1
|
Wang L, Zhang J, Zhao Y, Li J, Lu X, Song J, Zhang L, Niu Q. Nuclear factor kappa B (NF-κB) participates in the aluminum-induced down-regulation of miR29a/b1. J Trace Elem Med Biol 2023; 80:127309. [PMID: 37801786 DOI: 10.1016/j.jtemb.2023.127309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 08/11/2023] [Accepted: 09/20/2023] [Indexed: 10/08/2023]
Abstract
BACKGROUND Studies have shown that aluminum (Al) is one of the environmental risk factors leading to Alzheimer's disease (AD), and Al exposure can cause elevated levels of BACE1mRNA, β-secretase (BACE1), and amyloid beta (Aβ) in vivo and in vitro. Previous studies by our research group have shown that this is partly caused by the negative regulation of BACE1 by miRNA29a/b1 (miR29a/b1). Despite the observed the role of nuclear factor kappa B (NF-κB) on many miRNAs, the upstream regulation of NF-κB protein on miR29 remains poorly understood. The purpose of this study was to better define the relationship between NF-κB and miR29a/b1 and the potentially relevant signaling pathways. METHODS On the one hand, we constructed the animal model of Al exposure by the intraperitoneal injection of aluminum-maltolate (Al(mal)3) in rats. Conversely, NF- κB inhibitors were added to adrenal phaeochromocytoma (PC12) cells exposed to Al(mal)3. RESULTS We verified that NF-κB shows an increasing trend with Al accumulation in the brain of rats, which is accompanied by a downward trend of miR29a/b1. Notably, the suppression of NF-κB significantly increased miR29a/b1 and affected the expression of BACE1mRNA and downstream proteins. CONCLUSION Al-induced NF-κB can negatively regulate the expression of miR29a/b1, which then significantly enhances the expression of BACE1 and Aβ plaques.
Collapse
Affiliation(s)
- Linping Wang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; NHC Key Laboratory of Pneumoconiosis (Shanxi), Medical University, Taiyuan, 030001 Shanxi Province, China
| | - Jingqi Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; NHC Key Laboratory of Pneumoconiosis (Shanxi), Medical University, Taiyuan, 030001 Shanxi Province, China
| | - Yue Zhao
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; NHC Key Laboratory of Pneumoconiosis (Shanxi), Medical University, Taiyuan, 030001 Shanxi Province, China
| | - Juan Li
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; NHC Key Laboratory of Pneumoconiosis (Shanxi), Medical University, Taiyuan, 030001 Shanxi Province, China
| | - Xiaoting Lu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; NHC Key Laboratory of Pneumoconiosis (Shanxi), Medical University, Taiyuan, 030001 Shanxi Province, China
| | - Jing Song
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; NHC Key Laboratory of Pneumoconiosis (Shanxi), Medical University, Taiyuan, 030001 Shanxi Province, China
| | - Ling Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; NHC Key Laboratory of Pneumoconiosis (Shanxi), Medical University, Taiyuan, 030001 Shanxi Province, China
| | - Qiao Niu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, China; Key Lab of Environmental Hazard and Health of Shanxi Province, Shanxi Medical University, China; NHC Key Laboratory of Pneumoconiosis (Shanxi), Medical University, Taiyuan, 030001 Shanxi Province, China.
| |
Collapse
|
2
|
Han EJ, Zhang C, Kim HS, Kim JY, Park SM, Jung WK, Ahn G, Cha SH. Sargachromenol Isolated from Sargassum horneri Attenuates Glutamate-Induced Neuronal Cell Death and Oxidative Stress through Inhibition of MAPK/NF-κB and Activation of Nrf2/HO-1 Signaling Pathway. Mar Drugs 2022; 20:710. [PMID: 36421988 PMCID: PMC9695719 DOI: 10.3390/md20110710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 10/29/2023] Open
Abstract
Oxidative stress-induced neuronal cell loss is considered to be the major mechanism underlying the pathogenesis of neurodegenerative diseases, which could be induced by a high concentration of glutamate. In this study, sargachromenol (SC) was isolated from a marine brown seaweed Sargassum horneri (S. horneri) and its neuroprotective effects against glutamate-induced oxidative stress in HT22 cells were investigated. An MTT assay was applied to assess the cytotoxicity of the SC, and the efficacies of SC were determined by flow cytometry, an analysis of ROS production, quantitative Real-Time PCR, and the Western blot assay. Our results showed that the pretreatment of SC reduced glutamate-induced apoptosis in HT22 cells via inhibiting the sub-G1 population, DNA fragmentation, and nuclear condensation, as well as up-regulating anti-apoptotic protein (Bcl-2) and down-regulating apoptotic proteins (Bax, p53, cleaved-PARP, caspase-3, caspase-9, and cytochrome c). Additionally, SC attenuated glutamate-induced oxidative stress by suppressing mitogen-activated protein kinases (MAPKs;ERK, JNK, and p38) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling (IκBα and NF-κB p65), while activating nuclear factor erythroid-2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) signaling (Nrf2; HO-1, and NQO-1). Our results suggest that SC could be used as a pharmacological candidate for the prevention and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Eui-Jeong Han
- Department of Marine Bio-Food Sciences, Chonnam National University, Yeosu 59626, Korea
| | - Chunying Zhang
- Department of Marine Bio and Medical Sciences, Hanseo University, Seosan-si 32158, Korea
| | - Hyun-Soo Kim
- National Marine Biodiversity Institute of Korea, Seocheon-kun 33662, Korea
| | - Ji-Yul Kim
- National Marine Biodiversity Institute of Korea, Seocheon-kun 33662, Korea
| | - Sang-Muyn Park
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Korea
| | - Won-Kyo Jung
- Research Center for Marine Integrated Bionics Technology and Marine Integrated Biomedical Technology Center, Pukyong National University, Busan 48513, Korea
- Department of Biomedical Engineering, New Senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan 48513, Korea
| | - Ginnae Ahn
- Department of Marine Bio-Food Sciences, Chonnam National University, Yeosu 59626, Korea
| | - Seon-Heui Cha
- Department of Marine Bio and Medical Sciences, Hanseo University, Seosan-si 32158, Korea
| |
Collapse
|
3
|
Jover-Mengual T, Hwang JY, Byun HR, Court-Vazquez BL, Centeno JM, Burguete MC, Zukin RS. The Role of NF-κB Triggered Inflammation in Cerebral Ischemia. Front Cell Neurosci 2021; 15:633610. [PMID: 34040505 PMCID: PMC8141555 DOI: 10.3389/fncel.2021.633610] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 04/12/2021] [Indexed: 12/17/2022] Open
Abstract
Cerebral ischemia is a devastating disease that affects many people worldwide every year. The neurodegenerative damage as a consequence of oxygen and energy deprivation, to date, has no known effective treatment. The ischemic insult is followed by an inflammatory response that involves a complex interaction between inflammatory cells and molecules which play a role in the progression towards cell death. However, there is presently a matter of controversy over whether inflammation could either be involved in brain damage or be a necessary part of brain repair. The inflammatory response is triggered by inflammasomes, key multiprotein complexes that promote secretion of pro-inflammatory cytokines. An early event in post-ischemic brain tissue is the release of certain molecules and reactive oxygen species (ROS) from injured neurons which induce the expression of the nuclear factor-kappaB (NF-κB), a transcription factor involved in the activation of the inflammasome. There are conflicting observations related to the role of NF-κB. While some observe that NF-κB plays a damaging role, others suggest it to be neuroprotective in the context of cerebral ischemia, indicating the need for additional investigation. Here we discuss the dual role of the major inflammatory signaling pathways and provide a review of the latest research aiming to clarify the relationship between NF-κB mediated inflammation and neuronal death in cerebral ischemia.
Collapse
Affiliation(s)
- Teresa Jover-Mengual
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States.,Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe-Universidad de Valencia, Valencia, Spain.,Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| | - Jee-Yeon Hwang
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States.,Department of Pharmacology, Creighton University School of Medicine, Omaha, NE, United States
| | - Hyae-Ran Byun
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| | - Brenda L Court-Vazquez
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| | - José M Centeno
- Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| | - María C Burguete
- Unidad Mixta de Investigación Cerebrovascular, Instituto de Investigación Sanitaria La Fe-Universidad de Valencia, Valencia, Spain.,Departamento de Fisiología, Universidad de Valencia, Valencia, Spain
| | - R Suzanne Zukin
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, United States
| |
Collapse
|
4
|
Acute remote ischemic preconditioning alleviates free radical injury and inflammatory response in cerebral ischemia/reperfusion rats. Exp Ther Med 2019; 18:1953-1960. [PMID: 31410157 PMCID: PMC6676222 DOI: 10.3892/etm.2019.7797] [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: 01/19/2018] [Accepted: 08/31/2018] [Indexed: 12/12/2022] Open
Abstract
Remote ischemic preconditioning (IPreC) is an effective strategy to defend against cerebral ischemia/reperfusion (IR) injury; however, its mechanisms remain to be elucidated. The aim of the present study was to investigate the effect of IPreC on brain tissue following cerebral ischemia, as well as the underlying mechanisms. Adult male Sprague-Dawley rats were treated with IPreC for 72 h prior to the induction of transient cerebral ischemia and reperfusion. The results demonstrated that IPreC reduced the area of cerebral infarction in the IR rats by 2,3,5-triphenyl-tetrazolium chloride staining. In addition, cell apoptosis was markedly suppressed by IPreC with an increased expression of B-cell lymphoma 2 (Bcl-2)/Bcl-2-associatd X protein using Terminal deoxynucleotidyl-transferase-mediated dUTP nick end labeling assay and western blot analysis. IR induced a decrease in the level of superoxide dismutase, and IPreC significantly suppressed increased levels of malondialdehyde, lactate dehydrogenase and nitric oxide. The expression of CD11b and CD18 was markedly inhibited by IpreC unsing flow cytometry. Furthermore, IPreC markedly decreased the release of pro-inflammatory factors interleukin (IL)-6 and IL-1β, and enhanced the level of anti-inflammatory factors (IL-10 and IL-1 receptor antagonist) by ELISA assay. Finally, IPreC reduced the levels of transforming growth factor-β-activated kinase 1, phosphorylated-P65/P65, and tumor necrosis factor-α, indicating that the nuclear factor-κB pathway was involved in IPreC-mediated protection against cerebral ischemia. Taken together, the results suggested that IPreC decreased ischemic brain injury through alleviating free radical injury and the inflammatory response in cerebral IR rats.
Collapse
|
5
|
Chen C, Chu SF, Ai QD, Zhang Z, Guan FF, Wang SS, Dong YX, Zhu J, Jian WX, Chen NH. CKLF1 Aggravates Focal Cerebral Ischemia Injury at Early Stage Partly by Modulating Microglia/Macrophage Toward M1 Polarization Through CCR4. Cell Mol Neurobiol 2019; 39:651-669. [PMID: 30982091 PMCID: PMC11462892 DOI: 10.1007/s10571-019-00669-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 03/12/2019] [Indexed: 10/27/2022]
Abstract
CKLF1 is a chemokine with increased expression in ischemic brain, and targeting CKLF1 has shown therapeutic effects in cerebral ischemia model. Microglia/macrophage polarization is a mechanism involved in poststroke injury expansion. Considering the quick and obvious response of CKLF1 and expeditious evolution of stroke lesions, we focused on the effects of CKLF1 on microglial/macrophage polarization at early stage of ischemic stroke (IS). The present study is to investigate the CKLF1-mediated expression of microglia/macrophage phenotypes in vitro and in vivo, discussing the involved pathway. Primary microglia culture was used in vitro, and mice transient middle cerebral artery occlusion (MCAO) model was adopted to mimic IS. CKLF1 was added to the primary microglia for 24 h, and we found that CKLF1 modulated primary microglia skew toward M1 phenotype. In mice transient IS model, CKLF1 was stereotactically microinjected to the lateral ventricle of ischemic hemisphere. CKLF1 aggravated ischemic injury, accompanied by promoting microglia/macrophage toward M1 phenotypic polarization. Increased expression of pro-inflammatory cytokines and decreased expression of anti-inflammatory cytokines were observed in mice subjected to cerebral ischemia and administrated with CKLF1. CKLF1-/- mice were used to confirm the effects of CKLF1. CKLF1-/- mice showed lighter cerebral damage and decreased M1 phenotype of microglia/macrophage compared with the WT control subjected to cerebral ischemia. Moreover, NF-κB activation enhancement was detected in CKLF1 treatment group. Our results demonstrated that CKLF1 is an important mediator that skewing microglia/macrophage toward M1 phenotype at early stage of cerebral ischemic injury, which further deteriorates followed inflammatory response, contributing to early expansion of cerebral ischemia injury. Targeting CKLF1 may be a novel way for IS therapy.
Collapse
Affiliation(s)
- Chen Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Shi-Feng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Qi-Di Ai
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces & Hunan University of Chinese Medicine First-class Disciple Construction Project of Chinese Materia Medica, Changsha, 410208, China
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Fei-Fei Guan
- Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Science, NHFPC, Peking Union Medicine College and Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Sha-Sha Wang
- School of Basic Medicine, Shanxi University of Traditional Chinese Medicine, Taiyuan, 030619, China
| | - Yi-Xiao Dong
- Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Jie Zhu
- Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100050, China
| | - Wen-Xuan Jian
- DME Center, Clinical Pharmacology Institute, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Nai-Hong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces & Hunan University of Chinese Medicine First-class Disciple Construction Project of Chinese Materia Medica, Changsha, 410208, China.
| |
Collapse
|
6
|
Chen C, Ai Q, Chu S, Zhang Z, Zhou X, Luo P, Liu Y, Chen N. IMM-H004 protects against oxygen-glucose deprivation/reperfusion injury to BV2 microglia partly by modulating CKLF1 involved in microglia polarization. Int Immunopharmacol 2019; 70:69-79. [PMID: 30785093 DOI: 10.1016/j.intimp.2019.02.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/28/2018] [Accepted: 02/06/2019] [Indexed: 01/07/2023]
Abstract
BACKGROUND IMM-H004 is a novel compound that has been shown to protect against cerebral ischemia/reperfusion injury in our previous works. Chemokine-like factor 1 (CKLF1) is a chemokine that exhibits increased expression in the ischemic brain. Dysregulation of microglia polarization dynamics is a mechanism of injury expansion poststroke. PURPOSES The aim of present study was to investigate the effects of IMM-H004 on cell viability and microglia phenotypes in BV2 microglia suffering from oxygen-glucose deprivation/reperfusion and discussing the involvement of CKLF1 and possible mechanisms. RESULTS IMM-H004 protected BV2 microglia from oxygen-glucose deprivation/reperfusion-induced toxicity. We found that the expression of CKLF1 was increased in BV2 microglia with oxygen-glucose deprivation/reperfusion, and IMM-H004 decreased this specially increased expression. Moreover, oxygen-glucose deprivation/reperfusion induced the BV2 microglia to polarize toward an M1 phenotype, and IMM-H004 modulated the polarization shift from the M1 phenotype and skewed toward the M2 phenotype, followed by suppressing the excessive inflammatory response and improving recovery. CKLF1 modulated BV2 microglia toward M1 polarization and induced an inflammatory response. By using receptor inhibitors, we found that OGD/R induced microglia polarization partly through CC chemokine receptor 4. Furthermore, the Co-IP assay showed that IMM-H004 decreased the amount of CKLF1 binding to CC chemokine receptor 4 in the BV2 microglia oxygen-glucose deprivation/reperfusion model. CONCLUSIONS IMM-H004 protects BV2 microglia against oxygen-glucose deprivation/reperfusion injury partly by modulating microglia polarization and further regulating the inflammatory response. The CKLF1/CCR4 axis may be involved in the protective effects of IMM-H004 modulating microglia polarization.
Collapse
Affiliation(s)
- Chen Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Qidi Ai
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China; Hunan University of Traditional Chinese Medicine, Changsha 410208, China
| | - Shifeng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Xin Zhou
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China
| | - Piao Luo
- Hunan University of Traditional Chinese Medicine, Changsha 410208, China
| | - Yingjiao Liu
- Hunan University of Traditional Chinese Medicine, Changsha 410208, China
| | - Naihong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100050, China; Hunan University of Traditional Chinese Medicine, Changsha 410208, China.
| |
Collapse
|
7
|
Ai Q, Chen C, Chu S, Luo Y, Zhang Z, Zhang S, Yang P, Gao Y, Zhang X, Chen N. IMM-H004 Protects against Cerebral Ischemia Injury and Cardiopulmonary Complications via CKLF1 Mediated Inflammation Pathway in Adult and Aged Rats. Int J Mol Sci 2019; 20:E1661. [PMID: 30987181 PMCID: PMC6480569 DOI: 10.3390/ijms20071661] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/31/2019] [Accepted: 04/02/2019] [Indexed: 12/30/2022] Open
Abstract
(1) Background: Chemokine-like factor 1 (CKLF1) is a chemokine with potential to be a target for stroke therapy. Compound IMM-H004 is a novel coumarin derivative screened from a CKLF1/C-C chemokine receptor type 4 (CCR4) system and has been reported to improve cerebral ischemia/reperfusion injury. This study aims to investigate the protective effects of IMM-H004 on cerebral ischemia injury and its infectious cardiopulmonary complications in adult and aged rats from the CKLF1 perspective. (2) Methods: The effects of IMM-H004 on the protection was determined by 2,3,5-triphenyltetrazolium chloride (TTC) staining, behavior tests, magnetic resonance imaging (MRI) scans, enzyme-linked immunosorbent assay (ELISA), Nissl staining, histo-pathological examination, and cardiopulmonary function detection. Immunohistological staining, immunofluorescence staining, quantitative real-time PCR (qPCR), and western blotting were used to elucidate the underlying mechanisms. (3) Results: IMM-H004 protects against cerebral ischemia induced brain injury and its cardiopulmonary complications, inhibiting injury, and inflammation through CKLF1-dependent anti-inflammation pathway in adult and aged rats. IMM-H004 downregulates the amount of CKLF1, suppressing the followed inflammatory response, and further protects the damaged organs from ischemic injury. (4) Conclusions: The present study suggested that the protective mechanism of IMM-H004 is dependent on CKLF1, which will lead to excessive inflammatory response in cerebral ischemia. IMM-H004 could also be a therapeutic agent in therapy for ischemic stroke and cardiopulmonary complications in the aged population.
Collapse
Affiliation(s)
- Qidi Ai
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces & College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China.
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Chen Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Shifeng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yun Luo
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Shuai Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Pengfei Yang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yan Gao
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Xiaoling Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Naihong Chen
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces & College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China.
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
8
|
Gopalakrishnan P, Shrestha B, Kaskas AM, Green J, Alexander JS, Pattillo CB. Hydrogen sulfide: Therapeutic or injurious in ischemic stroke? PATHOPHYSIOLOGY : THE OFFICIAL JOURNAL OF THE INTERNATIONAL SOCIETY FOR PATHOPHYSIOLOGY 2019; 26:1-10. [PMID: 30528175 DOI: 10.1016/j.pathophys.2018.10.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 07/10/2018] [Accepted: 10/22/2018] [Indexed: 01/09/2023]
Abstract
Hydrogen sulfide (H2S) has been identified as a vasodilatory, neuromodulatory, and anti-inflammatory gasotransmitter with antioxidant properties. Studies focused in cardiac tissue suggest H2S functions as a protective agent; however in the central nervous system (CNS) the effects of H2S during states of stress or injury, such as stroke, remain controversial. Currently, the application of H2S donors and modulators in stroke depends on the type of H2S donor and the timing of the therapy.
Collapse
Affiliation(s)
- Priya Gopalakrishnan
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71130-3932, USA
| | - B Shrestha
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71130-3932, USA
| | - A M Kaskas
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71130-3932, USA
| | - J Green
- Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71130-3932, USA
| | - J S Alexander
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71130-3932, USA
| | - C B Pattillo
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71130-3932, USA.
| |
Collapse
|
9
|
Zhang H, Zhang D, Li H, Yan H, Zhang Z, Zhou C, Chen Q, Ye Z, Hang C. Biphasic activation of nuclear factor-κB and expression of p65 and c-Rel following traumatic neuronal injury. Int J Mol Med 2018; 41:3203-3210. [PMID: 29568960 PMCID: PMC5881643 DOI: 10.3892/ijmm.2018.3567] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 02/22/2018] [Indexed: 01/28/2023] Open
Abstract
The transcription factor nuclear factor-κB (NF-κB) has been shown to function as a key regulator of cell death or survival in neuronal cells. Previous studies indicate that the biphasic activation of NF-κB occurs following experimental neonatal hypoxia-ischemia and subarachnoid hemorrhage. However, the comprehensive understanding of NF-κB activity following traumatic brain injury (TBI) is incomplete. In the current study, an in vitro model of TBI was designed to investigate the NF-κB activity and expression of p65 and c-Rel subunits following traumatic neuronal injury. Primary cultured neurons were assigned to control and transected groups. NF-κB activity was detected by electrophoretic mobility shift assay. Western blotting and immunofluorescence were used to investigate the expression and distribution of p65 and c-Rel. Reverse transcription-quantitative polymerase chain reaction was performed to assess the downstream genes of NF-κB. Lactate dehydrogenase (LDH) quantification and trypan blue staining were used to estimate the neuronal injury. Double peaks of elevated NF-κB activity were observed at 1 and 24 h following transection. The expression levels of downstream genes exhibited similar changes. The protein levels of p65 also presented double peaks while c-Rel was elevated significantly in the late stage. The results of the trypan blue staining and LDH leakage assays indicated there was no sustained neuronal injury during the late peak of NF-κB activity. In conclusion, biphasic activation of NF-κB is induced following experimental traumatic neuronal injury. The elevation of p65 and c-Rel levels at different time periods suggests that within a single neuron, NF-κB may participate in different pathophysiological processes.
Collapse
Affiliation(s)
- Huasheng Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Dingding Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Hua Li
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Huiying Yan
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Zihuan Zhang
- Department of Neurosurgery, Zhongdu Hospital, Bengbu, Anhui 233004, P.R. China
| | - Chenhui Zhou
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| | - Qiang Chen
- Department of Neurosurgery, Southern Medical University (Guangzhou), Jinling Hospital, Nanjing, Jiangsu 210002, P.R. China
| | - Zhennan Ye
- Department of Neurosurgery, Southern Medical University (Guangzhou), Jinling Hospital, Nanjing, Jiangsu 210002, P.R. China
| | - Chunhua Hang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu 210002, P.R. China
| |
Collapse
|
10
|
Espinosa-Garcia C, Sayeed I, Yousuf S, Atif F, Sergeeva EG, Neigh GN, Stein DG. Stress primes microglial polarization after global ischemia: Therapeutic potential of progesterone. Brain Behav Immun 2017. [PMID: 28648389 DOI: 10.1016/j.bbi.2017.06.012] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Despite the fact that stress is associated with increased risk of stroke and worsened outcome, most preclinical studies have ignored this comorbid factor, especially in the context of testing neuroprotective treatments. Preclinical research suggests that stress primes microglia, resulting in an enhanced reactivity to a subsequent insult and potentially increasing vulnerability to stroke. Ischemia-induced activated microglia can be polarized into a harmful phenotype, M1, which produces pro-inflammatory cytokines, or a protective phenotype, M2, which releases anti-inflammatory cytokines and neurotrophic factors. Selective modulation of microglial polarization by inhibiting M1 or stimulating M2 may be a potential therapeutic strategy for treating cerebral ischemia. Our laboratory and others have shown progesterone to be neuroprotective against ischemic stroke in rodents, but it is not known whether it will be as effective under a comorbid condition of chronic stress. Here we evaluated the neuroprotective effect of progesterone on the inflammatory response in the hippocampus after exposure to stress followed by global ischemia. We focused on the effects of microglial M1/M2 polarization and pro- and anti-inflammatory mediators in stressed ischemic animals. Male Sprague-Dawley rats were exposed to 8 consecutive days of social defeat stress and then subjected to global ischemia or sham surgery. The rats received intraperitoneal injections of progesterone (8mg/kg) or vehicle at 2h post-ischemia followed by subcutaneous injections at 6h and once every 24h post-injury for 7days. The animals were killed at 7 and 14days post-ischemia, and brains were removed and processed to assess outcome measures using histological, immunohistochemical and molecular biology techniques. Pre-ischemic stress (1) exacerbated neuronal loss and neurodegeneration as well as microglial activation in the selectively vulnerable CA1 hippocampal region, (2) dysregulated microglial polarization, leading to upregulation of both M1 and M2 phenotype markers, (3) increased pro-inflammatory cytokine expression, and (4) reduced anti-inflammatory cytokine and neurotrophic factor expression in the ischemic hippocampus. Treatment with progesterone significantly attenuated stress-induced microglia priming by modulating polarized microglia and the inflammatory environment in the hippocampus, the area most vulnerable to ischemic injury. Our findings can be taken to suggest that progesterone holds potential as a candidate for clinical testing in ischemic stroke where high stress may be a contributing factor.
Collapse
Affiliation(s)
| | - Iqbal Sayeed
- Department of Emergency Medicine, Emory University, Atlanta, GA 30322, USA.
| | - Seema Yousuf
- Department of Emergency Medicine, Emory University, Atlanta, GA 30322, USA.
| | - Fahim Atif
- Department of Emergency Medicine, Emory University, Atlanta, GA 30322, USA.
| | - Elena G Sergeeva
- Department of Emergency Medicine, Emory University, Atlanta, GA 30322, USA.
| | - Gretchen N Neigh
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA 30322, USA.
| | - Donald G Stein
- Department of Emergency Medicine, Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
11
|
Zhang B, Song C, Feng B, Fan W. Neuroprotection by triptolide against cerebral ischemia/reperfusion injury through the inhibition of NF-κB/PUMA signal in rats. Ther Clin Risk Manag 2016; 12:817-24. [PMID: 27307742 PMCID: PMC4888863 DOI: 10.2147/tcrm.s106012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Triptolide, an active compound extracted from the Chinese herb thunder god vine (Tripterygium wilfordii Hook F.), has potent antitumor activity. Recently, triptolide was found to have protective effects against acute cerebral ischemia/reperfusion (I/R) injury through inhibition of cell apoptosis. However, the regulatory mechanism of the effect remains unclear. We hypothesize that the regulatory mechanisms of triptolide are mediated by nuclear factor κB (NF-κB) and p53-upregulated-modulator-of-apoptosis signal inhibition. To verify this hypothesis, we occluded the middle cerebral artery in male rats to establish focal cerebral I/R model. The rats received triptolide or vehicle at the onset of reperfusion following middle cerebral artery occlusion. At 24 hours after reperfusion, neurological deficits, infarct volume, and cell apoptosis were evaluated. The expression levels of NF-κBp65, PUMA, and caspase-3 were determined by Western blot. Real-time polymerase chain reaction was used to determine the levels of NF-κBp65 mRNA, PUMA mRNA, and caspase-3 mRNA. NF-κB activity was determined by electrophoretic mobility shift assay. Apoptotic cells were detected using terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) staining. In I/R group, neurological deficit scores, cerebral infarct volume, expression of NF-κBp65, PUMA, caspase-3, NF-κB activity, and TUNEL-positive cells were found to be increased at 24 hours after I/R injury. The I/R/triptolide rats showed significantly better neurological deficit scores, decreased neural apoptosis, and reduced cerebral infarct volume. In addition, the expression of NF-κBp65, PUMA, caspase-3, and NF-κB activity was suppressed in the I/R/triptolide rats. These results indicate that the neuroprotective effects of triptolide during acute cerebral I/R injury are possibly related to the inhibition of apoptosis through suppression of NF-κB/PUMA signaling pathway.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Neurology, The Third Hospital of Liaocheng, Liaocheng, Shandong, People's Republic of China
| | - Cunfeng Song
- Department of Neurology, The Third Hospital of Liaocheng, Liaocheng, Shandong, People's Republic of China
| | - Bo Feng
- Department of Neurology, The Third Hospital of Liaocheng, Liaocheng, Shandong, People's Republic of China
| | - Weibing Fan
- Department of Neurology, The Third Hospital of Changsha, Changsha, Hunan, People's Republic of China
| |
Collapse
|
12
|
Rolova T, Dhungana H, Korhonen P, Valonen P, Kolosowska N, Konttinen H, Kanninen K, Tanila H, Malm T, Koistinaho J. Deletion of Nuclear Factor kappa B p50 Subunit Decreases Inflammatory Response and Mildly Protects Neurons from Transient Forebrain Ischemia-induced Damage. Aging Dis 2015; 7:450-65. [PMID: 27493832 DOI: 10.14336/ad.2015.1123] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 11/23/2015] [Indexed: 12/16/2022] Open
Abstract
Transient forebrain ischemia induces delayed death of the hippocampal pyramidal neurons, particularly in the CA2 and medial CA1 area. Early pharmacological inhibition of inflammatory response can ameliorate neuronal death, but it also inhibits processes leading to tissue regeneration. Therefore, research efforts are now directed to modulation of post-ischemic inflammation, with the aim to promote beneficial effects of inflammation and limit adverse effects. Transcription factor NF-κB plays a key role in the inflammation and cell survival/apoptosis pathways. In the brain, NF-κB is predominantly found in the form of a heterodimer of p65 (RelA) and p50 subunit, where p65 has a transactivation domain while p50 is chiefly involved in DNA binding. In this study, we subjected middle-aged Nfkb1 knockout mice (lacking p50 subunit) and wild-type controls of both sexs to 17 min of transient forebrain ischemia and assessed mouse performance in a panel of behavioral tests after two weeks of post-operative recovery. We found that ischemia failed to induce clear memory and motor deficits, but affected spontaneous locomotion in genotype- and sex-specific way. We also show that both the lack of the NF-κB p50 subunit and female sex independently protected CA2 hippocampal neurons from ischemia-induced cell death. Additionally, the NF-κB p50 subunit deficiency significantly reduced ischemia-induced microgliosis, astrogliosis, and neurogenesis. Lower levels of hippocampal microgliosis significantly correlated with faster spatial learning. We conclude that NF-κB regulates the outcome of transient forebrain ischemia in middle-aged subjects in a sex-specific way, having an impact not only on neuronal death but also specific inflammatory responses and neurogenesis.
Collapse
Affiliation(s)
- Taisia Rolova
- 1Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland
| | - Hiramani Dhungana
- 1Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland
| | - Paula Korhonen
- 1Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland
| | - Piia Valonen
- 1Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland
| | - Natalia Kolosowska
- 1Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland
| | - Henna Konttinen
- 1Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland
| | - Katja Kanninen
- 1Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland
| | - Heikki Tanila
- 1Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland; 2Department of Neurology, Kuopio University Hospital, Kuopio, Finland
| | - Tarja Malm
- 1Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland
| | - Jari Koistinaho
- 1Department of Neurobiology, A.I. Virtanen Institute, University of Eastern Finland
| |
Collapse
|
13
|
Liang J, Luan Y, Lu B, Zhang H, Luo YN, Ge P. Protection of ischemic postconditioning against neuronal apoptosis induced by transient focal ischemia is associated with attenuation of NF-κB/p65 activation. PLoS One 2014; 9:e96734. [PMID: 24800741 PMCID: PMC4011781 DOI: 10.1371/journal.pone.0096734] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Accepted: 04/10/2014] [Indexed: 11/19/2022] Open
Abstract
Background and Purpose Accumulating evidences have demonstrated that nuclear factor κB/p65 plays a protective role in the protection of ischemic preconditioning and detrimental role in lethal ischemia-induced programmed cell death including apoptosis and autophagic death. However, its role in the protection of ischemic postconditioning is still unclear. Methods Rat MCAO model was used to produce transient focal ischemia. The procedure of ischemic postconditioning consisted of three cycles of 30 seconds reperfusion/reocclusion of MCA. The volume of cerebral infarction was measured by TTC staining and neuronal apoptosis was detected by TUNEL staining. Western blotting was used to analyze the changes in protein levels of Caspase-3, NF-κB/p65, phosphor- NF-κB/p65, IκBα, phosphor- IκBα, Noxa, Bim and Bax between rats treated with and without ischemic postconditioning. Laser scanning confocal microscopy was used to examine the distribution of NF-κB/p65 and Noxa. Results Ischemic postconditioning made transient focal ischemia-induced infarct volume decrease obviously from 38.6%±5.8% to 23.5%±4.3%, and apoptosis rate reduce significantly from 46.5%±6.2 to 29.6%±5.3% at reperfusion 24 h following 2 h focal cerebral ischemia. Western blotting analysis showed that ischemic postconditioning suppressed markedly the reduction of NF-κB/p65 in cytoplasm, but elevated its content in nucleus either at reperfusion 6 h or 24 h. Moreover, the decrease of IκBα and the increase of phosphorylated IκBα and phosphorylated NF-κB/p65 at indicated reperfusion time were reversed by ischemic postconditioning. Correspondingly, proapoptotic proteins Caspase-3, cleaved Caspase-3, Noxa, Bim and Bax were all mitigated significantly by ischemic postconditioning. Confocal microscopy revealed that ischemic postconditioning not only attenuated ischemia-induced translocation of NF-κB/p65 from neuronal cytoplasm to nucleus, but also inhibited the abnormal expression of proapoptotic protein Noxa within neurons. Conclusions We demonstrated in this study that the protection of ischemic postconditioning on neuronal apoptosis caused by transient focal ischemia is associated with attenuation of the activation of NF-κB/p65 in neurons.
Collapse
Affiliation(s)
- Jianmin Liang
- Department of Pediatrics, First hospital of Jilin University, Changchun, China
- Neuroscience Research Center, First hospital of Jilin University, Changchun, China
| | - Yongxin Luan
- Department of Neurosurgery, First hospital of Jilin University, Changchun, China
| | - Bin Lu
- Department of Neurosurgery, First hospital of Jilin University, Changchun, China
| | - Hongbo Zhang
- Department of Pediatrics, First hospital of Jilin University, Changchun, China
| | - Yi-nan Luo
- Department of Neurosurgery, First hospital of Jilin University, Changchun, China
- Neuroscience Research Center, First hospital of Jilin University, Changchun, China
| | - Pengfei Ge
- Department of Neurosurgery, First hospital of Jilin University, Changchun, China
- Neuroscience Research Center, First hospital of Jilin University, Changchun, China
- * E-mail:
| |
Collapse
|
14
|
Cheng CY, Lin JG, Tang NY, Kao ST, Hsieh CL. Electroacupuncture-like stimulation at the Baihui (GV20) and Dazhui (GV14) acupoints protects rats against subacute-phase cerebral ischemia-reperfusion injuries by reducing S100B-mediated neurotoxicity. PLoS One 2014; 9:e91426. [PMID: 24626220 PMCID: PMC3953388 DOI: 10.1371/journal.pone.0091426] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Accepted: 02/09/2014] [Indexed: 11/30/2022] Open
Abstract
Objectives The purpose of this study was to evaluate the effects of electroacupuncture-like stimulation at the Baihui (GV20) and Dazhui (GV14) acupoints (EA at acupoints) during the subacute phase of cerebral ischemia-reperfusion (I/R) injury and to establish the neuroprotective mechanisms involved in the modulation of the S100B-mediated signaling pathway. Methods The experimental rats were subjected to middle cerebral artery occlusion (MCAo) for 15 min followed by 1 d or 7 d of reperfusion. EA at acupoints was applied 1 d postreperfusion then once daily for 6 consecutive days. Results We observed that 15 min of MCAo caused delayed infarct expansion 7 d after reperfusion. EA at acupoints significantly reduced the cerebral infarct and neurological deficit scores. EA at acupoints also downregulated the expression of the glial fibrillary acidic protein (GFAP), S100B, nuclear factor-κB (NF-κB; p50), and tumor necrosis factor-α (TNF-α), and reduced the level of inducible nitric oxide synthase (iNOS) and apoptosis in the ischemic cortical penumbra 7 d after reperfusion. Western blot analysis showed that EA at acupoints significantly downregulated the cytosolic expression of phospho-p38 MAP kinase (p-p38 MAP kinase), tumor necrosis factor receptor type 1-associated death domain (TRADD), Fas-associated death domain (FADD), cleaved caspase-8, and cleaved caspase-3 in the ischemic cortical penumbra 7 d after reperfusion. EA at acupoints significantly reduced the numbers of GFAP/S100B and S100B/nitrotyrosine double-labeled cells. Conclusion Our study results indicate that EA at acupoints initiated 1 d postreperfusion effectively downregulates astrocytic S100B expression to provide neuroprotection against delayed infarct expansion by modulating p38 MAP kinase-mediated NF-κB expression. These effects subsequently reduce oxidative/nitrative stress and inhibit the TNF-α/TRADD/FADD/cleaved caspase-8/cleaved caspase-3 apoptotic pathway in the ischemic cortical penumbra 7 d after reperfusion.
Collapse
Affiliation(s)
- Chin-Yi Cheng
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Chinese Medicine, Hui-Sheng Hospital, Taichung, Taiwan
| | - Jaung-Geng Lin
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Nou-Ying Tang
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Shung-Te Kao
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Ching-Liang Hsieh
- Acupuncture Research Center, China Medical University, Taichung, Taiwan
- Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
15
|
|
16
|
Protective effect of Tanshinone IIA against infarct size and increased HMGB1, NFκB, GFAP and apoptosis consequent to transient middle cerebral artery occlusion. Neurochem Res 2013; 39:295-304. [PMID: 24362639 DOI: 10.1007/s11064-013-1221-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 11/18/2013] [Accepted: 12/07/2013] [Indexed: 10/25/2022]
Abstract
Acute inflammation plays an important role in brain damage following cerebral ischemia and reperfusion (I/R) injury. The present study employed a rat model of middle cerebral artery occlusion to explore the neuroprotective effects of tanshinone IIA (TSN), which is widely used in China for treating cerebrovascular and cardiovascular diseases. Rats were divided into a sham-operated group and I/R transiently occluded then reperfused groups. Some of the I/R animals were treated daily for 7 or 15 days with two different doses of TSN. After 15 days, triphenyl tetrazolium chloride staining revealed less unstained area indicating fewer lesions in the TSN-treated I/R group relative to the untreated corresponding I/R group. TSN treatment dramatically reduced infarct sizes and reduced content of high mobility group box 1 protein following I/R. Nuclear translocation of NFκB was also attenuated in I/R animals subsequently receiving TSN. Terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling staining revealed more apoptosis in the I/R model group and this was reduced in the I/R animals treated with TSN for 15 days. Thus, TSN mitigates the severity of damage effected by I/R.
Collapse
|
17
|
Peng L, Xu L, Ouyang W. Role of peripheral inflammatory markers in postoperative cognitive dysfunction (POCD): a meta-analysis. PLoS One 2013; 8:e79624. [PMID: 24236147 PMCID: PMC3827367 DOI: 10.1371/journal.pone.0079624] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Accepted: 10/03/2013] [Indexed: 12/23/2022] Open
Abstract
Background Postoperative cognitive dysfunction (POCD) is common following cardiac and non-cardiac surgery, but the pathogenic mechanisms remain unknown. Many studies suggest that an inflammatory response is a key contributor to POCD. The current meta-analysis shows that the levels of peripheral inflammatory markers are associated with POCD. Methods An online search was performed to identify peer-reviewed studies without language restriction that measured peripheral inflammatory markers of patients with and without POCD, using PubMed, ScienceDirect, SinoMed and the National Knowledge Infrastructure database. Extracted data were analyzed with STATA (version 12).The standardized mean difference (SMD) and the 95% confidence interval (95%CI) were calculated for each outcome using a random effect model. Tests of heterogeneity assessment of bias, and meta-regression were performed in the meta-analysis. Results A total of 13 studies that measured the concentrations of peripheral inflammatory markers were included. The current meta-analysis found significantly higher concentrations of S-100β(SMD[95%CI]) (1.377 [0.423, 2.331], p-value < 0.001, N [POCD/non-POCD] =178/391, 7 studies), and interleukin(IL)-6 (SMD[95%CI]) (1.614 [0.603,2.624], p-value < 0.001, N[POCD/non-POCD] = 91/99, 5 studies), but not of neuron specific enolase, interleukin-1β, or tumor necrosis factor-α , in POCD compared with patients without POCD. In meta-regression analyses, a significant positive association was found between the SMD and the preoperative interleukin-6 peripheral blood concentration in patients with POCD (Coef.= 0.0587, p-value=0.038, 5 studies). Conclusions This study shows that POCD is indeed correlated with the concentrations of peripheral inflammatory markers, particularly interleukin-6 and S-100β.
Collapse
Affiliation(s)
- Linying Peng
- Department of Anesthesiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Liwei Xu
- Department of Gastrointestinal Surgery, The First Xiangya Hospital of Central South University, Changsha, China
| | - Wen Ouyang
- Department of Anesthesiology, The Third Xiangya Hospital of Central South University, Changsha, China
- * E-mail:
| |
Collapse
|
18
|
Biphasic activation of nuclear factor kappa B and expression of p65 and c-Rel after traumatic brain injury in rats. Inflamm Res 2013; 63:109-15. [DOI: 10.1007/s00011-013-0677-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Revised: 10/01/2013] [Accepted: 10/11/2013] [Indexed: 10/26/2022] Open
|
19
|
Generation of hydrogen peroxide mediates hanging death-induced neuronal cell apoptosis in the dentate gyrus of the rat brain. Brain Res Bull 2013; 95:54-60. [DOI: 10.1016/j.brainresbull.2013.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 03/05/2013] [Accepted: 03/11/2013] [Indexed: 11/15/2022]
|
20
|
Guo Y, Zhang H, Yang J, Liu S, Bing L, Gao J, Hao A. Granulocyte colony-stimulating factor improves alternative activation of microglia under microenvironment of spinal cord injury. Neuroscience 2013; 238:1-10. [DOI: 10.1016/j.neuroscience.2013.01.047] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Revised: 01/19/2013] [Accepted: 01/22/2013] [Indexed: 10/27/2022]
|
21
|
Nitric oxide donors as neuroprotective agents after an ischemic stroke-related inflammatory reaction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:297357. [PMID: 23691263 PMCID: PMC3649699 DOI: 10.1155/2013/297357] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 02/26/2013] [Accepted: 02/27/2013] [Indexed: 12/17/2022]
Abstract
Cerebral ischemia initiates a cascade of detrimental events including glutamate-associated excitotoxicity, intracellular calcium accumulation, formation of Reactive oxygen species (ROS), membrane lipid degradation, and DNA damage, which lead to the disruption of cellular homeostasis and structural damage of ischemic brain tissue. Cerebral ischemia also triggers acute inflammation, which exacerbates primary brain damage. Therefore, reducing oxidative stress (OS) and downregulating the inflammatory response are options that merit consideration as potential therapeutic targets for ischemic stroke. Consequently, agents capable of modulating both elements will constitute promising therapeutic solutions because clinically effective neuroprotectants have not yet been discovered and no specific therapy for stroke is available to date. Because of their ability to modulate both oxidative stress and the inflammatory response, much attention has been focused on the role of nitric oxide donors (NOD) as neuroprotective agents in the pathophysiology of cerebral ischemia-reperfusion injury. Given their short therapeutic window, NOD appears to be appropriate for use during neurosurgical procedures involving transient arterial occlusions, or in very early treatment of acute ischemic stroke, and also possibly as complementary treatment for neurodegenerative diseases such as Parkinson or Alzheimer, where oxidative stress is an important promoter of damage. In the present paper, we focus on the role of NOD as possible neuroprotective therapeutic agents for ischemia/reperfusion treatment.
Collapse
|
22
|
Xu JH, Zhang TZ, Zhao YY, Wang JK, Yuan ZG. Protective effects of recombinant human bone morphogenetic protein-7 on focal cerebral ischemia-reperfusion injury. Int J Neurosci 2013; 123:375-84. [PMID: 23327662 DOI: 10.3109/00207454.2012.761614] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
This study was to investigate the protective effect of recombinant human bone morphogenetic protein-7 (rhBMP-7) on focal cerebral ischemia-reperfusion (IR) injuries and their underlying mechanisms. An intraluminal suture method was used to generate a middle cerebral artery occlusion model in rats, which was followed by reperfusion. A sham operation (SO) group underwent the procedure without occlusion, whereas an IR group and rhBMP-7 treated group (RT) underwent occlusion in the absence and presence of rhBMP-7 (250 μg/kg) administered via a femoral vein injection 30 minutes prior to reperfusion. Twenty-four hours after reperfusion, neurological function, brain water content and morphological alterations were examined. Apoptosis was detected using terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assays, and immunohistochemical staining and Western blot assays were used to detect nuclear nuclear factor-kappa B (NF-κB) p65 expression. Compared with the SO group, IR rats showed a decrease in neurological function, an increase in brain water content, and pathological and morphological damage (p < 0.05). Higher levels of apoptosis were also detected in the infarct region area. In contrast, RT rats had reduced injury after IR. In addition, while immunohistochemical staining and western blot assays consistently detected increased expression of nuclear NF-κB after IR, these levels were reduced in the RT group. Administration of rhBMP-7 prior to reperfusion effectively inhibited the extent of IR injury by attenuating cerebral edema and ameliorating ultrastructural damage. The underlying mechanisms responsible for these observations potentially involve the inhibition of apoptosis induced by IR by rhBMP-7 via an NF-κB-related signaling cascade.
Collapse
Affiliation(s)
- Ji-Hong Xu
- Department of Anesthesiology, General Hospital of Shenyang Military Region, Shenyang, China
| | | | | | | | | |
Collapse
|
23
|
Raza S, Khan M, Ahmad A, Ashafaq M, Islam F, Wagner A, Safhi M, Islam F. Neuroprotective effect of naringenin is mediated through suppression of NF-κB signaling pathway in experimental stroke. Neuroscience 2013; 230:157-71. [DOI: 10.1016/j.neuroscience.2012.10.041] [Citation(s) in RCA: 122] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 10/18/2012] [Accepted: 10/19/2012] [Indexed: 10/27/2022]
|
24
|
van Harten AE, Scheeren TWL, Absalom AR. A review of postoperative cognitive dysfunction and neuroinflammation associated with cardiac surgery and anaesthesia. Anaesthesia 2012; 67:280-93. [DOI: 10.1111/j.1365-2044.2011.07008.x] [Citation(s) in RCA: 188] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
25
|
Herkenham M, Rathore P, Brown P, Listwak SJ. Cautionary notes on the use of NF-κB p65 and p50 antibodies for CNS studies. J Neuroinflammation 2011; 8:141. [PMID: 21999414 PMCID: PMC3210105 DOI: 10.1186/1742-2094-8-141] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 10/14/2011] [Indexed: 01/02/2023] Open
Abstract
Background The characterization and cellular localization of transcription factors like NF-κB requires the use of antibodies for western blots and immunohistochemistry. However, if target protein levels are low and the antibodies not well characterized, false positive data can result. In studies of NF-κB activity in the CNS, antibodies detecting NF-κB proteins have been used to support the finding that NF-κB is constitutively active in neurons, and activity levels are further increased by neurotoxic treatments, glutamate stimulation, or elevated synaptic activity. The specificity of the antibodies used was analyzed in this study. Methods Selectivity and nonselectivity of commonly used commercial and non-commercial p50 and p65 antibodies were demonstrated in western blot assays conducted in tissues from mutant gene knockout mice lacking the target proteins. Results A few antibodies for p50 and p65 each mark a single band at the appropriate molecular weight in gels containing proteins from wildtype tissue, and this band is absent in proteins from knockout tissues. Several antibodies mark proteins that are present in knockout tissues, indicating that they are nonspecific. These include antibodies raised against the peptide sequence containing the nuclear localization signals of p65 (MAB3026; Chemicon) and p50 (sc-114; Santa Cruz). Some antibodies that recognize target proteins at the correct molecular weight still fail in western blot analysis because they also mark additional proteins and inconsistently so. We show that the criterion for validation by use of blocking peptides can still fail the test of specificity, as demonstrated for several antibodies raised against p65 phosphorylated at serine 276. Finally, even antibodies that show specificity in western blots produce nonspecific neuronal staining by immunohistochemistry. Conclusions We note that many of the findings in the literature about neuronal NF-κB are based on data garnered with antibodies that are not selective for the NF-κB subunit proteins p65 and p50. The data urge caution in interpreting studies of neuronal NF-κB activity in the brain.
Collapse
Affiliation(s)
- Miles Herkenham
- Section on Functional Neuroanatomy, Laboratory of Cellular & Molecular Regulation, National Institute of Mental Health, NIH, Bethesda, MD 20892, USA.
| | | | | | | |
Collapse
|
26
|
Chen MJ, Wong CHY, Peng ZF, Manikandan J, Melendez AJ, Tan TM, Crack PJ, Cheung NS. A global transcriptomic view of the multifaceted role of glutathione peroxidase-1 in cerebral ischemic-reperfusion injury. Free Radic Biol Med 2011; 50:736-48. [PMID: 21193029 DOI: 10.1016/j.freeradbiomed.2010.12.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 12/09/2010] [Accepted: 12/16/2010] [Indexed: 12/01/2022]
Abstract
Transient cerebral ischemia often results in secondary ischemic/reperfusion injury, the pathogenesis of which remains unclear. This study provides a comprehensive, temporal description of the molecular events contributing to neuronal injury after transient cerebral ischemia. Intraluminal middle cerebral artery occlusion (MCAO) was performed to induce a 2-h ischemia with reperfusion. Microarray analysis was then performed on the infarct cortex of wild-type (WT) and glutathione peroxidase-1 (a major antioxidant enzyme) knockout (Gpx1(-/-)) mice at 8 and 24h postreperfusion to identify differential gene expression profile patterns and potential alternative injury cascades in the absence of Gpx1, a crucial antioxidant enzyme, in cerebral ischemia. Genes with at least ±1.5-fold change in expression at either time point were considered significant. Global transcriptomic analyses demonstrated that 70% of the WT-MCAO profile overlapped with that of Gpx1(-/-)-MCAO, and 28% vice versa. Critical analysis of the 1034 gene probes specific to the Gpx1(-/-)-MCAO profile revealed regulation of additional novel pathways, including the p53-mediated proapoptotic pathway and Fas ligand (CD95/Apo1)-mediated pathways; downplay of the Nrf2 antioxidative cascade; and ubiquitin-proteasome system dysfunction. Therefore, this comparative study forms the foundation for the establishment of screening platforms for target definition in acute cerebral ischemia intervention.
Collapse
MESH Headings
- Animals
- Apoptosis/genetics
- Disease Models, Animal
- Fas Ligand Protein/genetics
- Gene Expression Profiling
- Genes, p53
- Glutathione Peroxidase/genetics
- Glutathione Peroxidase/metabolism
- Infarction, Middle Cerebral Artery/genetics
- Infarction, Middle Cerebral Artery/metabolism
- Infarction, Middle Cerebral Artery/pathology
- Inflammation
- Ischemic Attack, Transient/genetics
- Ischemic Attack, Transient/metabolism
- Ischemic Attack, Transient/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- NF-E2-Related Factor 2/genetics
- Oligonucleotide Array Sequence Analysis
- Oxidation-Reduction
- Polymerase Chain Reaction
- Reactive Oxygen Species
- Reperfusion Injury/genetics
- Reperfusion Injury/metabolism
- Reperfusion Injury/pathology
- Reperfusion Injury/surgery
- Signal Transduction
- Ubiquitin-Protein Ligase Complexes/genetics
- Glutathione Peroxidase GPX1
Collapse
|
27
|
Park IS, Jo JR, Hong H, Nam KY, Kim JB, Hwang SH, Choi MS, Ryu NH, Jang HJ, Lee SH, Kim CS, Kwon TG, Park GY, Park JW, Jang BC. Aspirin induces apoptosis in YD-8 human oral squamous carcinoma cells through activation of caspases, down-regulation of Mcl-1, and inactivation of ERK-1/2 and AKT. Toxicol In Vitro 2010; 24:713-20. [DOI: 10.1016/j.tiv.2010.01.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 11/03/2009] [Accepted: 01/25/2010] [Indexed: 01/04/2023]
|
28
|
Wang L, Zhang X, Liu L, Yang R, Cui L, Li M. Atorvastatin protects rat brains against permanent focal ischemia and downregulates HMGB1, HMGB1 receptors (RAGE and TLR4), NF-kappaB expression. Neurosci Lett 2010; 471:152-6. [PMID: 20100543 DOI: 10.1016/j.neulet.2010.01.030] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2009] [Revised: 01/14/2010] [Accepted: 01/14/2010] [Indexed: 01/26/2023]
Abstract
Inflammatory processes play a key, mainly detrimental role in the pathophysiology of ischemic stroke. Currently, HMGB1-induced NF-kappaB activation pathway has been recognized as a key contributor to the proinflammatory response. It has been proved that chronic administration and pre-treatment with statins could protect brain tissue against ischemic injury. However, little is known about the effects of statins in the acute phase after cerebral ischemia. Thus, this study investigated the atorvastatin's protective role and the underlying mechanisms in cerebral ischemia. After middle cerebral artery occlusion (MCAO), atorvastatin was administered immediately. We found that atorvastatin dramatically improved neurological deficits, reduced brain water contents and infarct sizes at 24h after stroke. Moreover, the over-expression of HMGB1, RAGE, TLR4 and NF-kappaB induced by ischemia was significantly attenuated by atorvastatin.
Collapse
Affiliation(s)
- Lina Wang
- Department of Neurology, Second Hospital of Hebei Medical University, No. 251 Hepingxi Road, Shijiazhuang, Hebei 050000, China
| | | | | | | | | | | |
Collapse
|
29
|
Wang L, Zhang X, Liu L, Cui L, Yang R, Li M, Du W. Tanshinone II A down-regulates HMGB1, RAGE, TLR4, NF-kappaB expression, ameliorates BBB permeability and endothelial cell function, and protects rat brains against focal ischemia. Brain Res 2010; 1321:143-51. [PMID: 20043889 DOI: 10.1016/j.brainres.2009.12.046] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2009] [Revised: 12/14/2009] [Accepted: 12/14/2009] [Indexed: 11/18/2022]
Abstract
Inflammatory damage plays an important role in cerebral ischemic pathogenesis. HMGB1-induced NF-kappaB activation pathway has been gaining recognition as a key contributor to the proinflammatory response. Tanshinone II A (Tan II A) has been proved to elicit a series of biologic effects through its antiinflammatory property. But the mechanism underlying is poorly understood. This study evaluated the Tan II A's protective role in cerebral ischemia and its potential mechanism. Male Sprague-Dawley rats were subjected to pMCAO. Experiment 1 was used to evaluate the longitudinal expression of HMGB1, RAGE and TLR4 and NF-kappaB in the cerebral ischemia. Experiment 2 was used to detect Tan II A's neuroprotection. At 24 h after pMCAO, neurologic deficit, brain water content and infarct size were measured. Immunohistochemistry, RT-PCR, Western blot and confocal microscope were used to analyze the expression of HMGB1, RAGE, TLR4 and NF-kappaB. Experiment 3 was used to detect Tan II A's influence on BBB. The expressions of HMGB1, TLR4, RAGE and NF-kappaB were up-regulated in ischemic brain. Compared with pMCAO group, the expressions of these factors significantly decreased in Tan II A-H group, the neurologic deficit, infarct volume and brain water content were alleviated (P<0.05) and claudin-5 was predominantly expressed in brain capillaries. Tan II A protected the brain from damage caused by pMCAO; this effect may be through down-regulation of HMGB1, RAGE and TLR4, NF-kappaB and up-regulation claudin-5 expression.
Collapse
Affiliation(s)
- Lina Wang
- Department of Neurology, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
| | | | | | | | | | | | | |
Collapse
|
30
|
Liu Y, Zhang XJ, Yang CH, Fan HG. Oxymatrine protects rat brains against permanent focal ischemia and downregulates NF-kappaB expression. Brain Res 2009; 1268:174-180. [PMID: 19285049 DOI: 10.1016/j.brainres.2009.02.069] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2008] [Revised: 02/15/2009] [Accepted: 02/20/2009] [Indexed: 12/14/2022]
Abstract
BACKGROUND Oxymatrine is proven to protect ischemic and reperfusion injury in liver, intestine and heart, this effect is via anti-inflammation and anti-apoptosis. Whether this protective effect applies to ischemic injury in brain, we therefore investigate the potential neuroprotective role of oxymatrine and the underlying mechanisms. METHODS Male, Sprague-Dawley rats were randomly assigned to four groups: permanent middle cerebral artery occlusion (pMCAO), high dose (pMCAO+oxymatrine 120 mg/kg), low dose (pMCAO+oxymatrine 60 mg/kg) and sham operated group. We used a permanent middle cerebral artery occlusion model and administered oxymatrine intraperitoneally immediately after cerebral ischemia and once daily on the following days. At 24 h after MCAO, neurological deficit was evaluated using a modified six point scale; brain water content was measured; NF-kappaB expression was measured by immunohistochemistry, Western blotting and RT-PCR. Infarct volume was analyzed with 2, 3, 5-triphenyltetrazolium chloride (TTC) staining at 72 h. RESULTS Compared with pMCAO group, neurological deficit in high dose group was improved (P<0.05), infarct volume was decreased (P<0.001) and cerebral edema was alleviated (P<0.05). Consistent with these indices, immunohistochemistry, Western blot and RT-PCR analysis indicated that NF-kappaB expression was significantly decreased in high dose group. Low dose of oxymatrine did not affect NF-kappaB expression in pMCAO rats. CONCLUSIONS Oxymatrine reduced infarct volume induced by pMCAO, this effect may be through the decreasing of NF-kappaB expression.
Collapse
Affiliation(s)
- Ying Liu
- Department of Neurology, Second Hospital of Hebei Medical University. Shijiazhuang 050000, China
| | - Xiang-Jian Zhang
- Department of Neurology, Second Hospital of Hebei Medical University. Shijiazhuang 050000, China.
| | - Chen-Hui Yang
- Department of Neurology, Second Hospital of Hebei Medical University. Shijiazhuang 050000, China
| | - Hong-Guang Fan
- Department of Neurology, Second Hospital of Hebei Medical University. Shijiazhuang 050000, China
| |
Collapse
|
31
|
Hua Q, Zhu XL, Li PT, Liu Y, Zhang N, Xu Y, Jia X. The inhibitory effects of cholalic acid and hyodeoxycholalic acid on the expression of TNFalpha and IL-1beta after cerebral ischemia in rats. Arch Pharm Res 2009; 32:65-73. [PMID: 19183878 DOI: 10.1007/s12272-009-1119-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2008] [Revised: 12/04/2008] [Accepted: 12/05/2008] [Indexed: 11/26/2022]
Abstract
Previous studies have shown that Qing Kai Ling, a traditional Chinese medicine, was able to effectively prevent the inflammation from cerebral ischemia (Chen et al., 2002). The cholalic acid and hyodeoxycholalic acid (cholalic acid mixture) was major active components in Qing Kai Ling. To study the effects of cholalic acid mixture on the damage cascade of cerebral ischemia, rat model of focal cerebral ischemia was established by permanent occlusion of left middle cerebral artery. We found that the administration of cholalic acid mixture could reduce the ischemic infarct size after 24 h of ischemia, and cholalic acid mixture could be detected in cerebrospinal fluid after 2h of administration. We also found that the concentrations of tumor necrosis factor-alpha and interlukin-1beta in rat brain were significantly lower when compared to the untreated animals after 12 h and 24 h of ischemia. The concentrations of von Willebrand factor and neuron specific enolase in the plasma were remarkably decreased in cholalic acid mixture treated animals than in the untreated ones after 12h of ischemia. Our results suggested that cholalic acid mixture is able to decrease the expression of inflammation factors including tumor necrosis factor-alpha and interlukin-1beta after focal cerebral ischemia.
Collapse
Affiliation(s)
- Qian Hua
- School of Preclinical Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing, 100029, China
| | | | | | | | | | | | | |
Collapse
|
32
|
Ji L, Nazarali AJ, Paterson PG. Protein-energy malnutrition increases activation of the transcription factor, nuclear factor kappaB, in the gerbil hippocampus following global ischemia. J Nutr Biochem 2008; 19:770-7. [PMID: 18430555 DOI: 10.1016/j.jnutbio.2007.09.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2007] [Revised: 08/12/2007] [Accepted: 09/14/2007] [Indexed: 12/20/2022]
Abstract
Protein-energy malnutrition (PEM) exacerbates functional impairment caused by brain ischemia. This is correlated with reactive gliosis, which suggests an increased inflammatory response. The objective of the current study was to investigate if PEM increases hippocampal activation of nuclear factor kappaB (NFkappaB), a transcription factor that amplifies the inflammatory response involved in ischemic brain injury. Mongolian gerbils (11-12 weeks old) were randomly assigned to control diet (12.5% protein) or protein-deficient diet (2%) for 4 weeks. The 2% protein group had a 15% decrease in voluntary food intake (P<.001; unpaired t test), resulting in PEM. Body weight after 4 weeks was 20% lower in the PEM group (P<.001). Gerbils were then exposed to sham surgery or global ischemia induced by 5-min bilateral common carotid artery occlusion. PEM independently increased hippocampal NFkappaB activation detected by electrophoretic mobility shift assay at 6 h after surgery (P=.014; 2-factor ANOVA). Ischemia did not significantly affect NFkappaB activation nor was there interaction between diet and ischemia. Serum glucose and cortisol concentrations at 6 h postischemia were unaltered by diet or ischemia. A second experiment using gerbils of the same age and feeding paradigm demonstrated that PEM also increases hippocampal NFkappaB activation in the absence of surgery. These findings suggest that PEM, which exists in 16% of elderly patients at admission for stroke, may worsen outcome by increasing activation of NFkappaB. Since PEM increased NFkappaB activation independent of ischemia or surgery, the data also have implications for the inflammatory response of the many individuals affected globally by PEM.
Collapse
Affiliation(s)
- Liang Ji
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Saskatchewan, Canada S7N 5C9
| | | | | |
Collapse
|
33
|
Yang S, Zhang D, Yang Z, Hu X, Qian S, Liu J, Wilson B, Block M, Hong JS. Curcumin protects dopaminergic neuron against LPS induced neurotoxicity in primary rat neuron/glia culture. Neurochem Res 2008; 33:2044-53. [PMID: 18368483 DOI: 10.1007/s11064-008-9675-z] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Accepted: 03/13/2008] [Indexed: 10/22/2022]
Abstract
Using primary rat mesencephalic neuron-glia cultures as an in vitro model of Parkinson's disease (PD), we tested the effect of curcumin, a natural dietary pigment with well-known anti-inflammation effects, on dopaminergic (DA) degeneration. Curcumin pretreatment mitigated LPS-induced DA neurotoxicity in a concentration-dependent manner and curcumin post-treatment also showed protective effect. Microglia depletion abolished this protective effect of curcumin, indicating that microglia play an important role in this effect. Supportively, observation by immunocytochemistry staining using OX-42 antibody showed that curcumin treatment inhibited LPS-induced morphological change of microglia. Besides, LPS-induced production of many proinflammatory factors and their gene expressions decreased dramatically after curcumin treatment. Results also revealed that curcumin treatment decreased LPS-induced activation of two transcription factors--nuclear factors kappaB (NF-kappaB) and activator protein-1 (AP-1). Taken together, our study implicated that curcumin might be a potential preventive and therapeutic strategy for inflammation-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Sufen Yang
- Laboratory of Pharmacology and Chemistry, National Institute of Environmental, Health Sciences, National Institutes of Health, PO Box 12233, Research Triangle Park, NC 27709, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Qin ZH, Tao LY, Chen X. Dual roles of NF-kappaB in cell survival and implications of NF-kappaB inhibitors in neuroprotective therapy. Acta Pharmacol Sin 2007; 28:1859-72. [PMID: 18031598 DOI: 10.1111/j.1745-7254.2007.00741.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
NF-kappaB is a well-characterized transcription factor with multiple physiological and pathological functions. NF-kappaB plays important roles in the development and maturation of lymphoids, regulation of immune and inflammatory response, and cell death and survival. The influence of NF-kappaB on cell survival could be protective or destructive, depending on types, developmental stages of cells, and pathological conditions. The complexity of NF-kappaB in cell death and survival derives from its multiple roles in regulating the expression of a broad array of genes involved in promoting cell death and survival. The activation of NF-kappaB has been found in many neurological disorders, but its actual roles in pathogenesis are still being debated. Many compounds with neuroprotective actions are strongly associated with the inhibition of NF-kappaB, leading to speculation that blocking the pathological activation of NF-kappaB could offer neuroprotective effects in certain neurodegenerative conditions. This paper reviews the recent developments in understanding the dual roles of NF-kappaB in cell death and survival and explores its possible usefulness in treating neurological diseases. This paper will summarize the genes regulated by NF-kappaB that are involved in cell death and survival to elucidate why NF-kappaB promotes cell survival in some conditions while facilitating cell death in other conditions. This paper will also focus on the effects of various NF-kappaB inhibitors on neuroprotection in certain pathological conditions to speculate if NF-kappaB is a potential target for neuroprotective therapy.
Collapse
Affiliation(s)
- Zheng-hong Qin
- Department of Pharmacology, Soochow University School of Medicine, Suzhou 215123, China.
| | | | | |
Collapse
|
35
|
Ang HL, Tergaonkar V. Notch and NFkappaB signaling pathways: Do they collaborate in normal vertebrate brain development and function? Bioessays 2007; 29:1039-47. [PMID: 17876798 DOI: 10.1002/bies.20647] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Both Notch and NFkappaB signaling pathways are well-known for regulating proliferation, differentiation and apoptosis. Recent studies have presented several lines of evidence supporting an integration of the Notch and NFkappaB signaling pathways in differentiation/maturation of a diverse range of cell types. It is notable that Notch and NFkappaB signaling pathways share many common features: (i) both are activated by common stimuli such as TNF-alpha and hypoxia, (ii) activated Notch (NICD) and NFkappaB mediate transcription by regulating corepressors such as SMRT/N-COR, and (iii) both regulate similar target genes such as Hes-1 and IkappaBalpha. This review expands on how the collaboration between these pathways may play an important role in the CNS. We will speculate on the mechanisms by which Notch and NFkappaB signaling may collaborate to regulate stem cell renewal and differentiation during brain development and function.
Collapse
Affiliation(s)
- Hwee-Luan Ang
- Laboratory of NFkappaB Signaling and Human Ailments, Institute of Molecular and Cell Biology, 61 Biopolis Drive (Proteos), Singapore 138673
| | | |
Collapse
|
36
|
Kim YS, Cho KO, Lee HJ, Kim SY, Sato Y, Cho YJ. Down syndrome candidate region 1 increases the stability of the IkappaBalpha protein: implications for its anti-inflammatory effects. J Biol Chem 2006; 281:39051-61. [PMID: 17062574 DOI: 10.1074/jbc.m604659200] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Down syndrome candidate region 1 (DSCR1), an endogenous inhibitor of calcineurin, inhibits the expression of genes involved in the inflammatory response. To elucidate the molecular basis of these anti-inflammatory effects, we analyzed the role of DSCR1 in the regulation of NF-kappaB transactivation using glioblastoma cells stably transfected with DSCR1.4 or its truncation mutants (DSCR1.4-(1-133) and DSCR1.4-(134-197)). Overexpression of DSCR1.4 significantly attenuated the induction of cyclooxygenase-2 (COX-2) expression by phorbol 12-myristate 13-acetate (PMA) via a calcineurin-independent mechanism. Experiments using inhibitors of the signaling molecules for NF-kappaB activation showed that NF-kappaB is responsible for the induction of COX-2. Full-length and truncated DSCR1.4 decreased the steady-state activity of NF-kappaB as well as PMA-induced activation of NF-kappaB, which correlated with attenuation of COX-2 induction. DSCR1.4 did not affect the PMA-stimulated phosphorylation or degradation kinetics of IkappaBalpha; however, DSCR1.4 significantly decreased the basal turnover rate of IkappaBalpha and consequently up-regulated its steady-state level. In the same context, knockdown of endogenous DSCR1.4 increased the turnover rate of IkappaBalpha as well as COX-2 induction. These results suggest that DSCR1 attenuates NF-kappaB-mediated transcriptional activation by stabilizing its inhibitory protein, IkappaBalpha.
Collapse
Affiliation(s)
- Young Sun Kim
- Department of Pharmacology, Catholic University of Korea, Seoul 137-701, Korea
| | | | | | | | | | | |
Collapse
|
37
|
Li DQ, Bao YM, Li Y, Wang CF, Liu Y, An LJ. Catalpol modulates the expressions of Bcl-2 and Bax and attenuates apoptosis in gerbils after ischemic injury. Brain Res 2006; 1115:179-85. [DOI: 10.1016/j.brainres.2006.07.063] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2006] [Revised: 07/15/2006] [Accepted: 07/20/2006] [Indexed: 11/28/2022]
|
38
|
Crack PJ, Taylor JM, Ali U, Mansell A, Hertzog PJ. Potential Contribution of NF-κB in Neuronal Cell Death in the Glutathione Peroxidase-1 Knockout Mouse in Response to Ischemia-Reperfusion Injury. Stroke 2006; 37:1533-8. [PMID: 16627788 DOI: 10.1161/01.str.0000221708.17159.64] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background and Purpose—
We have previously identified an increased susceptibility of Gpx1
−/−
mice to increased infarct size after middle cerebral artery occlusion (MCAO). This study was designed to elucidate the mechanisms involved in elevated neuronal cell death arising from an altered endogenous oxidant state.
Methods—
Gpx1
−/−
mice were exposed to transient MCAO and reperfusion by intraluminal suture blockade. Protein expression of the p65 subunit of transcription factor nuclear factor-κB (NF-κB) was examined by immunohistochemistry and Western Analysis. NF-κB DNA-protein activity was assessed by electrophoretic mobility shift assays (EMSA). Wild-type and Gpx1
−/−
mice were exposed to MCAO with or without the NF-κB inhibitor, pyrrolidinedithiocarbamate (PDTC).
Results—
Upregulation of the p65 subunit of NF-κB and subsequent p65 phosphorylation at serine 536 was detected in the Gpx1
−/−
brains after stroke. EMSAs revealed that increased ischemia-enhanced DNA binding of NF-κB was observed in Gpx1
−/−
mice compared with wild-type. Supershift assays indicated that the p50 and p65 subunits participated in the bound NF-κB complex. The NF-κB inhibitor PDTC, a potential antioxidant, was able to afford partial neuroprotection in the Gpx1
−/−
mice.
Conclusions—
NF-κB is upregulated in the Gpx1
−/−
mouse, and this upregulation contributes to the increased cell death seen in the Gpx1
−/−
after MCAO. The activation of NF-κB may increase the expression of downstream target genes that are involved in the progression of neural injury after MCAO.
Collapse
Affiliation(s)
- Peter J Crack
- Centre for Functional Genomics and Human Disease, Monash Institute of Medical Research, Monash University, Melbourne, Australia.
| | | | | | | | | |
Collapse
|
39
|
Nurmi A, Goldsteins G, Närväinen J, Pihlaja R, Ahtoniemi T, Gröhn O, Koistinaho J. Antioxidant pyrrolidine dithiocarbamate activates Akt-GSK signaling and is neuroprotective in neonatal hypoxia-ischemia. Free Radic Biol Med 2006; 40:1776-84. [PMID: 16678015 DOI: 10.1016/j.freeradbiomed.2006.01.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2005] [Revised: 01/02/2006] [Accepted: 01/10/2006] [Indexed: 12/29/2022]
Abstract
Pyrrolidine dithiocarbamate (PDTC), an antioxidant and inhibitor of transcription factor nuclear factor kappa-B (NF-kappaB), has been reported to reduce inflammation and apoptosis. Because PDTC was recently found to protect in various models of adult brain ischemia with a wide therapeutic time window, we tested the effect of PDTC in a rodent model of neonatal hypoxia-ischemia (HI) brain injury. T2-weighed magnetic resonance imaging (T2-MRI) 7 days after the insult showed that a single PDTC (50 mg/kg) injection 2.5 h after the HI reduced the mean brain infarct size by 59%. PDTC reduced the HI-induced dephosphorylation of Akt and glycogen synthase kinase-3beta (GSK-3beta), expression of cleaved caspase-3, and nuclear translocation of NF-kappaB in the neonatal brain. PDTC targeted directly neurons, as PDTC reduced hypoxia-reoxygenation-induced cell death in pure hippocampal neuronal cultures. It is suggested that in addition to the previously indicated NF-kappaB inhibition as a protective mechanism of PDTC treatment, PDTC may reduce HI-induced brain injury at least partially by acting as an antioxidant, which reduces the Akt-GSK-3beta pathway of apoptotic cell death. The clinically approved PDTC and its analogues may be beneficial after HI insults with a reasonable time window.
Collapse
Affiliation(s)
- Antti Nurmi
- Department of Neurobiology, A.I.Virtanen Institute of Molecular Sciences, University of Kuopio, Finland
| | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Perinatal hypoxic-ischaemic injury (HII) is a significant cause of neurodevelopmental impairment and disability. Studies employing 31P magnetic resonance spectroscopy to measure phosphorus metabolites in situ in the brains of newborn infants and animals have demonstrated that transient hypoxia-ischaemia leads to a delayed disruption in cerebral energy metabolism, the magnitude of which correlates with the subsequent neurodevelopmental impairment. Prominent among the biochemical features of HII is the loss of cellular ATP, resulting in increased intracellular Na+ and Ca2+, and decreased intracellular K+. These ionic imbalances, together with a breakdown in cellular defence systems following HII, can contribute to oxidative stress with a net increase in reactive oxygen species. Subsequent damage to lipids, proteins, and DNA and inactivation of key cellular enzymes leads ultimately to cell death. Although the precise mechanisms of neuronal loss are unclear, it is now clear both apoptosis and necrosis are the significant components of cell death following HII. A number of different factors influence whether a cell will undergo apoptosis or necrosis, including the stage of development, cell type, severity of mitochondrial injury and the availability of ATP for apoptotic execution. This review will focus on some pathological mechanisms of cell death in which there is a disruption to oxidative metabolism. The first sections will discuss the process of damage to oxidative metabolism, covering the data collected both from human infants and from animal models. Following sections will deal with the molecular mechanisms that may underlie cerebral energy failure and cell death in this form of brain injury, with a particular emphasis on the role of apoptosis and mitochondria.
Collapse
Affiliation(s)
- Deanna L. Taylor
- Weston Laboratory, Division of Paediatrics, Obstetrics and Gynaecology, Imperial College School of Medicine, Hammersmith Hospital, Du Cane Road, London W12 0NN, U.K
| | - A. David Edwards
- Weston Laboratory, Division of Paediatrics, Obstetrics and Gynaecology, Imperial College School of Medicine, Hammersmith Hospital, Du Cane Road, London W12 0NN, U.K
| | - Huseyin Mehmet
- Weston Laboratory, Division of Paediatrics, Obstetrics and Gynaecology, Imperial College School of Medicine, Hammersmith Hospital, Du Cane Road, London W12 0NN, U.K
| |
Collapse
|
41
|
Song YS, Lee YS, Chan PH. Oxidative stress transiently decreases the IKK complex (IKKalpha, beta, and gamma), an upstream component of NF-kappaB signaling, after transient focal cerebral ischemia in mice. J Cereb Blood Flow Metab 2005; 25:1301-11. [PMID: 15829915 DOI: 10.1038/sj.jcbfm.9600123] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Nuclear factor-kappaB (NF-kappaB) has a central role in coordinating the expression of a wide variety of genes that control cerebral ischemia. Although there has been intense research on NF-kappaB, its mechanisms in the ischemic brain have not been clearly elucidated. We investigated the temporal profile of NF-kappaB-related genes using a complementary DNA array method in wild-type mice and human copper/zinc-superoxide dismutase transgenic (SOD 1 Tg) mice that had low-level reactive oxygen species (ROS) by scavenging superoxide. Our DNA array showed that IkappaB kinase (IKK) complex (IKKalpha, beta, and gamma) mRNA in the wild-type mice was decreased as early as 1 h after reperfusion, after 30 mins of transient focal cerebral ischemia (tFCI). In contrast, tFCI in the SOD1 Tg mice caused an increase in the IKK complex. The IKK complex protein levels were also drastically decreased at 1 h in the wild-type mice, but did not change in the SOD 1 Tg mice throughout the 7 days. Electrophoretic mobility shift assay revealed activation of NF-kappaB DNA binding after tFCI in the wild-type mice. Nuclear factor-kappaB activation occurred at the same time, as did the phosphorylation and degradation of the inhibitory protein IkappaBalpha. However, SOD 1 prevented NF-kappaB activation, and phosphorylation and degradation of IkappaBalpha after tFCI. Superoxide production and ubiquitinated protein in the SOD 1 Tg mice were also lower than in the wild-type mice after tFCI. These results suggest that ROS are implicated in transient downregulation of IKKalpha, beta, and gamma in cerebral ischemia.
Collapse
Affiliation(s)
- Yun S Song
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
| | | | | |
Collapse
|
42
|
Yu CG, Yezierski RP. Activation of the ERK1/2 signaling cascade by excitotoxic spinal cord injury. ACTA ACUST UNITED AC 2005; 138:244-55. [PMID: 15922485 DOI: 10.1016/j.molbrainres.2005.04.013] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2004] [Revised: 03/14/2005] [Accepted: 04/18/2005] [Indexed: 12/17/2022]
Abstract
The role of the ERK1/2 signal transduction pathway and related transcription factors in the regulation of gene expression and pain behavior following excitotoxic spinal cord injury (SCI) was examined. Specifically, phosphorylation of ERK1/2, activation of transcription factors NF-kB, ELK-1, and CREB, and gene expression of the neurokinin-1 receptor and NMDA receptor subunits NR1 and NR-2A were investigated. Excitotoxic injury was produced by intraspinal injection of quisqualic acid (QUIS) in male Sprague-Dawley rats. Western blots were used to evaluate phosphorylation and activation of ERK1/2 and transcription factors using phospho-specific or total antibodies. Real-time PCR was used to evaluate gene expression of NK-1R, NR-1, and NR-2A. Assessment of excessive grooming behavior was used to evaluate the presence of spontaneous pain behavior. Excitotoxic spinal injury resulted in: (1) increased phosphorylation of ERK1/2; (2) increased activation of NF-kB and phosphorylation of ELK-1; and (3) increased gene expression for the NK-1 receptor and NR1 and NR-2A subunits of the NMDA receptor. Blockade of the ERK cascade with the MEK inhibitor PD98059 inhibited phosphorylation of ELK-1, activation of NF-kB and gene expression of NR1, NR-2A and NK-1R, and prevented the development of excessive grooming behavior. The results have shown that excitotoxic spinal injury leads to the injury-induced activation of the ERK-->ELK-1 and NF-kB signaling cascades and transcriptional regulation of receptors important in the development of chronic pain. Blockade of this intracellular kinase cascade prevented the onset of injury-induced pain behavior.
Collapse
Affiliation(s)
- Chen-Guang Yu
- Department of Orthodontics and Neuroscience, College of Dentistry and McKnight Brain Institute, Comprehensive Center for Pain Research, University of Florida, Gainesville, FL 32610, USA
| | | |
Collapse
|
43
|
Ishiyama T, Dharmarajan S, Hayama M, Moriya H, Grapperhaus K, Patterson GA. Inhibition of nuclear factor κB by IκB superrepressor gene transfer ameliorates ischemia-reperfusion injury after experimental lung transplantation. J Thorac Cardiovasc Surg 2005; 130:194-201. [PMID: 15999062 DOI: 10.1016/j.jtcvs.2005.02.040] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVES Ischemia-reperfusion injury after lung transplantation is associated with significant morbidity and mortality. The activation of the transcription factor nuclear factor kappaB is central to the 2 important pathways that characterize ischemia-reperfusion injury, namely the inflammatory response and apoptosis. The purpose of this study was to determine the effects of nuclear factor kappaB inhibition on experimental lung transplant ischemia-reperfusion injury with gene transfer of the nuclear factor kappaB inhibitor IkappaB in a superrepressor form (IkappaBSR). METHODS An orthotopic left lung transplant model in isogeneic rats was used, with 18 hours of prolonged cold storage of donor lung grafts used to create severe ischemia-reperfusion injury. Donor rats underwent endobronchial gene transfection with saline alone or adenovirus encoding either beta-galactosidase control or IkappaBSR 48 hours before harvest. The function of transplanted lung grafts was assessed on the basis of isolated graft oxygenation, wet/dry lung weight ratio, and myeloperoxidase activity. Nuclear factor kappaB activation was assessed by means of enzyme-linked immunosorbent assay. Apoptotic cell death was assessed by evaluating the levels of histone-associated DNA fragments and caspase-3 activity. RESULTS Treatment of donor lung grafts with IkappaBSR resulted in significantly improved oxygenation compared with that seen in control tissue 24 hours after transplantation. IkappaBSR-treated lungs also demonstrated less pulmonary edema and reduced neutrophil infiltration 24 hours after reperfusion. Nuclear factor kappaB activation and apoptotic cell death induction 2 hours after transplantation was significantly reduced in IkappaBSR-treated lungs compared with in control lungs. CONCLUSIONS Inhibition of nuclear factor kappaB activation by IkappaBSR gene transfer improves transplanted lung graft oxygenation, decreases pulmonary edema and neutrophil sequestration, and reduces apoptotic cell death after experimental lung transplantation.
Collapse
Affiliation(s)
- Takaaki Ishiyama
- Division of Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | |
Collapse
|
44
|
Zhang HL, Huang ZH, Zhu Y, Liang ZQ, Han R, Wang XX, Chase TN, Qin ZH. Neuroprotective effects of prostaglandin A1 in animal models of focal ischemia. Brain Res 2005; 1039:203-6. [PMID: 15781063 DOI: 10.1016/j.brainres.2005.01.046] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2004] [Revised: 01/12/2005] [Accepted: 01/13/2005] [Indexed: 11/18/2022]
Abstract
The present study evaluated the neuroprotective potential of prostaglandin A1 (PGA1) in rodent models of focal cerebral ischemia. PGA1 33 nmol reduced infarction volume by about 43% (P < 0.05) when administered intracerebroventricularly before and after transient ischemia in mice. PGA1 16.5-66 nmol diminished infarction volume by 18% to 27% (P < 0.01) when administered immediately following permanent ischemia in rats. PGA1 treatment also significantly ameliorated motor dysfunction after brain ischemia. These results suggest that PGA1 protects neurons from ischemic injury.
Collapse
Affiliation(s)
- Hui-Ling Zhang
- Department of Pharmacology, Soochow University School of Medicine, 48 Ren Ming Road, Suzhou 215007, PR China
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Qiu J, Hu X, Nesic O, Grafe MR, Rassin DK, Wood TG, Perez-Polo JR. Effects of NF-kappaB oligonucleotide "decoys" on gene expression in P7 rat hippocampus after hypoxia/ischemia. J Neurosci Res 2004; 77:108-18. [PMID: 15197744 DOI: 10.1002/jnr.20156] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
"Decoy" oligonucleotides can be used as gene-specific nuclear factor (NF-kappaB) inhibitors to regulate gene expression. We applied two different decoy oligonucleotides that contained the NF-kappaB binding consensus sequences present in the immunoglobulin G (IgG)-kappaB and Bcl-x promoter into 7-day-old (P7) rat lateral ventricles before hypoxia/ischemia (HI) and compared their effects on gene expression in hippocampi to saline-treated, scrambled decoy-treated, or untreated hippocampi exposed to HI. Left hippocampi were collected at 12 hr after HI. Electrophoretic mobility shift assays (EMSAs) showed that the two decoy treatments had different effects on NF-kappaB binding to the IgG-kappaB and Bcl-x promoter-specific consensus sequences, respectively. We assessed the decoys' effects on gene expression 12 hr after HI using ribonuclease protection assays (RPAs) and Affymetrix DNA microarrays. RPAs showed that both decoys significantly decreased interleukin (IL)-1alpha mRNA levels but had no impact on IL-1beta, IL-6, and IL-10 mRNA levels. IgG-kappaB decoys significantly decreased tumor necrosis factor (TNF)-alpha and TNF-beta mRNA levels compared to minimal changes after treatment with Bcl-x decoys. DNA microarray analyses showed that Bcl-x decoy treatment significantly decreased Bcl-x(L) mRNA levels. The decreased Bcl-x(L) mRNA levels after Bcl-x decoy treatment was confirmed by RPA analysis. DNA microarray data also indicated that several other genes were affected by both decoys. Our results suggest that different NF-kappaB decoy treatments could differentially regulate transcriptional responses to central nervous system trauma. Careful design of decoy sequences, however, is essential to acquire selective effects on cell death outcome.
Collapse
Affiliation(s)
- Jingxin Qiu
- Department of Human Biological Chemistry and Genetics, University of Texas Medical Branch, Galveston, Texas 77555-0652, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Aleyasin H, Cregan SP, Iyirhiaro G, O'Hare MJ, Callaghan SM, Slack RS, Park DS. Nuclear factor-(kappa)B modulates the p53 response in neurons exposed to DNA damage. J Neurosci 2004; 24:2963-73. [PMID: 15044535 PMCID: PMC6729853 DOI: 10.1523/jneurosci.0155-04.2004] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Previous studies have shown that DNA damage-evoked death of primary cortical neurons occurs in a p53 and cyclin-dependent kinase-dependent (CDK) manner. The manner by which these signals modulate death is unclear. Nuclear factor-kappaB (NF-kappaB) is a group of transcription factors that potentially interact with these pathways. Presently, we show that NF-kappaB is activated shortly after induction of DNA damage in a manner independent of the classic IkappaB kinase (IKK) activation pathway, CDKs, ATM, and p53. Acute inhibition of NF-kappaB via expression of a stable IkappaB mutant, downregulation of the p65 NF-kappaB subunit by RNA interference (RNAi), or pharmacological NF-kappaB inhibitors significantly protected against DNA damage-induced neuronal death. NF-kappaB inhibition also reduced p53 transcripts and p53 activity as measured by the p53-inducible messages, Puma and Noxa, implicating the p53 tumor suppressor in the mechanism of NF-kappaB-mediated neuronal death. Importantly, p53 expression still induces death in the presence of NF-kappaB inhibition, indicating that p53 acts downstream of NF-kappaB. Interestingly, neurons cultured from p65 or p50 NF-kappaB-deficient mice were not resistant to death and did not show diminished p53 activity, suggesting compensatory processes attributable to germline deficiencies, which allow p53 activation still to occur. In contrast to acute NF-kappaB inhibition, prolonged NF-kappaB inhibition caused neuronal death in the absence of DNA damage. These results uniquely define a signaling paradigm by which NF-kappaB serves both an acute p53-dependent pro-apoptotic function in the presence of DNA damage and an anti-apoptotic function in untreated normal neurons.
Collapse
Affiliation(s)
- Hossein Aleyasin
- Ottawa Health Research Institute, Neurosciences, East Division, Ottawa, Ontario, Canada K1H 8M5
| | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Hypothermia to mitigate ischemic brain tissue damage has a history of about six decades. Both in clinical and experimental studies of hypothermia, two principal arbitrary patterns of core temperature lowering have been defined: mild (32-35 degrees C) and moderate hypothermia (30-33 degrees C). The neuroprotective effectiveness of postischemic hypothermia is typically viewed with skepticism because of conflicting experimental data. The questions to be resolved include the: (i) postischemic delay; (ii) depth; and (iii) duration of hypothermia. However, more recent experimental data have revealed that a protected reduction in brain temperature can provide sustained behavioral and histological neuroprotection, especially when thermoregulatory responses are suppressed by sedation or anesthesia. Conversely, brief or very mild hypothermia may only delay neuronal damage. Accordingly, protracted hypothermia of 32-34 degrees C may be beneficial following acute cerebral ischemia. But the pathophysiological mechanism of this protection remains yet unclear. Although reduction of metabolism could explain protection by deep hypothermia, it does not explain the robust protection connected with mild hypothermia. A thorough understanding of the experimental data of postischemic hypothermia would lead to a more selective and effective clinical therapy. For this reason, we here summarize recent experimental data on the application of hypothermia in cerebral ischemia, discuss problems to be solved in the experimental field, and try to draw parallels to therapeutic potentials and limitations.
Collapse
Affiliation(s)
- B Schaller
- Max-Planck-Institute for Neurological Research, Cologne, Germany
| | | |
Collapse
|
48
|
Leker RR, Neufeld MY. Anti-epileptic drugs as possible neuroprotectants in cerebral ischemia. BRAIN RESEARCH. BRAIN RESEARCH REVIEWS 2003; 42:187-203. [PMID: 12791439 DOI: 10.1016/s0165-0173(03)00170-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Many similarities exist between cerebral ischemia and epilepsy regarding brain-damaging and auto-protective mechanisms that are activated following the injurious insult. Therefore, drugs that are effective in minimizing seizure-induced brain damage may also be useful in minimizing ischemic injury. Use of such drugs in stroke victims may have important clinical and financial advantages. Therefore, the authors conducted a Medline search of studies involving the use of anti-epileptic drugs (AEDs) as possible neuroprotectants and summarize the data. Most AEDs have been tested in animal models of focal or global ischemia and some were already tested in humans, for a possible neuroprotective effect. The existing data is rather scant and insufficient but it appears that only drugs that have multiple mechanisms of action seem to have some potential in conferring a degree of neuroprotection that could be clinically applicable to stroke patients. In conclusion, some of the newer AEDs show promise as possible neuroprotectants in the setup of acute ischemic stroke but more studies are needed before clinical trials in humans could be undertaken.
Collapse
Affiliation(s)
- R R Leker
- Department of Neurology and the Agnes Ginges Center for Human Neurogenetics, Hebrew University-Hadassah Medical School, Hadassah University Hospital, Jerusalem, Israel.
| | | |
Collapse
|
49
|
Kelly A, Vereker E, Nolan Y, Brady M, Barry C, Loscher CE, Mills KHG, Lynch MA. Activation of p38 plays a pivotal role in the inhibitory effect of lipopolysaccharide and interleukin-1 beta on long term potentiation in rat dentate gyrus. J Biol Chem 2003; 278:19453-62. [PMID: 12609991 DOI: 10.1074/jbc.m301938200] [Citation(s) in RCA: 142] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Lipopolysaccharide (LPS), a component of the cell wall of Gram-negative bacteria, has been shown to induce profound changes both peripherally and centrally. It has recently been reported that intraperitoneal injection of LPS inhibited long term potentiation (LTP) in perforant path-granule cell synapses and that this effect was coupled with an increase in the concentration of the proinflammatory cytokine, interleukin-1 beta (IL-1 beta). The LPS-induced effects were abrogated by inhibition of caspase-1, suggesting that IL-1 beta may mediate the effects of LPS. Here we report that the inhibition of LTP induced by LPS and IL-1 beta was coupled with stimulation of the stress-activated protein kinase p38 in hippocampus and entorhinal cortex and that this effect was abrogated by the p38 inhibitor SB203580, while the effect of LPS was markedly attenuated in C57BL/6 IL-1RI-/- mice. The data also indicate that activation of the transcription factor, nuclear factor kappa B (NF kappa B), may play a role, since the inhibitory effect of LPS and IL-1 beta on LTP was attenuated by the NF kappa B inhibitor, SN50; consistently, LPS and IL-1 beta led to activation of NF kappa B in entorhinal cortex. We suggest that one consequence of these LPS and IL-1 beta-induced changes is a compromise in glutamate release in dentate gyrus, which was coupled with the inhibition of LTP. The evidence is consistent with the idea that the LPS-induced impairment in LTP is mediated by IL-1 beta and is a consequence of activation of p38.
Collapse
Affiliation(s)
- Aine Kelly
- Trinity College Institute of Neuroscience, Department of Physiology, Dublin 2, Ireland.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Han HS, Karabiyikoglu M, Kelly S, Sobel RA, Yenari MA. Mild hypothermia inhibits nuclear factor-kappaB translocation in experimental stroke. J Cereb Blood Flow Metab 2003; 23:589-98. [PMID: 12771574 DOI: 10.1097/01.wcb.0000059566.39780.8d] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Nuclear factor-kappaB (NFkappaB) is a transcription factor that is activated after cerebral ischemia. NFkappaB activation leads to the expression of many inflammatory genes involved in the pathogenesis of stroke. The authors previously showed that mild hypothermia is protective even when cooling begins 2 h after stroke onset. In the present study, they examined the influence of hypothermia on NFkappaB activation. Rats underwent 2 h of transient middle cerebral artery occlusion. Brains were cooled to 33 degrees C immediately after or 2 h after occlusion, and maintained for 2 h. After normothermic ischemia (brain temperature at 38 degrees C), NFkappaB cytoplasmic expression, nuclear translocation, and binding activity were observed as early as 2 h in the ischemic hemisphere and persisted at 24 h. Hypothermia decreased NFkappaB translocation and binding activity but did not alter overall expression. Hypothermia also affected the levels of NFkappaB regulatory proteins by suppressing phosphorylation of NFkappaB's inhibitory protein (IkappaB-alpha) and IkappaB kinase (IKK-gamma) and decreasing IKK activity, but did not alter overall IKK levels. Hypothermia suppressed the expression of two NFkappaB target genes: inducible nitric oxide synthase and TNF-alpha. These data suggest that the protective effect of hypothermia on cerebral injury is, in part, related to NFkappaB inhibition due to decreased activity of IKK.
Collapse
Affiliation(s)
- Hyung Soo Han
- Department of Neurosurgery, Stanford University Medical Center, Stanford, California 94305, U.S.A
| | | | | | | | | |
Collapse
|