1
|
Shao L, Wang C, Xu G, Tu Z, Yu X, Weng C, Liu J, Jian Z. Utilizing reactive oxygen species-scavenging nanoparticles for targeting oxidative stress in the treatment of ischemic stroke: A review. Open Med (Wars) 2024; 19:20241041. [PMID: 39588390 PMCID: PMC11587925 DOI: 10.1515/med-2024-1041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/06/2024] [Accepted: 08/23/2024] [Indexed: 11/27/2024] Open
Abstract
Ischemic stroke, which accounts for the majority of stroke cases, triggers a complex series of pathophysiological events, prominently characterized by acute oxidative stress due to excessive production of reactive oxygen species (ROS). Oxidative stress plays a crucial role in driving cell death and inflammation in ischemic stroke, making it a significant target for therapeutic intervention. Nanomedicine presents an innovative approach to directly mitigate oxidative damage. This review consolidates existing knowledge on the role of oxidative stress in ischemic stroke and assesses the potential of various ROS-scavenging nanoparticles (NPs) as therapeutic agents. We explore the properties and mechanisms of metal, metal-oxide, and carbon-based NPs, emphasizing their catalytic activity and biocompatibility in scavenging free radicals and facilitating the delivery of therapeutic agents across the blood-brain barrier. Additionally, we address the challenges such as cytotoxicity, immunogenicity, and biodistribution that need to be overcome to translate these nanotechnologies from bench to bedside. The future of NP-based therapies for ischemic stroke holds promise, with the potential to enhance outcomes through targeted modulation of oxidative stress.
Collapse
Affiliation(s)
- Lingmin Shao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Can Wang
- Department of Neurosurgery, Ezhou Central Hospital, Ezhou, 436000, Hubei, China
| | - Gang Xu
- Department of Neurosurgery, Xiantao First People’s Hospital, Xiantao, 433000, Hubei, China
| | - Zewei Tu
- Department of Neurosurgery, Yale School of Medicine, New Haven, 06510, CT, United States of America
| | - Xinyuan Yu
- Department of Anesthesiology, Duke University Medical Center, Durham, 27710, NC, United States of America
| | - Chao Weng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Jia Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| |
Collapse
|
2
|
Zilberter Y, Tabuena DR, Zilberter M. NOX-induced oxidative stress is a primary trigger of major neurodegenerative disorders. Prog Neurobiol 2023; 231:102539. [PMID: 37838279 DOI: 10.1016/j.pneurobio.2023.102539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 10/10/2023] [Indexed: 10/16/2023]
Abstract
Neurodegenerative diseases (NDDs) causing cognitive impairment and dementia are difficult to treat due to the lack of understanding of primary initiating factors. Meanwhile, major sporadic NDDs share many risk factors and exhibit similar pathologies in their early stages, indicating the existence of common initiation pathways. Glucose hypometabolism associated with oxidative stress is one such primary, early and shared pathology, and a likely major cause of detrimental disease-associated cascades; targeting this common pathology may therefore be an effective preventative strategy for most sporadic NDDs. However, its exact cause and trigger remain unclear. Recent research suggests that early oxidative stress caused by NADPH oxidase (NOX) activation is a shared initiating mechanism among major sporadic NDDs and could prove to be the long-sought ubiquitous NDD trigger. We focus on two major NDDs - Alzheimer's disease (AD) and Parkinson's disease (PD), as well as on acquired epilepsy which is an increasingly recognized comorbidity in NDDs. We also discuss available data suggesting the relevance of the proposed mechanisms to other NDDs. We delve into the commonalities among these NDDs in neuroinflammation and NOX involvement to identify potential therapeutic targets and gain a deeper understanding of the underlying causes of NDDs.
Collapse
Affiliation(s)
- Yuri Zilberter
- Aix-Marseille Université, INSERM UMR1106, Institut de Neurosciences des Systèmes, Marseille, France
| | - Dennis R Tabuena
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Misha Zilberter
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA.
| |
Collapse
|
3
|
Martynov MY, Zhuravleva MV, Vasyukova NS, Kuznetsova EV, Kameneva TR. [Oxidative stress in the pathogenesis of stroke and its correction]. Zh Nevrol Psikhiatr Im S S Korsakova 2023; 123:16-27. [PMID: 36719115 DOI: 10.17116/jnevro202312301116] [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: 02/01/2023]
Abstract
We reviewed the role of oxidative stress (OS) in the pathogenesis of ischemic (IS) and hemorrhagic stroke (HS). OS plays a major role in programmed cell death, increased permeability of the blood-brain barrier, astroglial and microglial activation, and local inflammatory response. We also reviewed the current state of neuro- and cytoprotection studies and their translation in clinical practice. With respect to experimental and clinical data the efficacy of long term administration of multimodal cytoprotective drug with antioxidant effect - ethylmethylhydroxypyridine succinate (Mexidol) is discussed during the acute and early recovery period after stroke.
Collapse
Affiliation(s)
- M Yu Martynov
- Pirogov Russian National Research Medical University, Moscow, Russia.,Federal Center of Brain Research and Neurotechnologies, Moscow, Russia
| | - M V Zhuravleva
- Research Center for Examination of Medical Devices, Moscow, Russia.,Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - N S Vasyukova
- Skriabin and Kovalenko Institute of Experimental Veterinary Medicine, Moscow, Russia
| | - E V Kuznetsova
- Research Institute for Healthcare and Medical Management, Moscow, Russia
| | - T R Kameneva
- Konchalovsky City Clinical Hospital, Moscow, Russia
| |
Collapse
|
4
|
Insight into Glyproline Peptides' Activity through the Modulation of the Inflammatory and Neurosignaling Genetic Response Following Cerebral Ischemia-Reperfusion. Genes (Basel) 2022; 13:genes13122380. [PMID: 36553646 PMCID: PMC9777888 DOI: 10.3390/genes13122380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/10/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
Glyprolines are Gly-Pro (GP)- or Pro-Gly (PG)-containing biogenic peptides. These peptides can act as neutrophil chemoattractants, or atheroprotective, anticoagulant, and neuroprotective agents. The Pro-Gly-Pro (PGP) tripeptide is an active factor of resistance to the biodegradation of peptide drugs. The synthetic Semax peptide, which includes Met-Glu-His-Phe (MEHF) fragments of adrenocorticotropic hormone and the C-terminal tripeptide PGP, serves as a neuroprotective drug for the treatment of ischemic stroke. Previously, we revealed that Semax mostly prevented the disruption of the gene expression pattern 24 h after a transient middle cerebral artery occlusion (tMCAO) in a rat brain model. The genes of this pattern were grouped into an inflammatory cluster (IC) and a neurotransmitter cluster (NC). Here, using real-time RT-PCR, the effect of other PGP-containing peptides, PGP and Pro-Gly-Pro-Leu (PGPL), on the expression of a number of genes in the IC and NC was studied 24 h after tMCAO. Both the PGP and PGPL peptides showed Semax-unlike effects, predominantly without changing gene expression 24 h after tMCAO. Moreover, there were IC genes (iL1b, iL6, and Socs3) for PGP, as well as IC (iL6, Ccl3, Socs3, and Fos) and NC genes (Cplx2, Neurod6, and Ptk2b) for PGPL, that significantly changed in expression levels after peptide administration compared to Semax treatment under tMCAO conditions. Furthermore, gene enrichment analysis was carried out, and a regulatory gene network was constructed. Thus, the spectra of the common and unique effects of the PGP, PGPL, and Semax peptides under ischemia-reperfusion were distinguished.
Collapse
|
5
|
An Q, Su S, Chai L, Wang Y, Wang X, Li X, Liang T, Hu W, Song X, Li C. Imaging of peroxynitrite in mitochondria by a near-infrared fluorescent probe with a large Stokes shift. Talanta 2022. [DOI: 10.1016/j.talanta.2022.124073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
6
|
Su S, Chai L, An Q, Hu W, Wang L, Li X, Zhang H, Li C. Tracking Autophagy Process with a TBET and AIE-Based Ratiometric Two-Photon Viscosity Probe. Anal Chem 2022; 94:15146-15154. [PMID: 36260837 DOI: 10.1021/acs.analchem.2c03555] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Autophagy is a cellular self-degrading process that plays a key role in cellular health and functioning. Since autophagy disorder is related to many diseases, it is highly important to detect autophagy. This study aimed to establish a dual-sensing mechanism-based ratiometric viscosity-sensitive lysosome-targeted two-photon fluorescent probe Vis-sun to track the autophagy process (the increase in lysosome viscosity during autophagy) by combining through bond energy transfer (TBET) and aggregation-induced emission (AIE). The introduction of TBET not only overcame the interference of background signals but also achieved the baseline separation of two emission peaks, thus reducing the crosstalk between emissions, as well as the noninvasive bio-sensing of biological targets and long-term real-time tracer imaging by introducing AIE. In vitro experiments showed that the fluorescence intensity at 485 nm decreased gradually on increasing the volume ratio of water to tetrahydrofuran (Vwater/VTHF), while the fluorescence intensity at 605 nm increased significantly. Also, the fluorescence signal was maximized when the water content reached 100%. At the same time, the probe exhibited a significant dependence on the ambient viscosity. Therefore, the dynamic monitoring of lysosome viscosity during autophagy and the in situ imaging of autophagy fluctuations during stroke-induced neuroinflammation were successfully achieved by implementing Vis-sun lysosome anchoring with morpholine.
Collapse
Affiliation(s)
- Shengze Su
- Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, College of Chemistry and Material Science, South-central University for Nationalities, Wuhan 430074, China
| | - Li Chai
- Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, College of Chemistry and Material Science, South-central University for Nationalities, Wuhan 430074, China
| | - Qian An
- Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, College of Chemistry and Material Science, South-central University for Nationalities, Wuhan 430074, China
| | - Wei Hu
- Department of Chemistry, Xinzhou Normal University, Xinzhou 034000, Shanxi, China
| | - Lina Wang
- Department of Chemistry, Xinzhou Normal University, Xinzhou 034000, Shanxi, China
| | - Xingcan Li
- Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, College of Chemistry and Material Science, South-central University for Nationalities, Wuhan 430074, China
| | - Huijuan Zhang
- Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, College of Chemistry and Material Science, South-central University for Nationalities, Wuhan 430074, China
| | - Chunya Li
- Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, College of Chemistry and Material Science, South-central University for Nationalities, Wuhan 430074, China
| |
Collapse
|
7
|
Hu W, Qiang T, Li C, Ren L, Cheng F, Wang B, Li M, Song X, James TD. Imaging of hypochlorous acid in mitochondria using an asymmetric near-infrared fluorescent probe with large Stokes shift. Chem Sci 2022; 13:11140-11149. [PMID: 36320485 PMCID: PMC9516895 DOI: 10.1039/d2sc03833a] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 08/22/2022] [Indexed: 02/10/2024] Open
Abstract
Small-molecule near-infrared (NIR) imaging facilitates deep tissue penetration, low autofluorescence, non-invasive visualization, and a relatively simple operation. As such it has emerged as a popular technique for tracking biological species and events. However, the small Stokes shift of most NIR dyes often results in a low signal-to-noise ratio and self-quenching due to crosstalk between the excitation and emission spectra. With this research, we developed a NIR-based fluorescent probe WD-HOCl for hypochlorous acid (HOCl) detection using the NIR dye TJ730 as the fluorophore, which exhibits a large Stokes shift of 156 nm, with no crosstalk between the excitation and emission spectra. It contains acyl hydrazide as the responsive group and a pyridinium cation as the mitochondria-targeting group. The fluorescence intensity of WD-HOCl was enhanced by 30.1-fold after reacting with HOCl. Imaging studies performed using BV-2 cells indicated that WD-HOCl could be used for endogenous HOCl detection and imaging in living cells exposed to glucose and oxygen deprivation/reperfusion. Finally, we demonstrated that inhibiting the expression of NOX2 reduced the HOCl levels and the severity of oxidative stress during stroke in a mouse model.
Collapse
Affiliation(s)
- Wei Hu
- College of Bioresources and Materials Engineering, Shaanxi Collaborative Innovation Center of Industrial Auxiliary Chemistry & Technology, Shaanxi University of Science & Technology Xi'an 710021 China
- Department of Chemistry, University of Bath Bath BA27AY UK
| | - Taotao Qiang
- College of Bioresources and Materials Engineering, Shaanxi Collaborative Innovation Center of Industrial Auxiliary Chemistry & Technology, Shaanxi University of Science & Technology Xi'an 710021 China
| | - Chenchen Li
- Hubei Key Laboratory of Biological Resources Protection and Utilization, Hubei University for Nationalities Enshi 445000 China
| | - Longfang Ren
- College of Bioresources and Materials Engineering, Shaanxi Collaborative Innovation Center of Industrial Auxiliary Chemistry & Technology, Shaanxi University of Science & Technology Xi'an 710021 China
| | - Fei Cheng
- College of Bioresources and Materials Engineering, Shaanxi Collaborative Innovation Center of Industrial Auxiliary Chemistry & Technology, Shaanxi University of Science & Technology Xi'an 710021 China
| | - Baoshuai Wang
- College of Bioresources and Materials Engineering, Shaanxi Collaborative Innovation Center of Industrial Auxiliary Chemistry & Technology, Shaanxi University of Science & Technology Xi'an 710021 China
| | - Mingli Li
- College of Bioresources and Materials Engineering, Shaanxi Collaborative Innovation Center of Industrial Auxiliary Chemistry & Technology, Shaanxi University of Science & Technology Xi'an 710021 China
| | - Xinjian Song
- Hubei Key Laboratory of Biological Resources Protection and Utilization, Hubei University for Nationalities Enshi 445000 China
| | - Tony D James
- Department of Chemistry, University of Bath Bath BA27AY UK
- School of Chemistry and Chemical Engineering, Henan Normal University Xinxiang 453007 China
| |
Collapse
|
8
|
Cadiz Diaz A, Schmidt NA, Yamazaki M, Hsieh CJ, Lisse TS, Rieger S. Coordinated NADPH oxidase/hydrogen peroxide functions regulate cutaneous sensory axon de- and regeneration. Proc Natl Acad Sci U S A 2022; 119:e2115009119. [PMID: 35858442 PMCID: PMC9340058 DOI: 10.1073/pnas.2115009119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 03/30/2022] [Indexed: 01/21/2023] Open
Abstract
Tissue wounding induces cutaneous sensory axon regeneration via hydrogen peroxide (H2O2) that is produced by the epithelial NADPH oxidase, Duox1. Sciatic nerve injury instead induces axon regeneration through neuronal uptake of the NADPH oxidase, Nox2, from macrophages. We therefore reasoned that the tissue environment in which axons are damaged stimulates distinct regenerative mechanisms. Here, we show that cutaneous axon regeneration induced by tissue wounding depends on both neuronal and keratinocyte-specific mechanisms involving H2O2 signaling. Genetic depletion of H2O2 in sensory neurons abolishes axon regeneration, whereas keratinocyte-specific H2O2 depletion promotes axonal repulsion, a phenotype mirrored in duox1 mutants. Intriguingly, cyba mutants, deficient in the essential Nox subunit, p22Phox, retain limited axon regenerative capacity but display delayed Wallerian degeneration and axonal fusion, observed so far only in invertebrates. We further show that keratinocyte-specific oxidation of the epidermal growth factor receptor (EGFR) at a conserved cysteine thiol (C797) serves as an attractive cue for regenerating axons, leading to EGFR-dependent localized epidermal matrix remodeling via the matrix-metalloproteinase, MMP-13. Therefore, wound-induced cutaneous axon de- and regeneration depend on the coordinated functions of NADPH oxidases mediating distinct processes following injury.
Collapse
Affiliation(s)
| | | | - Mamiko Yamazaki
- Department of Regenerative Biology and Medicine, MDI Biological Laboratory, Bar Harbor, ME 04672
| | - Chia-Jung Hsieh
- Department of Biology, University of Miami, Coral Gables, FL 33146
| | - Thomas S. Lisse
- Department of Biology, University of Miami, Coral Gables, FL 33146
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, FL 33136
| | - Sandra Rieger
- Department of Biology, University of Miami, Coral Gables, FL 33146
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, Miami, FL 33136
| |
Collapse
|
9
|
Almeida C, Pongilio RP, Móvio MI, Higa GSV, Resende RR, Jiang J, Kinjo ER, Kihara AH. Distinct Cell-specific Roles of NOX2 and MyD88 in Epileptogenesis. Front Cell Dev Biol 2022; 10:926776. [PMID: 35859905 PMCID: PMC9289522 DOI: 10.3389/fcell.2022.926776] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 06/15/2022] [Indexed: 02/05/2023] Open
Abstract
It is well established that temporal lobe epilepsy (TLE) is often related to oxidative stress and neuroinflammation. Both processes subserve alterations observed in epileptogenesis and ultimately involve distinct classes of cells, including astrocytes, microglia, and specific neural subtypes. For this reason, molecules associated with oxidative stress response and neuroinflammation have been proposed as potential targets for therapeutic strategies. However, these molecules can participate in distinct intracellular pathways depending on the cell type. To illustrate this, we reviewed the potential role of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (NOX2) and myeloid differentiation primary response 88 (MyD88) in astrocytes, microglia, and neurons in epileptogenesis. Furthermore, we presented approaches to study genes in different cells, employing single-cell RNA-sequencing (scRNAseq) transcriptomic analyses, transgenic technologies and viral serotypes carrying vectors with specific promoters. We discussed the importance of identifying particular roles of molecules depending on the cell type, endowing more effective therapeutic strategies to treat TLE.
Collapse
Affiliation(s)
- Cayo Almeida
- Laboratório de Neurogenética, Universidade Federal do ABC, São Bernardo do Campo, Brazil
| | | | - Marília Inês Móvio
- Laboratório de Neurogenética, Universidade Federal do ABC, São Bernardo do Campo, Brazil
| | | | - Rodrigo Ribeiro Resende
- Laboratório de Sinalização Celular e Nanobiotecnologia, Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jianxiong Jiang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Erika Reime Kinjo
- Laboratório de Neurogenética, Universidade Federal do ABC, São Bernardo do Campo, Brazil
| | | |
Collapse
|
10
|
Zhu G, Wang X, Chen L, Lenahan C, Fu Z, Fang Y, Yu W. Crosstalk Between the Oxidative Stress and Glia Cells After Stroke: From Mechanism to Therapies. Front Immunol 2022; 13:852416. [PMID: 35281064 PMCID: PMC8913707 DOI: 10.3389/fimmu.2022.852416] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
Stroke is the second leading cause of global death and is characterized by high rates of mortality and disability. Oxidative stress is accompanied by other pathological processes that together lead to secondary brain damage in stroke. As the major component of the brain, glial cells play an important role in normal brain development and pathological injury processes. Multiple connections exist in the pathophysiological changes of reactive oxygen species (ROS) metabolism and glia cell activation. Astrocytes and microglia are rapidly activated after stroke, generating large amounts of ROS via mitochondrial and NADPH oxidase pathways, causing oxidative damage to the glial cells themselves and neurons. Meanwhile, ROS cause alterations in glial cell morphology and function, and mediate their role in pathological processes, such as neuroinflammation, excitotoxicity, and blood-brain barrier damage. In contrast, glial cells protect the Central Nervous System (CNS) from oxidative damage by synthesizing antioxidants and regulating the Nuclear factor E2-related factor 2 (Nrf2) pathway, among others. Although numerous previous studies have focused on the immune function of glial cells, little attention has been paid to the role of glial cells in oxidative stress. In this paper, we discuss the adverse consequences of ROS production and oxidative-antioxidant imbalance after stroke. In addition, we further describe the biological role of glial cells in oxidative stress after stroke, and we describe potential therapeutic tools based on glia cells.
Collapse
Affiliation(s)
- Ganggui Zhu
- Department of Neurosurgery, Hangzhou First People's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoyu Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Luxi Chen
- Department of Medical Genetics, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cameron Lenahan
- Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA, United States.,Department of Biomedical Science, Burrell College of Osteopathic Medicine, Las Cruces, NM, United States
| | - Zaixiang Fu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuanjian Fang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wenhua Yu
- Department of Neurosurgery, Hangzhou First People's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Chai L, Liang T, An Q, Hu W, Wang Y, Wang B, Su S, Li C. Near-Infrared in and out: Observation of Autophagy during Stroke via a Lysosome-Targeting Two-Photon Viscosity-Dependent Probe. Anal Chem 2022; 94:5797-5804. [PMID: 35380428 DOI: 10.1021/acs.analchem.1c05143] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Fluorescence imaging using probes with two-photon excitation and near-infrared emission is currently the most popular in situ method for monitoring biological species or events, with a large imaging depth, low background fluorescence, low optical damage, and high spatial and temporal resolution. Nevertheless, current fluorescent dyes with near-infrared emission still have some disadvantages such as poor water solubility, low fluorescence quantum yield, and small two-photon absorption cross sections. These drawbacks are mainly caused by the structural characteristics of dyes with large conjugation surfaces but lacking strong and rigid structures. Herein, a lysosome-targeted and viscosity-sensitive probe (NCIC-VIS) is designed and synthesized. The protonation of morpholine not only helps anchor NCIC-VIS to the lysosome but also significantly enhances its water solubility. More importantly, its viscosity can increase the rigid structure of NCIC-VIS, which will improve the fluorescence quantum yield and the two-photon absorption cross section due to the imposed restrictions on molecular torsion. Based on the abovementioned characteristics, the real-time imaging of cellular autophagy (could increase the viscosity of lysosomes) was realized using NCIC-VIS. The results demonstrated that the level of autophagy was significantly enhanced in mice during stroke, while the inhibition of oxidative stress significantly reduced the degree of autophagy. The study corroborates that oxidative stress induced by stroke can lead to the development of autophagy.
Collapse
Affiliation(s)
- Li Chai
- Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, College of Chemistry and Material Science, South-Central University for Nationalities, Wuhan 430074, China
| | - Tianyu Liang
- College of Bioresources and Materials Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Qian An
- Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, College of Chemistry and Material Science, South-Central University for Nationalities, Wuhan 430074, China
| | - Wei Hu
- College of Bioresources and Materials Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Yanying Wang
- Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, College of Chemistry and Material Science, South-Central University for Nationalities, Wuhan 430074, China
| | - Baoshuai Wang
- College of Bioresources and Materials Engineering, Shaanxi University of Science & Technology, Xi'an 710021, China
| | - Shengze Su
- Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, College of Chemistry and Material Science, South-Central University for Nationalities, Wuhan 430074, China
| | - Chunya Li
- Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, College of Chemistry and Material Science, South-Central University for Nationalities, Wuhan 430074, China
| |
Collapse
|
12
|
Wang L, Qiang T, Ren L, Cheng F, Hu W, Qu R. Observation of macrophage autophagy in the healing of diabetic ulcers via a lysosome-targeting polarity-specific two-photon probe. RSC Adv 2022; 12:3654-3661. [PMID: 35425343 PMCID: PMC8979232 DOI: 10.1039/d1ra08417h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/19/2022] [Indexed: 12/25/2022] Open
Abstract
As a disease with high incidence, mutilation, and fatality rates, diabetic ulcers (DUs) have become a difficult and complicated disease of widely concern in recent years due to the unclear healing mechanism. The main reason for the delayed healing in DU patients is the unduly long chronic inflammation window, and the polarization state of macrophages plays a key role in this process. Since autophagy is believed to be closely related to the polarization trend of macrophages, recent studies have shown that autophagy is closely related to the healing of DU. To this end, a lysosome-targeting polarity-sensitive probe, XZTU-VIS, was developed to monitor the changes in lysosomal polarity, thereby assessing the autophagy of macrophages in mice suffering from DU. The experimental results showed that under two-photon fluorescence microscopy, the green channel fluorescence signal of XZTU-VIS decreased significantly during autophagy. In the meantime, DU models established using BV-2 cells and mice showed a process that could cause inflammation and the release of ROS, thereby inducing autophagy. A polarity-dependent two-photon fluorescent probe for evaluation of autophagy in the process of diabetic mouse skin ulcer-induced inflammation was constructed.![]()
Collapse
Affiliation(s)
- Lina Wang
- College of Bioresources and Materials Engineering, Shaanxi Collaborative Innovation Center of Industrial Auxiliary Chemistry & Technology, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Taotao Qiang
- College of Bioresources and Materials Engineering, Shaanxi Collaborative Innovation Center of Industrial Auxiliary Chemistry & Technology, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Longfang Ren
- College of Bioresources and Materials Engineering, Shaanxi Collaborative Innovation Center of Industrial Auxiliary Chemistry & Technology, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Fei Cheng
- College of Bioresources and Materials Engineering, Shaanxi Collaborative Innovation Center of Industrial Auxiliary Chemistry & Technology, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Wei Hu
- College of Bioresources and Materials Engineering, Shaanxi Collaborative Innovation Center of Industrial Auxiliary Chemistry & Technology, Shaanxi University of Science & Technology, Xi'an, 710021, China
- Shaanxi Collaborative Innovation Center of Industrial Auxiliary Chemistry & Technology, Shaanxi University of Science & Technology, Xi'an, 710021, China
| | - Renyu Qu
- Jiangsu Sevencontinent Green Chemical Co., Ltd., Zhangjiagang, 215600, China
| |
Collapse
|
13
|
Fang J, Sheng R, Qin ZH. NADPH Oxidases in the Central Nervous System: Regional and Cellular Localization and the Possible Link to Brain Diseases. Antioxid Redox Signal 2021; 35:951-973. [PMID: 34293949 DOI: 10.1089/ars.2021.0040] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Significance: The significant role of reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox) in signal transduction is mediated by the production of reactive oxygen species (ROS), especially in the central nervous system (CNS). The pathogenesis of some neurologic and psychiatric diseases is regulated by ROS, acting as a second messenger or pathogen. Recent Advances: In the CNS, the involvement of Nox-derived ROS has been implicated in the regulation of multiple signals, including cell survival/apoptosis, neuroinflammation, migration, differentiation, proliferation, and synaptic plasticity, as well as the integrity of the blood/brain barrier. In these processes, the intracellular signals mediated by the members of the Nox family vary among different tissues. The present review illuminates the regions and cellular, subcellular localization of Nox isoforms in the brain, the signal transduction, and the role of NOX enzymes in pathophysiology, respectively. Critical Issues: Different signal transduction cascades are coupled to ROS derived from various Nox homologues with varying degrees. Therefore, a critical issue worth noting is the varied role of the homologues of NOX enzymes in different signaling pathways and also they mediate different phenotypes in the diverse pathophysiological condition. This substantiates the effectiveness of selective Nox inhibitors in the CNS. Future Directions: Further investigation to elucidate the role of various homologues of NOX enzymes in acute and chronic brain diseases and signaling mechanisms, and the development of more specific NOX inhibitors for the treatment of CNS disease are urgently needed. Antioxid. Redox Signal. 35, 951-973.
Collapse
Affiliation(s)
- Jie Fang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, China
| |
Collapse
|
14
|
Mielnicka A, Michaluk P. Exocytosis in Astrocytes. Biomolecules 2021; 11:1367. [PMID: 34572580 PMCID: PMC8471187 DOI: 10.3390/biom11091367] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/10/2021] [Accepted: 09/14/2021] [Indexed: 12/17/2022] Open
Abstract
Until recently, astrocytes were thought to be a part of a simple "brain glue" providing only a supporting role for neurons. However, the discoveries of the last two decades have proven astrocytes to be dynamic partners participating in brain metabolism and actively influencing communication between neurons. The means of astrocyte-neuron communication are diverse, although regulated exocytosis has received the most attention but also caused the most debate. Similar to most of eukaryotic cells, astrocytes have a complex range of vesicular organelles which can undergo exocytosis as well as intricate molecular mechanisms that regulate this process. In this review, we focus on the components needed for regulated exocytosis to occur and summarise the knowledge about experimental evidence showing its presence in astrocytes.
Collapse
Affiliation(s)
| | - Piotr Michaluk
- BRAINCITY, Laboratory of Neurobiology, The Nencki Institute of Experimental Biology, PAS, 02-093 Warsaw, Poland;
| |
Collapse
|
15
|
Li X, Wang W, Yan J, Zeng F. Glutamic Acid Transporters: Targets for Neuroprotective Therapies in Parkinson's Disease. Front Neurosci 2021; 15:678154. [PMID: 34220434 PMCID: PMC8242205 DOI: 10.3389/fnins.2021.678154] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/07/2021] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease in middle-aged and elderly individuals. At present, no effective drug has been developed to treat PD. Although a variety of drugs exist for the symptomatic treatment of PD, they all have strong side effects. Most studies on PD mainly focus on dopaminergic neurons. This review highlights the function of glutamic acid transporters (GLTs), including excitatory amino acid transporters (EAATs) and vesicular glutamate transporters (VGLUTs), during the development of PD. In addition, using bioinformatics, we compared the expression of different types of glutamate transporter genes in the cingulate gyrus of PD patients and healthy controls. More importantly, we suggest that the functional roles of glutamate transporters may prove beneficial in the treatment of PD. In summary, VGLUTs and EAATs may be potential targets in the treatment of PD. VGLUTs and EAATs can be used as clinical drug targets to achieve better efficacy. Through this review article, we hope to enable future researchers to improve the condition of PD patients.
Collapse
Affiliation(s)
- Xiang Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, China
| | - Wenjun Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, China.,Institute for Cancer Medicine and School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Jianghong Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, China
| | - Fancai Zeng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, China
| |
Collapse
|
16
|
Mao L, Zuo ML, Wang AP, Tian Y, Dong LC, Li TM, Kuang DB, Song GL, Yang ZB. Low expression of miR‑532‑3p contributes to cerebral ischemia/reperfusion oxidative stress injury by directly targeting NOX2. Mol Med Rep 2020; 22:2415-2423. [PMID: 32705253 PMCID: PMC7411405 DOI: 10.3892/mmr.2020.11325] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 05/21/2020] [Indexed: 12/16/2022] Open
Abstract
NADPH oxidase 2 (NOX2) is a major subtype of NOX and is responsible for the generation of reactive oxygen species (ROS) in brain tissues. MicroRNAs (miRNAs/miRs) are important epigenetic regulators of NOX2. The present study aimed to identify the role of NOX2 miRNA-targets in ischemic stroke (IS). A rat cerebral ischemia/reperfusion (CI/R) injury model and a SH-SY5Y cell hypoxia/reoxygenation (H/R) model were used to simulate IS. Gene expression levels, ROS production and apoptosis in tissue or cells were determined, and bioinformatic analysis was conducted for target prediction of miRNA. In vitro experiments, including function-gain and luciferase activity assays, were also performed to assess the roles of miRNAs. The results indicated that NOX2 was significantly increased in brain tissues subjected to I/R and in SH-SY5Y cells subjected to H/R, while the expression of miR-532-3p (putative target of NOX2) was significantly decreased in brain tissues and plasma. Overexpression of miR-532-3p significantly suppressed NOX2 expression and ROS generation in SH-SY5Y cells subjected to H/R, as well as reduced the relative luciferase activity of cells transfected with a reporter gene plasmid. Collectively, these data indicated that miR-532-3p may be a target of NOX2 and a biomarker for CI/R injury. Thus, the present study may provide a novel target for drug development and IS therapy.
Collapse
Affiliation(s)
- Li Mao
- Office of Good Clinical Practice, The Affiliated Changsha Hospital of Hunan, Normal University, Changsha, Hunan 410006, P.R. China
| | - Mei-Ling Zuo
- Office of Good Clinical Practice, The Affiliated Changsha Hospital of Hunan, Normal University, Changsha, Hunan 410006, P.R. China
| | - Ai-Ping Wang
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Ying Tian
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Li-Chen Dong
- Office of Good Clinical Practice, The Affiliated Changsha Hospital of Hunan, Normal University, Changsha, Hunan 410006, P.R. China
| | - Tao-Ming Li
- Office of Good Clinical Practice, The Affiliated Changsha Hospital of Hunan, Normal University, Changsha, Hunan 410006, P.R. China
| | - Da-Bin Kuang
- Office of Good Clinical Practice, The Affiliated Changsha Hospital of Hunan, Normal University, Changsha, Hunan 410006, P.R. China
| | - Gui-Lin Song
- Office of Good Clinical Practice, The Affiliated Changsha Hospital of Hunan, Normal University, Changsha, Hunan 410006, P.R. China
| | - Zhong-Bao Yang
- Office of Good Clinical Practice, The Affiliated Changsha Hospital of Hunan, Normal University, Changsha, Hunan 410006, P.R. China
| |
Collapse
|
17
|
Zuo ML, Wang AP, Song GL, Yang ZB. miR-652 protects rats from cerebral ischemia/reperfusion oxidative stress injury by directly targeting NOX2. Biomed Pharmacother 2020; 124:109860. [PMID: 32000043 DOI: 10.1016/j.biopha.2020.109860] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/30/2019] [Accepted: 01/06/2020] [Indexed: 10/25/2022] Open
Abstract
Ischemic stroke is a devastating central nervous disease associated with oxidative stress and NOX2 is the main source of ROS responsible for brain tissue. miRNAs are a class of negative regulator of genes in mammals and involves the pathogenesis of ischemic stroke. This study aims to observe the role of target miRNA(miR-652) of NOX2 in ischemic stroke. A rat cerebral ischemia/reperfusion (CI/R) injury model and an SH-SY5Y cell hypoxia/reoxygenation(H/R) model were used to simulate ischemic stroke, and corresponding gene expression, biochemical indicators and pathophysiological indicators were measured to observe the role of miR-652. NOX2 significantly increased in brain tissues subjected to I/R or in SH-SY5Y cells subjected to H/R, while the expression level of miR-652(potential target of NOX2) significantly decreased in both brain tissues and plasma. Overexpression of miR-652 significantly suppressed NOX2 expression and ROS generation in H/R treated SH-SY5Y cells and reduced the relative luciferase activity of cells transfected with plasmid NOX2-WT (reporter gene plasmid). MiR-652 agomir significantly decreased the expression of NOX2 and ROS generation in brain tissues of CIR rats, as well as tissue injury. These data indicated that miR-652 protected rats from cerebral ischemia reperfusion injury by directly targeting NOX2, is a novel target for ischemic stroke therapy.
Collapse
Affiliation(s)
- Mei-Ling Zuo
- Office of Good Clinical Practice, The Affiliated Changsha Hospital of Hunan Normal University, Changsha, 410006, Hunan, China
| | - Ai-Ping Wang
- Institute of Clinical Research, Affiliated Nanhua Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Gui-Lin Song
- Office of Good Clinical Practice, The Affiliated Changsha Hospital of Hunan Normal University, Changsha, 410006, Hunan, China; Institute of Emergency and Critical Care Medicine of Changsha, Changsha, China
| | - Zhong-Bao Yang
- Office of Good Clinical Practice, The Affiliated Changsha Hospital of Hunan Normal University, Changsha, 410006, Hunan, China; Institute of Emergency and Critical Care Medicine of Changsha, Changsha, China.
| |
Collapse
|
18
|
Barua S, Kim JY, Yenari MA, Lee JE. The role of NOX inhibitors in neurodegenerative diseases. IBRO Rep 2019; 7:59-69. [PMID: 31463415 PMCID: PMC6709343 DOI: 10.1016/j.ibror.2019.07.1721] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 07/27/2019] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress is a key player in both chronic and acute brain disease due to the higher metabolic demand of the brain. Among the producers of free radicals, NADPH-oxidase (NOX) is a major contributor to oxidative stress in neurological disorders. In the brain, the superoxide produced by NOX is mainly found in leukocytes. However, recent studies have reported that it can be found in several other cell types. NOX has been reported to regulate neuronal signaling, memory processing, and central cardiovascular homeostasis. However, overproduction of NOX can contribute to neurotoxicity, CNS degeneration, and cardiovascular disorders. Regarding the above functions, NOX has been shown to play a crucial role in chronic CNS diseases like Parkinson's disease (PD), Alzheimer's disease (AD), Huntington's disease (HD), multiple sclerosis (MS), and amyotrophic lateral sclerosis (ALS), and in acute CNS disorders such as stroke, spinal cord injury, traumatic brain injury (TBI), and related cerebrovascular diseases. NOX is a multi-subunit complex consisting of two membrane-associated and four cytosolic subunits. Thus, in recent years, inhibition of NOX activity has drawn a great deal of attention from researchers in the field of treating chronic and acute CNS disorders and preventing secondary complications. Mounting evidence has shown that NOX inhibition is neuroprotective and that inhibiting NOX in circulating immune cells can improve neurological disease conditions. This review summarizes recent studies on the therapeutic effects and pharmacological strategies regarding NOX inhibitors in chronic and acute brain diseases and focuses on the hurdles that should be overcome before their clinical implementation.
Collapse
Affiliation(s)
- Sumit Barua
- Department of Anatomy, College of Medicine, Yonsei University, Republic of Korea
| | - Jong Youl Kim
- Department of Anatomy, College of Medicine, Yonsei University, Republic of Korea
| | - Midori A Yenari
- Department of Neurology, San Francisco Veterans Affairs Medical Center, University of California, San Francisco, 4150 Clement Street, MS 127, San Francisco, CA, 94121, United States
| | - Jong Eun Lee
- Department of Anatomy, College of Medicine, Yonsei University, Republic of Korea.,Brain Korea 21, PLUS Project for Medical Science, College of Medicine, Yonsei University, Republic of Korea.,Brain Research Institute, College of Medicine, Yonsei University, Republic of Korea
| |
Collapse
|
19
|
Selenium Drives a Transcriptional Adaptive Program to Block Ferroptosis and Treat Stroke. Cell 2019; 177:1262-1279.e25. [PMID: 31056284 DOI: 10.1016/j.cell.2019.03.032] [Citation(s) in RCA: 632] [Impact Index Per Article: 105.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 01/29/2019] [Accepted: 03/13/2019] [Indexed: 02/07/2023]
Abstract
Ferroptosis, a non-apoptotic form of programmed cell death, is triggered by oxidative stress in cancer, heat stress in plants, and hemorrhagic stroke. A homeostatic transcriptional response to ferroptotic stimuli is unknown. We show that neurons respond to ferroptotic stimuli by induction of selenoproteins, including antioxidant glutathione peroxidase 4 (GPX4). Pharmacological selenium (Se) augments GPX4 and other genes in this transcriptional program, the selenome, via coordinated activation of the transcription factors TFAP2c and Sp1 to protect neurons. Remarkably, a single dose of Se delivered into the brain drives antioxidant GPX4 expression, protects neurons, and improves behavior in a hemorrhagic stroke model. Altogether, we show that pharmacological Se supplementation effectively inhibits GPX4-dependent ferroptotic death as well as cell death induced by excitotoxicity or ER stress, which are GPX4 independent. Systemic administration of a brain-penetrant selenopeptide activates homeostatic transcription to inhibit cell death and improves function when delivered after hemorrhagic or ischemic stroke.
Collapse
|
20
|
Role of NADPH oxidase-2 in the progression of the inflammatory response secondary to striatum excitotoxic damage. J Neuroinflammation 2019; 16:91. [PMID: 30995916 PMCID: PMC6471795 DOI: 10.1186/s12974-019-1478-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 04/03/2019] [Indexed: 01/11/2023] Open
Abstract
Background During excitotoxic damage, neuronal death results from the increase in intracellular calcium, the induction of oxidative stress, and a subsequent inflammatory response. NADPH oxidases (NOX) are relevant sources of reactive oxygen species (ROS) during excitotoxic damage. NADPH oxidase-2 (NOX-2) has been particularly related to neuronal damage and death, as well as to the resolution of the subsequent inflammatory response. As ROS are crucial components of the regulation of inflammatory response, in this work, we evaluated the role of NOX-2 in the progression of inflammation resulting from glutamate-induced excitotoxic damage of the striatum in an in vivo model. Methods The striata of wild-type C57BL/6 J and NOX-2 KO mice (gp91Cybbtm1Din/J) were stereotactically injected with monosodium glutamate either alone or in combination with IL-4 or IL-10. The damage was evaluated in histological sections stained with cresyl violet and Fluoro-Jade B. The enzymatic activity of caspase-3 and NOX were also measured. Additionally, the cytokine profile was identified by ELISA and motor activity was verified by the tests of the cylinder, the adhesive tape removal, and the inverted grid. Results Our results show a neuroprotective effect in mice with a genetic inhibition of NOX-2, which is partially due to a differential response to excitotoxic damage, characterized by the production of anti-inflammatory cytokines. In NOX-2 KO animals, the excitotoxic condition increased the production of interleukin-4, which could contribute to the production of interleukin-10 that decreased neuronal apoptotic death and the magnitude of striatal injury. Treatment with interleukin-4 and interleukin-10 protected from excitotoxic damage in wild-type animals. Conclusions The release of proinflammatory cytokines during the excitotoxic event promotes an additional apoptotic death of neurons that survived the initial damage. During the subsequent inflammatory response to excitotoxic damage, ROS generated by NOX-2 play a decisive role in the extension of the lesion and consequently in the severity of the functional compromise, probably by regulating the anti-inflammatory cytokines production. Electronic supplementary material The online version of this article (10.1186/s12974-019-1478-4) contains supplementary material, which is available to authorized users.
Collapse
|
21
|
Rana AK, Singh D. Targeting glycogen synthase kinase-3 for oxidative stress and neuroinflammation: Opportunities, challenges and future directions for cerebral stroke management. Neuropharmacology 2018; 139:124-136. [DOI: 10.1016/j.neuropharm.2018.07.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/02/2018] [Accepted: 07/05/2018] [Indexed: 12/15/2022]
|
22
|
Wei M, Lyu H, Huo K, Su H. Impact of Bone Fracture on Ischemic Stroke Recovery. Int J Mol Sci 2018; 19:ijms19051533. [PMID: 29786644 PMCID: PMC5983742 DOI: 10.3390/ijms19051533] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/17/2018] [Accepted: 05/18/2018] [Indexed: 01/21/2023] Open
Abstract
Stroke is one of the most devastating complications of bone fracture, occurring in up to 4% of patients after surgical repair for hip fracture. Bone fracture and ischemic stroke have many common risk factors. The impact of bone fracture on stroke recovery has not drawn much attention in the research field. Bone fracture could occur in stroke patients at different times during the recovery phase, which steepens the trajectory of cognitive decline, greatly affects the quality of life, and causes a heavy burden on healthcare resources. In this paper, we reviewed the growing information on the pathophysiological mechanisms by which bone fracture may affect ischemic stroke recovery process.
Collapse
Affiliation(s)
- Meng Wei
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94110, USA.
| | - Haiyian Lyu
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94110, USA.
| | - Kang Huo
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94110, USA.
| | - Hua Su
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California, San Francisco, CA 94110, USA.
| |
Collapse
|
23
|
Abstract
Reactive oxygen species (ROS) are well known for their role in mediating both physiological and pathophysiological signal transduction. Enzymes and subcellular compartments that typically produce ROS are associated with metabolic regulation, and diseases associated with metabolic dysfunction may be influenced by changes in redox balance. In this review, we summarize the current literature surrounding ROS and their role in metabolic and inflammatory regulation, focusing on ROS signal transduction and its relationship to disease progression. In particular, we examine ROS production in compartments such as the cytoplasm, mitochondria, peroxisome, and endoplasmic reticulum and discuss how ROS influence metabolic processes such as proteasome function, autophagy, and general inflammatory signaling. We also summarize and highlight the role of ROS in the regulation metabolic/inflammatory diseases including atherosclerosis, diabetes mellitus, and stroke. In order to develop therapies that target oxidative signaling, it is vital to understand the balance ROS signaling plays in both physiology and pathophysiology, and how manipulation of this balance and the identity of the ROS may influence cellular and tissue homeostasis. An increased understanding of specific sources of ROS production and an appreciation for how ROS influence cellular metabolism may help guide us in the effort to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Steven J Forrester
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta GA
| | - Daniel S Kikuchi
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta GA
| | - Marina S Hernandes
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta GA
| | - Qian Xu
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta GA
| | - Kathy K Griendling
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta GA.
| |
Collapse
|
24
|
Lee WJ, Ham SA, Yoo H, Hwang JS, Yoo T, Paek KS, Lim DS, Han SG, Lee CH, Hong K, Seo HG. Activation of PPARδ attenuates neurotoxicity by inhibiting lipopolysaccharide-triggered glutamate release in BV-2 microglial cells. J Cell Biochem 2018; 119:5609-5619. [PMID: 29388693 DOI: 10.1002/jcb.26732] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 01/29/2018] [Indexed: 12/25/2022]
Abstract
Neuroinflammation-associated release of glutamate from activated microglia has been implicated in the progression of neurodegenerative diseases. However, the regulatory mechanisms underlying this glutamate release are poorly understood. Here, we show that peroxisome proliferator-activated receptor delta (PPARδ) modulates neurotoxicity by inhibiting glutamate release in lipopolysaccharide (LPS)-activated BV-2 microglial cells. Activation of PPARδ by GW501516, a specific PPARδ agonist, inhibited glutamate release in BV-2 cells. This effect of GW501516 was significantly blocked by shRNA-mediated knockdown of PPARδ and by treatment with GSK0660, a specific PPARδ antagonist, indicating that PPARδ is associated with blockade of glutamate release. Additionally, GW501516-activated PPARδ suppressed generation of reactive oxygen species and expression of gp91phox, a functional subunit of NADPH oxidase 2, in BV-2 cells stimulated with LPS. The inhibitory effect of GW501516 on gp91phox expression and glutamate release was further potentiated in the presence of AG490, a specific inhibitor of janus kinase 2 (JAK2), leading to the inhibition of signal transducer and activator of transcription 1 (STAT1). By contrast, GW501516 upregulated the expression of suppressor of cytokine signaling 1 (SOCS1), an endogenous inhibitor of JAK2. Furthermore, neurotoxicity induced by conditioned media from LPS-stimulated BV-2 cells was significantly reduced when conditioned media from BV-2 cells treated with both LPS and GW501516 were used. These results indicate that PPARδ attenuates LPS-triggered neuroinflammation by enhancing SOCS1-mediated inhibition of JAK2/STAT1 signaling, thereby inhibiting neurotoxicity associated with glutamate release.
Collapse
Affiliation(s)
- Won Jin Lee
- Sanghuh College of Life Sciences, Konkuk University, Gwangjin-gu, Seoul, Korea
| | - Sun Ah Ham
- Sanghuh College of Life Sciences, Konkuk University, Gwangjin-gu, Seoul, Korea
| | - Hyunjin Yoo
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Gwangjin-gu, Seoul, Korea
| | - Jung Seok Hwang
- Sanghuh College of Life Sciences, Konkuk University, Gwangjin-gu, Seoul, Korea
| | - Taesik Yoo
- Sanghuh College of Life Sciences, Konkuk University, Gwangjin-gu, Seoul, Korea
| | | | - Dae-Seog Lim
- Department of Biotechnology, CHA University, Bundang-gu, Seongnam, Korea
| | - Sung Gu Han
- Sanghuh College of Life Sciences, Konkuk University, Gwangjin-gu, Seoul, Korea
| | - Chi-Ho Lee
- Sanghuh College of Life Sciences, Konkuk University, Gwangjin-gu, Seoul, Korea
| | - Kwonho Hong
- Department of Stem Cell & Regenerative Biotechnology, Konkuk University, Gwangjin-gu, Seoul, Korea
| | - Han Geuk Seo
- Sanghuh College of Life Sciences, Konkuk University, Gwangjin-gu, Seoul, Korea
| |
Collapse
|
25
|
Therapeutic Potential of Novel Twin Compounds Containing Tetramethylpyrazine and Carnitine Substructures in Experimental Ischemic Stroke. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7191856. [PMID: 29387294 PMCID: PMC5745738 DOI: 10.1155/2017/7191856] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 07/16/2017] [Accepted: 08/13/2017] [Indexed: 02/07/2023]
Abstract
Although studies have seen dramatic advances in the understanding of the pathogenesis of stroke such as oxidative stress, inflammation, excitotoxicity, calcium overload and apoptosis, the delivery of stroke therapies is still a great challenge. In this study, we designed and synthesized a series of novel twin compounds containing tetramethylpyrazine and carnitine substructures and explored their therapeutic potential and mechanism in stroke-related neuronal injury. We first screened the neuroprotective effects of candidate compounds and found that among the tested compounds, LR134 and LR143 exhibited significant neuroprotection as evidenced by reducing cerebral infarct and edema, improving neurological function as well as blood-brain barrier integrity in rats after cerebral ischemia/reperfusion injury. We further demonstrated that the neuroprotective effects of compounds LR134 and LR143 were associated with the reduced inflammatory responses and NADPH oxidase- (NOX2-) mediated oxidative stress and the protection of mitochondria accompanied by the improvement of energy supply. In summary, this study provides direct evidence showing that the novel twin compounds containing tetramethylpyrazine and carnitine substructures have neuroprotective effects with multiple therapeutic targets, suggesting that modulation of these chemical structures may be an innovative therapeutic strategy for treating patients with stroke.
Collapse
|
26
|
Pan Y, Wang N, Xia P, Wang E, Guo Q, Ye Z. Inhibition of Rac1 ameliorates neuronal oxidative stress damage via reducing Bcl-2/Rac1 complex formation in mitochondria through PI3K/Akt/mTOR pathway. Exp Neurol 2017; 300:149-166. [PMID: 29129468 DOI: 10.1016/j.expneurol.2017.10.030] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Revised: 10/20/2017] [Accepted: 10/30/2017] [Indexed: 01/28/2023]
Abstract
Although the neuroprotective effects of Rac1 inhibition have been reported in various cerebral ischemic models, the molecular mechanisms of action have not yet been fully elucidated. In this study, we investigated whether the inhibition of Rac1 provided neuroprotection in a diabetic rat model of focal cerebral ischemia and hyperglycemia-exposed PC-12 cells. Intracerebroventricular administration of lentivirus expressing the Rac1 small hairpin RNA (shRNA) and specific Rac1 inhibitor NSC23766 not only decreased the infarct volumes and improved neurologic deficits with a correlated significant activation of mitochondrial DNA specific proteins, such as OGG1 and POLG, but also elevated Bcl-2 S70 phosphorylation in mitochondria. Furthermore, the levels of p-PI3K, p-Akt and p-mTOR increased, while 8-OHdG, ROS production and Bcl-2/Rac1 complex formation in mitochondria reduced in both Rac1-shRNA- and NSC23766-treated rats. Moreover, to confirm our in vivo observations, inhibition of Rac1 activity by NSC23766 suppressed the interactions between Bcl-2 and Rac1 in the mitochondria of PC-12 cells cultured in high glucose conditions and protected PC-12 cells from high glucose-induced neurotoxicity. More importantly, these beneficial effects of Rac1 inhibition were abolished by PI3K inhibitor LY294002. In contrast to NSC23766 treatment, LY294002 had little effect on the decrement of p-PTEN level. Taken together, these findings revealed novel neuroprotective roles of Rac1 inhibition against cerebral ischemic reperfusion injury in vivo and high glucose-induced neurotoxicity in PC-12 cells in vitro, by reducing Bcl-2/Rac1 complex formation in mitochondria through the activation of PI3K/Akt/mTOR survival pathway.
Collapse
Affiliation(s)
- Yundan Pan
- Department of Anesthesiology, Affiliated Xiangya Hospital of Central South University, Changsha 410078, Hunan Province, China
| | - Na Wang
- Department of Anesthesiology, Affiliated Xiangya Hospital of Central South University, Changsha 410078, Hunan Province, China
| | - Pingping Xia
- Department of Anesthesiology, Affiliated Xiangya Hospital of Central South University, Changsha 410078, Hunan Province, China
| | - E Wang
- Department of Anesthesiology, Affiliated Xiangya Hospital of Central South University, Changsha 410078, Hunan Province, China
| | - Qulian Guo
- Department of Anesthesiology, Affiliated Xiangya Hospital of Central South University, Changsha 410078, Hunan Province, China
| | - Zhi Ye
- Department of Anesthesiology, Affiliated Xiangya Hospital of Central South University, Changsha 410078, Hunan Province, China.
| |
Collapse
|
27
|
Kim JY, Park J, Lee JE, Yenari MA. NOX Inhibitors - A Promising Avenue for Ischemic Stroke. Exp Neurobiol 2017; 26:195-205. [PMID: 28912642 PMCID: PMC5597550 DOI: 10.5607/en.2017.26.4.195] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 08/09/2017] [Accepted: 08/09/2017] [Indexed: 12/19/2022] Open
Abstract
NADPH-oxidase (NOX) mediated superoxide originally found on leukocytes, but now recognized in several types of cells in the brain. It has been shown to play an important role in the progression of stroke and related cerebrovascular disease. NOX is a multisubunit complex consisting of 2 membrane-associated and 4 cytosolic subunits. NOX activation occurs when cytosolic subunits translocate to the membrane, leading to transport electrons to oxygen, thus producing superoxide. Superoxide produced by NOX is thought to function in long-term potentiation and intercellular signaling, but excessive production is damaging and has been implicated to play an important role in the progression of ischemic brain. Thus, inhibition of NOX activity may prove to be a promising treatment for ischemic brain as well as an adjunctive agent to prevent its secondary complications. There is mounting evidence that NOX inhibition in the ischemic brain is neuroprotective, and targeting NOX in circulating immune cells will also improve outcome. This review will focus on therapeutic effects of NOX assembly inhibitors in brain ischemia and stroke. However, the lack of specificity and toxicities of existing inhibitors are clear hurdles that will need to be overcome before this class of compounds could be translated clinically.
Collapse
Affiliation(s)
- Jong Youl Kim
- Department of Anatomy, Yensei University College of Medicine, Seoul 03722, Korea
| | - Joohyun Park
- Department of Anatomy, Yensei University College of Medicine, Seoul 03722, Korea.,BK21 Plus Project for Medical Sciences and Brain Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Jong Eun Lee
- Department of Anatomy, Yensei University College of Medicine, Seoul 03722, Korea.,BK21 Plus Project for Medical Sciences and Brain Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Midori A Yenari
- Department of Neurology, University of California, San Francisco, and San Francisco Veterans Affairs Medical Center, San Francisco, California 94121, USA
| |
Collapse
|
28
|
Ovelleiro D, Blanco S, Hernández R, Peinado MÁ. Comparative proteomic study of early hypoxic response in the cerebral cortex of rats submitted to two different hypoxic models. Proteomics Clin Appl 2017; 11. [PMID: 28697276 DOI: 10.1002/prca.201700058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/21/2017] [Accepted: 06/30/2017] [Indexed: 11/07/2022]
Abstract
PURPOSE The present study analyses and compares the cortical brain proteomic profiles of two different models of cerebral hypoxic insult in rats (HH: hypobaric hypoxia and HHI: ischemia followed by hypobaric hypoxia) in an attempt to describe the alterations of the early molecular hypoxic adaptive response underlying each one. EXPERIMENTAL DESIGN A quantitative proteomic profile of left-brain cortices of rats under HH, HHI, and control conditions was determined using isobaric labeling (Tandem Mass Tag™) on the protein extracts from pools of five individuals. Data are available via ProteomeXchange with identifier PXD004091. RESULTS Altogether, 339 proteins were confidently quantified, 99 of them showing significant variations in the hypoxic conditions with respect to the control. The HHI model presents a global effect of protein downregulation while HH produces an overall increase of the protein levels. While HH mainly affecting oxidative and energetic metabolism, HHI also interferes with synaptic transmission, neurotransmitter secretion, substantia nigra development, and triggers apoptosis through mitochondrial pathway. CONCLUSIONS AND CLINICAL RELEVANCE The findings obtained show an overview of protein alterations under two hypoxic models of different aetiology and provide a basis for more detailed studies in order to unravel new specific mechanisms and therapies for hypoxic pathologies.
Collapse
Affiliation(s)
- David Ovelleiro
- Area of Bioinformatics, Instituto Maimónides de Investigación Biomédica de Córdoba, Jaén, Spain
| | - Santos Blanco
- Area of Cellular Biology, Department of Experimental Biology, University of Jaén, Jaén, Spain
| | - Raquel Hernández
- Area of Cellular Biology, Department of Experimental Biology, University of Jaén, Jaén, Spain
| | - María Ángeles Peinado
- Area of Cellular Biology, Department of Experimental Biology, University of Jaén, Jaén, Spain
| |
Collapse
|
29
|
Benusa SD, George NM, Sword BA, DeVries GH, Dupree JL. Acute neuroinflammation induces AIS structural plasticity in a NOX2-dependent manner. J Neuroinflammation 2017; 14:116. [PMID: 28595650 PMCID: PMC5465457 DOI: 10.1186/s12974-017-0889-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 05/25/2017] [Indexed: 02/08/2023] Open
Abstract
Background Chronic microglia-mediated inflammation and oxidative stress are well-characterized underlying factors in neurodegenerative disease, whereby reactive inflammatory microglia enhance ROS production and impact neuronal integrity. Recently, it has been shown that during chronic inflammation, neuronal integrity is compromised through targeted disruption of the axon initial segment (AIS), the axonal domain critical for action potential initiation. AIS disruption was associated with contact by reactive inflammatory microglia which wrap around the AIS, increasing association with disease progression. While it is clear that chronic microglial inflammation and enhanced ROS production impact neuronal integrity, little is known about how acute microglial inflammation influences AIS stability. Here, we demonstrate that acute neuroinflammation induces AIS structural plasticity in a ROS-mediated and calpain-dependent manner. Methods C57BL/6J and NOX2−/− mice were given a single injection of lipopolysaccharide (LPS; 5 mg/kg) or vehicle (0.9% saline, 10 mL/kg) and analyzed at 6 h–2 weeks post-injection. Anti-inflammatory Didox (250 mg/kg) or vehicle (0.9% saline, 10 mL/kg) was administered beginning 24 h post-LPS injection and continued for 5 days; animals were analyzed 1 week post-injection. Microglial inflammation was assessed using immunohistochemistry (IHC) and RT-qPCR, and AIS integrity was quantitatively analyzed using ankyrinG immunolabeling. Data were statistically compared by one-way or two-way ANOVA where mean differences were significant as assessed using Tukey’s post hoc analysis. Results LPS-induced neuroinflammation, characterized by enhanced microglial inflammation and increased expression of ROS-producing enzymes, altered AIS protein clustering. Importantly, inflammation-induced AIS changes were reversed following resolution of microglial inflammation. Modulation of the inflammatory response using anti-inflammatory Didox, even after significant AIS disruption occurred, increased the rate of AIS recovery. qPCR and IHC analysis revealed that expression of microglial NOX2, a ROS-producing enzyme, was significantly increased correlating with AIS disruption. Furthermore, ablation of NOX2 prevented inflammation-induced AIS plasticity, suggesting that ROS drive AIS structural plasticity. Conclusions In the presence of acute microglial inflammation, the AIS undergoes an adaptive change that is capable of spontaneous recovery. Moreover, recovery can be therapeutically accelerated. Together, these findings underscore the dynamic capabilities of this domain in the presence of a pathological insult and provide evidence that the AIS is a viable therapeutic target.
Collapse
Affiliation(s)
- S D Benusa
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980709, , 1101 East Marshall Street, Richmond, VA, 23298, USA.,Neuroscience Curriculum, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - N M George
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980709, , 1101 East Marshall Street, Richmond, VA, 23298, USA.,Neuroscience Graduate Program, University of Colorado, Denver, CO, 80204, USA
| | - B A Sword
- Research Service 151, Hunter Holmes McGuire Veterans Affairs Medical Center, Department of Veterans Affairs, 1201 Broad Rock Blvd, Richmond, VA, 23249, USA
| | - G H DeVries
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980709, , 1101 East Marshall Street, Richmond, VA, 23298, USA.,Research Service 151, Hunter Holmes McGuire Veterans Affairs Medical Center, Department of Veterans Affairs, 1201 Broad Rock Blvd, Richmond, VA, 23249, USA
| | - J L Dupree
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, PO Box 980709, , 1101 East Marshall Street, Richmond, VA, 23298, USA. .,Research Service 151, Hunter Holmes McGuire Veterans Affairs Medical Center, Department of Veterans Affairs, 1201 Broad Rock Blvd, Richmond, VA, 23249, USA.
| |
Collapse
|
30
|
Activation of p47phox as a Mechanism of Bupivacaine-Induced Burst Production of Reactive Oxygen Species and Neural Toxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:8539026. [PMID: 28751934 PMCID: PMC5480047 DOI: 10.1155/2017/8539026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/04/2017] [Indexed: 12/02/2022]
Abstract
Bupivacaine has been shown to induce neurotoxicity through inducing excessive reactive oxygen species (ROS), but the underlying mechanism remains unclear. NOX2 is one of the most important sources of ROS in the nervous system, and its activation requires the membrane translocation of subunit p47phox. However, the role of p47phox in bupivacaine-induced neurotoxicity has not been explored. In our in vitro study, cultured human SH-SY5Y neuroblastoma cells were treated with 1.5 mM bupivacaine to induce neurotoxicity. Membrane translocation of p47phox was assessed by measuring the cytosol/membrane ratio of p47phox. The effects of the NOX inhibitor VAS2870 and p47phox-siRNA on bupivacaine-induced neurotoxicity were investigated. Furthermore, the effect of VAS2870 on bupivacaine-induced neurotoxicity was assessed in vivo in rats. All these changes were reversed by pretreatment with VAS2870 or transfection with p47phox-siRNA in SH-SY5Y cells. Similarly, pretreatment with VAS2870 attenuated bupivacaine-induced neuronal toxicity in rats. It is concluded that enhancing p47phox membrane translocation is a major mechanism whereby bupivacaine induced neurotoxicity and that pretreatment with VAS2870 or local p47phox gene knockdown attenuated bupivacaine-induced neuronal cell injury.
Collapse
|
31
|
Wu J, Hu W, Gong Y, Wang P, Tong L, Chen X, Chen Z, Xu X, Yao W, Zhang W, Huang C. Current pharmacological developments in 2,3,4',5-tetrahydroxystilbene 2-O-β-D-glucoside (TSG). Eur J Pharmacol 2017; 811:21-29. [PMID: 28545778 DOI: 10.1016/j.ejphar.2017.05.037] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 05/11/2017] [Accepted: 05/17/2017] [Indexed: 12/18/2022]
Abstract
2,3,4',5-tetrahydroxystilbene 2-O-β-D-glucoside (TSG), a resveratrol analog with glucoside, is purified from a traditional Chinese herbal medicine polygonum multiflorum. It has been extensively studied in last decade and known to exert strong anti-inflammatory, anti-oxidative, anti-apoptotic, and free radical scavenging activities, and therefore has been listed as a potential agent for disease therapies. Recent studies extend well-beyond effects of TSG on the injury of neurons, cardiomyocytes and endothelial cells, and report important functions of TSG in a lot of pathophysiological conditions. For example, TSG has been shown to prevent the production of pro-inflammatory cytokines in microglia and macrophages in vitro, and ameliorate pro-inflammatory responses in animal models with neurodegeneration, atherosclerosis, and rat paw or ear oedema. TSG can prevent the proliferation of vascular smooth cells, gastrointestinal dysfunctions, platelet aggregation, osteoblastic injury, diabetic nephropathy and melanogenesis. TSG is also indicated to facilitate long-term potentiation and learning and memory in both normal and pathological conditions. These effects to some extent enrich the understanding about the role of TSG in disease prevention and therapy. However, to date, we still have no outlined knowledges about the pharmacological effects of TSG, though the role of TSG in aging and Alzheimer's disease has been reviewed in recent years. Here, we summarize the current pharmacological developments of TSG as well as its possible mechanisms in disease prevention and therapy, aiming to push the understanding about the protective role of TSG as well as its preclinical assessment of novel applications.
Collapse
Affiliation(s)
- Jingjing Wu
- Department of Cardiology, Suzhou Kowloon Hospital of Shanghai Jiaotong University School of Medicine, #118 Wansheng Street, Suzhou 215021, Jiangsu, China
| | - Wenfeng Hu
- Department of Pharmacology, School of Pharmacy, Nantong University,#19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Yu Gong
- Department of Pharmacology, School of Pharmacy, Nantong University,#19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Peng Wang
- Department of Pharmacology, School of Pharmacy, Nantong University,#19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Lijuan Tong
- Department of Pharmacology, School of Pharmacy, Nantong University,#19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Xiangfan Chen
- Department of Pharmacology, School of Pharmacy, Nantong University,#19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Zhuo Chen
- Invasive Technology Department, Nantong First People's Hospital, the Second Affiliated Hospital of Nantong University, #6 North Road Hai'er Xiang, Nantong 226001, Jiangsu, China
| | - Xiaole Xu
- Department of Pharmacology, School of Pharmacy, Nantong University,#19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Wenjuan Yao
- Department of Pharmacology, School of Pharmacy, Nantong University,#19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Wei Zhang
- Department of Pharmacology, School of Pharmacy, Nantong University,#19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University,#19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China.
| |
Collapse
|
32
|
Bohmbach K, Schwarz MK, Schoch S, Henneberger C. The structural and functional evidence for vesicular release from astrocytes in situ. Brain Res Bull 2017; 136:65-75. [PMID: 28122264 DOI: 10.1016/j.brainresbull.2017.01.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 01/17/2017] [Accepted: 01/19/2017] [Indexed: 12/31/2022]
Abstract
The concept of the tripartite synapse states that bi-directional signalling between perisynaptic astrocyte processes, presynaptic axonal boutons and postsynaptic neuronal structures defines the properties of synaptic information processing. Ca2+-dependent vesicular release from astrocytes, as one of the mechanisms of astrocyte-neuron communication, has attracted particular attention but has also been the subject of intense debate. In neurons, regulated vesicular release is a strongly coordinated process. It requires a complex release machinery comprised of many individual components ranging from vesicular neurotransmitter transporters and soluble NSF attachment protein receptors (SNARE) proteins to Ca2+-sensors and the proteins that spatially and temporally control exocytosis of synaptic vesicles. If astrocytes employ similar mechanisms to release neurotransmitters is less well understood. The aim of this review is therefore to discuss recent experimental evidence that sheds light on the central structural components responsible for vesicular release from astrocytes in situ.
Collapse
Affiliation(s)
- Kirsten Bohmbach
- Institute of Cellular Neurosciences, University of Bonn Medical School, Bonn, Germany.
| | - Martin K Schwarz
- Department of Epileptology, University of Bonn Medical School, Bonn, Germany
| | - Susanne Schoch
- Institute of Neuropathology, University of Bonn Medical School, Bonn, Germany
| | - Christian Henneberger
- Institute of Cellular Neurosciences, University of Bonn Medical School, Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany; Institute of Neurology, University College London, London, United Kingdom.
| |
Collapse
|
33
|
Ma MW, Wang J, Zhang Q, Wang R, Dhandapani KM, Vadlamudi RK, Brann DW. NADPH oxidase in brain injury and neurodegenerative disorders. Mol Neurodegener 2017; 12:7. [PMID: 28095923 PMCID: PMC5240251 DOI: 10.1186/s13024-017-0150-7] [Citation(s) in RCA: 299] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 01/05/2017] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress is a common denominator in the pathology of neurodegenerative disorders such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, and multiple sclerosis, as well as in ischemic and traumatic brain injury. The brain is highly vulnerable to oxidative damage due to its high metabolic demand. However, therapies attempting to scavenge free radicals have shown little success. By shifting the focus to inhibit the generation of damaging free radicals, recent studies have identified NADPH oxidase as a major contributor to disease pathology. NADPH oxidase has the primary function to generate free radicals. In particular, there is growing evidence that the isoforms NOX1, NOX2, and NOX4 can be upregulated by a variety of neurodegenerative factors. The majority of recent studies have shown that genetic and pharmacological inhibition of NADPH oxidase enzymes are neuroprotective and able to reduce detrimental aspects of pathology following ischemic and traumatic brain injury, as well as in chronic neurodegenerative disorders. This review aims to summarize evidence supporting the role of NADPH oxidase in the pathology of these neurological disorders, explores pharmacological strategies of targeting this major oxidative stress pathway, and outlines obstacles that need to be overcome for successful translation of these therapies to the clinic.
Collapse
Affiliation(s)
- Merry W Ma
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Jing Wang
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Quanguang Zhang
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Ruimin Wang
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Krishnan M Dhandapani
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neurosurgery, Medical College of Georgia, Augusta University, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Ratna K Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, 7703 Medical Drive, San Antonio, TX, 78229, USA
| | - Darrell W Brann
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA. .,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA.
| |
Collapse
|
34
|
Kim H, Wei Y, Lee JY, Wu Y, Zheng Y, Moskowitz MA, Chen JW. Myeloperoxidase Inhibition Increases Neurogenesis after Ischemic Stroke. J Pharmacol Exp Ther 2016; 359:262-272. [PMID: 27550713 DOI: 10.1124/jpet.116.235127] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 08/19/2016] [Indexed: 01/08/2023] Open
Abstract
The relationship between inflammation and neurogenesis in stroke is currently not well understood. Focal ischemia enhances cell proliferation and neurogenesis in the neurogenic regions, including the subventricular zone (SVZ), dentate gyrus, as well as the non-neurogenic striatum, and cortex in the ischemic hemisphere. Myeloperoxidase (MPO) is a potent oxidizing enzyme secreted during inflammation by activated leukocytes, and its enzymatic activity is highly elevated after stroke. In this study, we investigated whether the inhibition of MPO activity by a specific irreversible inhibitor, 4-aminobenzoic acid hydrazide (ABAH) (MPO-/- mice) can increase neurogenesis after transient middle cerebral artery occlusion in mice. ABAH administration increased the number of proliferating bromodeoxyuridine (BrdU)-positive cells expressing markers for neural stems cells, astrocytes, neuroprogenitor cells (Nestin), and neuroblasts (doublecortin) in the ischemic SVZ, anterior SVZ, striatum, and cortex. MPO inhibition also increased levels of brain-derived neurotrophic factor, phosphorylation of cAMP response element-binding protein (Ser133), acetylated H3, and NeuN to promote neurogenesis in the ischemic SVZ. ABAH treatment also increased chemokine CXC receptor 4 expression in the ischemic SVZ. MPO-deficient mice treated with vehicle or ABAH both showed similar effects on the number of BrdU+ cells in the ischemic hemisphere, demonstrating that ABAH is specific to MPO. Taken together, our results underscore a detrimental role of MPO activity to postischemia neurogenesis and that a strategy to inhibit MPO activity can increase cell proliferation and improve neurogenesis after ischemic stroke.
Collapse
Affiliation(s)
- HyeonJu Kim
- Center for Systems Biology and Institute for Innovation in Imaging (H.K., J.Y.L., J.W.C), and Neuroscience Center (Y. Wei, Y. Wu, Y.Z., M.A.M.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ying Wei
- Center for Systems Biology and Institute for Innovation in Imaging (H.K., J.Y.L., J.W.C), and Neuroscience Center (Y. Wei, Y. Wu, Y.Z., M.A.M.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ji Yong Lee
- Center for Systems Biology and Institute for Innovation in Imaging (H.K., J.Y.L., J.W.C), and Neuroscience Center (Y. Wei, Y. Wu, Y.Z., M.A.M.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yue Wu
- Center for Systems Biology and Institute for Innovation in Imaging (H.K., J.Y.L., J.W.C), and Neuroscience Center (Y. Wei, Y. Wu, Y.Z., M.A.M.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yi Zheng
- Center for Systems Biology and Institute for Innovation in Imaging (H.K., J.Y.L., J.W.C), and Neuroscience Center (Y. Wei, Y. Wu, Y.Z., M.A.M.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Michael A Moskowitz
- Center for Systems Biology and Institute for Innovation in Imaging (H.K., J.Y.L., J.W.C), and Neuroscience Center (Y. Wei, Y. Wu, Y.Z., M.A.M.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - John W Chen
- Center for Systems Biology and Institute for Innovation in Imaging (H.K., J.Y.L., J.W.C), and Neuroscience Center (Y. Wei, Y. Wu, Y.Z., M.A.M.), Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
35
|
Lu Y, Li CJ, Chen C, Luo P, Zhou M, Li C, Xu XL, Lu Q, He Z, Guo LJ. Activation of GABAB2 subunits alleviates chronic cerebral hypoperfusion-induced anxiety-like behaviours: A role for BDNF signalling and Kir3 channels. Neuropharmacology 2016; 110:308-321. [PMID: 27515806 DOI: 10.1016/j.neuropharm.2016.08.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 07/30/2016] [Accepted: 08/06/2016] [Indexed: 12/15/2022]
Abstract
Anxiety is an affective disorder that is commonly observed after irreversible brain damage induced by cerebral ischemia and can delay the physical and cognitive recovery, which affects the quality of life of both the patient and family members. However, anxiety after ischemia has received less attention, and mechanisms underlying anxiety-like behaviours induced by chronic cerebral ischemia are under-investigated. In the present study, the chronic cerebral hypoperfusion model was established by the permanent occlusion of the bilateral common carotid arteries (two-vessel occlusion, 2VO) in rats, and anxiety-related behaviours were evaluated. Results indicated that 2VO induced obvious anxiety-like behaviours; the surface expressions of GABAB2 subunits were down-regulated; Brain derived neurotrophic factor (BDNF), tyrosine kinase B (TrkB) and neural cell adhesion molecule (NCAM) were reduced; Meanwhile, the surface expressions of G protein-activated inwardly rectifying potassium (GIRK, Kir3) channels were up-regulated in hippocampal CA1 in 2VO rats. Baclofen, a GABAB receptor agonist, significantly ameliorated the anxiety-like behaviours. It also improved the down-regulation of GABAB2 surface expressions, restored the levels of BDNF, TrkB and NCAM, and reversed the increased surface expressions of Kir3 in hippocampal CA1 in 2VO rats. However, the effects of baclofen were absent in shRNA-GABAB2 infected 2VO rats. These results suggested that activation of GABAB2 subunits could improve BDNF signalling and reverse Kir3 channel surface expressions in hippocampal CA1, which may alleviate the anxiety-like behaviours in rats with chronic cerebral hypoperfusion.
Collapse
Affiliation(s)
- Yun Lu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chang-Jun Li
- Neurology Department, Tongji Medical College, Huazhong University of Science and Technology, The Central Hospital of Wuhan, Wuhan 430030, China
| | - Cheng Chen
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Pan Luo
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Mei Zhou
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Cai Li
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xu-Lin Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Institute of Brain Research, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qing Lu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Institute of Brain Research, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhi He
- Department of Neuropsychopharmacology, Medical School of China Three Gorges University, Yichang 443002, China.
| | - Lian-Jun Guo
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Institute of Brain Research, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
36
|
Shi W, Wei X, Wang Z, Han H, Fu Y, Liu J, Zhang Y, Guo J, Dong C, Zhou D, Zhou Q, Chen Y, Yi F. HDAC9 exacerbates endothelial injury in cerebral ischaemia/reperfusion injury. J Cell Mol Med 2016; 20:1139-49. [PMID: 26865248 PMCID: PMC4882992 DOI: 10.1111/jcmm.12803] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Accepted: 12/22/2015] [Indexed: 01/08/2023] Open
Abstract
Histone deacetylase (HDAC) 9, a member of class II HDACs, regulates a wide variety of normal and abnormal physiological functions, which is usually expressed at high levels in the brain and skeletal muscle. Although studies have highlighted the importance of HDAC-mediated epigenetic processes in the development of ischaemic stroke and very recent genome-wide association studies have identified a variant in HDAC9 associated with large-vessel ischemic stroke, the molecular events by which HDAC9 induces cerebral injury keep unclear. In this study, we found that HDAC9 was up-regulated in the ischaemic cerebral hemisphere after cerebral ischaemia/reperfusion (I/R) injury in rats and in vivo gene silencing of HDAC9 by recombinated lentivirus infection in the brain reduced cerebral injury in experimental stroke. We further demonstrated that HDAC9 contributed to oxygen-glucose deprivation-induced brain microvessel endothelial cell dysfunction as demonstrated by the increased inflammatory responses, cellular apoptosis and endothelial cell permeability dysfunction accompanied by reduced expression of tight-junction proteins. We further found that HDAC9 suppressed autophagy, which was associated with endothelial dysfunction. This study for the first time provides direct evidence that HDAC9 contributes to endothelial cell injury and demonstrates that HDAC9 is one of critical components of a signal transduction pathway that links cerebral injury to epigenetic modification in the brain.
Collapse
Affiliation(s)
- Weichen Shi
- Department of Pharmacology, Shandong University School of Medicine, Jinan, China.,Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Xinbing Wei
- Department of Pharmacology, Shandong University School of Medicine, Jinan, China
| | - Ziying Wang
- Department of Pharmacology, Shandong University School of Medicine, Jinan, China
| | - Huirong Han
- Department of Pharmacology, Shandong University School of Medicine, Jinan, China
| | - Yi Fu
- Department of Pharmacology, Shandong University School of Medicine, Jinan, China
| | - Jiang Liu
- Department of Pharmacology, Shandong University School of Medicine, Jinan, China
| | - Yan Zhang
- Department of Pharmacology, Shandong University School of Medicine, Jinan, China
| | - Jian Guo
- Department of Pharmacology, Shandong University School of Medicine, Jinan, China
| | - Chuanqiao Dong
- Department of Pharmacology, Shandong University School of Medicine, Jinan, China
| | - Di Zhou
- Department of Pharmacology, Shandong University School of Medicine, Jinan, China
| | - Quan Zhou
- Department of Pharmacology, Shandong University School of Medicine, Jinan, China
| | - Yuxin Chen
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Fan Yi
- Department of Pharmacology, Shandong University School of Medicine, Jinan, China
| |
Collapse
|
37
|
Rezq S, Abdel-Rahman AA. Central GPR109A Activation Mediates Glutamate-Dependent Pressor Response in Conscious Rats. J Pharmacol Exp Ther 2015; 356:456-65. [PMID: 26621144 DOI: 10.1124/jpet.115.229146] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/02/2015] [Indexed: 12/11/2022] Open
Abstract
G protein-coupled receptor 109A (GPR109A) activation by its ligand nicotinic acid (NA) in immune cells increases Ca(2+) levels, and Ca(2+) induces glutamate release and oxidative stress in central blood pressure (BP)-regulating nuclei, for example, the rostral ventrolateral medulla (RVLM), leading to sympathoexcitation. Despite NA's ability to reach the brain, the expression and function of its receptor GPR109A in the RVLM remain unknown. We hypothesized that NA activation of RVLM GPR109A causes Ca(2+)-dependent l-glutamate release and subsequently increases neuronal oxidative stress, sympathetic activity, and BP. To test this hypothesis, we adopted a multilevel approach, which included pharmacologic in vivo studies along with ex vivo and in vitro molecular studies in rat pheochromocytoma cell line (PC12) cells (which exhibit neuronal phenotype). We present the first evidence for GPR109A expression in the RVLM and in PC12 cells. Next, we showed that RVLM GPR109A activation (NA) caused pressor and bradycardic responses in conscious rats. The resemblance of these responses to those caused by intra-RVLM glutamate and their attenuation by NMDA receptor (NMDAR) blockade (2-amino-5-phosphonopentanoic acid) and enhancement by l-glutamate uptake inhibition (l-trans-pyrrolidine-2,4-dicarboxylic acid, PDC) supported our hypothesis. NA increased Ca(2+), glutamate, nitric oxide and reactive oxygen species (ROS) levels in PC12 cells and increased RVLM ROS levels. The inactive NA analog isonicotinic acid failed to replicate the cardiovascular and biochemical effects of NA. Further, GPR109A knockdown (siRNA) abrogated the biochemical effects of NA in PC12 cells. These novel findings yield new insight into the role of RVLM GPR109A in central BP control.
Collapse
Affiliation(s)
- Samar Rezq
- Department of Pharmacology, School of Medicine, East Carolina University, North Carolina
| | - Abdel A Abdel-Rahman
- Department of Pharmacology, School of Medicine, East Carolina University, North Carolina
| |
Collapse
|
38
|
Kleikers PWM, Hooijmans C, Göb E, Langhauser F, Rewell SSJ, Radermacher K, Ritskes-Hoitinga M, Howells DW, Kleinschnitz C, HHW Schmidt H. A combined pre-clinical meta-analysis and randomized confirmatory trial approach to improve data validity for therapeutic target validation. Sci Rep 2015; 5:13428. [PMID: 26310318 PMCID: PMC4550831 DOI: 10.1038/srep13428] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 07/27/2015] [Indexed: 12/30/2022] Open
Abstract
Biomedical research suffers from a dramatically poor translational success. For example, in ischemic stroke, a condition with a high medical need, over a thousand experimental drug targets were unsuccessful. Here, we adopt methods from clinical research for a late-stage pre-clinical meta-analysis (MA) and randomized confirmatory trial (pRCT) approach. A profound body of literature suggests NOX2 to be a major therapeutic target in stroke. Systematic review and MA of all available NOX2(-/y) studies revealed a positive publication bias and lack of statistical power to detect a relevant reduction in infarct size. A fully powered multi-center pRCT rejects NOX2 as a target to improve neurofunctional outcomes or achieve a translationally relevant infarct size reduction. Thus stringent statistical thresholds, reporting negative data and a MA-pRCT approach can ensure biomedical data validity and overcome risks of bias.
Collapse
Affiliation(s)
- Pamela WM. Kleikers
- Department of Pharmacology, CARIM, Faculty of Health, Medicine and Life Sciences, Maastricht University, The Netherlands
| | - Carlijn Hooijmans
- SYRCLE at Central Animal Laboratory, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Eva Göb
- Neurologische Klinik und Poliklinik der Universitätsklinik Würzburg, Würzburg, Germany
| | - Friederike Langhauser
- Neurologische Klinik und Poliklinik der Universitätsklinik Würzburg, Würzburg, Germany
| | - Sarah SJ. Rewell
- Florey Institute of Neuroscience and Mental Health, Austin Health, Melbourne, Victoria, Australia
| | - Kim Radermacher
- Department of Pharmacology, CARIM, Faculty of Health, Medicine and Life Sciences, Maastricht University, The Netherlands
| | - Merel Ritskes-Hoitinga
- SYRCLE at Central Animal Laboratory, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - David W. Howells
- Florey Institute of Neuroscience and Mental Health, Austin Health, Melbourne, Victoria, Australia
| | | | - Harald HHW Schmidt
- Department of Pharmacology, CARIM, Faculty of Health, Medicine and Life Sciences, Maastricht University, The Netherlands
| |
Collapse
|
39
|
Li M, Niu F, Zhu X, Wu X, Shen N, Peng X, Liu Y. PRRT2 Mutant Leads to Dysfunction of Glutamate Signaling. Int J Mol Sci 2015; 16:9134-51. [PMID: 25915028 PMCID: PMC4463582 DOI: 10.3390/ijms16059134] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Revised: 04/10/2015] [Accepted: 04/13/2015] [Indexed: 01/28/2023] Open
Abstract
Paroxysmal kinesigenic choreoathetosis (PKC) is an inherited disease of the nervous system. We previously identified PRRT2 as the causative gene of PKC. However, as little is known about the function of PRRT2, elucidating its function will benefit not only PKC studies, but also many other related disorders. Here, we reveal higher levels of glutamate in the plasma of PKC patients and the culture medium of neurons following knock-out Prrt2 expression. Using double immunostaining assays we confirm Prrt2 is located at the glutamatergic neurons in accordance with its function. Our co-immunoprecipitation assays reveal mutant PRRT2 interferes with SNAP25 and GRIA1 interactions, respectively. Furthermore, using live-labeling techniques, we confirmed co-transfection with mutant PRRT2 caused an increase in GRIA1 distribution on the cell surface. Therefore, our results suggest that mutant PRRT2, probably through its weakened interaction with SNAP25, affects glutamate signaling and glutamate receptor activity, resulting in the increase of glutamate release and subsequent neuronal hyperexcitability.
Collapse
Affiliation(s)
- Ming Li
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China.
| | - Fenghe Niu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China.
| | - Xilin Zhu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China.
| | - Xiaopan Wu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China.
| | - Ning Shen
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China.
| | - Xiaozhong Peng
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China.
| | - Ying Liu
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing 100005, China.
| |
Collapse
|
40
|
Katz PS, Sulzer JK, Impastato RA, Teng SX, Rogers EK, Molina PE. Endocannabinoid degradation inhibition improves neurobehavioral function, blood-brain barrier integrity, and neuroinflammation following mild traumatic brain injury. J Neurotrauma 2014; 32:297-306. [PMID: 25166905 DOI: 10.1089/neu.2014.3508] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Traumatic brain injury (TBI) is an increasingly frequent and poorly understood condition lacking effective therapeutic strategies. Inflammation and oxidative stress (OS) are critical components of injury, and targeted interventions to reduce their contribution to injury should improve neurobehavioral recovery and outcomes. Recent evidence reveals potential protective, yet short-lived, effects of the endocannabinoids (ECs), 2-arachidonoyl glycerol (2-AG) and N-arachidonoyl-ethanolamine (AEA), on neuroinflammatory and OS processes after TBI. The aim of this study was to determine whether EC degradation inhibition after TBI would improve neurobehavioral recovery by reducing inflammatory and oxidative damage. Adult male Sprague-Dawley rats underwent a 5-mm left lateral craniotomy, and TBI was induced by lateral fluid percussion. TBI produced apnea (17±5 sec) and a delayed righting reflex (479±21 sec). Thirty minutes post-TBI, rats were randomized to receive intraperitoneal injections of vehicle (alcohol, emulphor, and saline; 1:1:18) or a selective inhibitor of 2-AG (JZL184, 16 mg/kg) or AEA (URB597, 0.3 mg/kg) degradation. At 24 h post-TBI, animals showed significant neurological and -behavioral impairment as well as disruption of blood-brain barrier (BBB) integrity. Improved neurological and -behavioral function was observed in JZL184-treated animals. BBB integrity was protected in both JZL184- and URB597-treated animals. No significant differences in ipsilateral cortex messenger RNA expression of interleukin (IL)-1β, IL-6, chemokine (C-C motif) ligand 2, tumor necrosis factor alpha, cyclooxygenase 2 (COX2), or nicotinamide adenine dinucleotide phosphate oxidase (NOX2) and protein expression of COX2 or NOX2 were observed across experimental groups. Astrocyte and microglia activation was significantly increased post-TBI, and treatment with JZL184 or URB597 blocked activation of both cell types. These findings suggest that EC degradation inhibition post-TBI exerts neuroprotective effects. Whether repeated dosing would achieve greater protection remains to be examined.
Collapse
Affiliation(s)
- Paige S Katz
- Department of Physiology, Alcohol and Drug Abuse Center of Excellence, Louisiana State University Health Sciences Center , New Orleans, Louisiana
| | | | | | | | | | | |
Collapse
|
41
|
Han Z, Shen F, He Y, Degos V, Camus M, Maze M, Young WL, Su H. Activation of α-7 nicotinic acetylcholine receptor reduces ischemic stroke injury through reduction of pro-inflammatory macrophages and oxidative stress. PLoS One 2014; 9:e105711. [PMID: 25157794 PMCID: PMC4144901 DOI: 10.1371/journal.pone.0105711] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 07/23/2014] [Indexed: 12/17/2022] Open
Abstract
Activation of α-7 nicotinic acetylcholine receptor (α-7 nAchR) has a neuro-protective effect on ischemic and hemorrhagic stroke. However, the underlying mechanism is not completely understood. We hypothesized that α-7 nAchR agonist protects brain injury after ischemic stroke through reduction of pro-inflammatory macrophages (M1) and oxidative stress. C57BL/6 mice were treated with PHA568487 (PHA, α-7 nAchR agonist), methyllycaconitine (MLA, nAchR antagonist), or saline immediately and 24 hours after permanent occlusion of the distal middle cerebral artery (pMCAO). Behavior test, lesion volume, CD68+, M1 (CD11b+/Iba1+) and M2 (CD206/Iba1+) microglia/macrophages, and phosphorylated p65 component of NF-kB in microglia/macrophages were quantified using histological stained sections. The expression of M1 and M2 marker genes, anti-oxidant genes and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase were quantified using real-time RT-PCR. Compared to the saline-treated mice, PHA mice had fewer behavior deficits 3 and 7 days after pMCAO, and smaller lesion volume, fewer CD68+ and M1 macrophages, and more M2 macrophages 3 and 14 days after pMCAO, whereas MLA's effects were mostly the opposite in several analyses. PHA increased anti-oxidant genes and NADPH oxidase expression associated with decreased phosphorylation of NF-kB p65 in microglia/macrophages. Thus, reduction of inflammatory response and oxidative stress play roles in α-7 nAchR neuro-protective effect.
Collapse
Affiliation(s)
- Zhenying Han
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, United States of America
- Tianjin Medical University General Hospital, Tianjin, China
| | - Fanxia Shen
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, United States of America
| | - Yue He
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, United States of America
| | - Vincent Degos
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, United States of America
- Hôpital Pitié Salpetrière, Assistance Publique-Hopitaux de Paris (APHP), Université Pierre et Marie Curie-Paris VI and UMR INSERM 1141, Paris, France
| | - Marine Camus
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, United States of America
| | - Mervyn Maze
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, United States of America
| | - William L. Young
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, United States of America
- Department of Neurological Surgery, University of California San Francisco, San Francisco, California, United States of America
- Department of Neurology, University of California San Francisco, San Francisco, California, United States of America
| | - Hua Su
- Center for Cerebrovascular Research, Department of Anesthesia and Perioperative Care, University of California San Francisco, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
42
|
Han Z, Li L, Wang L, Degos V, Maze M, Su H. Alpha-7 nicotinic acetylcholine receptor agonist treatment reduces neuroinflammation, oxidative stress, and brain injury in mice with ischemic stroke and bone fracture. J Neurochem 2014; 131:498-508. [PMID: 25040630 DOI: 10.1111/jnc.12817] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 06/26/2014] [Accepted: 07/03/2014] [Indexed: 12/21/2022]
Abstract
Bone fracture at the acute stage of stroke exacerbates stroke injury by increasing neuroinflammation. We hypothesize that activation of α-7 nicotinic acetylcholine receptor (α-7 nAchR) attenuates neuroinflammation and oxidative stress, and reduces brain injury in mice with bone fracture and stroke. Permanent middle cerebral artery occlusion (pMCAO) was performed in C57BL/6J mice followed by tibia fracture 1 day later. Mice were treated with 0.8 mg/kg PHA 568487 (PHA, α-7 nAchR-specific agonist), 6 mg/kg methyllycaconitine (α-7 nAchR antagonist), or saline 1 and 2 days after pMCAO. Behavior was tested 3 days after pMCAO. Neuronal injury, CD68(+) , M1 (pro-inflammatory) and M2 (anti-inflammatory) microglia/macrophages, phosphorylated p65 component of nuclear factor kappa b in microglia/macrophages, oxidative and anti-oxidant gene expression were quantified. Compared to saline-treated mice, PHA-treated mice performed better in behavioral tests, had fewer apoptotic neurons (NeuN(+) TUNEL(+) ), fewer CD68(+) and M1 macrophages, and more M2 macrophages. PHA increased anti-oxidant gene expression and decreased oxidative stress and phosphorylation of nuclear factor kappa b p65. Methyllycaconitine had the opposite effects. Our data indicate that α-7 nAchR agonist treatment reduces neuroinflammation and oxidative stress, which are associated with reduced brain injury in mice with ischemic stroke plus tibia fracture. Bone fracture at the acute stage of stroke exacerbates neuroinflammation, oxidative stress, and brain injury, and our study has shown that the α-7 nAchR agonist, PHA (PHA 568487), attenuates neuroinflammation, oxidative stress, and brain injury in mice with stroke and bone fracture. Hence, PHA could provide an opportunity to develop a new strategy to reduce brain injury in patients suffering from stroke and bone fracture.
Collapse
Affiliation(s)
- Zhenying Han
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, San Francisco, California, USA; Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | | | | | | | | | | |
Collapse
|
43
|
Anandamide protects HT22 cells exposed to hydrogen peroxide by inhibiting CB1 receptor-mediated type 2 NADPH oxidase. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:893516. [PMID: 25136404 PMCID: PMC4127243 DOI: 10.1155/2014/893516] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 06/23/2014] [Accepted: 06/25/2014] [Indexed: 11/25/2022]
Abstract
Background. Endogenous cannabinoid anandamide (AEA) protects neurons from oxidative injury in rodent models; however the mechanism of AEA-induced neuroprotection remains to be determined. Activation of neuronal NADPH oxidase 2 (Nox2) contributes to oxidative damage of the brain, and inhibition of Nox2 can attenuate cerebral oxidative stress. We aimed to determine whether the neuronal Nox2 was involved in protection mediated by AEA. Methods. The mouse hippocampal neuron cell line HT22 was exposed to hydrogen peroxide (H2O2) to mimic oxidative injury of neurons. The protective effect of AEA was assessed by measuring cell metabolic activity, apoptosis, lactate dehydrogenase (LDH) release, cellular morphology, intracellular reactive oxygen species (ROS), and antioxidant and oxidant levels and Nox2 expression. Results. HT22 cells exposed to H2O2 demonstrated morphological changes, decreased LDH release, reduced metabolic activity, increased levels of intracellular ROS and oxidized glutathione (GSSG), reduced levels of superoxide dismutase (SOD), and reduced glutathione (GSH) and increased expression of Nox2. AEA prevented these effects, a property abolished by simultaneous administration of CB1 antagonist AM251 or CB1-siRNA. Conclusion. Nox2 inhibition is involved in AEA-induced cytoprotection against oxidative stress through CB1 activation in HT22 cells.
Collapse
|
44
|
Yang F, Wang Z, Wei X, Han H, Meng X, Zhang Y, Shi W, Li F, Xin T, Pang Q, Yi F. NLRP3 deficiency ameliorates neurovascular damage in experimental ischemic stroke. J Cereb Blood Flow Metab 2014; 34:660-7. [PMID: 24424382 PMCID: PMC3982086 DOI: 10.1038/jcbfm.2013.242] [Citation(s) in RCA: 332] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 12/18/2013] [Accepted: 12/19/2013] [Indexed: 01/10/2023]
Abstract
Although the innate immune response to induce postischemic inflammation is considered as an essential step in the progression of cerebral ischemia injury, the role of innate immunity mediator NLRP3 in the pathogenesis of ischemic stroke is unknown. In this study, focal ischemia was induced by middle cerebral artery occlusion in NLRP3(-/-), NOX2(-/-), or wild-type (WT) mice. By magnetic resonance imaging (MRI), Evans blue permeability, and electron microscopic analyses, we found that NLRP3 deficiency ameliorated cerebral injury in mice after ischemic stroke by reducing infarcts and blood-brain barrier (BBB) damage. We further showed that the contribution of NLRP3 to neurovascular damage was associated with an autocrine/paracrine pattern of NLRP3-mediated interleukin-1β (IL-1β) release as evidenced by increased brain microvessel endothelial cell permeability and microglia-mediated neurotoxicity. Finally, we found that NOX2 deficiency improved outcomes after ischemic stroke by mediating NLRP3 signaling. This study for the first time shows the contribution of NLRP3 to neurovascular damage and provides direct evidence that NLRP3 as an important target molecule links NOX2-mediated oxidative stress to neurovascular damage in ischemic stroke. Pharmacological targeting of NLRP3-mediated inflammatory response at multiple levels may help design a new approach to develop therapeutic strategies for prevention of deterioration of cerebral function and for the treatment of stroke.
Collapse
Affiliation(s)
- Fan Yang
- Department of Pharmacology, Shandong University School of Medicine, Jinan, PR China
| | - Ziying Wang
- Department of Pharmacology, Shandong University School of Medicine, Jinan, PR China
| | - Xinbing Wei
- Department of Pharmacology, Shandong University School of Medicine, Jinan, PR China
| | - Huirong Han
- Department of Pharmacology, Shandong University School of Medicine, Jinan, PR China
| | - Xianfang Meng
- Department of Neurobiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Yan Zhang
- Department of Pharmacology, Shandong University School of Medicine, Jinan, PR China
| | - Weichen Shi
- Department of Pharmacology, Shandong University School of Medicine, Jinan, PR China
| | - Fengli Li
- Department of Pharmacology, Shandong University School of Medicine, Jinan, PR China
| | - Tao Xin
- Department of Neurosurgery, Provincial Hospital Affiliated to Shandong University, Jinan, PR China
| | - Qi Pang
- Department of Neurosurgery, Provincial Hospital Affiliated to Shandong University, Jinan, PR China
| | - Fan Yi
- Department of Pharmacology, Shandong University School of Medicine, Jinan, PR China
| |
Collapse
|