1
|
Ahn W, Chi G, Kim S, Son Y, Zhang M. Substance P Reduces Infarct Size and Mortality After Ischemic Stroke, Possibly Through the M2 Polarization of Microglia/Macrophages and Neuroprotection in the Ischemic Rat Brain. Cell Mol Neurobiol 2023; 43:2035-2052. [PMID: 36112332 PMCID: PMC11412183 DOI: 10.1007/s10571-022-01284-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 09/08/2022] [Indexed: 12/12/2022]
Abstract
Substance-P (SP) is an 11 amino acid neuropeptide that is known to stimulate the peripheral mobilization of bone marrow mesenchymal stem cells and M2 polarization in monocytes/macrophages in a variety of acute and chronic tissue injuries. To examine the role of SP in protection and recovery from acute ischemic brain injury, experimental ischemic stroke was induced by transient middle cerebral artery occlusion (tMCAo) in rats for 1 h with subsequent reperfusion. Two injections of SP, immediately and one day post-tMCAo, resulted in approximately threefold lower mortality and 40% less infarct volume than those of saline-treated rats at seven days post-tMCAo. At 4.5 h, SP markedly increased CD11b/c+CD163+/CD 206+ cells in the blood, which were concomitantly decreased in the bone marrow, suggesting that SP preferentially mobilized M2-polarized monocytes. After two days, SP increased the expression of neuroprotective and anti-inflammatory genes in the ischemic brain and induced neuronal survival in the brain penumbra. Additionally, SP markedly increased CD68+CD163+ and CD68+CD206+ M2 microglia/macrophages in the ischemic brain during seven days post-tMCAo. Furthermore, SP preserved the blood‒brain barrier in the ischemic brain, which was confirmed by the abundant levels of SMI71+ brain endothelial cells that colocalized with α-SMA+ pericytes. The beneficial effects of SP on functional recovery and tissue preservation were maintained for six weeks. Collectively, SP treatment in the early phase of ischemic stroke markedly suppressed the destructive inflammatory response and improved the microenvironment for tissue protection and repair.
Collapse
Affiliation(s)
- Woosung Ahn
- Department of Genetics and Biotechnology, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Seocheon-dong, Kiheung-gu 446-701, Yongin-Si, Republic of Korea
| | - Guangfan Chi
- Department of Genetics and Biotechnology, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Seocheon-dong, Kiheung-gu 446-701, Yongin-Si, Republic of Korea
| | - Sumin Kim
- Department of Genetics and Biotechnology, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Seocheon-dong, Kiheung-gu 446-701, Yongin-Si, Republic of Korea
| | - Youngsook Son
- Department of Genetics and Biotechnology, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Seocheon-dong, Kiheung-gu 446-701, Yongin-Si, Republic of Korea.
| | - Mingzi Zhang
- Department of Genetics and Biotechnology, College of Life Science and Graduate School of Biotechnology, Kyung Hee University, Seocheon-dong, Kiheung-gu 446-701, Yongin-Si, Republic of Korea
| |
Collapse
|
2
|
Burda R, Burda J, Morochovič R. Ischemic Tolerance—A Way to Reduce the Extent of Ischemia–Reperfusion Damage. Cells 2023; 12:cells12060884. [PMID: 36980225 PMCID: PMC10047660 DOI: 10.3390/cells12060884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/08/2023] [Accepted: 03/11/2023] [Indexed: 03/14/2023] Open
Abstract
Individual tissues have significantly different resistance to ischemia–reperfusion damage. There is still no adequate treatment for the consequences of ischemia–reperfusion damage. By utilizing ischemic tolerance, it is possible to achieve a significant reduction in the extent of the cell damage due to ischemia–reperfusion injury. Since ischemia–reperfusion damage usually occurs unexpectedly, the use of preconditioning is extremely limited. In contrast, postconditioning has wider possibilities for use in practice. In both cases, the activation of ischemic tolerance can also be achieved by the application of sublethal stress on a remote organ. Despite very encouraging and successful results in animal experiments, the clinical results have been disappointing so far. To avoid the factors that prevent the activation of ischemic tolerance, the solution has been to use blood plasma containing tolerance effectors. This plasma is taken from healthy donors in which, after exposure to two sublethal stresses within 48 h, effectors of ischemic tolerance occur in the plasma. Application of this activated plasma to recipient animals after the end of lethal ischemia prevents cell death and significantly reduces the consequences of ischemia–reperfusion damage. Until there is a clear chemical identification of the end products of ischemic tolerance, the simplest way of enhancing ischemic tolerance will be the preparation of activated plasma from young healthy donors with the possibility of its immediate use in recipients during the initial treatment.
Collapse
Affiliation(s)
- Rastislav Burda
- Department of Trauma Surgery, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Rastislavova 43, 040 01 Košice, Slovakia
- Department of Trauma Surgery, Louis Pasteur University Hospital, Rastislavova 43, 040 01 Košice, Slovakia
- Correspondence:
| | - Jozef Burda
- Institute of Neurobiology, Slovak Academy of Sciences, 040 01 Košice, Slovakia
| | - Radoslav Morochovič
- Department of Trauma Surgery, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Rastislavova 43, 040 01 Košice, Slovakia
- Department of Trauma Surgery, Louis Pasteur University Hospital, Rastislavova 43, 040 01 Košice, Slovakia
| |
Collapse
|
3
|
Gao K, Liu M, Li Y, Wang L, Zhao C, Zhao X, Zhao J, Ding Y, Tang H, Jia Y, Wang J, Wen A. Lyciumamide A, a dimer of phenolic amide, protects against NMDA-induced neurotoxicity and potential mechanisms in vitro. J Mol Histol 2021; 52:449-459. [PMID: 33755822 DOI: 10.1007/s10735-020-09952-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 12/28/2020] [Indexed: 12/24/2022]
Abstract
Currently, the excessive activation of N-methyl-D-aspartate receptors (NMDARs) is considered to be a crucial mechanism of brain injury. Lycium barbarum A (LyA) is a dimer of phenol amides isolated from the fruit of Lycium barbarum. Our previous studies have shown that LyA has potential antioxidant activity. This study aimed to explore the neuroprotective effect of LyA and its potential mechanism. Firstly, the molecular docking was used to preliminarily explore the potential function of LyA to block NMDAR. Then, the ability of LyA was further verified by NMDA-induced human neuroblastoma SH-SY5Y cells in vivo. Treatment with LyA significantly attenuated NMDA-induced neuronal insults by increasing cell viability, reducing lactate dehydrogenase (LDH) release, and increasing cell survival. Meanwhile, LyA significantly reversed the increase in intracellular calcium and in ROS production induced by NMDA. Finally, the western blot indicated that LyA could suppress the Ca2+ influx and increase the p-NR2B, p-CaMKII, p-JNK, and p-p38 level induced by NMDA. These above findings provide evidence that LyA protect against brain injury, and restraining NMDARs and suppressing mitochondrial oxidative stress and inhibiting cell apoptosis may be involved in the protective mechanism.
Collapse
Affiliation(s)
- Kai Gao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Meiyou Liu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yuan Li
- Department of Pharmacy, Xi'an Children's Hospital, Xi'an, China
| | - Lei Wang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Chao Zhao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xian Zhao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jinyi Zhao
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yi Ding
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Haifeng Tang
- Institute of Materia Medica, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Yanyan Jia
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Jingwen Wang
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Aidong Wen
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
4
|
Hao Y, Xin M, Feng L, Wang X, Wang X, Ma D, Feng J. Review Cerebral Ischemic Tolerance and Preconditioning: Methods, Mechanisms, Clinical Applications, and Challenges. Front Neurol 2020; 11:812. [PMID: 33071923 PMCID: PMC7530891 DOI: 10.3389/fneur.2020.00812] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/29/2020] [Indexed: 12/13/2022] Open
Abstract
Stroke is one of the leading causes of morbidity and mortality worldwide, and it is increasing in prevalence. The limited therapeutic window and potential severe side effects prevent the widespread clinical application of the venous injection of thrombolytic tissue plasminogen activator and thrombectomy, which are regarded as the only approved treatments for acute ischemic stroke. Triggered by various types of mild stressors or stimuli, ischemic preconditioning (IPreC) induces adaptive endogenous tolerance to ischemia/reperfusion (I/R) injury by activating a multitude cascade of biomolecules, for example, proteins, enzymes, receptors, transcription factors, and others, which eventually lead to transcriptional regulation and epigenetic and genomic reprogramming. During the past 30 years, IPreC has been widely studied to confirm its neuroprotection against subsequent I/R injury, mainly including local ischemic preconditioning (LIPreC), remote ischemic preconditioning (RIPreC), and cross preconditioning. Although LIPreC has a strong neuroprotective effect, the clinical application of IPreC for subsequent cerebral ischemia is difficult. There are two main reasons for the above result: Cerebral ischemia is unpredictable, and LIPreC is also capable of inducing unexpected injury with only minor differences to durations or intensity. RIPreC and pharmacological preconditioning, an easy-to-use and non-invasive therapy, can be performed in a variety of clinical settings and appear to be more suitable for the clinical management of ischemic stroke. Hoping to advance our understanding of IPreC, this review mainly focuses on recent advances in IPreC in stroke management, its challenges, and the potential study directions.
Collapse
Affiliation(s)
| | | | | | | | | | - Di Ma
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jiachun Feng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
5
|
Qin Y, Zhang Q, Liu Y. Analysis of knowledge bases and research focuses of cerebral ischemia-reperfusion from the perspective of mapping knowledge domain. Brain Res Bull 2019; 156:15-24. [PMID: 31843561 DOI: 10.1016/j.brainresbull.2019.12.004] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 12/03/2019] [Indexed: 11/17/2022]
Abstract
Cerebral ischemia-reperfusion (IR) has attracted wide attention as a serious clinical problem. So far, the field has accumulated a large amount of scientific research literature. To clarify the temporal and spatial distribution characteristics of research resources, knowledge bases and research focuses, a visual analysis was performed on 5814 articles cited in the WoS databases from 2004 to 2019. This analysis was based on bibliometrics and mapping knowledge domain (MKD) analysis with VOSviewer, and CiteSpace 5.4.R4. The results can be elaborated from four aspects. First, the volume of publications in this area is on the rise. Second, the United States and China are the active regions. The USA is the central region of cerebral ischemia-reperfusion research. Third, the knowledge bases of IR have focused on five major areas of "Suitable small-animal models", "A framework with further study", "Molecular signaling targets by oxidative stress", "Finding new potential targets for therapy" and "Protective effect of multiple transient ischemia". Fourth, the research focuses consist of three representative areas: "Oxidative stress closelyd with cerebral ischemia-reperfusion", "Neuronal apoptosis and neuronal protection", and "Neuroprotective effect of the blood-brain barrier".
Collapse
Affiliation(s)
- Yi Qin
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China; Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China
| | - Qing Zhang
- No.4 Hospital Beijing University of Chinese Medicine, Zaozhuang, Shandong 277000
| | - Yaru Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
6
|
Huang L, Wu S, Li H, Dang Z, Wu Y. Hypoxic preconditioning relieved ischemic cerebral injury by promoting immunomodulation and microglia polarization after middle cerebral artery occlusion in rats. Brain Res 2019; 1723:146388. [PMID: 31421131 DOI: 10.1016/j.brainres.2019.146388] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 12/30/2022]
Abstract
OBJECTIVES This study was designed to investigate whether immunomodulation and Microglia polarization is involved in the anti-inflammatory and neuroprotective effect induced by hypoxic preconditioning (HPC) in the middle cerebral artery occlusion (MCAO) brain injury model. METHODS Longa method, (neurological disability status scale) NDSS method and TTC staining were used to evaluate the degree of cerebral infarction injury under different treatments (Sham, HPC, MCAO and co-treatment with HPC and MCAO). Western blot was used to detect expression profiles of apoptosis and related factors of neurological function. Flow cytometry was performed to analyze changes in the ratio of helper T cells, toxic T cells and NK cells in peripheral immune cells. And immunohistochemistry was used to examine the changes in microglial morphology. ELISA was used to evaluate the levels of nerve growth factors and neurogenesis conditions. Finally, RT-PCR was determined to analyze the transformation of microglia phenotype after HPC and MCAO treatment. RESULTS MCAO dramatically induced local formation of cerebral infarction. HPC relieved MCAO-induced cerebral infarction and increased rat cognition. HPC affected activation of microglia without significantly affecting in peripheral immune cell populations. After HPC co-treatment with MCAO, the M1 phenotype of microglia was changed and there was a transformation to M2. CONCLUSION The treatment of HPC remarkably affected the polarization of microglia cells in MCAO rats, and reduced the cerebral nerve injury and played a protective role in MCAO model.
Collapse
Affiliation(s)
- Lu Huang
- Research Center for High Altitude Medicine, Qinghai University, Xining, China; Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Loint Research Key Lab for High Altitude Medicine), Xining, China; Qinghai Provincial People's Hospital, Xining, China
| | - Shizheng Wu
- Qinghai Provincial People's Hospital, Xining, China.
| | - Hao Li
- Qinghai Provincial People's Hospital, Xining, China
| | - Zhancui Dang
- Qinghai University Medical College, Xining, China
| | - Yue Wu
- Qinghai University, Qinghai, China
| |
Collapse
|
7
|
Colpo GD, Venna VR, McCullough LD, Teixeira AL. Systematic Review on the Involvement of the Kynurenine Pathway in Stroke: Pre-clinical and Clinical Evidence. Front Neurol 2019; 10:778. [PMID: 31379727 PMCID: PMC6659442 DOI: 10.3389/fneur.2019.00778] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/03/2019] [Indexed: 12/27/2022] Open
Abstract
Background: Stroke is the second leading cause of death after ischemic heart disease and the third leading cause of disability-adjusted life-years lost worldwide. There is a great need for developing more effective strategies to treat stroke and its resulting impairments. Among several neuroprotective strategies tested so far, the kynurenine pathway (KP) seems to be promising, but the evidence is still sparse. Methods: Here, we performed a systematic review of preclinical and clinical studies evaluating the involvement of KP in stroke. We searched for the keywords: (“kynurenine” or “kynurenic acid” or “quinolinic acid”) AND (“ischemia” or “stroke” or “occlusion) in the electronic databases PubMed, Scopus, and Embase. A total of 1,130 papers was initially retrieved. Results: After careful screening, forty-five studies were included in this systematic review, being 39 pre-clinical and six clinical studies. Despite different experimental models of cerebral ischemia, the results are concordant in implicating the KP in the pathophysiology of stroke. Preclinical evidence also suggests that treatment with kynurenine and KMO inhibitors decrease infarct size and improve behavioral and cognitive outcomes. Few studies have investigated the KP in human stroke, and results are consistent with the experimental findings that the KP is activated after stroke. Conclusion: Well-designed preclinical studies addressing the expression of KP enzymes and metabolites in specific cell types and their potential effects at cellular levels alongside more clinical studies are warranted to confirm the translational potential of this pathway as a pharmacological target for stroke and related complications.
Collapse
Affiliation(s)
- Gabriela D Colpo
- Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Venugopal R Venna
- BRAINS Lab, Department of Neurology, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Louise D McCullough
- BRAINS Lab, Department of Neurology, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Antonio L Teixeira
- Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
8
|
Li S, Hafeez A, Noorulla F, Geng X, Shao G, Ren C, Lu G, Zhao H, Ding Y, Ji X. Preconditioning in neuroprotection: From hypoxia to ischemia. Prog Neurobiol 2017; 157:79-91. [PMID: 28110083 DOI: 10.1016/j.pneurobio.2017.01.001] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 01/08/2017] [Accepted: 01/13/2017] [Indexed: 01/05/2023]
Abstract
Sublethal hypoxic or ischemic events can improve the tolerance of tissues, organs, and even organisms from subsequent lethal injury caused by hypoxia or ischemia. This phenomenon has been termed hypoxic or ischemic preconditioning (HPC or IPC) and is well established in the heart and the brain. This review aims to discuss HPC and IPC with respect to their historical development and advancements in our understanding of the neurochemical basis for their neuroprotective role. Through decades of collaborative research and studies of HPC and IPC in other organ systems, our understanding of HPC and IPC-induced neuroprotection has expanded to include: early- (phosphorylation targets, transporter regulation, interfering RNA) and late- (regulation of genes like EPO, VEGF, and iNOS) phase changes, regulators of programmed cell death, members of metabolic pathways, receptor modulators, and many other novel targets. The rapid acceleration in our understanding of HPC and IPC will help facilitate transition into the clinical setting.
Collapse
Affiliation(s)
- Sijie Li
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuan Wu Hospital, Capital Medical University, Beijing, China; National Clinical Research Center for Geriatric Disorders, Beijing, China
| | - Adam Hafeez
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Fatima Noorulla
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Xiaokun Geng
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA; Department of Neurology, Luhe Hospital, Capital Medical University, Beijing, China
| | - Guo Shao
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Changhong Ren
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Guowei Lu
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuan Wu Hospital, Capital Medical University, Beijing, China
| | - Heng Zhao
- Department of Neurosurgery, Stanford University, CA, USA
| | - Yuchuan Ding
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuan Wu Hospital, Capital Medical University, Beijing, China; Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI, USA
| | - Xunming Ji
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuan Wu Hospital, Capital Medical University, Beijing, China; National Clinical Research Center for Geriatric Disorders, Beijing, China.
| |
Collapse
|
9
|
Wang W, Liu L, Jiang P, Chen C, Zhang T. Levodopa improves learning and memory ability on global cerebral ischemia-reperfusion injured rats in the Morris water maze test. Neurosci Lett 2016; 636:233-240. [PMID: 27856221 DOI: 10.1016/j.neulet.2016.11.026] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 11/01/2016] [Accepted: 11/10/2016] [Indexed: 11/24/2022]
Abstract
Previous studies have shown that levodopa (L-dopa) for 1-7days improved the consciousness level of certain patients who suffered from ischemia-reperfusion injury and were comatose for a long time period after cerebral resuscitation treatment. It also has an awakening effect on patients with disorders of consciousness. This study aimed to investigate whether L-dopa, which is used clinically to treat Parkinson's disease, might also ameliorate the behavior of rats following global cerebral ischemia-reperfusion injury. Fifty-six healthy adult male Sprague-Dawley rats were randomly divided into four groups: shamoperated, global cerebral ischemia mode, 25mg/kg/d L-dopa intervention, and 50mg/kg/d L-dopa intervention. The level of consciousness and modified neurological severity score (NSS) of the rats in each group were measured before reperfusion and 6, 24, and 72h and 1-4 weeks after reperfusion. The Morris water maze test was used to assess behavior of rats 1 week after reperfusion and 2 weeks after reperfusion in each group. The results showed that after global cerebral ischemiareperfusion injury, neurological deficits of rats are severe, and space exploration capacity and learning and memory capacity are significantly decreased. L-dopa can shorten the duration of coma in rats following global cerebral ischemia-reperfusion injury and improve the symptoms of neurological deficits and advanced learning and memory. In the range of the selected doses, the relationship between L-dopa and improvement of the neurological behavior in rats was not dose-dependent. Dopamine may be useful for treating severe ischemia-reperfusion brain injury.
Collapse
Affiliation(s)
- Wenzhu Wang
- Institute of Rehabilitation Medicine of China, Chinese Institute of Rehabilitation Science, China Rehabilitation Research Center, Beijing, PR China; Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing, PR China; Neural Injury and Repair Center, Beijing Institute for Brain Disorders, Beijing, PR China
| | - Lixu Liu
- School of Rehabilitation, Capital Medical University, Beijing, PR China; Beijing Bo'ai Hospital, China Rehabilitation Research Center, Beijing, PR China.
| | - Peng Jiang
- School of Rehabilitation, Capital Medical University, Beijing, PR China
| | - Chen Chen
- School of Rehabilitation, Capital Medical University, Beijing, PR China
| | - Tong Zhang
- School of Rehabilitation, Capital Medical University, Beijing, PR China; Beijing Bo'ai Hospital, China Rehabilitation Research Center, Beijing, PR China
| |
Collapse
|
10
|
Ahmed ME, Dong Y, Lu Y, Tucker D, Wang R, Zhang Q. Beneficial Effects of a CaMKIIα Inhibitor TatCN21 Peptide in Global Cerebral Ischemia. J Mol Neurosci 2016; 61:42-51. [PMID: 27604243 DOI: 10.1007/s12031-016-0830-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 08/30/2016] [Indexed: 12/16/2022]
Abstract
Aberrant calcium influx is a common feature following ischemic reperfusion (I/R) in transient global cerebral ischemia (GCI) and causes delayed neuronal cell death in the CA1 region of the hippocampus. Activation of calcium-calmodulin (CaM)-dependent protein kinase IIα (CaMKIIα) is a key event in calcium signaling in ischemic injury. The present study examined the effects of intracerebroventricular (icv) injection of tatCN21 in ischemic rats 3 h after GCI reperfusion. Cresyl violet and NeuN staining revealed that tatCN21 exerted neuroprotective effects against delayed neuronal cell death of hippocampal CA1 pyramidal neurons 10 days post-GCI. In addition, TatCN21 administration ameliorated GCI-induced spatial memory deficits in the Barnes maze task as well as anxiety-like behaviors and spontaneous motor activity in the elevated plus maze and open field test, respectively. Mechanistic studies showed that the administration of tatCN21 decreased GCI-induced phosphorylation, translocation, and membrane targeting of CaMKIIα. Treatment with tatCN21 also inhibited the level of CaMKIIα-NR2B interaction and NR2B phosphorylation. Our results revealed an important role of tatCN21 in inhibiting CaMKIIα activation and its beneficial effects in neuroprotection and memory preservation in an ischemic brain injury model.
Collapse
Affiliation(s)
- Mohammad Ejaz Ahmed
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Yan Dong
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Yujiao Lu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Donovan Tucker
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Ruimin Wang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Quanguang Zhang
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA.
| |
Collapse
|
11
|
Hye Kim I, Lee JC, Ha Park J, Hyeon Ahn J, Cho JH, Hui Chen B, Na Shin B, Chun Yan B, Rueol Ryu D, Hong S, Hwi Cho J, Lyul Lee Y, Kim YM, Cho BR, Won MH. Time interval after ischaemic preconditioning affects neuroprotection and gliosis in the gerbil hippocampal CA1 region induced by transient cerebral ischaemia. Neurol Res 2016; 38:210-9. [DOI: 10.1179/1743132815y.0000000098] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
12
|
Wang W, Zhao L, Bai F, Zhang T, Dong H, Liu L. The protective effect of dopamine against OGD/R injury-induced cell death in HT22 mouse hippocampal cells. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 42:176-182. [PMID: 26867202 DOI: 10.1016/j.etap.2016.01.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 01/21/2016] [Accepted: 01/23/2016] [Indexed: 06/05/2023]
Abstract
Previous studies have shown that levo-dopamine (L-dopa) can improve the consciousness of certain patients with prolonged coma after cerebral ischemia-reperfusion injury, and promote cell growth in vivo. This study aimed to investigate whether L-dopa, which is used clinically to treat Parkinson's disease, might also ameliorate ischemia-reperfusion injury-induced cell death. The oxygen-glucose deprivation and re-oxygenation (OGD/R) model was used to mimic the ischemia-reperfusion pathological process in vitro. HT22 cells were treated with dopamine hydrochloride at different times (i.e., 2 h prior to OGD, during the period of OGD, during the period of R, and throughout the period of OGD/R) and at different concentrations (i.e., 25 μM, 50 μM, 100 μM). Lactate dehydrogenase (LDH) release, flow cytometry-annexin V, and propidium iodide staining with light microscopy showed that dopamine hydrochloride (added during re-oxygenation) promoted cell proliferation and facilitated maintenance of normal cell morphology. However, when present during oxygen-glucose deprivation for 18 h and present throughout OGD/R, dopamine hydrochloride increased cell damage as manifested by shrinkage, rounding up, and reduced viability. In conclusion, dopamine protected HT22 cells from OGD/R injury-induced cell death only at a particular point in time, suggesting that it may be useful for treating severe ischemia-reperfusion brain injury.
Collapse
Affiliation(s)
- Wenzhu Wang
- Institute of Rehabilitation Medicine of China, Chinese Institute of Rehabilitation Science, China Rehabilitation Research Center, Beijing, PR China; Beijing Key Laboratory of Neural Injury and Rehabilitation, PR China
| | - Lixi Zhao
- Institute of Rehabilitation Medicine of China, Chinese Institute of Rehabilitation Science, China Rehabilitation Research Center, Beijing, PR China; Beijing Key Laboratory of Neural Injury and Rehabilitation, PR China
| | - Fan Bai
- Institute of Rehabilitation Medicine of China, Chinese Institute of Rehabilitation Science, China Rehabilitation Research Center, Beijing, PR China; Beijing Key Laboratory of Neural Injury and Rehabilitation, PR China
| | - Tong Zhang
- Beijing Bo Ai Hospital, China Rehabilitation Research Center, Beijing, PR China; School of Rehabilitation Medicine, Capital Medical University, Beijing, PR China
| | - Hao Dong
- Beijing Bo Ai Hospital, China Rehabilitation Research Center, Beijing, PR China; Institute of Rehabilitation Medicine of China, Chinese Institute of Rehabilitation Science, China Rehabilitation Research Center, Beijing, PR China
| | - Lixu Liu
- Beijing Bo Ai Hospital, China Rehabilitation Research Center, Beijing, PR China; School of Rehabilitation Medicine, Capital Medical University, Beijing, PR China.
| |
Collapse
|
13
|
Wang M, Qi DS, Zhou C, Han D, Li PP, Zhang F, Zhou XY, Han M, Di JH, Ye JS, Yu HM, Song YJ, Zhang GY. Ischemic preconditioning protects the brain against injury via inhibiting CaMKII-nNOS signaling pathway. Brain Res 2016; 1634:140-149. [PMID: 26794251 DOI: 10.1016/j.brainres.2016.01.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 12/31/2015] [Accepted: 01/06/2016] [Indexed: 01/23/2023]
Abstract
Although studies have shown that cerebral ischemic preconditioning (IPC) can ameliorate ischemia/reperfusion (I/R) induced brain damage, but its precise mechanisms remain unknown. Therefore, the aim of this study was to investigate the neuroprotective mechanisms of IPC against ischemic brain damage induced by cerebral I/R and to explore whether the Calcium/calmodulin-dependent protein kinase II (CaMKII)-mediated up-regulation of nNOS ser847-phosphorylation signaling pathway contributed to the protection provided by IPC. Transient global brain ischemia was induced by 4-vessel occlusion in adult male Sprague-Dawley rats. The rats were pretreated with 3 min of IPC alone or KN62 (selective antagonist of CaMKII) treatment before IPC, after reperfusion for 3 days, 6 min ischemia was induced. Cresyl violet staining was used to examine the survival of hippocampal CA1 pyramidal neurons. Immunoblotting was performed to measure the phosphorylation of CaMKII, nNOS, c-Jun and the expression of FasL. Immunoprecipitation was used to examine the binding between PSD95 and nNOS. The results showed that IPC could significantly protect neurons against cerebral I/R injury, furthermore, the combination of PSD95 and nNOS was increased, coinstantaneously the phosphorylation of CaMKII and nNOS (ser847) were up-regulated, however the activation of c-Jun and FasL were reduced. Conversely, KN62 treatment before IPC reversed all these effects of IPC. Taken together, the results suggest that IPC could diminish ischemic brain injury through CaMKII-mediated up-regulation of nNOS ser847-phosphorylation signaling pathway.
Collapse
Affiliation(s)
- Mei Wang
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, Jiangsu 221004, PR China; Laboratory of Morphology, Xuzhou Medical College, Xuzhou, Jiangsu 221004, PR China
| | - Da-Shi Qi
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, Jiangsu 221004, PR China; Department of Genetics, Xuzhou Medical College, Xuzhou, Jiangsu 221004, PR China
| | - Cui Zhou
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, Jiangsu 221004, PR China
| | - Dong Han
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, Jiangsu 221004, PR China
| | - Pei-Pei Li
- Department of Endocrine, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, PR China
| | - Fang Zhang
- Laboratory of Morphology, Xuzhou Medical College, Xuzhou, Jiangsu 221004, PR China
| | - Xiao-Yan Zhou
- Laboratory of Morphology, Xuzhou Medical College, Xuzhou, Jiangsu 221004, PR China
| | - Meng Han
- Department of Orthopaedics, Xuzhou Central Hospital, Xuzhou, Jiangsu 221009, PR China
| | - Jie-Hui Di
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, Jiangsu 221004, PR China
| | - Jun-Song Ye
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, Jiangsu 221004, PR China
| | - Hong-Min Yu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, Jiangsu 221004, PR China
| | - Yuan-Jian Song
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, Jiangsu 221004, PR China; Department of Genetics, Xuzhou Medical College, Xuzhou, Jiangsu 221004, PR China.
| | - Guang-Yi Zhang
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, Jiangsu 221004, PR China.
| |
Collapse
|