1
|
Tawbeh A, Gondcaille C, Saih FE, Raas Q, Loichot D, Hamon Y, Keime C, Benani A, Di Cara F, Cherkaoui-Malki M, Andreoletti P, Savary S. Impaired peroxisomal beta-oxidation in microglia triggers oxidative stress and impacts neurons and oligodendrocytes. Front Mol Neurosci 2025; 18:1542938. [PMID: 39958993 PMCID: PMC11826809 DOI: 10.3389/fnmol.2025.1542938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/10/2024] [Accepted: 01/15/2025] [Indexed: 02/18/2025] Open
Abstract
Microglia, the immune cells of the central nervous system, activate neuroinflammatory pathways in response to homeostatic disturbances, a process implicated in the pathogenesis of various neurodegenerative diseases. Emerging evidence identifies abnormal microglial activation as a causal factor at the onset of peroxisomal leukodystrophies, including X-linked adrenoleukodystrophy (X-ALD). This study investigates how primary peroxisomal deficiencies influence oxidative properties of microglia and examines the subsequent impact on neurons and oligodendrocytes. Using BV-2 microglial cells lacking ABCD1, ABCD2, or ACOX1, peroxisomal proteins that play key roles in the very-long-chain fatty acid beta-oxidation, we analyzed their response under basal condition and after stimulation by lipopolysaccharide (LPS). Transcriptomic analysis of the mutant microglial cells revealed numerous differentially expressed genes, particularly in redox-related pathways following LPS exposure. These changes are consistent with the increased production of reactive oxygen species (ROS) and nitric oxide (NO). Conditioned media (CM) from the mutant cells were then applied to cultures of neuron and oligodendrocyte cell lines. Exposure to CM from LPS-stimulated mutant microglial cells significantly increased apoptosis in both cell types. Furthermore, treated neurons exhibited a reduction in cell complexity and an increased ability to secrete neuropeptides. These findings demonstrate that peroxisomal impairments in microglia exacerbate inflammatory response and ROS/NO production, affecting the survival of neurons and oligodendrocytes, as well as neuronal morphology and function. This dysfunction might contribute to the early neurodegenerative events in X-ALD by triggering and sustaining neuroinflammatory cascades. Therapeutic strategies that target microglial activation and secretion profiles could hold promise in managing peroxisomal disorders such as X-ALD.
Collapse
Affiliation(s)
- Ali Tawbeh
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, University of Bourgogne, Dijon, France
| | - Catherine Gondcaille
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, University of Bourgogne, Dijon, France
| | - Fatima-Ezzahra Saih
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, University of Bourgogne, Dijon, France
| | - Quentin Raas
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, University of Bourgogne, Dijon, France
| | - Damien Loichot
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, University of Bourgogne, Dijon, France
| | - Yannick Hamon
- Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| | - Céline Keime
- Plateforme GenomEast, IGBMC, CNRS UMR, Inserm, University of Strasbourg, Strasbourg, France
| | - Alexandre Benani
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, University of Bourgogne, Dijon, France
| | - Francesca Di Cara
- Department of Microbiology and Immunology, IWK Health Centre, Dalhousie University, Halifax, NS, Canada
| | - Mustapha Cherkaoui-Malki
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, University of Bourgogne, Dijon, France
| | - Pierre Andreoletti
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, University of Bourgogne, Dijon, France
| | - Stéphane Savary
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut Agro, University of Bourgogne, Dijon, France
| |
Collapse
|
2
|
Rodriguez Fernandez V, Amato R, Piaggi S, Pinto B, Casini G, Bruschi F. A New Ex Vivo Model Based on Mouse Retinal Explants for the Study of Ocular Toxoplasmosis. Pathogens 2024; 13:701. [PMID: 39204301 PMCID: PMC11356793 DOI: 10.3390/pathogens13080701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/27/2024] [Revised: 08/05/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024] Open
Abstract
Ocular toxoplasmosis is the most prevalent clinical manifestation of T. gondii infection, which causes irreversible retinal damage. Different experimental models have been developed to study this pathology. In the present study, a new, ex vivo model is proposed to contribute to the elucidation of disease mechanisms and to possible therapeutic solutions. Ex-vivo retinal explants, prepared from mouse retinas following established protocols, were incubated with T. gondii tachyzoites maintained in Vero cells. At different times, starting at 12 h up to 10 days of incubation, the explants were analyzed with immunofluorescence and Western blot to investigate their responses to parasite infection. T. gondii invasion of the retinal thickness was evident after 3 days in culture, where parasites could be detected around retinal cell nuclei. This was paralleled by putative cyst formation and microglial activation. At the same time, an evident increase in inflammatory and oxidative stress markers was detected in infected explants compared to controls. Cell death also appeared to occur in retinal explants after 3 days of T. gondii infection, and it was characterized by increased necroptotic but not apoptotic markers. The proposed model recapitulates the main characteristics of T. gondii retinal infection within 3 days of incubation and, therefore, allows for studying the very early events of the process. In addition, it requires only a limited number of animals and offers easy manipulation and accessibility for setting up different experimental conditions and assessing the effects of putative drugs for therapy.
Collapse
Affiliation(s)
- Veronica Rodriguez Fernandez
- Department of Translational Research, School of Medicine, University of Pisa, 56126 Pisa, Italy
- Department of Infectious Diseases and Public Health, La Sapienza University, 00185 Rome, Italy
| | - Rosario Amato
- Department of Biology, University of Pisa, 56126 Pisa, Italy
| | - Simona Piaggi
- Department of Translational Research, School of Medicine, University of Pisa, 56126 Pisa, Italy
| | - Barbara Pinto
- Department of Translational Research, School of Medicine, University of Pisa, 56126 Pisa, Italy
| | - Giovanni Casini
- Department of Biology, University of Pisa, 56126 Pisa, Italy
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56126 Pisa, Italy
| | - Fabrizio Bruschi
- Department of Translational Research, School of Medicine, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
3
|
Munan S, Mondal A, Shailja S, Pati S, Samanta A. Unique Synthetic Strategy for Probing in Situ Lysosomal NO for Screening Neuroinflammatory Phenotypes against SARS-CoV-2 RNA in Phagocytotic Microglia. Anal Chem 2024; 96:7479-7486. [PMID: 38689560 DOI: 10.1021/acs.analchem.3c05981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 05/02/2024]
Abstract
In the pathogenesis of microglia, brain immune cells promote nitrergic stress by overproducing nitric oxide (NO), leading to neuroinflammation. Furthermore, NO has been linked to COVID-19 progression, which has caused significant morbidity and mortality. SARS-CoV-2 infection activates inflammation by releasing excess NO and causing cell death in human microglial clone 3 (HMC3). In addition, NO regulates lysosomal functions and complex machinery to neutralize pathogens through phagocytosis. Therefore, developing lysosome-specific NO probes to monitor phagocytosis in microglia during the COVID-19 infection would be a significant study. Herein, a unique synthetic strategy was adopted to develop a NO selective fluorescent probe, PDM-NO, which can discriminate activated microglia from their resting state. The nonfluorescent PDM-NO exhibits a turn-on response toward NO only at lysosomal pH (4.5-5.5). Quantum chemical calculations (DFT/TD-DFT/PCM) and photophysical study revealed that the photoinduced electron transfer (PET) process is pivotal in tuning optical properties. PDM-NO demonstrated good biocompatibility and lysosomal specificity in activated HMC3 cells. Moreover, it can effectively map the dynamics of lysosomal NO against SARS-CoV-2 RNA-induced neuroinflammation in HMC3. Thus, PDM-NO is a potential fluorescent marker for detecting RNA virus infection and monitoring phagocytosis in HMC3.
Collapse
Affiliation(s)
- Subrata Munan
- Molecular Sensors and Therapeutics (MST) Research Laboratory, Department of Chemistry, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University (SNIoE), Delhi NCR, NH 91, Tehsil Dadri, Greater Noida, Uttar Pradesh 201314, India
| | - Abir Mondal
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University (SNIoE), Delhi NCR, NH 91, Tehsil Dadri, Greater Noida, Uttar Pradesh 201314, India
| | - Singh Shailja
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| | - Soumya Pati
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University (SNIoE), Delhi NCR, NH 91, Tehsil Dadri, Greater Noida, Uttar Pradesh 201314, India
| | - Animesh Samanta
- Molecular Sensors and Therapeutics (MST) Research Laboratory, Department of Chemistry, School of Natural Sciences, Shiv Nadar Institution of Eminence Deemed to be University (SNIoE), Delhi NCR, NH 91, Tehsil Dadri, Greater Noida, Uttar Pradesh 201314, India
| |
Collapse
|
4
|
Meleady L, Towriss M, Kim J, Bacarac V, Dang V, Rowland ME, Ciernia AV. Histone deacetylase 3 regulates microglial function through histone deacetylation. Epigenetics 2023; 18:2241008. [PMID: 37506371 PMCID: PMC10392760 DOI: 10.1080/15592294.2023.2241008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/25/2022] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
As the primary innate immune cells of the brain, microglia respond to damage and disease through pro-inflammatory release of cytokines and neuroinflammatory molecules. Histone acetylation is an activating transcriptional mark that regulates inflammatory gene expression. Inhibition of histone deacetylase 3 (Hdac3) has been utilized in pre-clinical models of depression, stroke, and spinal cord injury to improve recovery following injury, but the molecular mechanisms underlying Hdac3's regulation of inflammatory gene expression in microglia is not well understood. To address this lack of knowledge, we examined how pharmacological inhibition of Hdac3 in an immortalized microglial cell line (BV2) impacted histone acetylation and gene expression of pro- and anti-inflammatory genes in response to immune challenge with lipopolysaccharide (LPS). Flow cytometry and cleavage under tags & release using nuclease (CUT & RUN) revealed that Hdac3 inhibition increases global and promoter-specific histone acetylation, resulting in the release of gene repression at baseline and enhanced responses to LPS. Hdac3 inhibition enhanced neuroprotective functions of microglia in response to LPS through reduced nitric oxide release and increased phagocytosis. The findings suggest Hdac3 serves as a regulator of microglial inflammation, and that inhibition of Hdac3 facilitates the microglial response to inflammation and its subsequent clearing of debris or damaged cells. Together, this work provides new mechanistic insights into therapeutic applications of Hdac3 inhibition which mediate reduced neuroinflammatory insults through microglial response.
Collapse
Affiliation(s)
- Laura Meleady
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Morgan Towriss
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Jennifer Kim
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
| | - Vince Bacarac
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Vivien Dang
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Megan E. Rowland
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Annie Vogel Ciernia
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| |
Collapse
|
5
|
Nelissen E, Schepers M, Ponsaerts L, Foulquier S, Bronckaers A, Vanmierlo T, Sandner P, Prickaerts J. Soluble guanylyl cyclase: A novel target for the treatment of vascular cognitive impairment? Pharmacol Res 2023; 197:106970. [PMID: 37884069 DOI: 10.1016/j.phrs.2023.106970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 06/19/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 10/28/2023]
Abstract
Vascular cognitive impairment (VCI) describes neurodegenerative disorders characterized by a vascular component. Pathologically, it involves decreased cerebral blood flow (CBF), white matter lesions, endothelial dysfunction, and blood-brain barrier (BBB) impairments. Molecularly, oxidative stress and inflammation are two of the major underlying mechanisms. Nitric oxide (NO) physiologically stimulates soluble guanylate cyclase (sGC) to induce cGMP production. However, under pathological conditions, NO seems to be at the basis of oxidative stress and inflammation, leading to a decrease in sGC activity and expression. The native form of sGC needs a ferrous heme group bound in order to be sensitive to NO (Fe(II)sGC). Oxidation of sGC leads to the conversion of ferrous to ferric heme (Fe(III)sGC) and even heme-loss (apo-sGC). Both Fe(III)sGC and apo-sGC are insensitive to NO, and the enzyme is therefore inactive. sGC activity can be enhanced either by targeting the NO-sensitive native sGC (Fe(II)sGC), or the inactive, oxidized sGC (Fe(III)sGC) and the heme-free apo-sGC. For this purpose, sGC stimulators acting on Fe(II)sGC and sGC activators acting on Fe(III)sGC/apo-sGC have been developed. These sGC agonists have shown their efficacy in cardiovascular diseases by restoring the physiological and protective functions of the NO-sGC-cGMP pathway, including the reduction of oxidative stress and inflammation, and improvement of vascular functioning. Yet, only very little research has been performed within the cerebrovascular system and VCI pathology when focusing on sGC modulation and its potential protective mechanisms on vascular and neural function. Therefore, within this review, the potential of sGC as a target for treating VCI is highlighted.
Collapse
Affiliation(s)
- Ellis Nelissen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands.
| | - Melissa Schepers
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands; Neuro-immune connect and repair lab, Biomedical Research Institute, Hasselt University, Hasselt 3500, Belgium
| | - Laura Ponsaerts
- Neuro-immune connect and repair lab, Biomedical Research Institute, Hasselt University, Hasselt 3500, Belgium; Department of Cardio & Organ Systems (COS), Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Sébastien Foulquier
- Department of Pharmacology and Toxicology, School for Mental Health and Neuroscience (MHeNS), School for Cardiovascular Diseases (CARIM), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| | - Annelies Bronckaers
- Department of Cardio & Organ Systems (COS), Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Tim Vanmierlo
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands; Neuro-immune connect and repair lab, Biomedical Research Institute, Hasselt University, Hasselt 3500, Belgium
| | - Peter Sandner
- Bayer AG, Pharmaceuticals R&D, Pharma Research Center, 42113 Wuppertal, Germany; Hannover Medical School, 30625 Hannover, Germany
| | - Jos Prickaerts
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, the Netherlands
| |
Collapse
|
6
|
Jehle A, Garaschuk O. The Interplay between cGMP and Calcium Signaling in Alzheimer's Disease. Int J Mol Sci 2022; 23:7048. [PMID: 35806059 PMCID: PMC9266933 DOI: 10.3390/ijms23137048] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/05/2022] [Revised: 05/31/2022] [Accepted: 06/22/2022] [Indexed: 02/04/2023] Open
Abstract
Cyclic guanosine monophosphate (cGMP) is a ubiquitous second messenger and a key molecule in many important signaling cascades in the body and brain, including phototransduction, olfaction, vasodilation, and functional hyperemia. Additionally, cGMP is involved in long-term potentiation (LTP), a cellular correlate of learning and memory, and recent studies have identified the cGMP-increasing drug Sildenafil as a potential risk modifier in Alzheimer's disease (AD). AD development is accompanied by a net increase in the expression of nitric oxide (NO) synthases but a decreased activity of soluble guanylate cyclases, so the exact sign and extent of AD-mediated imbalance remain unclear. Moreover, human patients and mouse models of the disease present with entangled deregulation of both cGMP and Ca2+ signaling, e.g., causing changes in cGMP-mediated Ca2+ release from the intracellular stores as well as Ca2+-mediated cGMP production. Still, the mechanisms governing such interplay are poorly understood. Here, we review the recent data on mechanisms underlying the brain cGMP signaling and its interconnection with Ca2+ signaling. We also discuss the recent evidence stressing the importance of such interplay for normal brain function as well as in Alzheimer's disease.
Collapse
Affiliation(s)
| | - Olga Garaschuk
- Department of Neurophysiology, Institute of Physiology, Eberhard Karls University of Tübingen, 72074 Tübingen, Germany;
| |
Collapse
|
7
|
Chen H, Siu SWI, Wong CTT, Qiu J, Cheung AKK, Lee SMY. Anti-epileptic Kunitz-like peptides discovered in the branching coral Acropora digitifera through transcriptomic analysis. Arch Toxicol 2022; 96:2589-2608. [PMID: 35604417 DOI: 10.1007/s00204-022-03311-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/24/2022] [Accepted: 04/27/2022] [Indexed: 11/29/2022]
Abstract
Approximately 50 million people are suffering from epilepsy worldwide. Corals have been used for treating epilepsy in traditional Chinese medicine, but the mechanism of this treatment is unknown. In this study, we analyzed the transcriptome of the branching coral Acropora digitifera and obtained its Kyoto Encyclopedia of Genes and Genomes (KEGG), EuKaryotic Orthologous Groups (KOG) and Gene Ontology (GO) annotation. Combined with multiple sequence alignment and phylogenetic analysis, we discovered three polypeptides, we named them AdKuz1, AdKuz2 and AdKuz3, from A. digitifera that showed a close relationship to Kunitz-type peptides. Molecular docking and molecular dynamics simulation indicated that AdKuz1 to 3 could interact with GABAA receptor but AdKuz2-GABAA remained more stable than others. The biological experiments showed that AdKuz1 and AdKuz2 exhibited an anti-inflammatory effect by decreasing the aberrant level of nitric oxide (NO), IL-6, TNF-α and IL-1β induced by LPS in BV-2 cells. In addition, the pentylenetetrazol (PTZ)-induced epileptic effect on zebrafish was remarkably suppressed by AdKuz1 and AdKuz2. AdKuz2 particularly showed superior anti-epileptic effects compared to the other two peptides. Furthermore, AdKuz2 significantly decreased the expression of c-fos and npas4a, which were up-regulated by PTZ treatment. In addition, AdKuz2 reduced the synthesis of glutamate and enhanced the biosynthesis of gamma-aminobutyric acid (GABA). In conclusion, the results indicated that AdKuz2 may affect the synthesis of glutamate and GABA and enhance the activity of the GABAA receptor to inhibit the symptoms of epilepsy. We believe, AdKuz2 could be a promising anti-epileptic agent and its mechanism of action should be further investigated.
Collapse
Affiliation(s)
- Hanbin Chen
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China.,Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, China
| | - Shirley Weng In Siu
- Institute of Science and Environment, University of Saint Joseph, Macao, China
| | - Clarence Tsun Ting Wong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, China
| | - Jianwen Qiu
- Department of Biology and Hong Kong Branch of the Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Hong Kong Baptist University, Hong Kong, China.,Southern Marine Science and Engineering Guangdong Laboratory (Guangzhou), Guangzhou, China
| | - Alex Kwok-Kuen Cheung
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Simon Ming Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China. .,Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao, China.
| |
Collapse
|
8
|
Xu H, Zhong Y, Yuan S, Wu Y, Ma Z, Hao Z, Ding H, Wu H, Liu G, Pang M, Liu N, Wang C, Zhang N. Nitric Oxide Synthase Type 1 Methylation Is Associated With White Matter Microstructure in the Corpus Callosum and Greater Panic Disorder Severity Among Panic Disorder Patients. Front Neurol 2021; 12:755270. [PMID: 34733233 PMCID: PMC8559336 DOI: 10.3389/fneur.2021.755270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/13/2021] [Accepted: 09/06/2021] [Indexed: 11/28/2022] Open
Abstract
Objectives: Methylation of the neuronal nitric oxide synthase (NOS1/nNOS) gene has recently been identified as a promising biomarker of psychiatric disorders. NOS1 plays an essential role in neurite outgrowth and may thus affect the microstructure development of white matter (WM) in the corpus callosum (CC), which is known to be altered in panic disorder (PD). We examined the relationship between NOS1 methylation, WM tracts in the CC, and symptoms based on this finding. Methods: Thirty-two patients with PD and 22 healthy controls (HCs) were recruited after age, gender, and the education level were matched. The cell type used was whole-blood DNA, and DNA methylation of NOS1 was measured at 20 CpG sites in the promoter region. Although 25 patients with PD were assessed with the Panic Disorder Severity Scale (PDSS), diffusion tensor imaging (DTI) scans were only collected from 16 participants with PD. Results: We observed that the PD group showed lower methylation than did the HCs group and positive correlations between the symptom severity of PD and methylation at CpG4 and CpG9. In addition, CpG9 methylation was significantly correlated with the fractional anisotropy (FA) and mean diffusivity (MD) values of the CC and its major components (the genu and the splenium) in the PD group. Furthermore, path analyses showed that CpG9 methylation offers a mediating effect for the association between the MD values of the genu of the CC and PD symptom severity (95% CI = −1.731 to −0.034). Conclusions: The results suggest that CpG9 methylation leads to atypical development of the genu of the CC, resulting in higher PD symptom severity, adding support for the methylation of NOS1 as a future prognostic indicator of PD.
Collapse
Affiliation(s)
- Huazhen Xu
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China.,The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Yuan Zhong
- School of Psychology, Nanjing Normal University, Nanjing, China.,Jiangsu Key Laboratory of Mental Health and Cognitive Science, Nanjing Normal University, Nanjing, China
| | - Shiting Yuan
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yun Wu
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Zijuan Ma
- School of Psychology, South China Normal University, Guangzhou, China
| | - Ziyu Hao
- School of Psychology, Nanjing Normal University, Nanjing, China
| | - Huachen Ding
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Huiqing Wu
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Gang Liu
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Manlong Pang
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Na Liu
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China.,Cognitive Behavioral Therapy Institute of Nanjing Medical University, Nanjing, China
| | - Chun Wang
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China.,School of Psychology, Nanjing Normal University, Nanjing, China.,Cognitive Behavioral Therapy Institute of Nanjing Medical University, Nanjing, China.,Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
| | - Ning Zhang
- Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing, China.,Cognitive Behavioral Therapy Institute of Nanjing Medical University, Nanjing, China.,Functional Brain Imaging Institute of Nanjing Medical University, Nanjing, China
| |
Collapse
|
9
|
Hsieh MH, Hsiao G, Chang CH, Yang YL, Ju YM, Kuo YH, Lee TH. Polyketides with Anti-neuroinflammatory Activity from Theissenia cinerea. JOURNAL OF NATURAL PRODUCTS 2021; 84:1898-1903. [PMID: 34185528 DOI: 10.1021/acs.jnatprod.0c01307] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 06/13/2023]
Abstract
Theissenia cinerea 89091602 is a previously reported plant-derived bioactive fungal strain, and the active principles separated from the extracts of its submerged culture were shown to exhibit potent anti-neuroinflammatory activities in both cellular study and animal testing. In a continuation of our previous investigation on the bioactive entities from this fungus, solid state fermentation was performed in an attempt to diversify the bioactive secondary metabolites. In the present study, five previously unreported polyketides, theissenophenol (1), theissenepoxide (2), theissenolactone D (3), theissenone (4), and theissenisochromanone (5), together with the known theissenolactone B (6), theissenolactone C (7), and arthrinone (8), were isolated and characterized through spectroscopic analysis and comparison with the literature data. The configurations of theissenepoxide (2) and theissenisochromanone (5) were further corroborated by single-crystal X-ray diffraction data analysis. Theissenone (4), theissenolactone B (6), theissenolactone C (7), and arthrinone (8) exhibited potent nitric oxide production inhibitory activities in murine brain microglial BV-2 cells with IC50 values of 5.0 ± 1.0, 4.5 ± 0.6, 1.1 ± 0.1, and 3.2 ± 0.3 μM, respectively, without any significant cytotoxic effects.
Collapse
Affiliation(s)
- Meng-Hsuan Hsieh
- Institute of Fisheries Science, National Taiwan University, Taipei 10617, Taiwan
| | - George Hsiao
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Pharmacology, School of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chia-Hao Chang
- Institute of Fisheries Science, National Taiwan University, Taipei 10617, Taiwan
| | - Yu-Liang Yang
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 11529, Taiwan
| | - Yu-Ming Ju
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei 11529, Taiwan
| | - Yueh-Hsiung Kuo
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung 40447, Taiwan
- Department of Biotechnology, Asia University, Taichung 41354, Taiwan
- Chinese Medical Research Center, China Medical University, Taichung 40447, Taiwan
| | - Tzong-Huei Lee
- Institute of Fisheries Science, National Taiwan University, Taipei 10617, Taiwan
| |
Collapse
|
10
|
Stefano GB, Esch T, Ptacek R, Kream RM. Dysregulation of Nitric Oxide Signaling in Microglia: Multiple Points of Functional Convergence in the Complex Pathophysiology of Alzheimer Disease. Med Sci Monit 2020; 26:e927739. [PMID: 32975239 PMCID: PMC7523423 DOI: 10.12659/msm.927739] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/18/2022] Open
Abstract
Current critical thinking has displaced the elaborated beta amyloid theory as the underlying unitary mechanism of Alzheimer disease (AD) in favor of concerted, long-term disruption or dysregulation of broad-based physiological processes. We present a critical discussion in which a chronic state of systemic proinflammation sustained over the course of several decades and engendered by ongoing metabolic or autoimmune disease is predicted to promote severe disruptions of central neurological processes. Specifically, long-term functional rundown of microglial-mediated phagocytic activity in concert with aberrant expression and cellular deposition of beta amyloid and tau protein facilitates formation of senile plaques and neurofibrillary tangles. Within this functional context, we hypothesize that early initiation events in the pathophysiology of AD may operationally involve a convergence of dysregulated peripheral and central constitutive nitric oxide signaling pathways resulting from a chronic state of systemic proinflammation and leading to severely dysfunctional “hyperactivated” microglia.
Collapse
Affiliation(s)
- George B Stefano
- Department of Psychiatry, First Faculty of Medicine Charles University in Prague and General University Hospital in Prague, Center for Cognitive and Molecular Neuroscience, Prague, Czech Republic
| | - Tobias Esch
- Faculty of Health, School of Medicine, University Clinic for Integrative Health Care, Institute for Integrative Health Care and Health Promotion, Witten/Herdecke University, Witten, Germany
| | - Radek Ptacek
- Department of Psychiatry, First Faculty of Medicine Charles University in Prague and General University Hospital in Prague, Center for Cognitive and Molecular Neuroscience, Prague, Czech Republic
| | - Richard M Kream
- Department of Psychiatry, First Faculty of Medicine Charles University in Prague and General University Hospital in Prague, Center for Cognitive and Molecular Neuroscience, Prague, Czech Republic
| |
Collapse
|
11
|
Zhang H, Yang X, Li X, Cheng Y, Zhang H, Chang L, Sun M, Zhang Z, Wang Z, Niu Q, Wang T. Oxidative and nitrosative stress in the neurotoxicity of polybrominated diphenyl ether-153: possible mechanism and potential targeted intervention. CHEMOSPHERE 2020; 238:124602. [PMID: 31545211 DOI: 10.1016/j.chemosphere.2019.124602] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 06/12/2019] [Revised: 07/30/2019] [Accepted: 08/15/2019] [Indexed: 06/10/2023]
Abstract
Polybrominated diphenyl ethers (PBDEs) have been known to exhibit neurotoxicity in rats; however, the underlying mechanism remains unknown and there is no available intervention. In this study, we aimed to investigate the role of oxidative and nitrosative stress in the neurotoxicity in the cerebral cortex and primary neurons in rats following the BDE-153 treatment. Compared to the untreated group, BDE-153 treatment significantly induced the neurotoxic effects in rats, as manifested by the increased lactate dehydrogenase (LDH) activities and cell apoptosis rates, and the decreased neurotrophic factor contents and cholinergic enzyme activities in rats' cerebral cortices and primary neurons. When compared to the untreated group, the oxidative and nitrosative stress had occurred in the cerebral cortex or primary neurons in rats following the BDE-153 treatment, as manifested by the increments in levels of reactive oxygenspecies (ROS), malondialdehyde (MDA), nitric oxide (NO), and neuronal nitric oxide synthase (nNOS) mRNA and protein expressions, along with the decline in levels of superoxide dismutase (SOD) activity, glutathione (GSH) content, and peroxiredoxin I (Prx I) and Prx II mRNA and protein expressions. In addition, the ROS scavenger N-acetyl-l-cysteine (NAC) or NO scavenger NG-Nitro-l-arginine (L-NNA) significantly rescued the LDH leakage and cell survival, reversed the neurotrophin contents and cholinergic enzymes, mainly via regaining balance between oxidation/nitrosation and antioxidation. Overall, our findings suggested that oxidative and nitrosative stresses are involved in the neurotoxicity induced by BDE-153, and that the antioxidation is a potential targeted intervention.
Collapse
Affiliation(s)
- Hongmei Zhang
- Department of Environmental Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Xiaorong Yang
- National Key Disciplines, Key Laboratory for Cellular Physiology of Ministry of Education, Department of Neurobiology, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Xin Li
- Center of Disease Control and Prevention, Taiyuan Iron and Steel Company, Taiyuan, 030003, Shanxi, China
| | - Yan Cheng
- Department of Nuclear Medicine, First Affiliated Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Huajun Zhang
- Department of Environmental Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Lijun Chang
- Department of Environmental Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Min Sun
- Department of Environmental Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Zhihong Zhang
- Department of Environmental Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Zemin Wang
- Department of Environmental Health, Indiana University School of Public Health, Bloomington, IN, 47408, USA
| | - Qiao Niu
- Department of Occupational Health, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| | - Tong Wang
- Department of Health Statistics, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
12
|
Maksoud MJE, Tellios V, An D, Xiang Y, Lu W. Nitric oxide upregulates microglia phagocytosis and increases transient receptor potential vanilloid type 2 channel expression on the plasma membrane. Glia 2019; 67:2294-2311. [DOI: 10.1002/glia.23685] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/06/2019] [Revised: 07/02/2019] [Accepted: 07/06/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Matthew J. E. Maksoud
- Graduate Program of Neuroscience The University of Western Ontario London Canada
- Department of Molecular Medicine Robarts Research Institute, The University of Western Ontario London Canada
| | - Vasiliki Tellios
- Graduate Program of Neuroscience The University of Western Ontario London Canada
- Department of Molecular Medicine Robarts Research Institute, The University of Western Ontario London Canada
| | - Dong An
- Department of Molecular Medicine Robarts Research Institute, The University of Western Ontario London Canada
| | - Yun‐Yan Xiang
- Department of Molecular Medicine Robarts Research Institute, The University of Western Ontario London Canada
| | - Wei‐Yang Lu
- Graduate Program of Neuroscience The University of Western Ontario London Canada
- Department of Molecular Medicine Robarts Research Institute, The University of Western Ontario London Canada
- Department of Physiology and Pharmacology The University of Western Ontario London Canada
| |
Collapse
|
13
|
Roy A, Banerjee S, Saqib U, Baig MS. NOS1-derived nitric oxide facilitates macrophage uptake of low-density lipoprotein. J Cell Biochem 2019; 120:11593-11603. [PMID: 30805961 DOI: 10.1002/jcb.28439] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/25/2018] [Revised: 11/29/2018] [Accepted: 12/06/2018] [Indexed: 01/24/2023]
Abstract
Foam cell formation is a hallmark event during atherosclerosis. The current paradigm is that lipid uptake by a scavenger receptor in macrophages initiates necrosis core formation that characterizes atherosclerosis. We report that NOS1-derived nitric oxide (NO) facilitates low-density lipoprotein (LDL) uptake by macrophages independent of the inflammatory response. LDL uptake could be dramatically suppressed by NOS1 specific inhibitor 1-(2-trifluoromethylphenyl) imidazole (TRIM). Importantly, the notion that NOS1 can mediate uptake of lipoproteins suggests that the foam cell formation is regulated by NOS1-derived NO-mediated mechanism. This is a novel study involving NOS1 as a critical player of foam cell formation and reveals much about the key molecular proteins involved in atherosclerosis. Targeting NOS1 would be a useful strategy in reducing LDL uptake by macrophages and hence dampening the atherosclerosis progression.
Collapse
Affiliation(s)
- Anjali Roy
- Discipline of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Sreeparna Banerjee
- Department of Biological Sciences, Orta Doğu Teknik Üniversitesi (ODTU/METU), Ankara, Turkey
| | - Uzma Saqib
- Discipline of Chemistry, School of Basic Sciences, Indian Institute of Technology Indore (IITI), Indore, India
| | - Mirza S Baig
- Discipline of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| |
Collapse
|
14
|
De Santa F, Vitiello L, Torcinaro A, Ferraro E. The Role of Metabolic Remodeling in Macrophage Polarization and Its Effect on Skeletal Muscle Regeneration. Antioxid Redox Signal 2019; 30:1553-1598. [PMID: 30070144 DOI: 10.1089/ars.2017.7420] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 12/18/2022]
Abstract
Significance: Macrophages are crucial for tissue homeostasis. Based on their activation, they might display classical/M1 or alternative/M2 phenotypes. M1 macrophages produce pro-inflammatory cytokines, reactive oxygen species (ROS), and nitric oxide (NO). M2 macrophages upregulate arginase-1 and reduce NO and ROS levels; they also release anti-inflammatory cytokines, growth factors, and polyamines, thus promoting angiogenesis and tissue healing. Moreover, M1 and M2 display key metabolic differences; M1 polarization is characterized by an enhancement in glycolysis and in the pentose phosphate pathway (PPP) along with a decreased oxidative phosphorylation (OxPhos), whereas M2 are characterized by an efficient OxPhos and reduced PPP. Recent Advances: The glutamine-related metabolism has been discovered as crucial for M2 polarization. Vice versa, flux discontinuities in the Krebs cycle are considered additional M1 features; they lead to increased levels of immunoresponsive gene 1 and itaconic acid, to isocitrate dehydrogenase 1-downregulation and to succinate, citrate, and isocitrate over-expression. Critical Issues: A macrophage classification problem, particularly in vivo, originating from a gap in the knowledge of the several intermediate polarization statuses between the M1 and M2 extremes, characterizes this field. Moreover, the detailed features of metabolic reprogramming crucial for macrophage polarization are largely unknown; in particular, the role of β-oxidation is highly controversial. Future Directions: Manipulating the metabolism to redirect macrophage polarization might be useful in various pathologies, including an efficient skeletal muscle regeneration. Unraveling the complexity pertaining to metabolic signatures that are specific for the different macrophage subsets is crucial for identifying new compounds that are able to trigger macrophage polarization and that might be used for therapeutical purposes.
Collapse
Affiliation(s)
- Francesca De Santa
- Institute of Cell Biology and Neurobiology (IBCN), National Research Council (CNR), Rome, Italy
| | - Laura Vitiello
- Laboratory of Pathophysiology of Cachexia and Metabolism of Skeletal Muscle, IRCCS San Raffaele Pisana, Rome, Italy
| | - Alessio Torcinaro
- Institute of Cell Biology and Neurobiology (IBCN), National Research Council (CNR), Rome, Italy.,Department of Biology and Biotechnology "Charles Darwin," Sapienza University, Rome, Italy
| | - Elisabetta Ferraro
- Laboratory of Pathophysiology of Cachexia and Metabolism of Skeletal Muscle, IRCCS San Raffaele Pisana, Rome, Italy
| |
Collapse
|
15
|
Antineuroinflammatory and Neuroprotective Effects of Gyejibokryeong-Hwan in Lipopolysaccharide-Stimulated BV2 Microglia. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:7585896. [PMID: 31057653 PMCID: PMC6463633 DOI: 10.1155/2019/7585896] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 07/20/2018] [Revised: 01/23/2019] [Accepted: 02/10/2019] [Indexed: 11/29/2022]
Abstract
Microglia, the central nervous system's innate immune cells, mediate neuroinflammation and are implicated in a variety of neuropathologies. The present study investigated the antineuroinflammatory and neuroprotective effects of Gyejibokryeong-hwan (GBH), a traditional Korean medicine, in lipopolysaccharide- (LPS-) stimulated murine BV2 microglia. BV2 cells were pretreated with GBH, fluoxetine (FXT), or amitriptyline (AMT) for 1 h and then stimulated with LPS (100 ng/mL). The expression levels of nitric oxide (NO), cytokines, and chemokines were determined by the Griess method, ELISA, or real-time PCR. Western blotting was used to measure various transcription factors and mitogen activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K)/Akt activity. GBH significantly reduced the levels of NO, inducible nitric oxide synthase (iNOS), cyclooxygenase- (COX-) 2, tumor necrosis factor- (TNF-) α, interleukin- (IL-) 1β, IL-6, macrophage inhibitory protein- (MIP-) 1α, macrophage chemoattractant protein- (MCP-) 1, and IFN-γ inducible protein- (IP-) 10, regulated upon activation normal T cell expressed sequence (RANTES) in a dose-dependent manner. Expression of nuclear factor- (NF-) κB p65 was significantly decreased and phosphorylation of extracellular signal-regulated kinase (Erk), c-Jun NH2-terminal kinase (JNK), and PI3K/Akt by GBH, but not p38 MAPK, was decreased. Furthermore, production of anti-inflammatory cytokine IL-10 was increased and Heme oxygenase-1 (HO-1) was upregulated via the nuclear factor-E2-related factor 2 (NRF2)/cAMP response element-binding protein (CREB) pathway, collectively indicating the neuroprotective effects of GBH. We concluded that GBH may suppress neuroinflammatory responses by inhibiting NF-κB activation and upregulating the neuroprotective factor, HO-1. These results suggest that GBH has potential as anti-inflammatory and neuroprotective agents against microglia-mediated neuroinflammatory disorders.
Collapse
|
16
|
Lan M, Tang X, Zhang J, Yao Z. Insights in pathogenesis of multiple sclerosis: nitric oxide may induce mitochondrial dysfunction of oligodendrocytes. Rev Neurosci 2018; 29:39-53. [PMID: 28822986 DOI: 10.1515/revneuro-2017-0033] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/15/2017] [Accepted: 06/15/2017] [Indexed: 01/01/2023]
Abstract
Demyelinating diseases, such as multiple sclerosis (MS), are kinds of common diseases in the central nervous system (CNS), and originated from myelin loss and axonal damage. Oligodendrocyte dysfunction is the direct reason of demyelinating lesions in the CNS. Nitric oxide (NO) plays an important role in the pathological process of demyelinating diseases. Although the neurotoxicity of NO is more likely mediated by peroxynitrite rather than NO itself, NO can impair oligodendrocyte energy metabolism through mediating the damaging of mitochondrial DNA, mitochondrial membrane and mitochondrial respiratory chain complexes. In the progression of MS, NO can mainly mediate demyelination, axonal degeneration and cell death. Hence, in this review, we extensively discuss endangerments of NO in oligodendrocytes (OLs), which is suggested to be the main mediator in demyelinating diseases, e.g. MS. We hypothesize that NO takes part in MS through impairing the function of monocarboxylate transporter 1, especially causing axonal degeneration. Then, it further provides a new insight that NO for OLs may be a reliable therapeutic target to ameliorate the course of demyelinating diseases.
Collapse
Affiliation(s)
- Minghong Lan
- Department of Physiology, Third Military Medical University, Chongqing 400038, China
| | - Xiaoyi Tang
- Department of Physiology, Third Military Medical University, Chongqing 400038, China
| | - Jie Zhang
- Department of Physiology, Third Military Medical University, Chongqing 400038, China
| | - Zhongxiang Yao
- Department of Physiology, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|
17
|
Rodrigues-Neves AC, Aires ID, Vindeirinho J, Boia R, Madeira MH, Gonçalves FQ, Cunha RA, Santos PF, Ambrósio AF, Santiago AR. Elevated Pressure Changes the Purinergic System of Microglial Cells. Front Pharmacol 2018; 9:16. [PMID: 29416510 PMCID: PMC5787565 DOI: 10.3389/fphar.2018.00016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/17/2017] [Accepted: 01/05/2018] [Indexed: 12/20/2022] Open
Abstract
Glaucoma is the second cause of blindness worldwide and is characterized by the degeneration of retinal ganglion cells (RGCs) and optic nerve atrophy. Increased microglia reactivity is an early event in glaucoma that may precede the loss of RGCs, suggesting that microglia and neuroinflammation are involved in the pathophysiology of this disease. Although global changes of the purinergic system have been reported in experimental and human glaucoma, it is not known if this is due to alterations of the purinergic system of microglial cells, the resident immune cells of the central nervous system. We now studied if elevated hydrostatic pressure (EHP), mimicking ocular hypertension, changed the extracellular levels of ATP and adenosine and the expression, density and activity of enzymes, transporters and receptors defining the purinergic system. The exposure of the murine microglial BV-2 cell line to EHP increased the extracellular levels of ATP and adenosine, increased the density of ecto-nucleoside triphosphate diphosphohydrolase 1 (E-NTPDase1, CD39) and decreased the density of the equilibrative nucleotide transporter 2 as well as the activity of adenosine deaminase. The expression of adenosine A1 receptor also decreased, but the adenosine A3 receptor was not affected. Notably, ATP and adenosine selectively control migration rather than phagocytosis, both bolstered by EHP. The results show that the purinergic system is altered in microglia in conditions of elevated pressure. Understanding the impact of elevated pressure on the purinergic system will help to unravel the mechanisms underlying inflammation and neurodegeneration associated with glaucoma.
Collapse
Affiliation(s)
- Ana C Rodrigues-Neves
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Inês D Aires
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Joana Vindeirinho
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Raquel Boia
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Maria H Madeira
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Francisco Q Gonçalves
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Rodrigo A Cunha
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal.,Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Paulo F Santos
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal.,Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal
| | - António F Ambrósio
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Association for Innovation and Biomedical Research on Light and Image, Coimbra, Portugal
| | - Ana R Santiago
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Association for Innovation and Biomedical Research on Light and Image, Coimbra, Portugal
| |
Collapse
|
18
|
Yin Y, Qiu S, Li X, Huang B, Xu Y, Peng Y. EZH2 suppression in glioblastoma shifts microglia toward M1 phenotype in tumor microenvironment. J Neuroinflammation 2017; 14:220. [PMID: 29132376 PMCID: PMC5684749 DOI: 10.1186/s12974-017-0993-4] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/06/2017] [Accepted: 11/02/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) induces tumor immunosuppression through interacting with tumor-infiltrating microglia or macrophages (TAMs) with an unclear pathogenesis. Enhancer of zeste homolog 2 (EZH2) is abundant in GBM samples and cell lines and is involved in GBM proliferation, cell cycle, and invasion, whereas its association with innate immune response is not yet reported. Herein, the aim of this study was to investigate the role of EZH2 in GBM immune. METHODS Co-culturing models of human/murine GBM cells with PBMC-derived macrophages/primary microglia were employed. EZH2 mRNAs and function were suppressed by siEZH2 and DZNep. Real-time PCR and flow cytometry were used to determine levels of microglia/macrophages markers. The fluorescence-labeled latex beads and flow cytometry were utilized to evaluate phagocytic abilities of microglia. CCK8 assay was performed to assess microglia proliferation. RESULTS EZH2 inhibition led to significant reduction of TGFβ1-3 and IL10 and elevation of IL1β and IL6 in human and murine GBM cells. More importantly, EZH2 suppression in GBM cells resulted in significant increase of M1 markers (TNFα and iNOS) and decrease of a pool of M2 markers in murine microglia. The proportion of CD206+ cells was decreased in PBMC-derived macrophages as co-incubated with EZH2-inhibited GBM cells. Functional researches showed that phagocytic capacities of microglia were significantly ameliorated after EZH2 inhibition in co-culturing GBM cells and microglia proliferation was declined after addition of TGFβ2 antibodies to co-incubated GBM cells with EZH2 inhibition. Besides, we found that EZH2 suppression in GBM cells enhanced co-culturing microglia engulfment through activation of iNOS. CONCLUSIONS Our data demonstrates that EZH2 participates in GBM-induced immune deficient and EZH2 suppression in GBM can remodel microglia immune functions, which is beneficial for understanding GBM pathogenesis and suggests potential targets for therapeutic approaches.
Collapse
Affiliation(s)
- Yatao Yin
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Yangjiang Xi Road 107, Guangzhou, China.,Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuwei Qiu
- Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan Road 321, Nanjing, China.
| | - Xiangpen Li
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Yangjiang Xi Road 107, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bo Huang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Yangjiang Xi Road 107, Guangzhou, China
| | - Yun Xu
- Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Zhongshan Road 321, Nanjing, China
| | - Ying Peng
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Yangjiang Xi Road 107, Guangzhou, China. .,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
19
|
Möhrle D, Reimann K, Wolter S, Wolters M, Varakina K, Mergia E, Eichert N, Geisler HS, Sandner P, Ruth P, Friebe A, Feil R, Zimmermann U, Koesling D, Knipper M, Rüttiger L. NO-Sensitive Guanylate Cyclase Isoforms NO-GC1 and NO-GC2 Contribute to Noise-Induced Inner Hair Cell Synaptopathy. Mol Pharmacol 2017; 92:375-388. [PMID: 28874607 DOI: 10.1124/mol.117.108548] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/07/2017] [Accepted: 07/18/2017] [Indexed: 02/14/2025] Open
Abstract
Nitric oxide (NO) activates the NO-sensitive soluble guanylate cyclase (NO-GC, sGC) and triggers intracellular signaling pathways involving cGMP. For survival of cochlear hair cells and preservation of hearing, NO-mediated cascades have both protective and detrimental potential. Here we examine the cochlear function of mice lacking one of the two NO-sensitive guanylate cyclase isoforms [NO-GC1 knockout (KO) or NO-GC2 KO]. The deletion of NO-GC1 or NO-GC2 did not influence electromechanical outer hair cell (OHC) properties, as measured by distortion product otoacoustic emissions, neither before nor after noise exposure, nor were click- or noise-burst-evoked auditory brainstem response thresholds different from controls. Yet inner hair cell (IHC) ribbons and auditory nerve responses showed significantly less deterioration in NO-GC1 KO and NO-GC2 KO mice after noise exposure. Consistent with a selective role of NO-GC in IHCs, NO-GC β1 mRNA was found in isolated IHCs but not in OHCs. Using transgenic mice expressing the fluorescence resonance energy transfer-based cGMP biosensor cGi500, NO-induced elevation of cGMP was detected in real-time in IHCs but not in OHCs. Pharmacologic long-term treatment with a NO-GC stimulator altered auditory nerve responses but did not affect OHC function and hearing thresholds. Interestingly, NO-GC stimulation exacerbated the loss of auditory nerve response in aged animals but attenuated the loss in younger animals. We propose NO-GC2 and, to some degree, NO-GC1 as targets for early pharmacologic prevention of auditory fiber loss (synaptopathy). Both isoforms provide selective benefits for hearing function by maintaining the functional integrity of auditory nerve fibers in early life rather than at old age.
Collapse
MESH Headings
- Animals
- Female
- Guanylate Cyclase/metabolism
- Hair Cells, Auditory, Inner/drug effects
- Hair Cells, Auditory, Inner/metabolism
- Hair Cells, Auditory, Inner/pathology
- Isoenzymes/metabolism
- Male
- Mice
- Mice, Knockout
- Mice, Transgenic
- Morpholines/pharmacology
- Nitric Oxide/metabolism
- Noise/adverse effects
- Pyrimidines/pharmacology
- Rats
- Rats, Wistar
- Receptors, Cell Surface/agonists
- Receptors, Cell Surface/metabolism
- Synapses/drug effects
- Synapses/metabolism
- Synapses/pathology
Collapse
Affiliation(s)
- Dorit Möhrle
- Department of Otolaryngology, Head and Neck Surgery, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Tübingen (D.M., K.R., S.W., K.V., N.E., H.-S.G., U.Z., M.K., L.R.), Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen (M.W., R.F.), Department of Pharmacology and Toxicology, University of Bochum, Bochum (E.M., D.K.), Bayer AG, Drug Discovery Pharma Research Centre Wuppertal, Wuppertal (P.S.), Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen (P.R.), and Department of Physiology, University of Würzburg, Würzburg (A.F.), Germany
| | - Katrin Reimann
- Department of Otolaryngology, Head and Neck Surgery, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Tübingen (D.M., K.R., S.W., K.V., N.E., H.-S.G., U.Z., M.K., L.R.), Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen (M.W., R.F.), Department of Pharmacology and Toxicology, University of Bochum, Bochum (E.M., D.K.), Bayer AG, Drug Discovery Pharma Research Centre Wuppertal, Wuppertal (P.S.), Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen (P.R.), and Department of Physiology, University of Würzburg, Würzburg (A.F.), Germany
| | - Steffen Wolter
- Department of Otolaryngology, Head and Neck Surgery, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Tübingen (D.M., K.R., S.W., K.V., N.E., H.-S.G., U.Z., M.K., L.R.), Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen (M.W., R.F.), Department of Pharmacology and Toxicology, University of Bochum, Bochum (E.M., D.K.), Bayer AG, Drug Discovery Pharma Research Centre Wuppertal, Wuppertal (P.S.), Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen (P.R.), and Department of Physiology, University of Würzburg, Würzburg (A.F.), Germany
| | - Markus Wolters
- Department of Otolaryngology, Head and Neck Surgery, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Tübingen (D.M., K.R., S.W., K.V., N.E., H.-S.G., U.Z., M.K., L.R.), Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen (M.W., R.F.), Department of Pharmacology and Toxicology, University of Bochum, Bochum (E.M., D.K.), Bayer AG, Drug Discovery Pharma Research Centre Wuppertal, Wuppertal (P.S.), Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen (P.R.), and Department of Physiology, University of Würzburg, Würzburg (A.F.), Germany
| | - Ksenya Varakina
- Department of Otolaryngology, Head and Neck Surgery, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Tübingen (D.M., K.R., S.W., K.V., N.E., H.-S.G., U.Z., M.K., L.R.), Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen (M.W., R.F.), Department of Pharmacology and Toxicology, University of Bochum, Bochum (E.M., D.K.), Bayer AG, Drug Discovery Pharma Research Centre Wuppertal, Wuppertal (P.S.), Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen (P.R.), and Department of Physiology, University of Würzburg, Würzburg (A.F.), Germany
| | - Evanthia Mergia
- Department of Otolaryngology, Head and Neck Surgery, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Tübingen (D.M., K.R., S.W., K.V., N.E., H.-S.G., U.Z., M.K., L.R.), Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen (M.W., R.F.), Department of Pharmacology and Toxicology, University of Bochum, Bochum (E.M., D.K.), Bayer AG, Drug Discovery Pharma Research Centre Wuppertal, Wuppertal (P.S.), Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen (P.R.), and Department of Physiology, University of Würzburg, Würzburg (A.F.), Germany
| | - Nicole Eichert
- Department of Otolaryngology, Head and Neck Surgery, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Tübingen (D.M., K.R., S.W., K.V., N.E., H.-S.G., U.Z., M.K., L.R.), Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen (M.W., R.F.), Department of Pharmacology and Toxicology, University of Bochum, Bochum (E.M., D.K.), Bayer AG, Drug Discovery Pharma Research Centre Wuppertal, Wuppertal (P.S.), Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen (P.R.), and Department of Physiology, University of Würzburg, Würzburg (A.F.), Germany
| | - Hyun-Soon Geisler
- Department of Otolaryngology, Head and Neck Surgery, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Tübingen (D.M., K.R., S.W., K.V., N.E., H.-S.G., U.Z., M.K., L.R.), Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen (M.W., R.F.), Department of Pharmacology and Toxicology, University of Bochum, Bochum (E.M., D.K.), Bayer AG, Drug Discovery Pharma Research Centre Wuppertal, Wuppertal (P.S.), Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen (P.R.), and Department of Physiology, University of Würzburg, Würzburg (A.F.), Germany
| | - Peter Sandner
- Department of Otolaryngology, Head and Neck Surgery, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Tübingen (D.M., K.R., S.W., K.V., N.E., H.-S.G., U.Z., M.K., L.R.), Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen (M.W., R.F.), Department of Pharmacology and Toxicology, University of Bochum, Bochum (E.M., D.K.), Bayer AG, Drug Discovery Pharma Research Centre Wuppertal, Wuppertal (P.S.), Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen (P.R.), and Department of Physiology, University of Würzburg, Würzburg (A.F.), Germany
| | - Peter Ruth
- Department of Otolaryngology, Head and Neck Surgery, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Tübingen (D.M., K.R., S.W., K.V., N.E., H.-S.G., U.Z., M.K., L.R.), Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen (M.W., R.F.), Department of Pharmacology and Toxicology, University of Bochum, Bochum (E.M., D.K.), Bayer AG, Drug Discovery Pharma Research Centre Wuppertal, Wuppertal (P.S.), Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen (P.R.), and Department of Physiology, University of Würzburg, Würzburg (A.F.), Germany
| | - Andreas Friebe
- Department of Otolaryngology, Head and Neck Surgery, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Tübingen (D.M., K.R., S.W., K.V., N.E., H.-S.G., U.Z., M.K., L.R.), Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen (M.W., R.F.), Department of Pharmacology and Toxicology, University of Bochum, Bochum (E.M., D.K.), Bayer AG, Drug Discovery Pharma Research Centre Wuppertal, Wuppertal (P.S.), Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen (P.R.), and Department of Physiology, University of Würzburg, Würzburg (A.F.), Germany
| | - Robert Feil
- Department of Otolaryngology, Head and Neck Surgery, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Tübingen (D.M., K.R., S.W., K.V., N.E., H.-S.G., U.Z., M.K., L.R.), Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen (M.W., R.F.), Department of Pharmacology and Toxicology, University of Bochum, Bochum (E.M., D.K.), Bayer AG, Drug Discovery Pharma Research Centre Wuppertal, Wuppertal (P.S.), Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen (P.R.), and Department of Physiology, University of Würzburg, Würzburg (A.F.), Germany
| | - Ulrike Zimmermann
- Department of Otolaryngology, Head and Neck Surgery, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Tübingen (D.M., K.R., S.W., K.V., N.E., H.-S.G., U.Z., M.K., L.R.), Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen (M.W., R.F.), Department of Pharmacology and Toxicology, University of Bochum, Bochum (E.M., D.K.), Bayer AG, Drug Discovery Pharma Research Centre Wuppertal, Wuppertal (P.S.), Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen (P.R.), and Department of Physiology, University of Würzburg, Würzburg (A.F.), Germany
| | - Doris Koesling
- Department of Otolaryngology, Head and Neck Surgery, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Tübingen (D.M., K.R., S.W., K.V., N.E., H.-S.G., U.Z., M.K., L.R.), Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen (M.W., R.F.), Department of Pharmacology and Toxicology, University of Bochum, Bochum (E.M., D.K.), Bayer AG, Drug Discovery Pharma Research Centre Wuppertal, Wuppertal (P.S.), Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen (P.R.), and Department of Physiology, University of Würzburg, Würzburg (A.F.), Germany
| | - Marlies Knipper
- Department of Otolaryngology, Head and Neck Surgery, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Tübingen (D.M., K.R., S.W., K.V., N.E., H.-S.G., U.Z., M.K., L.R.), Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen (M.W., R.F.), Department of Pharmacology and Toxicology, University of Bochum, Bochum (E.M., D.K.), Bayer AG, Drug Discovery Pharma Research Centre Wuppertal, Wuppertal (P.S.), Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen (P.R.), and Department of Physiology, University of Würzburg, Würzburg (A.F.), Germany
| | - Lukas Rüttiger
- Department of Otolaryngology, Head and Neck Surgery, Hearing Research Centre Tübingen, Molecular Physiology of Hearing, University of Tübingen, Tübingen (D.M., K.R., S.W., K.V., N.E., H.-S.G., U.Z., M.K., L.R.), Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen (M.W., R.F.), Department of Pharmacology and Toxicology, University of Bochum, Bochum (E.M., D.K.), Bayer AG, Drug Discovery Pharma Research Centre Wuppertal, Wuppertal (P.S.), Department of Pharmacology, Toxicology and Clinical Pharmacy, Institute of Pharmacy, University of Tübingen, Tübingen (P.R.), and Department of Physiology, University of Würzburg, Würzburg (A.F.), Germany
| |
Collapse
|
20
|
Effect of nitric oxide to axonal degeneration in multiple sclerosis via downregulating monocarboxylate transporter 1 in oligodendrocytes. Nitric Oxide 2017; 67:75-80. [PMID: 28392448 DOI: 10.1016/j.niox.2017.04.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/06/2017] [Revised: 04/05/2017] [Accepted: 04/05/2017] [Indexed: 12/21/2022]
Abstract
Multiple sclerosis (MS) is a neurodegenerative disease of the central nervous system (CNS). Axonal degeneration, one of the main pathological characteristics of MS, is affected by nitric oxide (NO). In turn, NO induces mitochondrial dysfunction of neurons and glial cells. Inadequate glucose causes monocarboxylate transporter 1 (MCT1) to transfer lactate from oligodendrocytes (OLs) to neurons, which decreases MCT1 and results in energy substrate deficit (mainly lactate) in axons. The condition gradually leads to axonal degeneration. This study proposes that NO-induced MCT1 down-regulation in OLs may be involved in the pathological process of axonal degeneration, which eventually leads to MS.
Collapse
|
21
|
Scheiblich H, Bicker G. Regulation of Microglial Phagocytosis by RhoA/ROCK-Inhibiting Drugs. Cell Mol Neurobiol 2017; 37:461-473. [PMID: 27178562 DOI: 10.1007/s10571-016-0379-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/01/2016] [Accepted: 04/25/2016] [Indexed: 01/08/2023]
Abstract
Inflammation within the central nervous system (CNS) is a major component of many neurodegenerative diseases. The underlying mechanisms of neuronal loss are not fully understood, but the activation of CNS resident phagocytic microglia seems to be a significant element contributing to neurodegeneration. At the onset of inflammation, high levels of microglial phagocytosis may serve as an essential prerequisite for creating a favorable environment for neuronal regeneration. However, the excessive and long-lasting activation of microglia and the augmented engulfment of neurons have been suggested to eventually govern widespread neurodegeneration. Here, we investigated in a functional assay of acute inflammation how the small GTPase RhoA and its main target the Rho kinase (ROCK) influence microglial phagocytosis of neuronal debris. Using BV-2 microglia and human NT2 model neurons, we demonstrate that the pain reliever Ibuprofen decreases RhoA activation and microglial phagocytosis of neuronal cell fragments. Inhibition of the downstream effector ROCK with the small-molecule agents Y-27632 and Fasudil reduces the engulfment of neuronal debris and attenuates the production of the inflammatory mediator nitric oxide during stimulation with lipopolysaccharide. Our results support a therapeutic potential for RhoA/ROCK-inhibiting agents as an effective treatment of excessive inflammation and the resulting progression of microglia-mediated neurodegeneration in the CNS.
Collapse
Affiliation(s)
- Hannah Scheiblich
- Division of Cell Biology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15/102, 30173, Hannover, Germany
| | - Gerd Bicker
- Division of Cell Biology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15/102, 30173, Hannover, Germany.
- Center for Systems Neuroscience, Hannover, Germany.
| |
Collapse
|