101
|
Li X, Liu H, Han C, Luo J, Guan X, Wang L, Li Y, Wang J, Piao H, Zou W, Liu J. A Human Brain Model Mimicking Umbilical Cord Mesenchymal Stem Cells for the Treatment of Hypoxic-Ischemic Brain Injury. Int J Mol Sci 2023; 24:14208. [PMID: 37762511 PMCID: PMC10532043 DOI: 10.3390/ijms241814208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
We used an in vitro model of the human brain immune microenvironment to simulate hypoxic-ischemic brain injury (HIBI) and treatment with human umbilical cord mesenchymal stem cells (hUMSCs) to address the transformation barriers of gene differences between animals and humans in preclinical research. A co-culture system, termed hNAME, consisted of human hippocampal neurons (N), astrocytes (A), microglia (M), and brain microvascular endothelial cells (E). Flow cytometry measured the apoptosis rates of neurons and endothelial cells. hNAME-neurons and endothelial cells experienced more severe damage than monolayer cells, particularly after 48 h and 24 h of reoxygenation (OGD48/R24). Western blotting identified neuroinflammatory response markers, including HIF-1α, C1q, C3, TNF-α, and iNOS. Inflammatory factors originated from the glial chamber rather than the neurons and vascular endothelial chambers. A gradual increase in the release of inflammatory factors was observed as the OGD and reoxygenation times increased, peaking at OGD48/R24. The hNAME value was confirmed in human umbilical cord mesenchymal stem cells (hUMSCs). Treatment with hUMSCs resulted in a notable decrease in the severity of neuronal and endothelial cell damage in hNAME. The hNAME is an ideal in vitro model for simulating the immune microenvironment of the human brain because of the interactions between neurons, vessels, astrocytes, and microglia.
Collapse
Affiliation(s)
- Xidan Li
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (X.L.); (H.L.); (C.H.); (J.L.); (X.G.); (L.W.); (Y.L.); (J.W.)
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Engineering Research Center for Genetic Variation Detection of Infectious Pathogenic Microorganisms, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116085, China; (H.P.); (W.Z.)
| | - Haijing Liu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (X.L.); (H.L.); (C.H.); (J.L.); (X.G.); (L.W.); (Y.L.); (J.W.)
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Engineering Research Center for Genetic Variation Detection of Infectious Pathogenic Microorganisms, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116085, China; (H.P.); (W.Z.)
| | - Chao Han
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (X.L.); (H.L.); (C.H.); (J.L.); (X.G.); (L.W.); (Y.L.); (J.W.)
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Engineering Research Center for Genetic Variation Detection of Infectious Pathogenic Microorganisms, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116085, China; (H.P.); (W.Z.)
| | - Jianglin Luo
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (X.L.); (H.L.); (C.H.); (J.L.); (X.G.); (L.W.); (Y.L.); (J.W.)
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Engineering Research Center for Genetic Variation Detection of Infectious Pathogenic Microorganisms, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116085, China; (H.P.); (W.Z.)
| | - Xin Guan
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (X.L.); (H.L.); (C.H.); (J.L.); (X.G.); (L.W.); (Y.L.); (J.W.)
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Engineering Research Center for Genetic Variation Detection of Infectious Pathogenic Microorganisms, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116085, China; (H.P.); (W.Z.)
| | - Liang Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (X.L.); (H.L.); (C.H.); (J.L.); (X.G.); (L.W.); (Y.L.); (J.W.)
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Engineering Research Center for Genetic Variation Detection of Infectious Pathogenic Microorganisms, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116085, China; (H.P.); (W.Z.)
| | - Ying Li
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (X.L.); (H.L.); (C.H.); (J.L.); (X.G.); (L.W.); (Y.L.); (J.W.)
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Engineering Research Center for Genetic Variation Detection of Infectious Pathogenic Microorganisms, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116085, China; (H.P.); (W.Z.)
| | - Jiayi Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (X.L.); (H.L.); (C.H.); (J.L.); (X.G.); (L.W.); (Y.L.); (J.W.)
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Engineering Research Center for Genetic Variation Detection of Infectious Pathogenic Microorganisms, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116085, China; (H.P.); (W.Z.)
| | - Hua Piao
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Engineering Research Center for Genetic Variation Detection of Infectious Pathogenic Microorganisms, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116085, China; (H.P.); (W.Z.)
- College of Basic Medical Science, Dalian Medical University, Dalian 116027, China
| | - Wei Zou
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Engineering Research Center for Genetic Variation Detection of Infectious Pathogenic Microorganisms, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116085, China; (H.P.); (W.Z.)
| | - Jing Liu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (X.L.); (H.L.); (C.H.); (J.L.); (X.G.); (L.W.); (Y.L.); (J.W.)
- Liaoning Key Laboratory of Frontier Technology of Stem Cell and Precision Medicine, Dalian Engineering Research Center for Genetic Variation Detection of Infectious Pathogenic Microorganisms, Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian 116085, China; (H.P.); (W.Z.)
| |
Collapse
|
102
|
Kong F, Wu T, Dai J, Zhai Z, Cai J, Zhu Z, Xu Y, Sun T. Glucagon-like peptide 1 (GLP-1) receptor agonists in experimental Alzheimer's disease models: a systematic review and meta-analysis of preclinical studies. Front Pharmacol 2023; 14:1205207. [PMID: 37771725 PMCID: PMC10525376 DOI: 10.3389/fphar.2023.1205207] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 09/04/2023] [Indexed: 09/30/2023] Open
Abstract
Alzheimer's disease (AD) is a degenerative disease of the nervous system. Glucagon-like peptide-1 receptor agonists (GLP-1 RAs), a drug used to treat type 2 diabetes, have been shown to have neuroprotective effects. This systematic review and meta-analysis evaluated the effects and potential mechanisms of GLP-1 RAs in AD animal models. 26 studies were included by searching relevant studies from seven databases according to a predefined search strategy and inclusion criteria. Methodological quality was assessed using SYRCLE's risk of bias tool, and statistical analysis was performed using ReviewManger 5.3. The results showed that, in terms of behavioral tests, GLP-1 RAs could improve the learning and memory abilities of AD rodents; in terms of pathology, GLP-1 RAs could reduce Aβ deposition and phosphorylated tau levels in the brains of AD rodents. The therapeutic potential of GLP-1 RAs in AD involves a range of mechanisms that work synergistically to enhance the alleviation of various pathological manifestations associated with the condition. A total of five clinical trials were retrieved from ClinicalTrials.gov. More large-scale and high-quality preclinical trials should be conducted to more accurately assess the therapeutic effects of GLP-1 RAs on AD.
Collapse
Affiliation(s)
- Fanjing Kong
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tianyu Wu
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jingyi Dai
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhenwei Zhai
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jie Cai
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhishan Zhu
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying Xu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tao Sun
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
103
|
Yan L, Xuan FL, Chen S, Gou M, Chen W, Li Y, Wang Z, Wang L, Xie T, Fan F, Zharkovsky A, Tan Y, Tian L. Replenished microglia partially rescue schizophrenia-related stress response. Front Cell Neurosci 2023; 17:1254923. [PMID: 37771931 PMCID: PMC10522857 DOI: 10.3389/fncel.2023.1254923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 08/15/2023] [Indexed: 09/30/2023] Open
Abstract
Background Microglia play an important role in the maintenance of brain and behavioral homeostasis. The protective effect of microglial replenishment was reported in neurological diseases, but whether microglial therapy would benefit psychiatric disorders such as schizophrenia has been unclear. As schizophrenia is a stress-vulnerable disorder and psychosocial stress promotes inflammation and microglial activation, we aim to understand how microglial replenishment works in stress-associated schizophrenia. Methods We used a CSF1R-mediated pharmacological approach to study repopulated microglia (repMg) in a cohort of mice (n = 10/group) undergoing chronic unpredictable stress (CUS). We further studied a cohort of first-episode schizophrenia (FES, n = 74) patients who had higher perceived stress scores (PSS) than healthy controls (HCs, n = 68). Results Reborn microglia attenuated CUS-induced learned hopelessness and social withdrawal but not anxiety in mice. Compared to control, CUS- or repMg-induced differentially expressed genes (DEGs) in the prefrontal cortex regulated nervous system development and axonal guidance. CUS also caused microglial hyper-ramification and increased engulfment of synaptophysin and vesicular glutamate transporter-2 by microglia and astrocytes, which were recovered in CUS + repMg (all p < 0.05). Moreover, FES patients had smaller hippocampal fimbria than HCs (p < 1e-7), which were negatively associated with PSS (r = -0.397, p = 0.003). Blood DEGs involved in immune system development were also associated with PSS and the right fimbria more prominently in FES patients than HCs (Zr, p < 0.0001). The KCNQ1 was a partial mediator between PSS and fimbria size (β = -0.442, 95% CI: -1.326 ~ -0.087). Conclusion Microglial replenishment may potentially benefit psychiatric disorders such as schizophrenia.
Collapse
Affiliation(s)
- Ling Yan
- Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia
| | - Fang-Ling Xuan
- Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia
| | - Song Chen
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China
| | - Mengzhuang Gou
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China
| | - Wenjin Chen
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China
| | - Yanli Li
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China
| | - Zhiren Wang
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China
| | - Leilei Wang
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China
| | - Ting Xie
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China
| | - Fengmei Fan
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China
| | - Alexander Zharkovsky
- Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia
| | - Yunlong Tan
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China
| | - Li Tian
- Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia
- Psychiatry Research Centre, Beijing Huilongguan Hospital, Peking University Health Science Center, Beijing, China
| |
Collapse
|
104
|
Lana D, Branca JJV, Delfino G, Giovannini MG, Casamenti F, Nardiello P, Bucciantini M, Stefani M, Zach P, Zecchi-Orlandini S, Nosi D. Morphofunctional Investigation in a Transgenic Mouse Model of Alzheimer's Disease: Non-Reactive Astrocytes Are Involved in Aβ Load and Reactive Astrocytes in Plaque Build-Up. Cells 2023; 12:2258. [PMID: 37759482 PMCID: PMC10526848 DOI: 10.3390/cells12182258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
The term neuroinflammation defines the reactions of astrocytes and microglia to alterations in homeostasis in the diseased central nervous system (CNS), the exacerbation of which contributes to the neurodegenerative effects of Alzheimer's disease (AD). Local environmental conditions, such as the presence of proinflammatory molecules, mechanical properties of the extracellular matrix (ECM), and local cell-cell interactions, are determinants of glial cell phenotypes. In AD, the load of the cytotoxic/proinflammatory amyloid β (Aβ) peptide is a microenvironmental component increasingly growing in the CNS, imposing time-evolving challenges on resident cells. This study aimed to investigate the temporal and spatial variations of the effects produced by this process on astrocytes and microglia, either directly or by interfering in their interactions. Ex vivo confocal analyses of hippocampal sections from the mouse model TgCRND8 at different ages have shown that overproduction of Aβ peptide induced early and time-persistent disassembly of functional astroglial syncytium and promoted a senile phenotype of reactive microglia, hindering Aβ clearance. In the late stages of the disease, these patterns were altered in the presence of Aβ-plaques, surrounded by typically reactive astrocytes and microglia. Morphofunctional characterization of peri-plaque gliosis revealed a direct contribution of astrocytes in plaque buildup that might result in shielding Aβ-peptide cytotoxicity and, as a side effect, in exacerbating neuroinflammation.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, University of Florence, 50134 Florence, Italy; (D.L.); (M.G.G.)
| | - Jacopo Junio Valerio Branca
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (J.J.V.B.); (S.Z.-O.)
| | - Giovanni Delfino
- Department of Biology, University of Florence, 50121 Florence, Italy;
| | - Maria Grazia Giovannini
- Department of Health Sciences, University of Florence, 50134 Florence, Italy; (D.L.); (M.G.G.)
| | - Fiorella Casamenti
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, 50134 Florence, Italy;
| | - Pamela Nardiello
- General Laboratory, Careggi University Hospital, 50134 Florence, Italy;
| | - Monica Bucciantini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (M.B.); (M.S.)
| | - Massimo Stefani
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, 50134 Florence, Italy; (M.B.); (M.S.)
| | - Petr Zach
- Department of Anatomy, Third Faculty of Medicine, Charles University, 100 00 Prague, Czech Republic;
| | - Sandra Zecchi-Orlandini
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (J.J.V.B.); (S.Z.-O.)
| | - Daniele Nosi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (J.J.V.B.); (S.Z.-O.)
- DMSC Imaging Platform, 50134 Florence, Italy
| |
Collapse
|
105
|
Novakovic MM, Korshunov KS, Grant RA, Martin ME, Valencia HA, Budinger GRS, Radulovic J, Prakriya M. Astrocyte reactivity and inflammation-induced depression-like behaviors are regulated by Orai1 calcium channels. Nat Commun 2023; 14:5500. [PMID: 37679321 PMCID: PMC10485021 DOI: 10.1038/s41467-023-40968-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023] Open
Abstract
Astrocytes contribute to brain inflammation in neurological disorders but the molecular mechanisms controlling astrocyte reactivity and their relationship to neuroinflammatory endpoints are complex and poorly understood. In this study, we assessed the role of the calcium channel, Orai1, for astrocyte reactivity and inflammation-evoked depression behaviors in mice. Transcriptomics and metabolomics analysis indicated that deletion of Orai1 in astrocytes downregulates genes in inflammation and immunity, metabolism, and cell cycle pathways, and reduces cellular metabolites and ATP production. Systemic inflammation by peripheral lipopolysaccharide (LPS) increases hippocampal inflammatory markers in WT but not in astrocyte Orai1 knockout mice. Loss of Orai1 also blunts inflammation-induced astrocyte Ca2+ signaling and inhibitory neurotransmission in the hippocampus. In line with these cellular changes, Orai1 knockout mice showed amelioration of LPS-evoked depression-like behaviors including anhedonia and helplessness. These findings identify Orai1 as an important signaling hub controlling astrocyte reactivity and astrocyte-mediated brain inflammation that is commonly observed in many neurological disorders.
Collapse
Affiliation(s)
- Michaela M Novakovic
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Kirill S Korshunov
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Rogan A Grant
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Megan E Martin
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Hiam A Valencia
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - G R Scott Budinger
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Jelena Radulovic
- Department of Neuroscience, Albert Einstein School of Medicine, Bronx, NY, 10461, USA
| | - Murali Prakriya
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
106
|
Lana D, Magni G, Landucci E, Wenk GL, Pellegrini-Giampietro DE, Giovannini MG. Phenomic Microglia Diversity as a Druggable Target in the Hippocampus in Neurodegenerative Diseases. Int J Mol Sci 2023; 24:13668. [PMID: 37761971 PMCID: PMC10531074 DOI: 10.3390/ijms241813668] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/31/2023] [Accepted: 09/02/2023] [Indexed: 09/29/2023] Open
Abstract
Phenomics, the complexity of microglia phenotypes and their related functions compels the continuous study of microglia in disease animal models to find druggable targets for neurodegenerative disorders. Activation of microglia was long considered detrimental for neuron survival, but more recently it has become apparent that the real scenario of microglia morphofunctional diversity is far more complex. In this review, we discuss the recent literature on the alterations in microglia phenomics in the hippocampus of animal models of normal brain aging, acute neuroinflammation, ischemia, and neurodegenerative disorders, such as AD. Microglia undergo phenomic changes consisting of transcriptional, functional, and morphological changes that transform them into cells with different properties and functions. The classical subdivision of microglia into M1 and M2, two different, all-or-nothing states is too simplistic, and does not correspond to the variety of phenotypes recently discovered in the brain. We will discuss the phenomic modifications of microglia focusing not only on the differences in microglia reactivity in the diverse models of neurodegenerative disorders, but also among different areas of the brain. For instance, in contiguous and highly interconnected regions of the rat hippocampus, microglia show a differential, finely regulated, and region-specific reactivity, demonstrating that microglia responses are not uniform, but vary significantly from area to area in response to insults. It is of great interest to verify whether the differences in microglia reactivity may explain the differential susceptibility of different brain areas to insults, and particularly the higher sensitivity of CA1 pyramidal neurons to inflammatory stimuli. Understanding the spatiotemporal heterogeneity of microglia phenomics in health and disease is of paramount importance to find new druggable targets for the development of novel microglia-targeted therapies in different CNS disorders. This will allow interventions in three different ways: (i) by suppressing the pro-inflammatory properties of microglia to limit the deleterious effect of their activation; (ii) by modulating microglia phenotypic change to favor anti-inflammatory properties; (iii) by influencing microglia priming early in the disease process.
Collapse
Affiliation(s)
- Daniele Lana
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Giada Magni
- Institute of Applied Physics “Nello Carrara”, National Research Council (IFAC-CNR), Via Madonna del Piano 10, 50019 Florence, Italy;
| | - Elisa Landucci
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Gary L. Wenk
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA;
| | - Domenico Edoardo Pellegrini-Giampietro
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| | - Maria Grazia Giovannini
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Viale Pieraccini 6, 50139 Florence, Italy; (E.L.); (D.E.P.-G.); (M.G.G.)
| |
Collapse
|
107
|
Panchenko PE, Hippauf L, Konsman JP, Badaut J. Do astrocytes act as immune cells after pediatric TBI? Neurobiol Dis 2023; 185:106231. [PMID: 37468048 PMCID: PMC10530000 DOI: 10.1016/j.nbd.2023.106231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/28/2023] [Accepted: 07/15/2023] [Indexed: 07/21/2023] Open
Abstract
Astrocytes are in contact with the vasculature, neurons, oligodendrocytes and microglia, forming a local network with various functions critical for brain homeostasis. One of the primary responders to brain injury are astrocytes as they detect neuronal and vascular damage, change their phenotype with morphological, proteomic and transcriptomic transformations for an adaptive response. The role of astrocytic responses in brain dysfunction is not fully elucidated in adult, and even less described in the developing brain. Children are vulnerable to traumatic brain injury (TBI), which represents a leading cause of death and disability in the pediatric population. Pediatric brain trauma, even with mild severity, can lead to long-term health complications, such as cognitive impairments, emotional disorders and social dysfunction later in life. To date, the underlying pathophysiology is still not fully understood. In this review, we focus on the astrocytic response in pediatric TBI and propose a potential immune role of the astrocyte in response to trauma. We discuss the contribution of astrocytes in the local inflammatory cascades and secretion of various immunomodulatory factors involved in the recruitment of local microglial cells and peripheral immune cells through cerebral blood vessels. Taken together, we propose that early changes in the astrocytic phenotype can alter normal development of the brain, with long-term consequences on neurological outcomes, as described in preclinical models and patients.
Collapse
Affiliation(s)
| | - Lea Hippauf
- CNRS UMR 5536 RMSB-University of Bordeaux, Bordeaux, France
| | | | - Jerome Badaut
- CNRS UMR 5536 RMSB-University of Bordeaux, Bordeaux, France; Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| |
Collapse
|
108
|
Tsesmelis K, Maity‐Kumar G, Croner D, Sprissler J, Tsesmelis M, Hein T, Baumann B, Wirth T. Accelerated aging in mice with astrocytic redox imbalance as a consequence of SOD2 deletion. Aging Cell 2023; 22:e13911. [PMID: 37609868 PMCID: PMC10497807 DOI: 10.1111/acel.13911] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/08/2023] [Accepted: 05/31/2023] [Indexed: 08/24/2023] Open
Abstract
Aging of the central nervous system (CNS) leads to motoric and cognitive decline and increases the probability for neurodegenerative disease development. Astrocytes fulfill central homeostatic functions in the CNS including regulation of immune responses and metabolic support of neurons and oligodendrocytes. In this study, we investigated the effect of redox imbalance in astrocytes by using a conditional astrocyte-specific SOD2-deficient mouse model (SOD2ako ) and analyzed these animals at different stages of their life. SOD2ako mice did not exhibit any overt phenotype within the first postnatal weeks. However, already as young adults, they displayed progressive motoric impairments. Moreover, as these mice grew older, they exhibited signs of a progeroid phenotype and early death. Histological analysis in moribund SOD2ako mice revealed the presence of age-related brain alterations, neuroinflammation, neuronal damage and myelin impairment in brain and spinal cord. Additionally, transcriptome analysis of primary astrocytes revealed that SOD2 deletion triggered a hypometabolic state and promoted polarization toward A1-neurotoxic status, possibly underlying the neuronal and myelin deficits. Conclusively, our study identifies maintenance of ROS homeostasis in astrocytes as a critical prerequisite for physiological CNS aging.
Collapse
Affiliation(s)
| | - Gandhari Maity‐Kumar
- Institute of Physiological ChemistryUniversity of UlmUlmGermany
- Institute for Diabetes and ObesityHelmholtz Diabetes Center at Helmholtz Zentrum MünchenNeuherbergGermany
| | - Dana Croner
- Institute of Physiological ChemistryUniversity of UlmUlmGermany
| | - Jasmin Sprissler
- Institute of Physiological ChemistryUniversity of UlmUlmGermany
- Department of Pediatrics and Adolescent MedicineUlm University Medical CenterUlmGermany
| | | | - Tabea Hein
- Institute of Physiological ChemistryUniversity of UlmUlmGermany
| | - Bernd Baumann
- Institute of Physiological ChemistryUniversity of UlmUlmGermany
| | - Thomas Wirth
- Institute of Physiological ChemistryUniversity of UlmUlmGermany
| |
Collapse
|
109
|
Zelenka L, Jarek M, Pägelow D, Geffers R, van Vorst K, Fulde M. Crosstalk of Highly Purified Microglia and Astrocytes in the Frame of Toll-like Receptor (TLR)2/1 Activation. Neuroscience 2023; 526:256-266. [PMID: 37391121 DOI: 10.1016/j.neuroscience.2023.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 04/26/2023] [Accepted: 05/02/2023] [Indexed: 07/02/2023]
Abstract
The major immune cells of the central nervous systems (CNS) are microglia and astrocytes, subsets of the glial cell population. The crosstalk between glia via soluble signaling molecules plays an indispensable role for neuropathologies, brain development as well as homeostasis. However, the investigation of the microglia-astrocyte crosstalk has been hampered due to the lack of suitable glial isolation methods. In this study, we investigated for the first time the crosstalk between highly purified Toll-like receptor (TLR)2-knock out (TLR2-KO) and wild-type (WT) microglia and astrocytes. We examined the crosstalk of TLR2-KO microglia and astrocytes in the presence of WT supernatants of the respective other glial cell type. Interestingly, we observed a significant TNF release by TLR2-KO astrocytes, which were activated with Pam3CSK4-stimulated WT microglial supernatants, strongly indicating a crosstalk between microglia and astrocytes after TLR2/1 activation. Furthermore, transcriptome analysis using RNA-seq revealed a wide range of significant up- and down-regulated genes such as Cd300, Tnfrsf9 or Lcn2, which might be involved in the molecular conversation between microglia and astrocytes. Finally, co-culturing microglia and astrocytes confirmed the prior results by demonstrating a significant TNF release by WT microglia co-cultured with TLR2-KO astrocytes. Our findings suggest a molecular TLR2/1-dependent conversation between highly pure activated microglia and astrocytes via signaling molecules. Furthermore, we demonstrate the first crosstalk experiments using ∼100% pure microglia and astrocyte mono-/co-cultures derived from mice with different genotypes highlighting the urgent need of efficient glial isolation protocols, which particularly holds true for astrocytes.
Collapse
Affiliation(s)
- Laura Zelenka
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, Berlin, Germany
| | - Michael Jarek
- Helmholtz Centre for Infection Research, Research Group Genome Analytics (GMAK), Braunschweig, Germany
| | - Dennis Pägelow
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, Berlin, Germany
| | - Robert Geffers
- Helmholtz Centre for Infection Research, Research Group Genome Analytics (GMAK), Braunschweig, Germany
| | - Kira van Vorst
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, Berlin, Germany
| | - Marcus Fulde
- Centre for Infection Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, Berlin, Germany.
| |
Collapse
|
110
|
Lee AG, Kang S, Yoon HJ, Im S, Oh SJ, Pak YK. Polystyrene Microplastics Exacerbate Systemic Inflammation in High-Fat Diet-Induced Obesity. Int J Mol Sci 2023; 24:12421. [PMID: 37569796 PMCID: PMC10419071 DOI: 10.3390/ijms241512421] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/13/2023] Open
Abstract
Microplastics (MPs) are recognized as environmental pollutants with potential implications for human health. Considering the rapid increase in obesity rates despite stable caloric intake, there is a growing concern about the link between obesity and exposure to environmental pollutants, including MPs. In this study, we conducted a comprehensive investigation utilizing in silico, in vitro, and in vivo approaches to explore the brain distribution and physiological effects of MPs. Molecular docking simulations were performed to assess the binding affinity of three plastic polymers (ethylene, propylene, and styrene) to immune cells (macrophages, CD4+, and CD8+ lymphocytes). The results revealed that styrene exhibited the highest binding affinity for macrophages. Furthermore, in vitro experiments employing fluorescence-labeled PS-MPs (fPS-MPs) of 1 μm at various concentrations demonstrated a dose-dependent binding of fPS-MPs to BV2 murine microglial cells. Subsequent oral administration of fPS-MPs to high-fat diet-induced obese mice led to the co-existence of fPS-MPs with immune cells in the blood, exacerbating impaired glucose metabolism and insulin resistance and promoting systemic inflammation. Additionally, fPS-MPs were detected throughout the brain, with increased activation of microglia in the hypothalamus. These findings suggest that PS-MPs significantly contribute to the exacerbation of systemic inflammation in high-fat diet-induced obesity by activating peripheral and central inflammatory immune cells.
Collapse
Affiliation(s)
| | - Sora Kang
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.K.); (H.J.Y.)
| | - Hye Ji Yoon
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.K.); (H.J.Y.)
| | - Suyeol Im
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447,Republic of Korea; (S.I.); (S.J.O.)
| | - Seung Jun Oh
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447,Republic of Korea; (S.I.); (S.J.O.)
| | - Youngmi Kim Pak
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.K.); (H.J.Y.)
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447,Republic of Korea; (S.I.); (S.J.O.)
- Department of Physiology, School of Medicine, Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
111
|
Çarçak N, Onat F, Sitnikova E. Astrocytes as a target for therapeutic strategies in epilepsy: current insights. Front Mol Neurosci 2023; 16:1183775. [PMID: 37583518 PMCID: PMC10423940 DOI: 10.3389/fnmol.2023.1183775] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 07/12/2023] [Indexed: 08/17/2023] Open
Abstract
Astrocytes are specialized non-neuronal glial cells of the central nervous system, contributing to neuronal excitability and synaptic transmission (gliotransmission). Astrocytes play a key roles in epileptogenesis and seizure generation. Epilepsy, as a chronic disorder characterized by neuronal hyperexcitation and hypersynchronization, is accompanied by substantial disturbances of glial cells and impairment of astrocytic functions and neuronal signaling. Anti-seizure drugs that provide symptomatic control of seizures primarily target neural activity. In epileptic patients with inadequate control of seizures with available anti-seizure drugs, novel therapeutic candidates are needed. These candidates should treat epilepsy with anti-epileptogenic and disease-modifying effects. Evidence from human and animal studies shows that astrocytes have value for developing new anti-seizure and anti-epileptogenic drugs. In this review, we present the key functions of astrocytes contributing to neuronal hyperexcitability and synaptic activity following an etiology-based approach. We analyze the role of astrocytes in both development (epileptogenesis) and generation of seizures (ictogenesis). Several promising new strategies that attempted to modify astroglial functions for treating epilepsy are being developed: (1) selective targeting of glia-related molecular mechanisms of glutamate transport; (2) modulation of tonic GABA release from astrocytes; (3) gliotransmission; (4) targeting the astrocytic Kir4.1-BDNF system; (5) astrocytic Na+/K+/ATPase activity; (6) targeting DNA hypo- or hypermethylation of candidate genes in astrocytes; (7) targeting astrocytic gap junction regulators; (8) targeting astrocytic adenosine kinase (the major adenosine-metabolizing enzyme); and (9) targeting microglia-astrocyte communication and inflammatory pathways. Novel disease-modifying therapeutic strategies have now been developed, such as astroglia-targeted gene therapy with a broad spectrum of genetic constructs to target astroglial cells.
Collapse
Affiliation(s)
- Nihan Çarçak
- Department of Pharmacology, Faculty of Pharmacy, Istanbul University, Istanbul, Turkey
- Institute of Health Sciences, Department of Neuroscience, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Filiz Onat
- Institute of Health Sciences, Department of Neuroscience, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
- Department of Medical Pharmacology, Faculty of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Evgenia Sitnikova
- Institute of Higher Nervous Activity and Neurophysiology of Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
112
|
Marghani BH, Rezk S, Ateya AI, Alotaibi BS, Othman BH, Sayed SM, Alshehri MA, Shukry M, Mansour MM. The Effect of Cerebrolysin in an Animal Model of Forebrain Ischemic-Reperfusion Injury: New Insights into the Activation of the Keap1/Nrf2/Antioxidant Signaling Pathway. Int J Mol Sci 2023; 24:12080. [PMID: 37569457 PMCID: PMC10418386 DOI: 10.3390/ijms241512080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Forebrain ischemia-reperfusion (IR) injury causes neurological impairments due to decreased cerebral autoregulation, hypoperfusion, and edema in the hours to days following the restoration of spontaneous circulation. This study aimed to examine the protective and/or therapeutic effects of cerebrolysin (CBL) in managing forebrain IR injury and any probable underlying mechanisms. To study the contribution of reperfusion to forebrain injury, we developed a transient dual carotid artery ligation (tDCAL/IR) mouse model. Five equal groups of six BLC57 mice were created: Group 1: control group (no surgery was performed); Group 2: sham surgery (surgery was performed without IR); Group 3: tDCAL/IR (surgery with IR via permanently ligating the left CA and temporarily closing the right CA for 30 min, followed by reperfusion for 72 h); Group 4: CBL + tDCAL/IR (CBL was given intravenously at a 60 mg/kg BW dose 30 min before IR); and Group 5: tDCAL/IR + CBL (CBL was administered i.v. at 60 mg/kg BW three hours after IR). At 72 h following IR, the mice were euthanized. CBL administration 3 h after IR improved neurological functional recovery, enhanced anti-inflammatory and antioxidant activities, alleviated apoptotic neuronal death, and inhibited reactive microglial and astrocyte activation, resulting in neuroprotection after IR injury in the tDCAL/IR + CBL mice group as compared to the other groups. Furthermore, CBL reduced the TLRs/NF-kB/cytokines while activating the Keap1/Nrf2/antioxidant signaling pathway. These results indicate that CBL may improve neurologic function in mice following IR.
Collapse
Affiliation(s)
- Basma H. Marghani
- Department of Physiology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
- Department of Biochemistry, Physiology, and Pharmacology, Faculty of Veterinary Medicine, King Salman International University, El Tor 46612, Egypt
| | - Shaymaa Rezk
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Ahmed I. Ateya
- Department of Husbandry and Development of Animal Wealth, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Badriyah S. Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Basma H. Othman
- Medical Experimental Research Center, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt;
| | - Samy M. Sayed
- Faculty of Agriculture, Cairo University, Giza 12613, Egypt;
- Department of Science and Technology, Ranyah University College, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Mohammed Ali Alshehri
- Biology Department, College of Science, University of Tabuk, Tabuk 71491, Saudi Arabia;
| | - Mustafa Shukry
- Physiology Department, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
| | - Mohamed M. Mansour
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
113
|
Sun M, You H, Hu X, Luo Y, Zhang Z, Song Y, An J, Lu H. Microglia-Astrocyte Interaction in Neural Development and Neural Pathogenesis. Cells 2023; 12:1942. [PMID: 37566021 PMCID: PMC10417796 DOI: 10.3390/cells12151942] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/22/2023] [Accepted: 07/24/2023] [Indexed: 08/12/2023] Open
Abstract
The interaction between microglia and astrocytes exhibits a relatively balanced state in order to maintain homeostasis in the healthy central nervous system (CNS). Disease stimuli alter microglia-astrocyte interaction patterns and elicit cell-type-specific responses, resulting in their contribution to various pathological processes. Here, we review the similarities and differences in the activation modes between microglia and astrocytes in various scenarios, encompassing different stages of neural development and a wide range of neural disorders. The aim is to provide a comprehensive understanding of their roles in neural development and regeneration and guiding new strategies for restoring CNS homeostasis.
Collapse
Affiliation(s)
- Meiqi Sun
- Department/Institute of Neurobiology, School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (M.S.); (H.Y.); (X.H.); (Y.L.); (Z.Z.); (Y.S.)
| | - Hongli You
- Department/Institute of Neurobiology, School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (M.S.); (H.Y.); (X.H.); (Y.L.); (Z.Z.); (Y.S.)
| | - Xiaoxuan Hu
- Department/Institute of Neurobiology, School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (M.S.); (H.Y.); (X.H.); (Y.L.); (Z.Z.); (Y.S.)
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Yujia Luo
- Department/Institute of Neurobiology, School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (M.S.); (H.Y.); (X.H.); (Y.L.); (Z.Z.); (Y.S.)
| | - Zixuan Zhang
- Department/Institute of Neurobiology, School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (M.S.); (H.Y.); (X.H.); (Y.L.); (Z.Z.); (Y.S.)
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Yiqun Song
- Department/Institute of Neurobiology, School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (M.S.); (H.Y.); (X.H.); (Y.L.); (Z.Z.); (Y.S.)
| | - Jing An
- Department/Institute of Neurobiology, School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (M.S.); (H.Y.); (X.H.); (Y.L.); (Z.Z.); (Y.S.)
| | - Haixia Lu
- Department/Institute of Neurobiology, School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (M.S.); (H.Y.); (X.H.); (Y.L.); (Z.Z.); (Y.S.)
| |
Collapse
|
114
|
Da Silva IO, Crespo-Lopez ME, Augusto-Oliveira M, Arrifano GDP, Ramos-Nunes NR, Gomes EB, da Silva FRP, de Sousa AA, Leal ALAB, Damasceno HC, de Oliveira ACA, Souza-Monteiro JR. What We Know about Euterpe Genus and Neuroprotection: A Scoping Review. Nutrients 2023; 15:3189. [PMID: 37513607 PMCID: PMC10384735 DOI: 10.3390/nu15143189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/05/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023] Open
Abstract
The Euterpe genus (mainly Euterpe oleracea Martius, Euterpe precatoria Martius, and Euterpe edulis Martius) has recently gained commercial and scientific notoriety due to the high nutritional value of its fruits, which are rich in polyphenols (phenolic acids and anthocyanins) and have potent antioxidant activity. These characteristics have contributed to the increased number of neuropharmacological evaluations of the three species over the last 10 years, especially açaí of the species Euterpe oleracea Martius. The fruits of the three species exert neuroprotective effects through the modulation of inflammatory and oxidative pathways and other mechanisms, including the inhibition of the mTOR pathway and protection of the blood-brain barrier, all of them intimately involved in several neuropathologies. Thus, a better understanding of the neuropharmacological properties of these three species may open new paths for the development of therapeutic tools aimed at preventing and treating a variety of neurological conditions.
Collapse
Affiliation(s)
- Ilano Oliveira Da Silva
- Medicine College, Altamira Campus, Federal University of Pará (UFPA), Altamira 68372-040, PA, Brazil; (I.O.D.S.); (A.A.d.S.); (A.L.A.B.L.); (H.C.D.); (A.C.A.d.O.)
| | - Maria Elena Crespo-Lopez
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (M.E.C.-L.); (M.A.-O.); (G.d.P.A.)
| | - Marcus Augusto-Oliveira
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (M.E.C.-L.); (M.A.-O.); (G.d.P.A.)
| | - Gabriela de Paula Arrifano
- Laboratory of Molecular Pharmacology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (M.E.C.-L.); (M.A.-O.); (G.d.P.A.)
| | - Natália Raphaela Ramos-Nunes
- Medicine College, Altamira Campus, Federal University of Pará (UFPA), Altamira 68372-040, PA, Brazil; (I.O.D.S.); (A.A.d.S.); (A.L.A.B.L.); (H.C.D.); (A.C.A.d.O.)
| | - Elielton Barreto Gomes
- Medicine College, Altamira Campus, Federal University of Pará (UFPA), Altamira 68372-040, PA, Brazil; (I.O.D.S.); (A.A.d.S.); (A.L.A.B.L.); (H.C.D.); (A.C.A.d.O.)
| | - Felipe Rodolfo Pereira da Silva
- Medicine College, Altamira Campus, Federal University of Pará (UFPA), Altamira 68372-040, PA, Brazil; (I.O.D.S.); (A.A.d.S.); (A.L.A.B.L.); (H.C.D.); (A.C.A.d.O.)
| | - Aline Andrade de Sousa
- Medicine College, Altamira Campus, Federal University of Pará (UFPA), Altamira 68372-040, PA, Brazil; (I.O.D.S.); (A.A.d.S.); (A.L.A.B.L.); (H.C.D.); (A.C.A.d.O.)
| | - Alessandro Luiz Araújo Bentes Leal
- Medicine College, Altamira Campus, Federal University of Pará (UFPA), Altamira 68372-040, PA, Brazil; (I.O.D.S.); (A.A.d.S.); (A.L.A.B.L.); (H.C.D.); (A.C.A.d.O.)
| | - Helane Conceição Damasceno
- Medicine College, Altamira Campus, Federal University of Pará (UFPA), Altamira 68372-040, PA, Brazil; (I.O.D.S.); (A.A.d.S.); (A.L.A.B.L.); (H.C.D.); (A.C.A.d.O.)
| | - Ana Carolina Alves de Oliveira
- Medicine College, Altamira Campus, Federal University of Pará (UFPA), Altamira 68372-040, PA, Brazil; (I.O.D.S.); (A.A.d.S.); (A.L.A.B.L.); (H.C.D.); (A.C.A.d.O.)
| | - José Rogério Souza-Monteiro
- Medicine College, Altamira Campus, Federal University of Pará (UFPA), Altamira 68372-040, PA, Brazil; (I.O.D.S.); (A.A.d.S.); (A.L.A.B.L.); (H.C.D.); (A.C.A.d.O.)
| |
Collapse
|
115
|
Carata E, Muci M, Di Giulio S, Mariano S, Panzarini E. Looking to the Future of the Role of Macrophages and Extracellular Vesicles in Neuroinflammation in ALS. Int J Mol Sci 2023; 24:11251. [PMID: 37511010 PMCID: PMC10379393 DOI: 10.3390/ijms241411251] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Neuroinflammation is a common pathological feature of amyotrophic lateral sclerosis (ALS). Although scientific evidence to date does not allow defining neuroinflammation as an ALS trigger, its role in exacerbating motor neuron (MNs) degeneration and disease progression is attracting research interest. Activated CNS (Central Nervous System) glial cells, proinflammatory peripheral and infiltrated T lymphocytes and monocytes/macrophages, as well as the immunoreactive molecules they release, represent the active players for the role of immune dysregulation enhancing neuroinflammation. The crosstalk between the peripheral and CNS immune cells significantly correlates with the survival of ALS patients since the modification of peripheral macrophages can downregulate inflammation at the periphery along the nerves and in the CNS. As putative vehicles for misfolded protein and inflammatory mediators between cells, extracellular vesicles (EVs) have also drawn particular attention in the field of ALS. Both CNS and peripheral immune cells release EVs, which are able to modulate the behavior of neighboring recipient cells; unfortunately, the mechanisms involved in EVs-mediated communication in neuroinflammation remain unclear. This review aims to synthesize the current literature regarding EV-mediated cell-to-cell communication in the brain under ALS, with a particular point of view on the role of peripheral macrophages in responding to inflammation to understand the biological process and exploit it for ALS management.
Collapse
Affiliation(s)
- Elisabetta Carata
- Department of Biological Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy
| | - Marco Muci
- Department of Biological Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy
| | - Simona Di Giulio
- Department of Biological Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy
| | - Stefania Mariano
- Department of Biological Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy
| | - Elisa Panzarini
- Department of Biological Sciences and Technologies (Di.S.Te.B.A.), University of Salento, 73100 Lecce, Italy
| |
Collapse
|
116
|
Barthels D, Prateeksha P, Nozohouri S, Villalba H, Zhang Y, Sharma S, Anderson S, Howlader MSI, Nambiar A, Abbruscato TJ, Das H. Dental Pulp-Derived Stem Cells Preserve Astrocyte Health During Induced Gliosis by Modulating Mitochondrial Activity and Functions. Cell Mol Neurobiol 2023; 43:2105-2127. [PMID: 36201091 PMCID: PMC11412198 DOI: 10.1007/s10571-022-01291-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/27/2022] [Indexed: 11/03/2022]
Abstract
Astrocytes have been implicated in the onset and complication of various central nervous system (CNS) injuries and disorders. Uncontrolled astrogliosis (gliosis), while a necessary process for recovery after CNS trauma, also causes impairments in CNS performance and functions. The ability to preserve astrocyte health and better regulate the gliosis process could play a major role in controlling damage in the aftermath of acute insults and during chronic dysfunction. Here in, we demonstrate the ability of dental pulp-derived stem cells (DPSCs) in protecting the health of astrocytes during induced gliosis. First of all, we have characterized the expression of genes in primary astrocytes that are relevant to the pathological conditions of CNS by inducing gliosis. Subsequently, we found that astrocytes co-cultured with DPSCs reduced ROS production, NRF2 and GCLM expressions, mitochondrial membrane potential, and mitochondrial functions compared to the astrocytes that were not co-cultured with DPSCs in gliosis condition. In addition, hyperactive autophagy was also decreased in astrocytes that were co-cultured with DPSCs compared to the astrocytes that were not co-cultured with DPSCs during gliosis. This reversal and mitigation of gliosis in astrocytes were partly due to induction of neurogenesis in DPSCs through enhanced expressions of the neuronal genes like GFAP, NeuN, and Synapsin in DPSCs and by secretion of higher amounts of neurotropic factors, such as BDNF, GDNF, and TIMP-2. Protein-Protein docking analysis suggested that BDNF and GDNF can bind with CSPG4 and block the downstream signaling. Together these findings demonstrate novel functions of DPSCs to preserve astrocyte health during gliosis.
Collapse
Affiliation(s)
- Derek Barthels
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, ARB Suite 2116, 1406 South Coulter Street, Amarillo, TX, 79106, USA
| | - Prateeksha Prateeksha
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, ARB Suite 2116, 1406 South Coulter Street, Amarillo, TX, 79106, USA
| | - Saeideh Nozohouri
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, ARB Suite 2116, 1406 South Coulter Street, Amarillo, TX, 79106, USA
| | - Heidi Villalba
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, ARB Suite 2116, 1406 South Coulter Street, Amarillo, TX, 79106, USA
| | - Yong Zhang
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, ARB Suite 2116, 1406 South Coulter Street, Amarillo, TX, 79106, USA
| | - Sejal Sharma
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, ARB Suite 2116, 1406 South Coulter Street, Amarillo, TX, 79106, USA
| | - Sarah Anderson
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, ARB Suite 2116, 1406 South Coulter Street, Amarillo, TX, 79106, USA
| | - Md Sariful Islam Howlader
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, ARB Suite 2116, 1406 South Coulter Street, Amarillo, TX, 79106, USA
| | - Adarsh Nambiar
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, ARB Suite 2116, 1406 South Coulter Street, Amarillo, TX, 79106, USA
| | - Thomas J Abbruscato
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, ARB Suite 2116, 1406 South Coulter Street, Amarillo, TX, 79106, USA
| | - Hiranmoy Das
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, ARB Suite 2116, 1406 South Coulter Street, Amarillo, TX, 79106, USA.
| |
Collapse
|
117
|
Cain A, Taga M, McCabe C, Green GS, Hekselman I, White CC, Lee DI, Gaur P, Rozenblatt-Rosen O, Zhang F, Yeger-Lotem E, Bennett DA, Yang HS, Regev A, Menon V, Habib N, De Jager PL. Multicellular communities are perturbed in the aging human brain and Alzheimer's disease. Nat Neurosci 2023; 26:1267-1280. [PMID: 37336975 PMCID: PMC10789499 DOI: 10.1038/s41593-023-01356-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 05/10/2023] [Indexed: 06/21/2023]
Abstract
The role of different cell types and their interactions in Alzheimer's disease (AD) is a complex and open question. Here, we pursued this question by assembling a high-resolution cellular map of the aging frontal cortex using single-nucleus RNA sequencing of 24 individuals with a range of clinicopathologic characteristics. We used this map to infer the neocortical cellular architecture of 638 individuals profiled by bulk RNA sequencing, providing the sample size necessary for identifying statistically robust associations. We uncovered diverse cell populations associated with AD, including a somatostatin inhibitory neuronal subtype and oligodendroglial states. We further identified a network of multicellular communities, each composed of coordinated subpopulations of neuronal, glial and endothelial cells, and we found that two of these communities are altered in AD. Finally, we used mediation analyses to prioritize cellular changes that might contribute to cognitive decline. Thus, our deconstruction of the aging neocortex provides a roadmap for evaluating the cellular microenvironments underlying AD and dementia.
Collapse
Affiliation(s)
- Anael Cain
- Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Mariko Taga
- Center for Translational & Computational Immunology, Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Cristin McCabe
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Gilad S Green
- Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Idan Hekselman
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | - Dylan I Lee
- Center for Translational & Computational Immunology, Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Pallavi Gaur
- Center for Translational & Computational Immunology, Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA
| | - Orit Rozenblatt-Rosen
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Genentech, South San Francisco, CA, USA
| | - Feng Zhang
- Broad Institute, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Esti Yeger-Lotem
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Hyun-Sik Yang
- Broad Institute, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biology, Koch Institute of Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Genentech, South San Francisco, CA, USA
| | - Vilas Menon
- Center for Translational & Computational Immunology, Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA.
| | - Naomi Habib
- Edmond & Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Philip L De Jager
- Center for Translational & Computational Immunology, Department of Neurology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University Irving Medical Center, New York, NY, USA.
- Broad Institute, Cambridge, MA, USA.
| |
Collapse
|
118
|
Escobar AP, Bonansco C, Cruz G, Dagnino-Subiabre A, Fuenzalida M, Negrón I, Sotomayor-Zárate R, Martínez-Pinto J, Jorquera G. Central and Peripheral Inflammation: A Common Factor Causing Addictive and Neurological Disorders and Aging-Related Pathologies. Int J Mol Sci 2023; 24:10083. [PMID: 37373230 PMCID: PMC10298583 DOI: 10.3390/ijms241210083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Many diseases and degenerative processes affecting the nervous system and peripheral organs trigger the activation of inflammatory cascades. Inflammation can be triggered by different environmental conditions or risk factors, including drug and food addiction, stress, and aging, among others. Several pieces of evidence show that the modern lifestyle and, more recently, the confinement associated with the COVID-19 pandemic have contributed to increasing the incidence of addictive and neuropsychiatric disorders, plus cardiometabolic diseases. Here, we gather evidence on how some of these risk factors are implicated in activating central and peripheral inflammation contributing to some neuropathologies and behaviors associated with poor health. We discuss the current understanding of the cellular and molecular mechanisms involved in the generation of inflammation and how these processes occur in different cells and tissues to promote ill health and diseases. Concomitantly, we discuss how some pathology-associated and addictive behaviors contribute to worsening these inflammation mechanisms, leading to a vicious cycle that promotes disease progression. Finally, we list some drugs targeting inflammation-related pathways that may have beneficial effects on the pathological processes associated with addictive, mental, and cardiometabolic illnesses.
Collapse
Affiliation(s)
- Angélica P. Escobar
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (A.P.E.); (C.B.); (G.C.); (M.F.); (I.N.); (R.S.-Z.)
| | - Christian Bonansco
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (A.P.E.); (C.B.); (G.C.); (M.F.); (I.N.); (R.S.-Z.)
| | - Gonzalo Cruz
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (A.P.E.); (C.B.); (G.C.); (M.F.); (I.N.); (R.S.-Z.)
| | - Alexies Dagnino-Subiabre
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (A.P.E.); (C.B.); (G.C.); (M.F.); (I.N.); (R.S.-Z.)
| | - Marco Fuenzalida
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (A.P.E.); (C.B.); (G.C.); (M.F.); (I.N.); (R.S.-Z.)
| | - Ignacio Negrón
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (A.P.E.); (C.B.); (G.C.); (M.F.); (I.N.); (R.S.-Z.)
| | - Ramón Sotomayor-Zárate
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (A.P.E.); (C.B.); (G.C.); (M.F.); (I.N.); (R.S.-Z.)
| | - Jonathan Martínez-Pinto
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (A.P.E.); (C.B.); (G.C.); (M.F.); (I.N.); (R.S.-Z.)
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Gonzalo Jorquera
- Centro de Neurobiología y Fisiopatología Integrativa (CENFI), Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; (A.P.E.); (C.B.); (G.C.); (M.F.); (I.N.); (R.S.-Z.)
- Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago 7830490, Chile
| |
Collapse
|
119
|
Gao J, Su G, Chen W, Wu Q, Liu J, Liu J, Chai M, Dong Y, Wang H, Chen L, Zhang Z, Wang M. Mechanism of ligusticum cycloprolactam against neuroinflammation based on network pharmacology and experimental verification. Clin Exp Pharmacol Physiol 2023. [PMID: 37308175 DOI: 10.1111/1440-1681.13784] [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: 10/06/2022] [Revised: 04/05/2023] [Accepted: 04/26/2023] [Indexed: 06/14/2023]
Abstract
Ligustilide, a natural phthalide mainly derived from chuanxiong rhizomes and Angelica Sinensis roots, possesses anti-inflammatory activity, particularly in the context of the nervous system. However, its application is limited because of its unstable chemical properties. To overcome this limitation, ligusticum cycloprolactam (LIGc) was synthesized through structural modification of ligustilide. In this study, we combined network pharmacological methods with experimental verification to investigate the anti-neuroinflammatory effects and mechanisms of ligustilide and LIGc. Based on our network pharmacology analysis, we identified four key targets of ligustilide involved in exerting an anti-inflammatory effect, with the nuclear factor (NF)-κB signal pathway suggested as the main signalling pathway. To verify these results, we examined the expression of inflammatory cytokines and inflammation-related proteins, analysed the phosphorylation level of NF-κB, inhibitor of κBα (IκBα) and inhibitor of κB kinase α and β (IKKα+β), and evaluated the effect of BV2 cell-conditioned medium on HT22 cells in vitro. Our results, demonstrate for the first time that LIGc can downregulate the activation of the NF-κB signal pathway in BV2 cells induced by lipopolysaccharide, suppress the production of inflammatory cytokines and reduce nerve injury in HT22 cells mediated by BV2 cells. These findings suggest that LIGc inhibits the neuroinflammatory response mediated by BV2 cells, providing strong scientific support for the development of anti-inflammatory drugs based on natural ligustilide or its derivatives. However, there are some limitations to our current study. In the future, further experiments using in vivo models may provide additional evidence to support our findings.
Collapse
Affiliation(s)
- Juan Gao
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Gang Su
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Wei Chen
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Qionghui Wu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Junxi Liu
- Chinese Academy of Sciences Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Jifei Liu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Miao Chai
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Ying Dong
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - He Wang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Lixia Chen
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhenchang Zhang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Manxia Wang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
120
|
Qi C, Feng Y, Jiang Y, Chen W, Vakal S, Chen JF, Zheng W. A 2AR antagonist treatment for multiple sclerosis: Current progress and future prospects. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 170:185-223. [PMID: 37741692 DOI: 10.1016/bs.irn.2023.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2023]
Abstract
Emerging evidence suggests that both selective and non-selective Adenosine A2A receptor (A2AR) antagonists could effectively protect mice from experimental autoimmune encephalomyelitis (EAE), which is the most commonly used animal model for multiple sclerosis (MS) research. Meanwhile, the recent FDA approval of Nourianz® (istradefylline) in 2019 as an add-on treatment to levodopa in Parkinson's disease (PD) with "OFF" episodes, along with its proven clinical safety, has prompted us to explore the potential of A2AR antagonists in treating multiple sclerosis (MS) through clinical trials. However, despite promising findings in experimental autoimmune encephalomyelitis (EAE), the complex and contradictory role of A2AR signaling in EAE pathology has raised concerns about the feasibility of using A2AR antagonists as a therapeutic approach for MS. This review addresses the potential effect of A2AR antagonists on EAE/MS in both the peripheral immune system (PIS) and the central nervous system (CNS). In brief, A2AR antagonists had a moderate effect on the proliferation and inflammatory response, while exhibiting a potent anti-inflammatory effect in the CNS through their impact on microglia, astrocytes, and the endothelial cells/epithelium of the blood-brain barrier. Consequently, A2AR signaling remains an essential immunomodulator in EAE/MS, suggesting that A2AR antagonists hold promise as a drug class for treating MS.
Collapse
Affiliation(s)
- Chenxing Qi
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, P.R. China; Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, P.R. China
| | - Yijia Feng
- Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Yiwei Jiang
- Alberta Institute, Wenzhou Medical University, Wenzhou, P.R. China
| | - Wangchao Chen
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, P.R. China
| | - Serhii Vakal
- Structural Bioinformatics Laboratory, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Jiang-Fan Chen
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, P.R. China; Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, P.R. China
| | - Wu Zheng
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, P.R. China; Oujiang Laboratory (Zhejiang Laboratory for Regenerative Medicine, Vision and Brain Health), School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, P.R. China.
| |
Collapse
|
121
|
Latham AS, Geer CE, Ackart DF, Anderson IK, Vittoria KM, Podell BK, Basaraba RJ, Moreno JA. Gliosis, misfolded protein aggregation, and neuronal loss in a guinea pig model of pulmonary tuberculosis. Front Neurosci 2023; 17:1157652. [PMID: 37274195 PMCID: PMC10235533 DOI: 10.3389/fnins.2023.1157652] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 04/24/2023] [Indexed: 06/06/2023] Open
Abstract
Tuberculosis, caused by Mycobacterium tuberculosis infection, is an ongoing epidemic with an estimated ten million active cases of the disease worldwide. Pulmonary tuberculosis is associated with cognitive and memory deficits, and patients with this disease are at an increased risk for Parkinson's disease and dementia. Although epidemiological data correlates neurological effects with peripheral disease, the pathology in the central nervous system is unknown. In an established guinea pig model of low-dose, aerosolized Mycobacterium tuberculosis infection, we see behavior changes and memory loss in infected animals. We correlate these findings with pathological changes within brain regions related to motor, cognition, and sensation across disease progression. This includes microglial and astrocytic proliferation and reactivity. These cellular changes are followed by the aggregation of neurotoxic amyloid β and phosphorylated tau and, ultimately, neuronal degeneration in the hippocampus. Through these data, we have obtained a greater understanding of the neuropathological effects of a peripheral disease that affects millions of persons worldwide.
Collapse
Affiliation(s)
- Amanda S. Latham
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Brain Research Center, Colorado State University, Fort Collins, CO, United States
| | - Charlize E. Geer
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - David F. Ackart
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Isla K. Anderson
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Department of Biomedical Science, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Kaley M. Vittoria
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Brendan K. Podell
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Randall J. Basaraba
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Julie A. Moreno
- Department of Environmental and Radiological Health Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Brain Research Center, Colorado State University, Fort Collins, CO, United States
- Center for Healthy Aging, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
122
|
Xingi E, Koutsoudaki PN, Thanou I, Phan MS, Margariti M, Scheller A, Tinevez JY, Kirchhoff F, Thomaidou D. LPS-Induced Systemic Inflammation Affects the Dynamic Interactions of Astrocytes and Microglia with the Vasculature of the Mouse Brain Cortex. Cells 2023; 12:1418. [PMID: 37408252 DOI: 10.3390/cells12101418] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/05/2023] [Accepted: 05/13/2023] [Indexed: 07/07/2023] Open
Abstract
The Neurovascular Unit (NVU), composed of glia (astrocytes, oligodendrocytes, microglia), neurons, pericytes and endothelial cells, is a dynamic interface ensuring the physiological functioning of the central nervous system (CNS), which gets affected and contributes to the pathology of several neurodegenerative diseases. Neuroinflammation is a common feature of neurodegenerative diseases and is primarily related to the activation state of perivascular microglia and astrocytes, which constitute two of its major cellular components. Our studies focus on monitoring in real time the morphological changes of perivascular astrocytes and microglia, as well as their dynamic interactions with the brain vasculature, under physiological conditions and following systemic neuroinflammation triggering both microgliosis and astrogliosis. To this end, we performed 2-photon laser scanning microscopy (2P-LSM) for intravital imaging of the cortex of transgenic mice visualizing the dynamics of microglia and astroglia following neuroinflammation induced by systemic administration of the endotoxin lipopolysaccharide (LPS). Our results indicate that following neuroinflammation the endfeet of activated perivascular astrocytes lose their close proximity and physiological cross-talk with vasculature, an event that most possibly contributes to a loss of blood-brain barrier (BBB) integrity. At the same time, microglial cells become activated and exhibit a higher extent of physical contact with the blood vessels. These dynamic responses of perivascular astrocytes and microglia are peaking at 4 days following LPS administration; however, they still persist at a lower level at 8 days after LPS injection, revealing incomplete reversal of inflammation affecting the glial properties and interactions within the NVU.
Collapse
Affiliation(s)
- Evangelia Xingi
- Light Microscopy Unit, Hellenic Pasteur Institute, 11521 Athens, Greece
| | - Paraskevi N Koutsoudaki
- Neural Stem Cells and Neuroimaging Group, Department of Neurobiology, Hellenic Pasteur Institute, 11521 Athens, Greece
| | - Irini Thanou
- Neural Stem Cells and Neuroimaging Group, Department of Neurobiology, Hellenic Pasteur Institute, 11521 Athens, Greece
| | - Minh-Son Phan
- Institut Pasteur, Université de Paris, Image Analysis Hub, F-75015 Paris, France
| | - Maria Margariti
- Neural Stem Cells and Neuroimaging Group, Department of Neurobiology, Hellenic Pasteur Institute, 11521 Athens, Greece
| | - Anja Scheller
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421 Homburg, Germany
| | - Jean-Yves Tinevez
- Institut Pasteur, Université de Paris, Image Analysis Hub, F-75015 Paris, France
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, 66421 Homburg, Germany
| | - Dimitra Thomaidou
- Light Microscopy Unit, Hellenic Pasteur Institute, 11521 Athens, Greece
- Neural Stem Cells and Neuroimaging Group, Department of Neurobiology, Hellenic Pasteur Institute, 11521 Athens, Greece
| |
Collapse
|
123
|
Bourque M, Morissette M, Soulet D, Di Paolo T. Impact of Sex on Neuroimmune contributions to Parkinson's disease. Brain Res Bull 2023:110668. [PMID: 37196734 DOI: 10.1016/j.brainresbull.2023.110668] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/27/2023] [Accepted: 05/13/2023] [Indexed: 05/19/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder after Alzheimer's disease. Inflammation has been observed in both the idiopathic and familial forms of PD. Importantly, PD is reported more often in men than in women, men having at least 1.5- fold higher risk to develop PD than women. This review summarizes the impact of biological sex and sex hormones on the neuroimmune contributions to PD and its investigation in animal models of PD. Innate and peripheral immune systems participate in the brain neuroinflammation of PD patients and is reproduced in neurotoxin, genetic and alpha-synuclein based models of PD. Microglia and astrocytes are the main cells of the innate immune system in the central nervous system and are the first to react to restore homeostasis in the brain. Analysis of serum immunoprofiles in female and male control and PD patients show that a great proportion of these markers differ between male and female. The relationship between CSF inflammatory markers and PD clinical characteristics or PD biomarkers shows sex differences. Conversely, in animal models of PD, sex differences in inflammation are well documented and the beneficial effects of endogenous and exogenous estrogenic modulation in inflammation have been reported. Targeting neuroinflammation in PD is an emerging therapeutic option but gonadal drugs have not yet been investigated in this respect, thus offering new opportunities for sex specific treatments.
Collapse
Affiliation(s)
- Mélanie Bourque
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, (Québec), G1V4G2, Canada.
| | - Marc Morissette
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, (Québec), G1V4G2, Canada.
| | - Denis Soulet
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, (Québec), G1V4G2, Canada; Faculté de Pharmacie, Pavillon Ferdinand-Vandry, 1050, avenue de la Médecine, Université Laval, Québec (Québec) G1V 0A6, Canada.
| | - Thérèse Di Paolo
- Centre de Recherche du CHU de Québec, Axe Neurosciences, 2705, Boulevard Laurier, Québec, (Québec), G1V4G2, Canada; Faculté de Pharmacie, Pavillon Ferdinand-Vandry, 1050, avenue de la Médecine, Université Laval, Québec (Québec) G1V 0A6, Canada.
| |
Collapse
|
124
|
Belo TCA, Santos GX, da Silva BEG, Rocha BLG, Abdala DW, Freire LAM, Rocha FS, Galdino G. IL-10/β-Endorphin-Mediated Neuroimmune Modulation on Microglia during Antinociception. Brain Sci 2023; 13:brainsci13050789. [PMID: 37239261 DOI: 10.3390/brainsci13050789] [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: 04/13/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Microglia are glial cells centrally related to pathophysiology and neuroimmunological regulation of pain through microglia-neuron crosstalk mechanisms. In contrast, anti-inflammatory mechanisms guided by immunological effectors such as IL-10 trigger the secretion of analgesic substances, culminating in the differential expression of genes encoding endogenous opioid peptides, especially β-endorphin. Thus, when β-endorphin binds to the µ-opioid receptor, it generates neuronal hyperpolarization, inhibiting nociceptive stimuli. This review aimed to summarize the recent advances in understanding the mechanism by which IL-10/β-endorphin can reduce pain. For this, databases were searched for articles from their inception up until November 2022. Two independent reviewers extracted the data and assessed the methodological quality of the included studies, and seventeen studies were considered eligible for this review. Several studies have demonstrated the impact of IL-10/β-endorphin in reducing pain, where IL-10 can stimulate GLP-1R, GRP40, and α7nAChR receptors, as well as intracellular signaling pathways, such as STAT3, resulting in increased β-endorphin expression and secretion. In addition, molecules such as gabapentinoids, thalidomide, cynandione A, morroniside, lemairamin, and cinobufagin, as well as non-pharmacological treatments such as electroacupuncture, reduce pain through IL-10 mediated mechanisms, reflecting a microglia-dependent β-endorphin differential increase. This process represents a cornerstone in pain neuroimmunology knowledge, and the results obtained by different studies about the theme are presented in this review.
Collapse
Affiliation(s)
| | - Gabriela Xavier Santos
- Laboratory of Neuroimmunobiology of Pain, Federal University of Alfenas, Alfenas 37130-001, Brazil
| | | | | | - Dennis William Abdala
- Laboratory of Movement Analysis, Federal University of Alfenas, Alfenas 37130-001, Brazil
| | - Larissa Alves Moreira Freire
- Laboratory of Neuroscience, Neuroimmunomodulation and Pain Study, Federal University of Alfenas, Alfenas 37130-001, Brazil
| | - Fernanda Santos Rocha
- Laboratory of Neuroimmunobiology of Pain, Federal University of Alfenas, Alfenas 37130-001, Brazil
| | - Giovane Galdino
- Laboratory of Neuroimmunobiology of Pain, Federal University of Alfenas, Alfenas 37130-001, Brazil
| |
Collapse
|
125
|
Hua F, Zhu H, Yu W, Zheng Q, Zhang L, Liang W, Lin Y, Xiao F, Yi P, Xiong Y, Dong Y, Li H, Fang L, Liu H, Ying J, Wang X. β-arrestin1 regulates astrocytic reactivity via Drp1-dependent mitochondrial fission: implications in postoperative delirium. J Neuroinflammation 2023; 20:113. [PMID: 37170230 PMCID: PMC10173541 DOI: 10.1186/s12974-023-02794-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 04/24/2023] [Indexed: 05/13/2023] Open
Abstract
Postoperative delirium (POD) is a frequent and debilitating complication, especially amongst high risk procedures, such as orthopedic surgery. This kind of neurocognitive disorder negatively affects cognitive domains, such as memory, awareness, attention, and concentration after surgery; however, its pathophysiology remains unknown. Multiple lines of evidence supporting the occurrence of inflammatory events have come forward from studies in human patients' brain and bio-fluids (CSF and serum), as well as in animal models for POD. β-arrestins are downstream molecules of guanine nucleotide-binding protein (G protein)-coupled receptors (GPCRs). As versatile proteins, they regulate numerous pathophysiological processes of inflammatory diseases by scaffolding with inflammation-linked partners. Here we report that β-arrestin1, one type of β-arrestins, decreases significantly in the reactive astrocytes of a mouse model for POD. Using β-arrestin1 knockout (KO) mice, we find aggravating effect of β-arrestin1 deficiency on the cognitive dysfunctions and inflammatory phenotype of astrocytes in POD model mice. We conduct the in vitro experiments to investigate the regulatory roles of β-arrestin1 and demonstrate that β-arrestin1 in astrocytes interacts with the dynamin-related protein 1 (Drp1) to regulate mitochondrial fusion/fission process. β-arrestin1 deletion cancels the combination of β-arrestin1 and cellular Drp1, thus promoting the translocation of Drp1 to mitochondrial membrane to provoke the mitochondrial fragments and the subsequent mitochondrial malfunctions. Using β-arrestin1-biased agonist, cognitive dysfunctions of POD mice and pathogenic activation of astrocytes in the POD-linked brain region are reduced. We, therefore, conclude that β-arrestin1 is a promising target for the understanding of POD pathology and development of POD therapeutics.
Collapse
Affiliation(s)
- Fuzhou Hua
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, 1# Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
- Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Hong Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, 330006, Nanchang, Jiangxi, People's Republic of China
| | - Wen Yu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, 1# Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
- Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Qingcui Zheng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, 1# Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
- Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Lieliang Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, 1# Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
- Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Weidong Liang
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Yue Lin
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, 1# Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
- Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Fan Xiao
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, 1# Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
- Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Pengcheng Yi
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, 1# Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
- Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Yanhong Xiong
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, 1# Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
- Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Yao Dong
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, 1# Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
- Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Hua Li
- Department of Anesthesiology, First People's Hospital of Yihuang County, Fuzhou, 344400, Jiangxi, People's Republic of China
| | - Lanran Fang
- Department of Statistics, Jiangxi University of Finance and Economics, Nanchang, 330013, Jiangxi, People's Republic of China
| | - Hailin Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, 1# Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
- Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China
| | - Jun Ying
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, 1# Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China.
- Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, Nanchang, 330006, Jiangxi, People's Republic of China.
| | - Xifeng Wang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, 17# Yong Wai Zheng Street, Nanchang, 330006, Jiangxi, People's Republic of China.
| |
Collapse
|
126
|
Gao L, Pan X, Zhang JH, Xia Y. Glial cells: an important switch for the vascular function of the central nervous system. Front Cell Neurosci 2023; 17:1166770. [PMID: 37206667 PMCID: PMC10188976 DOI: 10.3389/fncel.2023.1166770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/13/2023] [Indexed: 05/21/2023] Open
Abstract
In this review, we first describe the current understanding of glial-mediated vascular function affecting the role of the blood-brain barrier (BBB) in central nervous system (CNS) disorders. BBB, mainly composed of glial and endothelial cells (ECs), is the protective structure that orchestrates the transport of substances, including ions, molecules, and cells from brain vessels into or out of the CNS. Then, we display the multiple communication between glial and vascular function based on angiogenesis, vascular wrapping, and blood perfusion in the brain. Glial can support microvascular ECs to form a blood network connecting to neurons. Astrocytes, microglia, and oligodendrocytes are the common types of glial surrounding the brain vessel. Glial-vessel interaction is required for the permeability and integrity of BBB. Glial cells surrounding the cerebral blood vessels can transmit communication signals to ECs and regulate the activity of vascular endothelial growth factor (VEGF) or Wnt-dependent endothelial angiogenesis mechanism. In addition, these glial cells monitor the blood flow in the brain via Ca2+/K+-dependent pathways. Finally, we provide a potential research direction for the glial-vessel axis in CNS disorders. Microglial activation can trigger astrocyte activation, which suggests that microglia-astrocyte interaction may play a key role in monitoring cerebral blood flow. Thus, microglia-astrocyte interaction can be the key point of follow-up studies focusing on the microglia-blood mechanism. More investigations focus on the mechanism of how oligodendrocyte progenitor cells communicate and interact with ECs. The direct role of oligodendrocytes in modulating vascular function needs to be explored in the future.
Collapse
Affiliation(s)
- Ling Gao
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou, China
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Xuezhen Pan
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou, China
| | - John H. Zhang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Ying Xia
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou, China
| |
Collapse
|
127
|
Kim JH, Han J, Afridi R, Kim JH, Rahman MH, Park DH, Lee WS, Song GJ, Suk K. A multiplexed siRNA screen identifies key kinase signaling networks of brain glia. Life Sci Alliance 2023; 6:e202201605. [PMID: 36878638 PMCID: PMC9990460 DOI: 10.26508/lsa.202201605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 02/22/2023] [Accepted: 02/22/2023] [Indexed: 03/08/2023] Open
Abstract
The dynamic behaviors of brain glial cells in various neuroinflammatory conditions and neurological disorders have been reported; however, little is known about the underlying intracellular signaling pathways. Here, we developed a multiplexed kinome-wide siRNA screen to identify the kinases regulating several inflammatory phenotypes of mouse glial cells in culture, including inflammatory activation, migration, and phagocytosis of glia. Subsequent proof-of-concept experiments involving genetic and pharmacological inhibitions indicated the importance of T-cell receptor signaling components in microglial activation and a metabolic shift from glycolysis to oxidative phosphorylation in astrocyte migration. This time- and cost-effective multiplexed kinome siRNA screen efficiently provides exploitable drug targets and novel insight into the mechanisms underlying the phenotypic regulation of glial cells and neuroinflammation. Moreover, the kinases identified in this screen may be relevant in other inflammatory diseases and cancer, wherein kinases play a critical role in disease signaling pathways.
Collapse
Affiliation(s)
- Jong-Heon Kim
- Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Jin Han
- Department of Biomedical Science, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Ruqayya Afridi
- Department of Biomedical Science, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jae-Hong Kim
- Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Md Habibur Rahman
- Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dong Ho Park
- Department of Ophthalmology, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Won Suk Lee
- Neuracle Science Co., Ltd. Seoul, Republic Korea
| | - Gyun Jee Song
- Department of Medical Science, College of Medicine, Catholic Kwandong University, Gangneung-si, Republic Korea; Translational Brain Research Center, International St. Mary's Hospital, Catholic Kwandong University, Incheon, Republic Korea
| | - Kyoungho Suk
- Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
- Department of Biomedical Science, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
128
|
Shi L, Xia Z, Guo J, Wang L, Peng Z, Qiu D, Zhou Y, Zhou D, Kuang L, Qiu T. Maresin-1 improves LPS-induced depressive-like behavior by inhibiting hippocampal microglial activation. J Affect Disord 2023; 328:261-272. [PMID: 36813041 DOI: 10.1016/j.jad.2023.02.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/22/2023]
Abstract
Maresin-1 is an antiphlogistic agonist synthesized by macrophages from docosahexaenoic acid (DHA). It has both anti-inflammatory and pro-inflammatory properties and has been found to enhance neuroprotection and cognitive function. However, there is limited knowledge of its effects on depression and the potential mechanism remains unclear. In this study, the effects of Maresin-1 on lipopolysaccharide (LPS)-induced depressive symptoms and neuroinflammation were investigated in mice and the possible cellular and molecular mechanisms were further clarified. Maresin-1 treatment (5 μg/kg, i.p.) led to improved tail suspension times, as well as distances moved in an open-field test but it did not improve reductions in sugar-water consumption in mice with depressive-like behaviors induced by LPS (1 mg/kg, i.p.); TSPO PETCT scanning showed that Maresin-1 reduced the standardized uptake value (SUV) of [18 F] DPA-714 in brain regions associated with depression (e.g., hippocampus and pre-frontal cortex), while immunofluorescence of hippocampal and indicated that Maresin-1 inhibited microglial activation reducing the expression of the pro-inflammatory cytokine IL-1β and NLRP3. The RNA sequencing of mouse hippocampi showed that genes expressed differentially between Maresin-1-treated and LPS-treated tissue were associated with tight connections between cells and the stress-activated MAPK cascade negative regulatory pathways. Overall, this study demonstrates that peripheral application of Maresin-1 could partially relieve LPS-induced depressive-like behaviors and showed for the first time that this effect was related to its anti-inflammatory action on microglia, thus providing new clues for the pharmacological mechanism underlying the anti-depression properties of Maresin-1.
Collapse
Affiliation(s)
- Lei Shi
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Zhu Xia
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiamei Guo
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Lixia Wang
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Zhiping Peng
- Department of Radiological Medicine, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Dachuan Qiu
- Department of Radiological Medicine, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Yi Zhou
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Dongdong Zhou
- Mental Health Center, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Li Kuang
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Tian Qiu
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
129
|
Hanin A, Cespedes J, Huttner A, Strelnikov D, Gopaul M, DiStasio M, Vezzani A, Hirsch LJ, Aronica E. Neuropathology of New-Onset Refractory Status Epilepticus (NORSE). J Neurol 2023:10.1007/s00415-023-11726-x. [PMID: 37079033 DOI: 10.1007/s00415-023-11726-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023]
Abstract
New-Onset Refractory Status Epilepticus (NORSE), including its subtype with a preceding febrile illness known as FIRES (Febrile Infection-Related Epilepsy Syndrome), is one of the most severe forms of status epilepticus. Despite an extensive workup (clinical evaluation, EEG, imaging, biological tests), the majority of NORSE cases remain unexplained (i.e., "cryptogenic NORSE"). Understanding the pathophysiological mechanisms underlying cryptogenic NORSE and the related long-term consequences is crucial to improve patient management and preventing secondary neuronal injury and drug-resistant post-NORSE epilepsy. Previously, neuropathological evaluations conducted on biopsies or autopsies have been found helpful for identifying the etiologies of some cases that were previously of unknown cause. Here, we summarize the findings of studies reporting neuropathology findings in patients with NORSE, including FIRES. We identified 64 cryptogenic cases and 66 neuropathology tissue samples, including 37 biopsies, 18 autopsies, and seven epilepsy surgeries (the type of tissue sample was not detailed for 4 cases). We describe the main neuropathology findings and place a particular emphasis on cases for which neuropathology findings helped establish a diagnosis or elucidate the pathophysiology of cryptogenic NORSE, or on described cases in which neuropathology findings supported the selection of specific treatments for patients with NORSE.
Collapse
Affiliation(s)
- Aurélie Hanin
- Department of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
- Institut du Cerveau, Paris Brain Institute, ICM, Inserm, CNRS, AP-HP, Hôpital de La Pitié-Salpêtrière, Sorbonne Université, DMU Neurosciences 6, Paris, France.
- Epilepsy Unit and Department of Clinical Neurophysiology, AP-HP, Hôpital de La Pitié-Salpêtrière, DMU Neurosciences 6, Paris, France.
| | - Jorge Cespedes
- Comprehensive Epilepsy Center, Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
- School of Medicine, Universidad Autonoma de Centro America, San Jose, Costa Rica
| | - Anita Huttner
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - David Strelnikov
- Comprehensive Epilepsy Center, Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Margaret Gopaul
- Comprehensive Epilepsy Center, Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Marcello DiStasio
- Department of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Annamaria Vezzani
- Department of Acute Brain Injury, Istituto di Recerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Lawrence J Hirsch
- Comprehensive Epilepsy Center, Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Meibergdreef 9, 1105, Amsterdam, The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| |
Collapse
|
130
|
Pearson A, Ortiz C, Eisenbaum M, Arrate C, Browning M, Mullan M, Bachmeier C, Crawford F, Ojo JO. Deletion of PTEN in microglia ameliorates chronic neuroinflammation following repetitive mTBI. Mol Cell Neurosci 2023; 125:103855. [PMID: 37084991 DOI: 10.1016/j.mcn.2023.103855] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/25/2023] [Accepted: 04/17/2023] [Indexed: 04/23/2023] Open
Abstract
Traumatic brain injury is a leading cause of morbidity and mortality in adults and children in developed nations. Following the primary injury, microglia, the resident innate immune cells of the CNS, initiate several inflammatory signaling cascades and pathophysiological responses that may persist chronically; chronic neuroinflammation following TBI has been closely linked to the development of neurodegeneration and neurological dysfunction. Phosphoinositide 3-kinases (PI3Ks) are a family of lipid kinases that have been shown to regulate several key mechanisms in the inflammatory response to TBI. Increasing evidence has shown that the modulation of the PI3K/AKT signaling pathway has the potential to influence the cellular response to inflammatory stimuli. However, directly targeting PI3K signaling poses several challenges due to its regulatory role in several cell survival pathways. We have previously identified that the phosphatase and tensin homolog deleted on chromosome 10 (PTEN), the major negative regulator of PI3K/AKT signaling, is dysregulated following exposure to repetitive mild traumatic brain injury (r-mTBI). Moreover, this dysregulated PI3K/AKT signaling was correlated with chronic microglial-mediated neuroinflammation. Therefore, we interrogated microglial-specific PTEN as a therapeutic target in TBI by generating a microglial-specific, Tamoxifen inducible conditional PTEN knockout model using a CX3CR1 Cre recombinase mouse line PTENfl/fl/CX3CR1+/CreERT2 (mcg-PTENcKO), and exposed them to our 20-hit r-mTBI paradigm. Animals were treated with tamoxifen at 76 days post-last injury, and the effects of microglia PTEN deletion on immune-inflammatory responses were assessed at 90-days post last injury. We observed that the deletion of microglial PTEN ameliorated the proinflammatory response to repetitive brain trauma, not only reducing chronic microglial activation and proinflammatory cytokine production but also rescuing TBI-induced reactive astrogliosis, demonstrating that these effects extended beyond microglia alone. Additionally, we observed that the pharmacological inhibition of PTEN with BpV(HOpic) ameliorated the LPS-induced activation of microglial NFκB signaling in vitro. Together, these data provide support for the role of PTEN as a regulator of chronic neuroinflammation following repetitive mild TBI.
Collapse
Affiliation(s)
- Andrew Pearson
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL 34243, USA; The Open University, Walton Hall, Kents Hill, Milton Keynes MK7 6AA, United Kingdom.
| | - Camila Ortiz
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL 34243, USA; The Open University, Walton Hall, Kents Hill, Milton Keynes MK7 6AA, United Kingdom
| | - Max Eisenbaum
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL 34243, USA; The Open University, Walton Hall, Kents Hill, Milton Keynes MK7 6AA, United Kingdom
| | - Clara Arrate
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL 34243, USA
| | | | - Michael Mullan
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL 34243, USA; The Open University, Walton Hall, Kents Hill, Milton Keynes MK7 6AA, United Kingdom
| | - Corbin Bachmeier
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL 34243, USA; The Open University, Walton Hall, Kents Hill, Milton Keynes MK7 6AA, United Kingdom
| | - Fiona Crawford
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL 34243, USA; The Open University, Walton Hall, Kents Hill, Milton Keynes MK7 6AA, United Kingdom; James A. Haley Veterans' Hospital, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA
| | - Joseph O Ojo
- The Roskamp Institute, 2040 Whitfield Avenue, Sarasota, FL 34243, USA; The Open University, Walton Hall, Kents Hill, Milton Keynes MK7 6AA, United Kingdom
| |
Collapse
|
131
|
Liu Z, Shen C, Li H, Tong J, Wu Y, Ma Y, Wang J, Wang Z, Li Q, Zhang X, Dong H, Yang Y, Yu M, Wang J, Zhou R, Fei J, Huang F. NOD-like receptor NLRC5 promotes neuroinflammation and inhibits neuronal survival in Parkinson's disease models. J Neuroinflammation 2023; 20:96. [PMID: 37072793 PMCID: PMC10111753 DOI: 10.1186/s12974-023-02755-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 03/02/2023] [Indexed: 04/20/2023] Open
Abstract
Parkinson's disease (PD) is mainly characterized by the progressive degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and neuroinflammation mediated by overactivated microglia and astrocytes. NLRC5 (nucleotide-binding oligomerization domain-like receptor family caspase recruitment domain containing 5) has been reported to participate in various immune disorders, but its role in neurodegenerative diseases remains unclear. In the current study, we found that the expression of NLRC5 was increased in the nigrostriatal axis of mice with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP)-induced PD, as well as in primary astrocytes, microglia and neurons exposed to different neurotoxic stimuli. In an acute MPTP-induced PD model, NLRC5 deficiency significantly reduced dopaminergic system degeneration and ameliorated motor deficits and striatal inflammation. Furthermore, we found that NLRC5 deficiency decreased the expression of the proinflammatory genes IL-1β, IL-6, TNF-α and COX2 in primary microglia and primary astrocytes treated with neuroinflammatory stimuli and reduced the inflammatory response in mixed glial cells in response to LPS treatment. Moreover, NLRC5 deficiency suppressed activation of the NF-κB and MAPK signaling pathways and enhanced the activation of AKT-GSK-3β and AMPK signaling in mixed glial cells. Furthermore, NLRC5 deficiency increased the survival of primary neurons treated with MPP+ or conditioned medium from LPS-stimulated mixed glial cells and promoted activation of the NF-κB and AKT signaling pathways. Moreover, the mRNA expression of NLRC5 was decreased in the blood of PD patients compared to healthy subjects. Therefore, we suggest that NLRC5 promotes neuroinflammation and dopaminergic degeneration in PD and may serve as a marker of glial activation.
Collapse
Affiliation(s)
- Zhaolin Liu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Chenye Shen
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Heng Li
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Jiabin Tong
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Yufei Wu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Yuanyuan Ma
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Jinghui Wang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Zishan Wang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Qing Li
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Xiaoshuang Zhang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Hongtian Dong
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Yufang Yang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Mei Yu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China
| | - Jian Wang
- Department of Neurology, Huashan Hospital, Fudan University, 12 Wulumuqi Zhong Road, Shanghai, 200040, China
| | - Renyuan Zhou
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China.
| | - Jian Fei
- School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China.
- Shanghai Engineering Research Center for Model Organisms, Shanghai Model Organisms Center, INC., Shanghai, 201203, China.
| | - Fang Huang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, 138 Yixueyuan Road, Shanghai, 200032, China.
| |
Collapse
|
132
|
Li J, Tong L, Schock BC, Ji LL. Post-traumatic Stress Disorder: Focus on Neuroinflammation. Mol Neurobiol 2023; 60:3963-3978. [PMID: 37004607 DOI: 10.1007/s12035-023-03320-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 03/09/2023] [Indexed: 04/04/2023]
Abstract
Post-traumatic stress disorder (PTSD), gaining increasing attention, is a multifaceted psychiatric disorder that occurs following a stressful or traumatic event or series of events. Recently, several studies showed a close relationship between PTSD and neuroinflammation. Neuroinflammation, a defense response of the nervous system, is associated with the activation of neuroimmune cells such as microglia and astrocytes and with changes in inflammatory markers. In this review, we first analyzed the relationship between neuroinflammation and PTSD: the effect of stress-derived activation of the hypothalamic-pituitary-adrenal (HPA) axis on the main immune cells in the brain and the effect of stimulated immune cells in the brain on the HPA axis. We then summarize the alteration of inflammatory markers in brain regions related to PTSD. Astrocytes are neural parenchymal cells that protect neurons by regulating the ionic microenvironment around neurons. Microglia are macrophages of the brain that coordinate the immunological response. Recent studies on these two cell types provided new insight into neuroinflammation in PTSD. These contribute to promoting comprehension of neuroinflammation, which plays a pivotal role in the pathogenesis of PTSD.
Collapse
Affiliation(s)
- Jimeng Li
- Department of 2nd Clinical College, China Medical University, Shenyang, Liaoning, China
| | - Lei Tong
- Department of Anatomy, College of Basic Sciences, China Medical University, Shenyang, Liaoning, China
| | - Bettina C Schock
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast Faculty of Medicine Health and Life Sciences, Belfast, UK
| | - Li-Li Ji
- Department of Anatomy, College of Basic Sciences, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
133
|
VanderZwaag J, Halvorson T, Dolhan K, Šimončičová E, Ben-Azu B, Tremblay MÈ. The Missing Piece? A Case for Microglia's Prominent Role in the Therapeutic Action of Anesthetics, Ketamine, and Psychedelics. Neurochem Res 2023; 48:1129-1166. [PMID: 36327017 DOI: 10.1007/s11064-022-03772-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 08/25/2022] [Accepted: 09/27/2022] [Indexed: 11/06/2022]
Abstract
There is much excitement surrounding recent research of promising, mechanistically novel psychotherapeutics - psychedelic, anesthetic, and dissociative agents - as they have demonstrated surprising efficacy in treating central nervous system (CNS) disorders, such as mood disorders and addiction. However, the mechanisms by which these drugs provide such profound psychological benefits are still to be fully elucidated. Microglia, the CNS's resident innate immune cells, are emerging as a cellular target for psychiatric disorders because of their critical role in regulating neuroplasticity and the inflammatory environment of the brain. The following paper is a review of recent literature surrounding these neuropharmacological therapies and their demonstrated or hypothesized interactions with microglia. Through investigating the mechanism of action of psychedelics, such as psilocybin and lysergic acid diethylamide, ketamine, and propofol, we demonstrate a largely under-investigated role for microglia in much of the emerging research surrounding these pharmacological agents. Among others, we detail sigma-1 receptors, serotonergic and γ-aminobutyric acid signalling, and tryptophan metabolism as pathways through which these agents modulate microglial phagocytic activity and inflammatory mediator release, inducing their therapeutic effects. The current review includes a discussion on future directions in the field of microglial pharmacology and covers bidirectional implications of microglia and these novel pharmacological agents in aging and age-related disease, glial cell heterogeneity, and state-of-the-art methodologies in microglial research.
Collapse
Affiliation(s)
- Jared VanderZwaag
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Torin Halvorson
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Kira Dolhan
- Department of Psychology, University of Victoria, Vancouver, BC, Canada
- Department of Biology, University of Victoria, Vancouver, BC, Canada
| | - Eva Šimončičová
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Benneth Ben-Azu
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Pharmacology, Faculty of Basic Medical Sciences, College of Health Sciences, Delta State University, Abraka, Delta State, Nigeria
| | - Marie-Ève Tremblay
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada.
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Département de médecine moléculaire, Université Laval, Québec City, QC, Canada.
- Axe Neurosciences, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada.
- Neurology and Neurosurgery Department, McGill University, Montreal, QC, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
- Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
134
|
Sonnefeld L, Rohmann N, Geisler C, Laudes M. Is human obesity an inflammatory disease of the hypothalamus? Eur J Endocrinol 2023; 188:R37-R45. [PMID: 36883605 DOI: 10.1093/ejendo/lvad030] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/23/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023]
Abstract
Obesity and its comorbidities are long-standing, challenging global health problems. Lack of exercise, overnutrition, and especially the consumption of fat-rich foods are some of the most important factors leading to an increase in prevalence in modern society. The pathophysiology of obesity as a metabolic inflammatory disease has moved into focus since new therapeutic approaches are required. The hypothalamus, a brain area responsible for energy homeostasis, has recently received special attention in this regard. Hypothalamic inflammation was identified to be associated with diet-induced obesity and new evidence suggests that it may be, beyond that, a pathological mechanism of the disease. This inflammation impairs the local signaling of insulin and leptin leading to dysfunction of the regulation of energy balance and thus, weight gain. After a high-fat diet consumption, activation of inflammatory mediators such as the nuclear factor κB or c-Jun N-terminal kinase pathway can be observed, accompanied by elevated secretion of pro-inflammatory interleukins and cytokines. Brain resident glia cells, especially microglia and astrocytes, initiate this release in response to the flux of fatty acids. The gliosis occurs rapidly before the actual weight gain. Dysregulated hypothalamic circuits change the interaction between neuronal and non-neuronal cells, contributing to the establishment of inflammatory processes. Several studies have reported reactive gliosis in obese humans. Although there is evidence for a causative role of hypothalamic inflammation in the obesity development, data on underlying molecular pathways in humans are limited. This review discusses the current state of knowledge on the relationship between hypothalamic inflammation and obesity in humans.
Collapse
Affiliation(s)
- Lena Sonnefeld
- Institute of Diabetes and Clinical Metabolic Research, University Medical Centre Schleswig-Holstein, Kiel 24105, Germany
| | - Nathalie Rohmann
- Institute of Diabetes and Clinical Metabolic Research, University Medical Centre Schleswig-Holstein, Kiel 24105, Germany
| | - Corinna Geisler
- Institute of Diabetes and Clinical Metabolic Research, University Medical Centre Schleswig-Holstein, Kiel 24105, Germany
| | - Matthias Laudes
- Institute of Diabetes and Clinical Metabolic Research, University Medical Centre Schleswig-Holstein, Kiel 24105, Germany
- Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Medicine 1, University Medical Centre Schleswig-Holstein, Kiel 24105, Germany
| |
Collapse
|
135
|
Wang J, Men Y, Wang Z. Polydatin Alleviates Chronic Stress-Induced Depressive and Anxiety-like Behaviors in a Mouse Model. ACS Chem Neurosci 2023; 14:977-987. [PMID: 36802487 DOI: 10.1021/acschemneuro.2c00758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023] Open
Abstract
We aimed to investigate whether polydatin could suppress stress-induced depression- and anxiety-like behaviors in a mouse model. Mice were divided into the control group, chronic unpredictable mild stress (CUMS) exposure group, and CUMS mice treated with polydatin group. Following CUMS exposure and polydatin treatment, mice were subjected to behavioral assays to assess depressive-like and anxiety-like behaviors. Synaptic function was determined by the levels of brain-derived neurotrophic factor (BDNF), postsynaptic density protein 95 (PSD95), and synaptophysin (SYN) in the hippocampus and cultured hippocampal neurons. The number and length of dendrites were assessed in cultured hippocampal neurons. Finally, we investigated the effect of polydatin on CUMS-induced inflammation and oxidative stress in the hippocampus by measuring inflammatory cytokine levels, oxidative stress markers such as reactive oxygen species, glutathione peroxidase, catalase, and superoxide dismutase, as well as components of the Nrf2 signaling pathway. Polydatin alleviated CUMS-induced depressive-like behaviors in forced swimming, tail suspension and sucrose preference tests, and anxiety-like behaviors in marble-burying and elevated plus maze tests. Polydatin increased the number and length of dendrites of cultured hippocampal neurons from mice exposed to CUMS and alleviated CUMS-induced synaptic deficits by restoring BDNF, PSD95, and SYN levels in vivo and in vitro. Importantly, polydatin inhibited CUMS-induced hippocampal inflammation and oxidative stress and suppressed the activation of NFκB and Nrf2 pathways. Our study suggests that polydatin may be an effective drug for the treatment of affective disorders through inhibiting neuroinflammation and oxidative stress. Our current findings warrant further study to investigate the potential clinical application of polydatin.
Collapse
Affiliation(s)
- Jianping Wang
- Department of Neurology, Cangzhou Central Hospital, Xinhua West Road, Cangzhou 061000, Hebei, China
| | - Yujiao Men
- Department of Neurology, Cangzhou Central Hospital, Xinhua West Road, Cangzhou 061000, Hebei, China
| | - Zeyu Wang
- Department of Neurology, Cangzhou Central Hospital, Xinhua West Road, Cangzhou 061000, Hebei, China
| |
Collapse
|
136
|
The Pathological Activation of Microglia Is Modulated by Sexually Dimorphic Pathways. Int J Mol Sci 2023; 24:ijms24054739. [PMID: 36902168 PMCID: PMC10003784 DOI: 10.3390/ijms24054739] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/11/2023] [Accepted: 02/22/2023] [Indexed: 03/05/2023] Open
Abstract
Microglia are the primary immunocompetent cells of the central nervous system (CNS). Their ability to survey, assess and respond to perturbations in their local environment is critical in their role of maintaining CNS homeostasis in health and disease. Microglia also have the capability of functioning in a heterogeneous manner depending on the nature of their local cues, as they can become activated on a spectrum from pro-inflammatory neurotoxic responses to anti-inflammatory protective responses. This review seeks to define the developmental and environmental cues that support microglial polarization towards these phenotypes, as well as discuss sexually dimorphic factors that can influence this process. Further, we describe a variety of CNS disorders including autoimmune disease, infection, and cancer that demonstrate disparities in disease severity or diagnosis rates between males and females, and posit that microglial sexual dimorphism underlies these differences. Understanding the mechanism behind differential CNS disease outcomes between men and women is crucial in the development of more effective targeted therapies.
Collapse
|
137
|
Chen YH, Jin SY, Yang JM, Gao TM. The Memory Orchestra: Contribution of Astrocytes. Neurosci Bull 2023; 39:409-424. [PMID: 36738435 PMCID: PMC10043126 DOI: 10.1007/s12264-023-01024-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 01/03/2023] [Indexed: 02/05/2023] Open
Abstract
For decades, memory research has centered on the role of neurons, which do not function in isolation. However, astrocytes play important roles in regulating neuronal recruitment and function at the local and network levels, forming the basis for information processing as well as memory formation and storage. In this review, we discuss the role of astrocytes in memory functions and their cellular underpinnings at multiple time points. We summarize important breakthroughs and controversies in the field as well as potential avenues to further illuminate the role of astrocytes in memory processes.
Collapse
Affiliation(s)
- Yi-Hua Chen
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Shi-Yang Jin
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Jian-Ming Yang
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Tian-Ming Gao
- State Key Laboratory of Organ Failure Research, Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
138
|
Hu X, Li S, Shi Z, Lin WJ, Yang Y, Li Y, Li H, Xu Y, Zhou M, Tang Y. Partial Ablation of Astrocytes Exacerbates Cerebral Infiltration of Monocytes and Neuronal Loss After Brain Stab Injury in Mice. Cell Mol Neurobiol 2023; 43:893-905. [PMID: 35437650 PMCID: PMC11415208 DOI: 10.1007/s10571-022-01224-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/31/2022] [Indexed: 11/03/2022]
Abstract
In traumatic brain injury (TBI), mechanical injury results in instantaneous tissue damages accompanied by subsequent pro-inflammatory cascades composed of microgliosis and astrogliosis. However, the interactive roles between microglia and astrocytes during the pathogenesis of TBI remain unclear and sometimes debatable. In this study, we used a forebrain stab injury mouse model to investigate the pathological role of reactive astrocytes in cellular and molecular changes of inflammatory response following TBI. In the ipsilateral hemisphere of stab-injured brain, monocyte infiltration and neuronal loss, as well as increased elevated astrogliosis, microglia activation and inflammatory cytokines were observed. To verify the role of reactive astrocytes in TBI, local and partial ablation of astrocytes was achieved by stereotactic injection of diphtheria toxin in the forebrain of Aldh1l1-CreERT2::Ai9::iDTR transgenic mice which expressed diphtheria toxin receptor (DTR) in astrocytes after tamoxifen induction. This strategy achieved about 20% of astrocytes reduction at the stab site as validated by immunofluorescence co-staining of GFAP with tdTomato-positive astrocytes. Interestingly, reduction of astrocytes showed increased microglia activation and monocyte infiltration, accompanied with increased severity in stab injury-induced neuronal loss when compared with DTR-/- mice, together with elevation of inflammatory chemokines such as CCL2, CCL5 and CXCL10 in astrogliosis-reduced mice. Collectively, our data verified the interactive role of astrocytes as an immune modulator in suppressing inflammatory responses in the injured brain. Schematic diagram shows monocyte infiltration and neuronal loss, as well as increased elevated astrogliosis, microglia activation and chemokines were observed in the injured site after stab injury. Local and partial ablation of astrocytes led to increased microglia activation and monocyte infiltration, accompanied with increased severity in neuronal loss together with elevation of inflammatory chemokines as compared with control mice subjected stab injury.
Collapse
Affiliation(s)
- Xia Hu
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Shaojian Li
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat‑Sen University, Guangzhou, 510120, China
| | - Zhongshan Shi
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat‑Sen University, Guangzhou, 510120, China
| | - Wei-Jye Lin
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Yuhua Yang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat‑Sen University, Guangzhou, 510120, China
| | - Yi Li
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat‑Sen University, Guangzhou, 510120, China
| | - Honghong Li
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat‑Sen University, Guangzhou, 510120, China
| | - Yongteng Xu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat‑Sen University, Guangzhou, 510120, China
| | - Meijuan Zhou
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China.
| | - Yamei Tang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat‑Sen University, Guangzhou, 510120, China.
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-Sen University, Guangzhou, 510275, China.
| |
Collapse
|
139
|
Henao‐Restrepo J, López‐Murillo C, Valderrama‐Carmona P, Orozco‐Santa N, Gomez J, Gutiérrez‐Vargas J, Moraga R, Toledo J, Littau JL, Härtel S, Arboleda‐Velásquez JF, Sepulveda‐Falla D, Lopera F, Cardona‐Gómez GP, Villegas A, Posada‐Duque R. Gliovascular alterations in sporadic and familial Alzheimer's disease: APOE3 Christchurch homozygote glioprotection. Brain Pathol 2023; 33:e13119. [PMID: 36130084 PMCID: PMC10041169 DOI: 10.1111/bpa.13119] [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: 01/21/2022] [Accepted: 08/30/2022] [Indexed: 11/28/2022] Open
Abstract
In response to brain insults, astrocytes become reactive, promoting protection and tissue repair. However, astroglial reactivity is typical of brain pathologies, including Alzheimer's disease (AD). Considering the heterogeneity of the reactive response, the role of astrocytes in the course of different forms of AD has been underestimated. Colombia has the largest human group known to have familial AD (FAD). This group carries the autosomal dominant and fully penetrant mutation E280A in PSEN1, which causes early-onset AD. Recently, our group identified an E280A carrier who did not develop FAD. The individual was homozygous for the Christchurch mutation R136S in APOE3 (APOEch). Remarkably, APOE is the main genetic risk factor for developing sporadic AD (SAD) and most of cerebral ApoE is produced by astroglia. Here, we characterized astrocyte properties related to reactivity, glutamate homeostasis, and structural integrity of the gliovascular unit (GVU), as factors that could underlie the pathogenesis or protection of AD. Specifically, through histological and 3D microscopy analyses of postmortem samples, we briefly describe the histopathology and cytoarchitecture of the frontal cortex of SAD, FAD, and APOEch, and demonstrate that, while astrodegeneration and vascular deterioration are prominent in SAD, FAD is characterized by hyperreactive-like glia, and APOEch displays the mildest astrocytic and vascular alterations despite having the highest burden of Aβ. Notably, astroglial, gliovascular, and vascular disturbances, as well as brain cell death, correlate with the specific astrocytic phenotypes identified in each condition. This study provides new insights into the potential relevance of the gliovasculature in the development and protection of AD. To our knowledge, this is the first study assessing the components of the GVU in human samples of SAD, FAD, and APOEch.
Collapse
Affiliation(s)
- Julián Henao‐Restrepo
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Carolina López‐Murillo
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Pablo Valderrama‐Carmona
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Natalia Orozco‐Santa
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Johana Gomez
- Grupo de Neurociencias de Antioquia, Facultad de MedicinaSIU, Universidad de AntioquiaMedellínColombia
| | - Johanna Gutiérrez‐Vargas
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Health Sciences FacultyRemington University CorporationMedellínColombia
| | - Renato Moraga
- Biomedical Neuroscience Institute BNI, Faculty of MedicineUniversity of ChileSantiagoChile
| | - Jorge Toledo
- Biomedical Neuroscience Institute BNI, Faculty of MedicineUniversity of ChileSantiagoChile
| | - Jessica Lisa Littau
- Molecular Neuropathology of Alzheimer's DiseaseInstitute of Neuropathology, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Steffen Härtel
- Biomedical Neuroscience Institute BNI, Faculty of MedicineUniversity of ChileSantiagoChile
| | - Joseph F. Arboleda‐Velásquez
- Schepens Eye Research Institute of Mass Eye and Ear, Department of OphthalmologyHarvard Medical SchoolBostonMassachusettsUSA
| | - Diego Sepulveda‐Falla
- Molecular Neuropathology of Alzheimer's DiseaseInstitute of Neuropathology, University Medical Center Hamburg‐EppendorfHamburgGermany
| | - Francisco Lopera
- Grupo de Neurociencias de Antioquia, Facultad de MedicinaSIU, Universidad de AntioquiaMedellínColombia
| | - Gloria Patricia Cardona‐Gómez
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| | - Andrés Villegas
- Grupo de Neurociencias de Antioquia, Facultad de MedicinaSIU, Universidad de AntioquiaMedellínColombia
| | - Rafael Posada‐Duque
- Instituto de Biología, Facultad de Ciencias Exactas y NaturalesUniversidad de AntioquiaMedellínColombia
- Área de Neurobiología Celular y Molecular, Grupo de Neurociencias de AntioquiaUniversidad de AntioquiaMedellínColombia
| |
Collapse
|
140
|
Olufunmilayo EO, Gerke-Duncan MB, Holsinger RMD. Oxidative Stress and Antioxidants in Neurodegenerative Disorders. Antioxidants (Basel) 2023; 12:antiox12020517. [PMID: 36830075 PMCID: PMC9952099 DOI: 10.3390/antiox12020517] [Citation(s) in RCA: 219] [Impact Index Per Article: 109.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Neurodegenerative disorders constitute a substantial proportion of neurological diseases with significant public health importance. The pathophysiology of neurodegenerative diseases is characterized by a complex interplay of various general and disease-specific factors that lead to the end point of neuronal degeneration and loss, and the eventual clinical manifestations. Oxidative stress is the result of an imbalance between pro-oxidant species and antioxidant systems, characterized by an elevation in the levels of reactive oxygen and reactive nitrogen species, and a reduction in the levels of endogenous antioxidants. Recent studies have increasingly highlighted oxidative stress and associated mitochondrial dysfunction to be important players in the pathophysiologic processes involved in neurodegenerative conditions. In this article, we review the current knowledge of the general effects of oxidative stress on the central nervous system, the different specific routes by which oxidative stress influences the pathophysiologic processes involved in Alzheimer's disease, Parkinson's disease, Amyotrophic Lateral Sclerosis and Huntington's disease, and how oxidative stress may be therapeutically reversed/mitigated in order to stall the pathological progression of these neurodegenerative disorders to bring about clinical benefits.
Collapse
Affiliation(s)
- Edward O. Olufunmilayo
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Department of Medicine, University College Hospital, Queen Elizabeth Road, Oritamefa, Ibadan 5116, PMB, Nigeria
| | - Michelle B. Gerke-Duncan
- Education Innovation, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - R. M. Damian Holsinger
- Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Neuroscience, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Correspondence:
| |
Collapse
|
141
|
Chang J, Qian Z, Wang B, Cao J, Zhang S, Jiang F, Kong R, Yu X, Cao X, Yang L, Chen H. Transplantation of A2 type astrocytes promotes neural repair and remyelination after spinal cord injury. Cell Commun Signal 2023; 21:37. [PMID: 36797790 PMCID: PMC9936716 DOI: 10.1186/s12964-022-01036-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/28/2022] [Indexed: 02/18/2023] Open
Abstract
BACKGROUND Limited progress in terms of an effective treatment for spinal cord injury (SCI) emphasizes the urgent need for novel therapies. As a vital central nervous system component, the resident astrocytes play crucial roles in regulating recovery after SCI. In this study, recovery after SCI was compared following the transplantation of either A1 or A2 astrocytes. A1 astrocytes are harmful as they upregulate the neurotoxic classical complement cascade genes. Conversely, A2 astrocytes are characterized as neuroprotective as they upregulate the production of many neurotrophic factors. METHODS We used different supernatant obtained from microglia stimulated with lipopolysaccharide or interleukin-4 to generate A1 and A2 astrocytes. We detected the influence of astrocytes on neurons by co-culturing A1 and A2 astrocytes with neurons. We transplanted astrocytes into the lesion site of the spinal cord and assessed lesion progression, neural restoration, glia formation and locomotor recovery. RESULTS Astrocytes were polarized into A1 and A2 phenotypes following culture in the supernatant obtained from microglia stimulated with lipopolysaccharide or interleukin-4, respectively. Furthermore, co-culturing A2 astrocytes with neurons significantly suppressed glutamate-induced neuronal apoptosis and promoted the degree of neuron arborization. Transplantation of these A2 astrocytes into the lesion site of the spinal cord of mice significantly improved motor function recovery, preserved spared supraspinal pathways, decreased glia scar deposition, and increased neurofilament formation at the site of injury compared to the transplantation of A1 astrocytes. Additionally, enhanced A2 astrocytes with potentially beneficial A2-like genes were also detected in the A2 group. Moreover, luxol fast blue staining and electron microscopy indicated increased preservation of myelin with organized structure after transplantation of A2 astrocytes than of A1 astrocytes. CONCLUSIONS A2 astrocyte transplantation could be a promising potential therapy for SCI. Video abstract.
Collapse
Affiliation(s)
- Jie Chang
- Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China.,Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Zhanyang Qian
- Spine Center, Zhongda Hospital of Southeast University, Nanjing, Jiangsu, China
| | - Binyu Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Jiang Cao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Sheng Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Fan Jiang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Renyi Kong
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Xiao Yu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China
| | - Xiaojian Cao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, Jiangsu, China.
| | - Lei Yang
- Department of Orthopedics, Taizhou People's Hospital, Nanjing Medical University, No. 366 Taihu Road, Taizhou, 225300, Jiangsu, China. .,School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Hongtao Chen
- Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
142
|
Cai W, Wu T, Chen N. The Amyloid-Beta Clearance: From Molecular Targets to Glial and Neural Cells. Biomolecules 2023; 13:313. [PMID: 36830682 PMCID: PMC9953441 DOI: 10.3390/biom13020313] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 02/03/2023] [Accepted: 02/04/2023] [Indexed: 02/11/2023] Open
Abstract
The deposition of amyloid-beta (Aβ) plaques in the brain is one of the primary pathological characteristics of Alzheimer's disease (AD). It can take place 20-30 years before the onset of clinical symptoms. The imbalance between the production and the clearance of Aβ is one of the major causes of AD. Enhancing Aβ clearance at an early stage is an attractive preventive and therapeutic strategy of AD. Direct inhibition of Aβ production and aggregation using small molecules, peptides, and monoclonal antibody drugs has not yielded satisfactory efficacy in clinical trials for decades. Novel approaches are required to understand and combat Aβ deposition. Neurological dysfunction is a complex process that integrates the functions of different types of cells in the brain. The role of non-neurons in AD has not been fully elucidated. An in-depth understanding of the interactions between neurons and non-neurons can contribute to the elucidation of Aβ formation and the identification of effective drug targets. AD patient-derived pluripotent stem cells (PSCs) contain complete disease background information and have the potential to differentiate into various types of neurons and non-neurons in vitro, which may bring new insight into the treatment of AD. Here, we systematically review the latest studies on Aβ clearance and clarify the roles of cell interactions among microglia, astroglia and neurons in response to Aβ plaques, which will be beneficial to explore methods for reconstructing AD disease models using inducible PSCs (iPSCs) through cell differentiation techniques and validating the applications of models in understanding the formation of Aβ plaques. This review may provide the most promising directions of finding the clues for preventing and delaying the development of AD.
Collapse
Affiliation(s)
| | | | - Ning Chen
- Tianjiu Research and Development Center for Exercise Nutrition and Foods, Hubei Key Laboratory of Exercise Training and Monitoring, College of Sports Medicine, Wuhan Sports University, Wuhan 430079, China
| |
Collapse
|
143
|
Zhang T, Zhang M, Cui S, Liang W, Jia Z, Guo F, Ou W, Wu Y, Zhang S. The core of maintaining neuropathic pain: Crosstalk between glial cells and neurons (neural cell crosstalk at spinal cord). Brain Behav 2023; 13:e2868. [PMID: 36602945 PMCID: PMC9927860 DOI: 10.1002/brb3.2868] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/29/2022] [Accepted: 12/06/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Neuropathic pain (NP) caused by the injury or dysfunction of the nervous system is a chronic pain state accompanied by hyperalgesia, and the available clinical treatment is relatively scarce. Hyperalgesia mediated by pro-inflammatory factors and chemokines plays an important role in the occurrence and maintenance of NP. DATA TREATMENT Therefore, we conducted a systematic literature review of experimental NP (PubMed Medline), in order to find the mechanism of inducing central sensitization and explore the intervention methods of hyperalgesia caused by real or simulated injury. RESULT In this review, we sorted out the activation pathways of microglia, astrocytes and neurons, and the process of crosstalk among them. It was found that in NP, the microglia P2X4 receptor is the key target, which can activate the mitogen-activated protein kinase pathway inward and then activate astrocytes and outwardly activate neuronal tropomyosin receptor kinase B receptor to activate neurons. At the same time, activated neurons continue to maintain the activation of astrocytes and microglia through chemokines on CXCL13/CXCR5 and CX3CL1/CX3CR1. This crosstalk process is the key to maintaining NP. CONCLUSION We summarize the further research on crosstalk among neurons, microglia, and astrocytes in the central nervous system, elaborate the ways and connections of relevant crosstalk, and find potential crosstalk targets, which provides a reference for drug development and preclinical research.
Collapse
Affiliation(s)
- Tianrui Zhang
- Department of Pharmacology of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Mingqian Zhang
- Department of Pharmacology of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shuang Cui
- Department of Pharmacology of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wulin Liang
- Department of Pharmacology of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhanhong Jia
- Department of Pharmacology of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Fanfan Guo
- Department of Pharmacology of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wenjing Ou
- Department of Pharmacology of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yonghong Wu
- Department of Pharmacology of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shuofeng Zhang
- Department of Pharmacology of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
144
|
Knockout of transient receptor potential ankyrin 1 (TRPA1) modulates the glial phenotype and alleviates perihematomal neuroinflammation after intracerebral hemorrhage in mice via MAPK/NF-κB signaling. Neuroreport 2023; 34:81-92. [PMID: 36608163 PMCID: PMC9815814 DOI: 10.1097/wnr.0000000000001862] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The objective is to explore the role of astrocytic transient receptor potential ankyrin 1 (TRPA1) in glial phenotype transformation in neuroinflammation after intracerebral hemorrhage (ICH). Wild-type astrocytes and TRPA1-/- astrocytes were subjected to 6-h hemin treatment, and the calcium ions and transcriptome sequencing were assessed. A mouse autologous blood injection ICH model was established to evaluate the proliferation and phenotypes of astrocytes and microglia around the hematoma. The neuroinflammation and behavioral performance of wild-type ICH mice and TRPA1-/- ICH mice were assessed. Knockout of astrocytic TRPA1 decreased calcium ions of astrocytes after hemin treatment in-vitro, and microglial and astrocytes around the hematoma proliferated after the ICH model. Furthermore, RNA-sequencing (RNA-seq), immunofluorescence, and Western blotting results showed that the activated astrocytes transformed into the A2 phenotype in TRPA1-/- ICH mice. The 'ameboid' microglia were observed around the hematoma in TRPA1-/- ICH mice. The proliferation of A2 astrocytes and 'ameboid' microglia ameliorated the neuroinflammation after ICH. The inflammatory response was reduced by inhibiting the mitogen-activated protein kinase/nuclear factor kappa-B signaling pathway, and neurologic deficits were improved in TRPA1-/- ICH mice compared with wild-type ICH mice. This research suggests that astrocytic TRPA1 is a new therapeutic target to rescue neuroinflammation by modulating the glial phenotype after ICH.
Collapse
|
145
|
Ghandy N, Ebrahimzadeh-Bideskan A, Gorji A, Negah SS. Co-transplantation of novel Nano-SDF scaffold with human neural stem cells attenuates inflammatory responses and apoptosis in traumatic brain injury. Int Immunopharmacol 2023; 115:109709. [PMID: 36638659 DOI: 10.1016/j.intimp.2023.109709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/02/2023] [Accepted: 01/05/2023] [Indexed: 01/12/2023]
Abstract
Traumatic brain injury (TBI) causes long-term disability and mortality worldwide. The prime pathological players in TBI are neuroinflammation and apoptosis. These pathological changes lead to a limited capacity of regeneration after TBI. To alleviate inflammatory responses and apoptosis triggered by TBI, developing bioactive scaffolds conjoined with stem cells is a decisive approach in neural tissue engineering. The aim of this study was to fabricate a novel nano-scaffold made of RADA-16 with a bioactive motif of stromal cell-derived factor-1 α (SDF-1α) and evaluate its effects with stem cell transplantation on inflammatory pathways, reactive gliosis, and apoptosis after TBI. Co-transplantation of Nano-SDF and human neural stem cells (hNSCs) derived from fetus brain in adult rats subjected to TBI led to the improvement of motor activitycompared with the control group. The treated animals with hNSCs + Nano-SDF had a significantly lower expression of toll-like receptor 4 and nuclear factor-kappa B at the injury site than the control animals. A significant reduction in the number of reactive astrocytes was also observed in rats that received hNSCs + Nano-SDF compared with the vehicle and Nano-SDF groups. Furthermore, the TUNEL assay indicated a significant reduction in TUNEL positive cells in the hNSCs + Nano-SDF group compared with the TBI, vehicle, and Nano-SDF groups. These data demonstrated co-transplantation of hNSCs with Nano-SDF can reduce inflammatory responses and cell death after TBI via creating a more supportive microenvironment. Further research is required to establish the therapeutic efficacy of Nano-SDF with stem cells for TBI.
Collapse
Affiliation(s)
- Nasibeh Ghandy
- Department of Anatomy and Cell Biology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Alireza Ebrahimzadeh-Bideskan
- Department of Anatomy and Cell Biology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Ali Gorji
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran; Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Münster, Germany.
| | - Sajad Sahab Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran; Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
146
|
Myers AJ, Brahimi A, Jenkins IJ, Koob AO. The Synucleins and the Astrocyte. BIOLOGY 2023; 12:biology12020155. [PMID: 36829434 PMCID: PMC9952504 DOI: 10.3390/biology12020155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023]
Abstract
Synucleins consist of three proteins exclusively expressed in vertebrates. α-Synuclein (αS) has been identified as the main proteinaceous aggregate in Lewy bodies, a pathological hallmark of many neurodegenerative diseases. Less is understood about β-synuclein (βS) and γ-synuclein (γS), although it is known βS can interact with αS in vivo to inhibit aggregation. Likewise, both γS and βS can inhibit αS's propensity to aggregate in vitro. In the central nervous system, βS and αS, and to a lesser extent γS, are highly expressed in the neural presynaptic terminal, although they are not strictly located there, and emerging data have shown a more complex expression profile. Synapse loss and astrocyte atrophy are early aspects of degenerative diseases of the brain and correlate with disease progression. Synucleins appear to be involved in synaptic transmission, and astrocytes coordinate and organize synaptic function, with excess αS degraded by astrocytes and microglia adjacent to the synapse. βS and γS have also been observed in the astrocyte and may provide beneficial roles. The astrocytic responsibility for degradation of αS as well as emerging evidence on possible astrocytic functions of βS and γS, warrant closer inspection on astrocyte-synuclein interactions at the synapse.
Collapse
Affiliation(s)
- Abigail J. Myers
- Neuroscience Program, Health Science Research Facility, University of Vermont, 149 Beaumont Ave., Burlington, VT 05405, USA
| | - Ayat Brahimi
- Biology Department, University of Hartford, 200 Bloomfield Ave., West Hartford, CT 06117, USA
| | - Imani J. Jenkins
- Biology Department, University of Hartford, 200 Bloomfield Ave., West Hartford, CT 06117, USA
| | - Andrew O. Koob
- Biology Department, University of Hartford, 200 Bloomfield Ave., West Hartford, CT 06117, USA
- Correspondence: ; Tel.: +1-860-768-5780
| |
Collapse
|
147
|
Zhang W, Wu Q, Hao S, Chen S. The hallmark and crosstalk of immune cells after intracerebral hemorrhage: Immunotherapy perspectives. Front Neurosci 2023; 16:1117999. [PMID: 36711145 PMCID: PMC9877537 DOI: 10.3389/fnins.2022.1117999] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 12/30/2022] [Indexed: 01/13/2023] Open
Abstract
Intracerebral hemorrhage (ICH) is one of the most dangerous types of strokes with a high morbidity and mortality rate. Currently, the treatment of ICH is not well developed, mainly because its mechanisms are still unclear. Inflammation is one of the main types of secondary injury after ICH and catalyzes the adverse consequences of ICH. A large number of immune cells are involved in neuroinflammation, such as microglia, astrocytes, oligodendrocytes, lymphocytes, macrophages, and neutrophils. Nevertheless, the characteristics and crosstalk of immune cells have not been fully elucidated. In this review, we endeavor to delve into the respective characteristics of immune cells and their interactions in neuroimmune inflammation, and further elucidate favorable immunotherapeutic approaches regarding ICH, and finally present an outlook.
Collapse
Affiliation(s)
- Wenqing Zhang
- School of Medicine, Chongqing University, Chongqing, China,Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Qingyuan Wu
- Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Shilei Hao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China,*Correspondence: Shilei Hao,
| | - Shengli Chen
- Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, China,Shengli Chen,
| |
Collapse
|
148
|
Lin MS, Wang YC, Chen WJ, Kung WM. Impact of gut-brain interaction in emerging neurological disorders. World J Clin Cases 2023; 11:1-6. [PMID: 36687174 PMCID: PMC9846976 DOI: 10.12998/wjcc.v11.i1.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/29/2022] [Accepted: 12/15/2022] [Indexed: 01/04/2023] Open
Abstract
The central nervous system (CNS) is a reservoir of immune privilege. Specialized immune glial cells are responsible for maintenance and defense against foreign invaders. The blood-brain barrier (BBB) prevents detrimental pathogens and potentially overreactive immune cells from entering the periphery. When the double-edged neuroinflammatory response is overloaded, it no longer has the protective function of promoting neuroregeneration. Notably, microbiota and its derivatives may emerge as pathogen-associated molecular patterns of brain pathology, causing microbiome-gut-brain axis dysregulation from the bottom-up. When dysbiosis of the gastrointestinal flora leads to subsequent alterations in BBB permeability, peripheral immune cells are recruited to the brain. This results in amplification of neuroinflammatory circuits in the brain, which eventually leads to specific neurological disorders. Aggressive treatment strategies for gastrointestinal disorders may protect against specific immune responses to gastrointestinal disorders, which can lead to potential protective effects in the CNS. Accordingly, this study investigated the mutual effects of microbiota and the gut-brain axis, which may provide targeting strategies for future disease treatment.
Collapse
Affiliation(s)
- Muh-Shi Lin
- Division of Neurosurgery, Department of Surgery, Kuang Tien General Hospital, Taichung 43303, Taiwan
- Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, Yilan 26047, Taiwan
- Department of Biotechnology, College of Medical and Health Care, Hung Kuang University, Taichung 43302, Taiwan
- Department of Health Business Administration, College of Medical and Health Care, Hung Kuang University, Taichung 43302, Taiwan
| | - Yao-Chin Wang
- Department of Emergency, Min-Sheng General Hospital, Taoyuan 33044, Taiwan
- Graduate Institute of Injury Prevention and Control, College of Public Health, Taipei Medical University, Taipei 11031, Taiwan
| | - Wei-Jung Chen
- Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, Yilan 26047, Taiwan
| | - Woon-Man Kung
- Division of Neurosurgery, Department of Surgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan
- Department of Exercise and Health Promotion, College of Kinesiology and Health, Chinese Culture University, Taipei 11114, Taiwan
| |
Collapse
|
149
|
Xiong Y, Chen J, Li Y. Microglia and astrocytes underlie neuroinflammation and synaptic susceptibility in autism spectrum disorder. Front Neurosci 2023; 17:1125428. [PMID: 37021129 PMCID: PMC10067592 DOI: 10.3389/fnins.2023.1125428] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 03/03/2023] [Indexed: 04/07/2023] Open
Abstract
Autism spectrum disorder (ASD) is a common neurodevelopmental disorder with onset in childhood. The mechanisms underlying ASD are unclear. In recent years, the role of microglia and astrocytes in ASD has received increasing attention. Microglia prune the synapses or respond to injury by sequestrating the injury site and expressing inflammatory cytokines. Astrocytes maintain homeostasis in the brain microenvironment through the uptake of ions and neurotransmitters. However, the molecular link between ASD and microglia and, or astrocytes remains unknown. Previous research has shown the significant role of microglia and astrocytes in ASD, with reports of increased numbers of reactive microglia and astrocytes in postmortem tissues and animal models of ASD. Therefore, an enhanced understanding of the roles of microglia and astrocytes in ASD is essential for developing effective therapies. This review aimed to summarize the functions of microglia and astrocytes and their contributions to ASD.
Collapse
|
150
|
Wen L, Miao X, Ding J, Tong X, Wu Y, He Y, Zheng F. Pesticides as a risk factor for cognitive impairment: Natural substances are expected to become alternative measures to prevent and improve cognitive impairment. Front Nutr 2023; 10:1113099. [PMID: 36937345 PMCID: PMC10016095 DOI: 10.3389/fnut.2023.1113099] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/01/2023] [Indexed: 03/08/2023] Open
Abstract
Pesticides are the most effective way to control diseases, insects, weeds, and fungi. The central nervous system (CNS) is damaged by pesticide residues in various ways. By consulting relevant databases, the systemic relationships between the possible mechanisms of pesticides damage to the CNS causing cognitive impairment and related learning and memory pathways networks, as well as the structure-activity relationships between some natural substances (such as polyphenols and vitamins) and the improvement were summarized in this article. The mechanisms of cognitive impairment caused by pesticides are closely related. For example, oxidative stress, mitochondrial dysfunction, and neuroinflammation can constitute three feedback loops that interact and restrict each other. The mechanisms of neurotransmitter abnormalities and intestinal dysfunction also play an important role. The connection between pathways is complex. NMDAR, PI3K/Akt, MAPK, Keap1/Nrf2/ARE, and NF-κB pathways can be connected into a pathway network by targets such as Ras, Akt, and IKK. The reasons for the improvement of natural substances are related to their specific structure, such as polyphenols with different hydroxyl groups. This review's purpose is to lay a foundation for exploring and developing more natural substances that can effectively improve the cognitive impairment caused by pesticides.
Collapse
Affiliation(s)
- Liankui Wen
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
| | - Xiwen Miao
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
| | - Jia Ding
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
| | - Xuewen Tong
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
| | - Yuzhu Wu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- National Engineering Research Center for Wheat and Corn Deep Processing, Changchun, China
- *Correspondence: Yuzhu Wu, ✉
| | - Yang He
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Yang He, ✉
| | - Fei Zheng
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, China
- Fei Zheng, ✉
| |
Collapse
|