrocyte-rich primary cultures (APCs) are frequently used as a model system for the investigation of properties of brain astrocytes. However, as APCs contain a substantial number of microglial and oligodendroglial cells, biochemical parameters determined for such cultures may at least in part reflect also the presence of the contaminating cell types. To lower the potential contributions of microglial and oligodendroglial cells on properties of the astrocytes in APCs we prepared rat astrocyte-rich secondary cultures (ASCs) by subculturing of APCs and compared these ASCs with APCs regarding basal metabolic parameters, specific enzyme activities and the accumulation of iron oxide nanoparticles. Immunocytochemical characterization revealed that ASCs contained only minute amounts of microglial and oligodendroglial cells. ASCs and APCs did not significantly differ in their specific glucose consumption and lactate production rates, in their specific iron and glutathione contents, in their specific activities of various enzymes involved in glucose and glutathione metabolism nor in their accumulation of iron oxide nanoparticles. Thus, the absence or presence of some contaminating microglial and oligodendroglial cells appears not to substantially modulate the investigated metabolic parameters of astrocyte cultures.
Collapse
Affiliation(s)
- Charlotte Petters
- Center for Biomolecular Interactions Bremen, University of Bremen, PO. Box 330440, 28334, Bremen, Germany
| | | |
Collapse
|
44
|
Weaver-Mikaere L, Gunn AJ, Bennet L, Mitchell MD, Fraser M. Inhibition of matrix metalloproteinases-2/-9 transiently reduces pre-oligodendrocyte loss during lipopolysaccharide- but not tumour necrosis factor-alpha-induced inflammation in fetal ovine glial culture. Dev Neurosci 2013; 35:461-73. [PMID: 24193164 DOI: 10.1159/000354862] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 08/07/2013] [Indexed: 11/19/2022] Open
Abstract
To determine whether increased matrix metalloproteinase (MMP) proteolytic activity plays a pathological role in infection/inflammation-induced preterm brain injury, primary cultures of preterm (day 90 of gestation; term 145 days) fetal ovine mixed glia were exposed to 24-96 h of lipopolysaccharide (LPS, 1 μg/ml) or tumour necrosis factor-α (TNF-α, 100 ng/ml). MMP-2 mRNA levels were significantly increased after TNF-α (96 h) and LPS exposure (48 and 96 h), and MMP-9 mRNA levels were significantly increased at 48 and 96 h after TNF-α. On zymography, the active form of secreted MMP-2 was significantly increased 24 h after LPS, but not TNF-α. Both active and latent forms of MMP-9 gelatinolytic activity were significantly increased by TNF-α (96 h) and LPS (72 and 96 h). On reverse zymography, inhibitory activity of TIMP-1 but not TIMP-2 was significantly increased by TNF-α and LPS. SB-3CT-mediated MMP-2 and MMP-9 inhibition transiently reduced LPS-induced oligodendrocyte cell death but had no effect during TNF-α exposure. Collectively, these observations suggest a limited, transient effect of MMPs on immature white matter damage associated with infection but not TNF-α-mediated inflammation.
Collapse
|
45
|
Lively S, Schlichter LC. The microglial activation state regulates migration and roles of matrix-dissolving enzymes for invasion. J Neuroinflammation 2013; 10:75. [PMID: 23786632 PMCID: PMC3693964 DOI: 10.1186/1742-2094-10-75] [Citation(s) in RCA: 147] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 06/06/2013] [Indexed: 12/30/2022] Open
Abstract
Background Microglial cells are highly mobile under many circumstances and, after central nervous system (CNS) damage, they must contend with the dense extracellular matrix (ECM) in order to reach their target sites. In response to damage or disease, microglia undergo complex activation processes that can be modulated by environmental cues and culminate in either detrimental or beneficial outcomes. Thus, there is considerable interest in comparing their pro-inflammatory (‘classical’ activation) and resolving ‘alternative’ activation states. Almost nothing is known about how these activation states affect the ability of microglia to migrate and degrade ECM, or the enzymes used for substrate degradation. This is the subject of the present study. Methods Primary cultured rat microglial cells were exposed to lipopolysaccharide (LPS) to evoke classical activation or IL4 to evoke alternative activation. High-resolution microscopy was used to monitor changes in cell morphology and aspects of the cytoskeleton. We quantified migration in a scratch-wound assay and through open filter holes, and invasion through Matrigel™. A panel of inhibitors was used to analyze contributions of different matrix-degrading enzymes to migration and invasion, and quantitative real-time reverse transcriptase PCR (qRT-PCR) was used to assess changes in their expression. Results Vinculin- and F-actin-rich lamellae were prominent in untreated and IL4-treated microglia (but not after LPS). IL4 increased the migratory capacity of microglia but eliminated the preferential anterior nuclear-centrosomal axis polarity and location of the microtubule organizing center (MTOC). Microglia degraded fibronectin, regardless of treatment, but LPS-treated cells were relatively immobile and IL4-treated cells invaded much more effectively through Matrigel™. For invasion, untreated microglia primarily used cysteine proteases, but IL4-treated cells used a wider range of enzymes (cysteine proteases, cathepsin S and K, heparanase, and matrix metalloproteases). Untreated microglia expressed MMP2, MMP12, heparanase, and four cathepsins (B, K, L1, and S). Each activation stimulus upregulated a different subset of enzymes. IL4 increased MMP2 and cathepsins S and K; whereas LPS increased MMP9, MMP12, MMP14 (MT1-MMP), heparanase, and cathepsin L1. Conclusions Microglial cells migrate during CNS development and after CNS damage or disease. Thus, there are broad implications of the finding that classically and alternatively activated microglia differ in morphology, cytoskeleton, migratory and invasive capacity, and in the usage of ECM-degrading enzymes.
Collapse
Affiliation(s)
- Starlee Lively
- Toronto Western Research Institute, Room MC9-417, 399 Bathurst Street, Toronto, ON M5T 2S8, Canada
| | | |
Collapse
|
46
|
Schwartz L, Spitsin SV, Meshki J, Tuluc F, Douglas SD, Wolfe JH. Substance P enhances HIV-1 infection in human fetal brain cell cultures expressing full-length neurokinin-1 receptor. J Neurovirol 2013; 19:219-27. [PMID: 23765222 PMCID: PMC3719168 DOI: 10.1007/s13365-013-0166-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 04/05/2013] [Accepted: 04/16/2013] [Indexed: 12/12/2022]
Abstract
The associations between the neurokinin-1 receptor (NK-1R), substance P (SP), and HIV-1 were investigated in neurosphere-derived cultures of microglial-depleted human fetal brain cells (HFBC). Full-length NK-1R was identified in HFBC cultures. SP treatment of the HFBC increased intracellular calcium mobilization and decreased electrical impedance, both of which were blocked by the NK-1R antagonist aprepitant. SP treatment of HIV-1-infected HFBC upregulated HIV-1 expression. These data show that human neural cells grown from neurospheres express functional full length NK-1R that is responsive to SP, and that SP enhanced HIV-1 infection in HBFC.
Collapse
Affiliation(s)
- Lynnae Schwartz
- Research Institute, Children's Hospital of Philadelphia, Suite 1208, 3615 Civic Center Boulevard, Philadelphia, PA 19104, USA.
| | | | | | | | | | | |
Collapse
|
47
|
Neria F, del Carmen Serrano-Perez M, Velasco P, Urso K, Tranque P, Cano E. NFATc3 promotes Ca(2+) -dependent MMP3 expression in astroglial cells. Glia 2013; 61:1052-66. [PMID: 23625833 DOI: 10.1002/glia.22494] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 02/14/2013] [Indexed: 12/30/2022]
Abstract
Increase in intracellular calcium ([Ca(2+) ]i ) is a key mediator of astrocyte signaling, important for activation of the calcineurin (CN)/nuclear factor of activated T cells (NFAT) pathway, a central mediator of inflammatory events. We analyzed the expression of matrix metalloproteinase 3 (Mmp3) in response to increases in [Ca(2+) ]i and the role of the CN/NFAT pathway in this regulation. Astrocyte Mmp3 expression was induced by overexpression of a constitutively active form of NFATc3, whereas other MMPs and tissue inhibitor of metalloproteinases (TIMP) were unaffected. Mmp3 mRNA and protein expression was also induced by calcium ionophore (Io) and 2'(3')-O-(4-benzoylbenzoyl) adenosine 5'-triphosphate (Bz-ATP) and Mmp3 upregulation was prevented by the CN inhibitor cyclosporin A (CsA). Ca(2+) -dependent astrocyte Mmp3 expression was also inhibited by actinomycin D, and a Mmp3 promoter luciferase reporter was efficiently activated by increased [Ca(2+) ]i , indicating regulation at the transcriptional level. Furthermore, Ca(2+) /CN/NFAT dependent Mmp3 expression was confirmed in pure astrocyte cultures derived from neural stem cells (Ast-NSC), demonstrating that the induced Mmp3 expression occurs in astrocytes, and not microglial cells. In an in vivo stab-wound model of brain injury, MMP3 expression was detected in NFATc3-positive scar-forming astrocytes. Because [Ca(2+) ]i increase is an early event in most brain injuries, these data support an important role for Ca(2+) /CN/NFAT-induced astrocyte MMP3 expression in the early neuroinflammatory response. Understanding the molecular pathways involved in this regulation could provide novel therapeutic targets and approaches to promoting recovery of the injured brain.
Collapse
Affiliation(s)
- Fernando Neria
- Unidad de Neuroinflamación, Área de Biología Celular y del Desarrollo, Unidad Funcional de Investigación en Enfermedades Crónicas, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
48
|
Welser-Alves JV, Milner R. Microglia are the major source of TNF-α and TGF-β1 in postnatal glial cultures; regulation by cytokines, lipopolysaccharide, and vitronectin. Neurochem Int 2013; 63:47-53. [PMID: 23619393 DOI: 10.1016/j.neuint.2013.04.007] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 03/22/2013] [Accepted: 04/15/2013] [Indexed: 12/15/2022]
Abstract
Damage to the central nervous system (CNS) leads to increased production of TNF-α and TGF-β1 cytokines that have pro- or anti-inflammatory actions, respectively. To define whether astrocytes or microglia express these cytokines, prior studies have used mixed glial cultures (MGC) to represent astrocytes, thought these results are inevitably complicated by the presence of contaminating microglia within MGC. To clarify the cellular source of these cytokines, here we employed a recently described method of preparing microglia-free astrocyte cultures, in which neural stem cells (NSC) are differentiated into astrocytes. Using ELISA to quantify cytokine production in three types of glial culture: MGC, pure microglia or pure astrocytes, this showed that microglia but not astrocytes, produce TNF-α, and that this expression is increased by LPS, IFN-γ, and to a lesser extent by vitronectin, but decreased by TGF-β1. In contrast, TGF-β1 was produced by microglia and astrocytes, though at 10-fold higher levels by microglia. TGF-β1 expression in microglia was increased by vitronectin and to a lesser extent by TNF-α and LPS, but astrocyte TGF-β1 expression was not regulated by any factor tested. In summary, our data reveal that microglia, not astrocytes are the major source of TNF-α and TGF-β1 in postnatal glial cultures, and that microglial production of these antagonistic cytokines is tightly regulated by cytokines, LPS, and vitronectin.
Collapse
Affiliation(s)
- Jennifer V Welser-Alves
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | |
Collapse
|
49
|
Liu Y, Zhang M, Hao W, Mihaljevic I, Liu X, Xie K, Walter S, Fassbender K. Matrix metalloproteinase-12 contributes to neuroinflammation in the aged brain. Neurobiol Aging 2013; 34:1231-9. [DOI: 10.1016/j.neurobiolaging.2012.10.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Revised: 10/09/2012] [Accepted: 10/19/2012] [Indexed: 01/02/2023]
|
50
|
Sinno M, Biagioni S, Ajmone-Cat MA, Pafumi I, Caramanica P, Medda V, Tonti G, Minghetti L, Mannello F, Cacci E. The matrix metalloproteinase inhibitor marimastat promotes neural progenitor cell differentiation into neurons by gelatinase-independent TIMP-2-dependent mechanisms. Stem Cells Dev 2012; 22:345-58. [PMID: 23098139 DOI: 10.1089/scd.2012.0299] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Metalloproteinases (MMPs) and their endogenous inhibitors (TIMPs), produced in the brain by cells of non-neural and neural origin, including neural progenitors (NPs), are emerging as regulators of nervous system development and adult brain functions. In the present study, we explored whether MMP-2, MMP-9, and TIMP-2, abundantly produced in the brain, modulate NP developmental properties. We found that treatment of NPs, isolated from the murine fetal cerebral cortex or adult subventricular zone, with the clinically tested broad-spectrum MMP inhibitor Marimastat profoundly affected the NP differentiation fate. Marimastat treatment allowed for an enrichment of our cultures in neuronal cells, inducing NPs to generate higher percentage of neurons and a lower percentage of astrocytes, possibly affecting NP commitment. Consistently with its proneurogenic effect, Marimastat early downregulated the expression of Notch target genes, such as Hes1 and Hes5. MMP-2 and MMP-9 profiling on proliferating and differentiating NPs revealed that MMP-9 was not expressed under these conditions, whereas MMP-2 increased in the medium as pro-MMP-2 (72 kDa) during differentiation; its active form (62 kDa) was not detectable by gel zymography. MMP-2 silencing or administration of recombinant active MMP-2 demonstrated that MMP-2 does not affect NP neuronal differentiation, nor it is involved in the Marimastat proneurogenic effect. We also found that TIMP-2 is expressed in NPs and increases during late differentiation, mainly as a consequence of astrocyte generation. Endogenous TIMP-2 did not modulate NP neurogenic potential; however, the proneurogenic action of Marimastat was mediated by TIMP-2, as demonstrated by silencing experiments. In conclusion, our data exclude a major involvement of MMP-2 and MMP-9 in the regulation of basal NP differentiation, but highlight the ability of TIMP-2 to act as key effector of the proneurogenic response to an inducing stimulus such as Marimastat.
Collapse
Affiliation(s)
- Maddalena Sinno
- Department of Biology and Biotechnology Charles Darwin, Sapienza, University of Rome, Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|