1
|
Li T, Li H, Zhang S, Wang Y, He J, Kang J. Transcriptome Sequencing-Based Screening of Key Melatonin-Related Genes in Ischemic Stroke. Int J Mol Sci 2024; 25:11620. [PMID: 39519172 PMCID: PMC11547107 DOI: 10.3390/ijms252111620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/23/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Ischemic stroke (IS) is a complex syndrome of neurological deficits due to stenosis or occlusion of the carotid and vertebral arteries for which there is still no effective treatment. Melatonin, a hormone secreted by the pineal gland, has multiple biological effects, such as antioxidant and anti-inflammatory properties, circadian rhythm regulation, and tissue regeneration, demonstrating potential applications in the treatment of IS. The aim of this study was to investigate key melatonin-regulated genes associated with IS using transcriptome sequencing and bioinformatics analyses and to explore their potential mechanisms of action in the disease process. We obtained gene expression data related to ischemic stroke (IS) from the Gene Expression Omnibus (GEO) database and identified candidate genes using machine learning algorithms. We then assessed the predictive power of these genes using PPI network analysis and diagnostic models. Finally, a series of enrichment analyses identified four key genes: ADM, PTGS2, MMP9, and VCAN. In addition, we determined the mRNA levels of these four key genes in an IS rat model using qPCR and found that all of these genes were significantly upregulated in the IS model compared to the control group, which is consistent with the results of previous analyses. Meanwhile, these genes have biological functions such as regulating vascular tone, participating in the inflammatory response, influencing tissue remodeling, and regulating cell adhesion and proliferation, playing key roles in the pathogenesis of IS. Therefore, we suggest that these four key genes may serve as prospective biomarkers for IS and help predict the risk of developing IS. In conclusion, this study elucidates for the first time the potential role of melatonin in the pathogenesis of IS and lays the foundation for in-depth studies on the functions of these key genes in the pathophysiology of IS and their potential applications in clinical diagnosis and treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Jingsong Kang
- Key Laboratory of Pathobiology, Department of Pathophysiology, Ministry of Education, College of Basical Medical Sciences, Jilin University, 126 Xinmin Street, Changchun 130012, China; (T.L.); (H.L.); (S.Z.); (Y.W.); (J.H.)
| |
Collapse
|
2
|
Schanbacher C, Bieber M, Reinders Y, Cherpokova D, Teichert C, Nieswandt B, Sickmann A, Kleinschnitz C, Langhauser F, Lorenz K. ERK1/2 Activity Is Critical for the Outcome of Ischemic Stroke. Int J Mol Sci 2022; 23:ijms23020706. [PMID: 35054890 PMCID: PMC8776221 DOI: 10.3390/ijms23020706] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/04/2022] [Accepted: 01/05/2022] [Indexed: 12/02/2022] Open
Abstract
Ischemic disorders are the leading cause of death worldwide. The extracellular signal-regulated kinases 1 and 2 (ERK1/2) are thought to affect the outcome of ischemic stroke. However, it is under debate whether activation or inhibition of ERK1/2 is beneficial. In this study, we report that the ubiquitous overexpression of wild-type ERK2 in mice (ERK2wt) is detrimental after transient occlusion of the middle cerebral artery (tMCAO), as it led to a massive increase in infarct volume and neurological deficits by increasing blood–brain barrier (BBB) leakiness, inflammation, and the number of apoptotic neurons. To compare ERK1/2 activation and inhibition side-by-side, we also used mice with ubiquitous overexpression of the Raf-kinase inhibitor protein (RKIPwt) and its phosphorylation-deficient mutant RKIPS153A, known inhibitors of the ERK1/2 signaling cascade. RKIPwt and RKIPS153A attenuated ischemia-induced damages, in particular via anti-inflammatory signaling. Taken together, our data suggest that stimulation of the Raf/MEK/ERK1/2-cascade is severely detrimental and its inhibition is rather protective. Thus, a tight control of the ERK1/2 signaling is essential for the outcome in response to ischemic stroke.
Collapse
Affiliation(s)
- Constanze Schanbacher
- Institute of Pharmacology and Toxicology, University of Würzburg, 97078 Würzburg, Germany;
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44139 Dortmund, Germany; (Y.R.); (C.T.); (A.S.)
| | - Michael Bieber
- Department of Neurology, University Hospital Würzburg, 97080 Würzburg, Germany;
| | - Yvonne Reinders
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44139 Dortmund, Germany; (Y.R.); (C.T.); (A.S.)
| | - Deya Cherpokova
- Institute of Experimental Biomedicine I, University Hospital Würzburg, 97080 Würzburg, Germany; (D.C.); (B.N.)
- Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Christina Teichert
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44139 Dortmund, Germany; (Y.R.); (C.T.); (A.S.)
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine I, University Hospital Würzburg, 97080 Würzburg, Germany; (D.C.); (B.N.)
- Rudolf Virchow Center, University of Würzburg, 97080 Würzburg, Germany
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44139 Dortmund, Germany; (Y.R.); (C.T.); (A.S.)
| | - Christoph Kleinschnitz
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, 45147 Essen, Germany;
| | - Friederike Langhauser
- Department of Neurology and Center for Translational Neuro- and Behavioral Sciences (C-TNBS), University Hospital Essen, 45147 Essen, Germany;
- Correspondence: (F.L.); (K.L.)
| | - Kristina Lorenz
- Institute of Pharmacology and Toxicology, University of Würzburg, 97078 Würzburg, Germany;
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., 44139 Dortmund, Germany; (Y.R.); (C.T.); (A.S.)
- Correspondence: (F.L.); (K.L.)
| |
Collapse
|
3
|
Elastase inhibitor agaphelin protects from acute ischemic stroke in mice by reducing thrombosis, blood-brain barrier damage, and inflammation. Brain Behav Immun 2021; 93:288-298. [PMID: 33401017 PMCID: PMC7979502 DOI: 10.1016/j.bbi.2020.12.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/14/2020] [Accepted: 12/22/2020] [Indexed: 12/23/2022] Open
Abstract
Recently it was shown that the hematophagous salivary gland protein agaphelin exhibits multiple antithrombotic effects without promoting the risk of bleeding. Agaphelin inhibits neutrophil elastase and thereby reduces cathepsin G-induced platelet aggregation. However, it is still unclear, whether pharmacological treatment with agaphelin in brain ischemia is protective and, regarding its bleeding risk, safe. To elucidate this issue, male C57BL/6 mice were subjected to 60 min of transient middle cerebral artery occlusion (tMCAO) and treated with 0.25 mg/kg agaphelin intravenously immediately after tMCAO. On day 1 and 7, infarct volume and functional neurological outcome were assessed by behavioural tests, histochemistry and magnetic resonance imaging. Thrombus formation, intracerebral bleeding risk, blood-brain barrier damage and the local inflammatory response were determined on day 1. This study shows for the first time a protective effect of agaphelin characterized by smaller infarct volume, reduced neurological deficits and reduced animal mortality. This protective effect was associated with reduced local thrombus formation, increased blood-brain barrier integrity and reduced brain inflammatory response. It is essential to mention that the protective effect of agaphelin was not linked to an increased risk of intracerebral bleeding. The promotion of brain tissue survival and inhibition of thromboinflammation identifies agaphelin as a promising treatment option in ischemic stroke, which considering the lack of bleeding risk should potentially be safe.
Collapse
|
4
|
Yu M, Zheng N, Jiang D, Wang L, Zhan Q, Zhao J. Chemokine C-C motif ligand 2 suppressed the growth of human brain astrocytes under Ischemic/hypoxic conditions via regulating ERK1/2 pathway. Brain Inj 2020; 34:1277-1282. [PMID: 32749897 DOI: 10.1080/02699052.2020.1797167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PRIMARY OBJECTIVE Chemokine C-C motif ligand 2 (CCL2) plays a critical role in inflammation-related diseases in the central nervous system (CNS). However, the role of CCL2 in ischemic stroke remains unclear. RESEARCH DESIGN To investigate the role of CCL2 in ischemic stroke, we performed oxygen-glucose deprivation (OGD) on human brain astrocytes. METHODS AND PROCEDURES To assess cell proliferation, the CCK-8 assay was performed. Cell apoptosis was determined using flow cytometry. qRT-PCR and western blotting were utilized to measure gene expression. MAIN OUTCOMES AND RESULTS Our results suggest that CCL2 and its receptor CCR2 are upregulated in OGD cells. Moreover, a CCL2 antibody significantly alleviated the ischemic/hypoxic-induced suppression of growth in human brain astrocytes. Human recombinant protein, CCL2, inhibited the growth of human brain astrocytes under normoxia conditions. These results demonstrate that CCL2 upregulation suppresses the recovery of human brain astrocytes under ischemic/hypoxic conditions. This effect was abolished by the ERK inhibitor PD98059. Therefore, CCL2/CCR2 activation may suppress the growth of human brain astrocytes through enhancing the activity of ERK1/2. CONCLUSIONS Our results not only developed a deeper understanding of the role of CCL2 in human brain astrocytes but also provided novel insight into potential treatments for ischemic stroke.
Collapse
Affiliation(s)
- Min Yu
- Department of Neurology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine , Shanghai, China
| | - Ni Zheng
- Department of Nuclear Medicine, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine , Shanghai, China
| | - Dudu Jiang
- Department of Neurology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine , Shanghai, China
| | - Lijing Wang
- Department of Neurology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine , Shanghai, China
| | - Qing Zhan
- Department of Neurology, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine , Shanghai, China
| | - Jiangmin Zhao
- Department of Radiology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai, China
| |
Collapse
|
5
|
Yong YX, Yang H, Lian J, Xu XW, Han K, Hu MY, Wang HC, Zhou LM. Up-regulated microRNA-199b-3p represses the apoptosis of cerebral microvascular endothelial cells in ischemic stroke through down-regulation of MAPK/ERK/EGR1 axis. Cell Cycle 2019; 18:1868-1881. [PMID: 31204565 DOI: 10.1080/15384101.2019.1632133] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
MicroRNAs (miRNAs) have emerged as key mediators of posttranscriptional gene silencing in both pathogenic and pathological aspects of ischemic stroke biology. Therefore, the purpose of present study was to explore the effect of microRNA-199b-3p (miR-199b-3p) on the cerebral microvascular endothelial cells (CMECs) in middle cerebral artery occlusion-reperfusion (MCAO-R) mice by regulating MAPK/ERK/EGR1 axis. Mice were used to establish MCAO-R models and to measure the expression of miR-199b-3p and the MAPK/ERK/EGR1 axis-related genes. CMECs were extracted from the MCAO-R mice. A series of mimic or inhibitor for miR-199b-3p, or U0126 (an inhibitor for the MAPK/ERK/EGR1 axis) were introduced to treat these CMECs. The levels of miR-199b-3p and MAPK/ERK/EGR1 axis-related genes in tissues and cells were detected. The effects miR-199b-3p on the process of CMECs, including cell viability, cell cycle and cell apoptosis were evaluated. miR-199b-3p expressed poorly in the brain tissues after MCAO-R, along with activated MAPK/ERK/EGR1 axis and increased CMECs apoptosis. CMECs transfected with miR-199b-3p mimics and U0126 manifested with increased cell viability, more cells arrested at the S stage, and inhibited apoptosis of CMECs. In conclusion, these key results demonstrated up-regulated miR-199b-3p could protect mice against ischemic stroke by inhibiting the apoptosis of CMECs through blockade of MAPK/ERK/EGR1 axis.
Collapse
Affiliation(s)
- Ya-Xiong Yong
- a Guizhou Medical University , Guiyang , P. R. China.,b Department of Neurology, the Seventh Affiliated Hospital of Sun Yat-sen University , Shenzhen , P. R. China
| | - Hua Yang
- a Guizhou Medical University , Guiyang , P. R. China.,c Institute of Medical Sciences, Guizhou Medical University , Guiyang , P.R. China.,d Department of Neurosurgery, the Affiliated Hospital of Guizhou Medical University , Guiyang , P. R. China
| | - Jia Lian
- e Department of Neurology, the Seventh Affiliated Hospital of Sun Yat-sen University , Shenzhen , P.R. China
| | - Xiao-Wei Xu
- b Department of Neurology, the Seventh Affiliated Hospital of Sun Yat-sen University , Shenzhen , P. R. China
| | - Ke Han
- b Department of Neurology, the Seventh Affiliated Hospital of Sun Yat-sen University , Shenzhen , P. R. China
| | - Ming-Yi Hu
- b Department of Neurology, the Seventh Affiliated Hospital of Sun Yat-sen University , Shenzhen , P. R. China
| | - Hua-Cheng Wang
- b Department of Neurology, the Seventh Affiliated Hospital of Sun Yat-sen University , Shenzhen , P. R. China
| | - Lie-Min Zhou
- b Department of Neurology, the Seventh Affiliated Hospital of Sun Yat-sen University , Shenzhen , P. R. China
| |
Collapse
|
6
|
Zhao H, Zheng T, Yang X, Fan M, Zhu L, Liu S, Wu L, Sun C. Cryptotanshinone Attenuates Oxygen-Glucose Deprivation/ Recovery-Induced Injury in an in vitro Model of Neurovascular Unit. Front Neurol 2019; 10:381. [PMID: 31057477 PMCID: PMC6482155 DOI: 10.3389/fneur.2019.00381] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 03/28/2019] [Indexed: 12/11/2022] Open
Abstract
Cryptotanshinone (CTs), an active component isolated from the root of Salvia miltiorrhiza (SM), has been shown to exert potent neuroprotective property. We here established an oxygen-glucose deprivation/recovery (OGD/R)-injured Neurovascular Unit (NVU) model in vitro to observe the neuroprotective effects of CTs on cerebral ischemia/reperfusion injury (CIRI), and explore the underlying mechanisms. CTs was observed to significantly inhibit the OGD/R-induced neuronal apoptosis, and decease the activation of Caspase-3 and the degradation of poly-ADP-ribose polymerase (PARP), as well as the increase of Bax/Bcl-2 ratio in neurons under OGD/R condition. The inhibitory effects of CTs on neuron apoptosis were associated with the blocking of mitogen-activated protein kinase (MAPK) signaling pathway. CTs also remarkably ameliorated OGD/R-induced reduction of transepithelial electrical resistance (TEER) values and the increase of transendothelial permeability coefficient (Pe) of sodium fluorescein (SF) by upregulating the expression of ZO-1, Claudin-5, and Occludin in brain microvascular endothelial cells (BMECs), which might be related to the down-regulation of matrix metalloproteinase (MMP)-9 expression. Based on these findings, CTs may play a neuroprotective role in OGD/R injure in NVU models in vitro by inhibiting cell apoptosis and alleviating the damage of blood-brain barrier (BBB).
Collapse
Affiliation(s)
- Hongye Zhao
- Department of Physiology and Key Laboratory of Brain Diseases of Liaoning Province, School of Basic Medical Sciences, Dalian Medical University, Dalian, China.,Department of Physiology, School of Basic Medical Sciences, Qiqihar Medical University, Qiqihar, China
| | - Tiezheng Zheng
- Department of Physiology and Key Laboratory of Brain Diseases of Liaoning Province, School of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiaohan Yang
- Department of Physiology and Key Laboratory of Brain Diseases of Liaoning Province, School of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Ming Fan
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Lingling Zhu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Shuhong Liu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Liying Wu
- Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Changkai Sun
- Department of Physiology and Key Laboratory of Brain Diseases of Liaoning Province, School of Basic Medical Sciences, Dalian Medical University, Dalian, China.,Department of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering & Research Center for the Control Engineering of Translational Precision Medicine, Dalian University of Technology, Dalian, China
| |
Collapse
|
7
|
Hoque A, Williamson NA, Ameen SS, Ciccotosto GD, Hossain MI, Oakhill JS, Ng DCH, Ang CS, Cheng HC. Quantitative proteomic analyses of dynamic signalling events in cortical neurons undergoing excitotoxic cell death. Cell Death Dis 2019; 10:213. [PMID: 30824683 PMCID: PMC6397184 DOI: 10.1038/s41419-019-1445-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/20/2019] [Accepted: 02/01/2019] [Indexed: 12/23/2022]
Abstract
Excitotoxicity, caused by overstimulation or dysregulation of ionotropic glutamate receptors (iGluRs), is a pathological process directing neuronal death in many neurological disorders. The aberrantly stimulated iGluRs direct massive influx of calcium ions into the affected neurons, leading to changes in expression and phosphorylation of specific proteins to modulate their functions and direct their participation in the signalling pathways that induce excitotoxic neuronal death. To define these pathways, we used quantitative proteomic approaches to identify these neuronal proteins (referred to as the changed proteins) and determine how their expression and/or phosphorylation dynamically changed in association with excitotoxic cell death. Our data, available in ProteomeXchange with identifier PXD008353, identified over 100 changed proteins exhibiting significant alterations in abundance and/or phosphorylation levels at different time points (5–240 min) in neurons after glutamate overstimulation. Bioinformatic analyses predicted that many of them are components of signalling networks directing defective neuronal morphology and functions. Among them, the well-known neuronal survival regulators including mitogen-activated protein kinases Erk1/2, glycogen synthase kinase 3 (GSK3) and microtubule-associated protein (Tau), were selected for validation by biochemical approaches, which confirmed the findings of the proteomic analysis. Bioinformatic analysis predicted Protein Kinase B (Akt), c-Jun kinase (JNK), cyclin-dependent protein kinase 5 (Cdk5), MAP kinase kinase (MEK), Casein kinase 2 (CK2), Rho-activated protein kinase (Rock) and Serum/glucocorticoid-regulated kinase 1 (SGK1) as the potential upstream kinases phosphorylating some of the changed proteins. Further biochemical investigation confirmed the predictions of sustained changes of the activation states of neuronal Akt and CK2 in excitotoxicity. Thus, future investigation to define the signalling pathways directing the dynamic alterations in abundance and phosphorylation of the identified changed neuronal proteins will help elucidate the molecular mechanism of neuronal death in excitotoxicity.
Collapse
Affiliation(s)
- Ashfaqul Hoque
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia.,Cell Signalling Research Laboratories, University of Melbourne, Parkville, VIC, 3010, Australia.,Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia.,Metabolic Signalling Laboratory, St. Vincent's Institute for Medical Research, University of Melbourne, Fitzroy, VIC, 3065, Australia
| | - Nicholas A Williamson
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - S Sadia Ameen
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia.,Cell Signalling Research Laboratories, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Giuseppe D Ciccotosto
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, VIC, 3010, Australia
| | - M Iqbal Hossain
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia.,Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Jonathan S Oakhill
- Metabolic Signalling Laboratory, St. Vincent's Institute for Medical Research, University of Melbourne, Fitzroy, VIC, 3065, Australia.,Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, 3000, Australia
| | - Dominic C H Ng
- School of Biomedical Sciences, University of Queensland, St. Lucia, QLD, Australia
| | - Ching-Seng Ang
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Heung-Chin Cheng
- Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, 3010, Australia. .,Cell Signalling Research Laboratories, University of Melbourne, Parkville, VIC, 3010, Australia. .,Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
8
|
Tau exacerbates excitotoxic brain damage in an animal model of stroke. Nat Commun 2017; 8:473. [PMID: 28883427 PMCID: PMC5589746 DOI: 10.1038/s41467-017-00618-0] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 07/13/2017] [Indexed: 02/07/2023] Open
Abstract
Neuronal excitotoxicity induced by aberrant excitation of glutamatergic receptors contributes to brain damage in stroke. Here we show that tau-deficient (tau−/−) mice are profoundly protected from excitotoxic brain damage and neurological deficits following experimental stroke, using a middle cerebral artery occlusion with reperfusion model. Mechanistically, we show that this protection is due to site-specific inhibition of glutamate-induced and Ras/ERK-mediated toxicity by accumulation of Ras-inhibiting SynGAP1, which resides in a post-synaptic complex with tau. Accordingly, reducing SynGAP1 levels in tau−/− mice abolished the protection from pharmacologically induced excitotoxicity and middle cerebral artery occlusion-induced brain damage. Conversely, over-expression of SynGAP1 prevented excitotoxic ERK activation in wild-type neurons. Our findings suggest that tau mediates excitotoxic Ras/ERK signaling by controlling post-synaptic compartmentalization of SynGAP1. Excitotoxicity contributes to neuronal injury following stroke. Here the authors show that tau promotes excitotoxicity by a post-synaptic mechanism, involving site-specific control of ERK activation, in a mouse model of stroke.
Collapse
|
9
|
Yang CH, Yen TL, Hsu CY, Thomas PA, Sheu JR, Jayakumar T. Multi-Targeting Andrographolide, a Novel NF-κB Inhibitor, as a Potential Therapeutic Agent for Stroke. Int J Mol Sci 2017; 18:ijms18081638. [PMID: 28749412 PMCID: PMC5578028 DOI: 10.3390/ijms18081638] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 07/24/2017] [Accepted: 07/26/2017] [Indexed: 12/13/2022] Open
Abstract
A key focus in the field of drug discovery has been motivated by the neuroprotection of natural compounds. Cerebral ischemia is a multifaceted pathological process with a series of mechanisms, and a perspective for the development of neuroprotectants from traditional herbal medicine or natural products is a promising treatment for this disease. Natural compounds with the effects of anti-oxidation, anti-inflammation, anti-apoptosis, and neurofunctional regulation exhibit therapeutic effects on experimental ischemic brain injury. Conferring to the pharmacological mechanisms underlying neuroprotection, a study found that androgapholide, a diterpene lactone compound, exhibits varying degrees of neuroprotective activities in both in vitro and in vivo experimental models of stroke. The neuroprotective mechanisms of andrographolide are suggested as: (I) increasing nuclear factor E2-related factor 2-heme oxygenase (Nrf2-HO-1) expression through p38-mitogen activated protein kinase (MAPK) regulation, (II) inducing cerebral endothelial cells (CEC) apoptosis and caspase-3 activation, (III) down regulating Bax, inducible nitric oxide synthase (iNOS), and (IV) inhibiting hydroxyl radical (OH−) formation, and activating transcription factor NF-κB signaling pathways. Recently, several researchers have also been trying to unveil the principal mechanisms involved in the neuroprotective effects of andrographolide. Therefore, this review aims to summarize an overview on the neuroprotective effects of andrographolide and exemplifies the essential mechanisms involved. This paper can provide information that andrographolide drug discovery may be a promising strategy for the development of a novel class of neuroprotective drug.
Collapse
Affiliation(s)
- Chih-Hao Yang
- Department of Pharmacology, Taipei Medical University, Taipei 110, Taiwan.
| | - Ting-Lin Yen
- Department of Pharmacology, Taipei Medical University, Taipei 110, Taiwan.
- Division of Cardiology, Department of Internal Medicine, Cathay General Hospital, Taipei 200, Taiwan.
| | - Chia-Yuan Hsu
- Department of Life Science, College of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan.
| | - Philip-Aloysius Thomas
- Department of Ocular Microbiology, Institute of Ophthalmology, Joseph Eye Hospital, Tiruchirappalli 620001, Tamil Nadu, India.
| | - Joen-Rong Sheu
- Department of Pharmacology, Taipei Medical University, Taipei 110, Taiwan.
- Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | | |
Collapse
|
10
|
Wang T, Liao Y, Sun Q, Tang H, Wang G, Zhao F, Jin Y. Upregulation of Matrix Metalloproteinase-9 in Primary Cultured Rat Astrocytes Induced by 2-Chloroethanol Via MAPK Signal Pathways. Front Cell Neurosci 2017; 11:218. [PMID: 28769771 PMCID: PMC5516094 DOI: 10.3389/fncel.2017.00218] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 07/06/2017] [Indexed: 01/25/2023] Open
Abstract
2-Chloroethanol (2-CE) is one of the reactive metabolites of 1,2-DCE in vivo, which might contribute to brain edema formation induced by 1,2-dichloroethane (1,2-DCE) poisoning. Thus, the purpose of this study was to explore the roles of mitogen-activated protein kinase (MAPK) signal pathways in upregulation of matrix metalloproteinase-9 (MMP-9) in 2-CE exposed rat astrocytes. Expression of p38 MAPK (p38), extracellular signal regulated protein kinase (ERK), c-Jun N-terminal kinase (JNK) and MMP-9 at both protein and gene levels in rat astrocytes were determined using western blot and real-time RT-PCR methods. The results showed that both protein and mRNA levels of MMP-9 in 2-CE exposed astrocytes significantly increased. Meanwhile, protein levels of phosphorylated p38 (p-p38), ERK1/2 (p-ERK1/2) and JNK1/2 (p-JNK1/2) in 2-CE exposed astrocytes also significantly increased. In addition, both protein and mRNA levels of MMP-9 significantly decreased in response to reduced protein levels of p-p38, p-ERK1/2 and p-JNK1/2 achieved by supplement with their specific inhibitors, indicating that activation of MAPK signal pathways might play an important role in upregulation of MMP-9 expression at the transcriptional level in 2-CE exposed astrocytes. Furthermore, since pretreatment of n-acetyl-l-cysteine (NAC), a powerful antioxidant amino acid, could attenuate the elevated levels of MMP-9, p-p38, p-ERK2 and p-JNK1/2 in 2-CE exposed astrocytes, activation of MAPK signal pathways in 2-CE exposed astrocytes could be mediated partially by reactive oxygen species (ROS), which was most likely generated in the metabolism of 2-CE.
Collapse
Affiliation(s)
- Tong Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| | - Yingjun Liao
- Department of Physiology, China Medical UniversityShenyang, China
| | - Qi Sun
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| | - Hongge Tang
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| | - Gaoyang Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| | - Fenghong Zhao
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| | - Yaping Jin
- Department of Occupational and Environmental Health, School of Public Health, China Medical UniversityShenyang, China
| |
Collapse
|
11
|
Sun Q, Liao Y, Wang T, Tang H, Wang G, Zhao F, Jin Y. 2-Chloroethanol Induced Upregulation of Matrix Metalloproteinase-2 in Primary Cultured Rat Astrocytes Via MAPK Signal Pathways. Front Neurosci 2017; 10:593. [PMID: 28101000 PMCID: PMC5209348 DOI: 10.3389/fnins.2016.00593] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Accepted: 12/12/2016] [Indexed: 01/10/2023] Open
Abstract
This study was to explore the mechanisms underlying 1,2-dichloroethane (1,2-DCE) induced brain edema by focusing on alteration of matrix metalloproteinase-2 (MMP-2) in rat astrocytes induced by 2-chloroethanol (2-CE), an intermediate metabolite of 1,2-DCE in vivo. Protein and mRNA levels of MMP-2, and the phosphorylated protein levels of p38 MAPK (p-p38), extracellular signal regulated protein kinase (p-ERK1/2) and c-Jun N-terminal kinase (p-JNK1/2) in astrocytes were examined by immunostaining, western blot or real-time RT-PCR analysis. Findings from this study disclosed that protein levels of MMP-2 were upregulated by 2-CE in astrocytes. Meanwhile, protein levels of p-p38, p-ERK1/2 and p-JNK1/2 were also increased apparently in the cells treated with 2-CE. Moreover, pretreatment of astrocytes with SB202190 (inhibitor of p38 MAPK), U0126 (inhibitor of ERK1/2) or SP600125 (inhibitor of JNK1/2) could suppress the upregulated expression of p-p38, p-ERK1/2, and p-JNK1/2. In response to suppressed protein levels of p-p38 and p-JNK1/2, the protein levels of MMP-2 also decreased significantly, indicating that activation of MAPK signal pathways were involved in the mechanisms underlying 2-CE-induced upregulation of MMP-2 expression.
Collapse
Affiliation(s)
- Qi Sun
- Department of Occupational and Environmental Health, School of Public Health, China Medical University Shenyang, China
| | - Yingjun Liao
- Department of Physiology, China Medical University Shenyang, China
| | - Tong Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University Shenyang, China
| | - Hongge Tang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University Shenyang, China
| | - Gaoyang Wang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University Shenyang, China
| | - Fenghong Zhao
- Department of Occupational and Environmental Health, School of Public Health, China Medical University Shenyang, China
| | - Yaping Jin
- Department of Occupational and Environmental Health, School of Public Health, China Medical University Shenyang, China
| |
Collapse
|
12
|
Sun J, Nan G. The Mitogen-Activated Protein Kinase (MAPK) Signaling Pathway as a Discovery Target in Stroke. J Mol Neurosci 2016; 59:90-8. [PMID: 26842916 DOI: 10.1007/s12031-016-0717-8] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 01/12/2016] [Indexed: 01/08/2023]
Abstract
Protein kinases are critical modulators of a variety of intracellular and extracellular signal transduction pathways, and abnormal phosphorylation events can contribute to disease progression in a variety of diseases. As a result, protein kinases have emerged as important new drug targets for small molecule therapeutics. The mitogen-activated protein kinase (MAPK) signaling pathway transmits signals from the cell membrane to the nucleus in response to a variety of different stimuli. Because this pathway controls a broad spectrum of cellular processes, including growth, inflammation, and stress responses, it is accepted as a therapeutic target for cancer and peripheral inflammatory disorders. There is also increasing evidence that MAPK is an important regulator of ischemic and hemorrhagic cerebral vascular disease, raising the possibility that it might be a drug discovery target for stroke. In this review, we discuss the MAPK signaling pathway in association with its activation in stroke-induced brain injury.
Collapse
Affiliation(s)
- Jing Sun
- Department of Neurology, China-Japan Union Hospital of Jilin University, No. 126 Xiantai Street, Changchun, 130000, Jilin, China
| | - Guangxian Nan
- Department of Neurology, China-Japan Union Hospital of Jilin University, No. 126 Xiantai Street, Changchun, 130000, Jilin, China.
| |
Collapse
|
13
|
Kargl J, Andersen L, Hasenöhrl C, Feuersinger D, Stančić A, Fauland A, Magnes C, El-Heliebi A, Lax S, Uranitsch S, Haybaeck J, Heinemann A, Schicho R. GPR55 promotes migration and adhesion of colon cancer cells indicating a role in metastasis. Br J Pharmacol 2015; 173:142-54. [PMID: 26436760 DOI: 10.1111/bph.13345] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 08/31/2015] [Accepted: 09/24/2015] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND AND PURPOSE Tumour cell migration and adhesion constitute essential features of metastasis. G-protein coupled receptor 55 (GPR55), a lysophospholipid receptor, has been shown to play an important role in carcinogenesis. Here, we investigated the involvement of GPR55 in migration and metastasis of colon cancer cells. EXPERIMENTAL APPROACH Adhesion and migration assays using the highly metastatic colon cancer cell line HCT116 and an in vivo assay of liver metastasis were performed. The GPR55 antagonist CID16020046, cannabidiol, a putative GPR55 antagonist and GPR55 siRNA were used to block GPR55 activity in HCT116 colon cancer cells. KEY RESULTS HCT116 cells showed a significant decrease in adhesion to endothelial cells and in migration after blockade with CID16020046 or cannabidiol. The inhibitory effects of CID16020046 or cannabidiol were averted by GPR55 siRNA knock down in cancer cells. The integrity of endothelial cell monolayers was increased after pretreatment of HCT116 cells with the antagonists or after GPR55 siRNA knockdown while pretreatment with lysophosphatidylinositol (LPI), the endogenous ligand of GPR55, decreased integrity of the monolayers. LPI also induced migration in GPR55 overexpressing HCT116 cells that was blocked by GPR55 antagonists. In a mouse model of metastasis, the arrest of HCT116 cancer cells in the liver was reduced after treatment with CID16020046 or cannabidiol. Increased levels of LPI (18:0) were found in colon cancer patients when compared with healthy individuals. CONCLUSIONS AND IMPLICATIONS GPR55 is involved in the migratory behaviour of colon carcinoma cells and may serve as a pharmacological target for the prevention of metastasis. © 2015 The British Pharmacological Society.
Collapse
Affiliation(s)
- J Kargl
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - L Andersen
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - C Hasenöhrl
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - D Feuersinger
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - A Stančić
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - A Fauland
- HEALTH - Institute for Biomedicine and Health Sciences, Joanneum Research Forschungsgesellschaft m.b.H., Graz, Austria
| | - C Magnes
- HEALTH - Institute for Biomedicine and Health Sciences, Joanneum Research Forschungsgesellschaft m.b.H., Graz, Austria
| | - A El-Heliebi
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria.,Biobank Graz, Medical University of Graz, Graz, Austria
| | - S Lax
- Department of Pathology, General Hospital Graz West, Graz, Austria
| | - S Uranitsch
- Department of Surgery, St John of God Hospital Graz, Graz, Austria
| | - J Haybaeck
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - A Heinemann
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| | - R Schicho
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Graz, Austria
| |
Collapse
|
14
|
Chuang DY, Cui J, Simonyi A, Engel VA, Chen S, Fritsche KL, Thomas AL, Applequist WL, Folk WR, Lubahn DB, Sun AY, Sun GY, Gu Z. Dietary Sutherlandia and elderberry mitigate cerebral ischemia-induced neuronal damage and attenuate p47phox and phospho-ERK1/2 expression in microglial cells. ASN Neuro 2014; 6:6/6/1759091414554946. [PMID: 25324465 PMCID: PMC4271764 DOI: 10.1177/1759091414554946] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Sutherlandia (Sutherlandia frutescens) and elderberry
(Sambucus spp.) are used to promote health and for treatment of a
number of ailments. Although studies with cultured cells have demonstrated antioxidative
and anti-inflammatory properties of these botanicals, little is known about their ability
to mitigate brain injury. In this study, C57BL/6 J male mice were fed AIN93G diets without
or with Sutherlandia or American elderberry for 2 months prior to a 30-min global cerebral
ischemia induced by occlusion of the bilateral common carotid arteries (BCCAs), followed
by reperfusion for 3 days. Accelerating rotarod assessment at 24 h after BCCA occlusion
showed amelioration of sensorimotor impairment in the mice fed the supplemented diets as
compared with the ischemic mice fed the control diet. Quantitative digital pathology
assessment of brain slides stained with cresyl violet at 3 days after ischemia/reperfusion
(I/R) revealed significant reduction in neuronal cell death in both dietary groups.
Immunohistochemical staining for ionized calcium-binding adapter molecule-1 demonstrated
pronounced activation of microglia in the hippocampus and striatum in the ischemic brains
3 days after I/R, and microglial activation was significantly reduced in animals fed
supplemented diets. Mitigation of microglial activation by the supplements was further
supported by the decrease in expression of p47phox, a cytosolic subunit of NADPH oxidase,
and phospho-ERK1/2, a mitogen-activated protein kinase known to mediate a number of
cytoplasmic processes including oxidative stress and neuroinflammatory responses. These
results demonstrate neuroprotective effect of Sutherlandia and American elderberry
botanicals against oxidative and inflammatory responses to cerebral I/R.
Collapse
Affiliation(s)
- Dennis Y Chuang
- Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO, USA Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA Center for Botanical Interaction Studies, University of Missouri, Columbia, MO, USA
| | - Jiankun Cui
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA Center for Botanical Interaction Studies, University of Missouri, Columbia, MO, USA Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, USA Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| | - Agnes Simonyi
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA Center for Botanical Interaction Studies, University of Missouri, Columbia, MO, USA Department of Biochemistry, University of Missouri, Columbia, MO, USA
| | - Victoria A Engel
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, USA
| | - Shanyan Chen
- Interdisciplinary Neuroscience Program, University of Missouri, Columbia, MO, USA Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, USA
| | - Kevin L Fritsche
- Center for Botanical Interaction Studies, University of Missouri, Columbia, MO, USA Department of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Andrew L Thomas
- Center for Botanical Interaction Studies, University of Missouri, Columbia, MO, USA Southwest Research Center, University of Missouri, Mt. Vernon, MO, USA
| | - Wendy L Applequist
- Center for Botanical Interaction Studies, University of Missouri, Columbia, MO, USA Missouri Botanical Garden, St. Louis, MO, USA
| | - William R Folk
- Center for Botanical Interaction Studies, University of Missouri, Columbia, MO, USA Department of Biochemistry, University of Missouri, Columbia, MO, USA
| | - Dennis B Lubahn
- Center for Botanical Interaction Studies, University of Missouri, Columbia, MO, USA Department of Biochemistry, University of Missouri, Columbia, MO, USA Department of Animal Sciences, University of Missouri, Columbia, MO, USA
| | - Albert Y Sun
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA Center for Botanical Interaction Studies, University of Missouri, Columbia, MO, USA Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, USA
| | - Grace Y Sun
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA Center for Botanical Interaction Studies, University of Missouri, Columbia, MO, USA Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, USA Department of Biochemistry, University of Missouri, Columbia, MO, USA
| | - Zezong Gu
- Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA Center for Botanical Interaction Studies, University of Missouri, Columbia, MO, USA Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, MO, USA Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA
| |
Collapse
|