1
|
Bagheri SM, Allahtavakoli M, Hakimizadeh E. Neuroprotective effect of ischemic postconditioning against hyperperfusion and its mechanisms of neuroprotection. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2024; 29:31. [PMID: 39239075 PMCID: PMC11376715 DOI: 10.4103/jrms.jrms_341_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/13/2023] [Accepted: 06/27/2023] [Indexed: 09/07/2024]
Abstract
Background In recent years, stroke and ischemia-reperfusion injury has motivated researchers to find new ways to reduce the complications. Although reperfusion is essential for brain survival, it is like a double-edged sword that may cause further damage to the brain. Ischemic postconditioning (IPostC) refers to the control of blood flow in postischemia-reperfusion that can reduce ischemia-reperfusion injuries. Materials and Methods Articles were collected by searching for the terms: Ischemic postconditioning and neuroprotective and ischemic postconditioning and hyperperfusion. Suitable articles were collected from electronic databases, including ISI Web of Knowledge, Medline/PubMed, ScienceDirect, Embase, Scopus, Biological Abstract, Chemical Abstract, and Google Scholar. Results New investigations show that IPostC has protection against hyperperfusion by reducing the amount of blood flow during reperfusion and thus reducing infarction volume, preventing the blood-brain barrier damage, and reducing the rate of apoptosis through the activation of innate protective systems. Numerous mechanisms have been suggested for IPostC, which include reduction of free radical production, apoptosis, inflammatory factors, and activation of endogenous protective pathways. Conclusion It seems that postconditioning can prevent damage to the brain by reducing the flow and blood pressure caused by hyperperfusion. It can protect the brain against damages such as stroke and hyperperfusion by activating various endogenous protection systems. In the present review article, we tried to evaluate both useful aspects of IPostC, neuroprotective effects, and fight against hyperperfusion.
Collapse
Affiliation(s)
- Seyyed Majid Bagheri
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Allahtavakoli
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Physiology and Pharmacology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Elham Hakimizadeh
- Physiology-Pharmacology Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
2
|
Sun L, Wu G, Zhou Y, Deng A, Chen Z. Prospective study on ultrasound-guided stellate ganglion block improves cerebral blood flow in patients with stroke. J Stroke Cerebrovasc Dis 2024; 33:107593. [PMID: 38290686 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 02/01/2024] Open
Abstract
OBJECTIVES The effect of routine internal medicine and stroke rehabilitation treatment was not good. To confirm that ultrasound-guided stellate ganglion block (SGB) can improve cerebral blood flow in patients with stroke, Transcranial Doppler (TCD) and carotid ultrasound were used to monitor the cerebral blood flow parameters of ultrasound-guided SGB in patients with stroke. METHODS A prospective study of 40 patients with stroke from January 2021 to October 2022 randomly divided into two groups (group SGB: undergoing ultrasound-guided SGB and standard medical procedures, control group: undergoing standard medical procedures) with 20 cases in each was conducted in People's Hospital of Chongqing Liang Jiang New Area. TCD and carotid artery ultrasound were monitored before and after treatment. There were no significant differences in general data on age, gender, disease course, and stroke type between two groups (P>0.05). RESULTS After treatment, the bilateral ACA Vm of group SGB was significantly higher, the bilateral internal carotid artery RI and left VA RI were significantly lower than in control group (P<0.05). In group SGB, the Vm of bilateral MCA, bilateral PCA, right ACA, bilateral VA, and BA after treatment were significantly (P<0.05) increased compared to before treatment. PI of bilateral MCA, right ACA, and left VA after treatment were significantly (P<0.05) decreased compared to before treatment. RI of bilateral MCA, bilateral PCA, and bilateral VA after treatment were significantly (P<0.05) decreased compared to before treatment. Right internal carotid artery D after treatment was significantly (P<0.05) higher than before treatment. RI of bilateral internal carotid artery after treatment was significantly (P<0.05) lower than before treatment. CONCLUSIONS Ultrasound-guided stellate ganglion block could improve local cerebral blood flow and vascular compliance in patients with stroke, and reduce vascular resistance.
Collapse
Affiliation(s)
- Lin Sun
- Department of anesthesiology and pain, People's Hospital of Chongqing Liang Jiang New Area; Liangjiang New Area Chongqing 401121, China
| | - Gu Wu
- Department of anesthesiology and pain, People's Hospital of Chongqing Liang Jiang New Area; Liangjiang New Area Chongqing 401121, China
| | - Yuan Zhou
- Department of anesthesiology and pain, People's Hospital of Chongqing Liang Jiang New Area; Liangjiang New Area Chongqing 401121, China
| | - Ansong Deng
- Department of anesthesiology and pain, People's Hospital of Chongqing Liang Jiang New Area; Liangjiang New Area Chongqing 401121, China
| | - Zongjie Chen
- Department of anesthesiology and pain, People's Hospital of Chongqing Liang Jiang New Area; Liangjiang New Area Chongqing 401121, China.
| |
Collapse
|
3
|
Saccaro LF, Aimo A, Emdin M, Pico F. Remote Ischemic Conditioning in Ischemic Stroke and Myocardial Infarction: Similarities and Differences. Front Neurol 2021; 12:716316. [PMID: 34764925 PMCID: PMC8576053 DOI: 10.3389/fneur.2021.716316] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 09/23/2021] [Indexed: 11/13/2022] Open
Abstract
Acute myocardial infarction and ischemic stroke are leading causes of morbidity and mortality worldwide. Although reperfusion therapies have greatly improved the outcomes of patients with these conditions, many patients die or are severely disabled despite complete reperfusion. It is therefore important to identify interventions that can prevent progression to ischemic necrosis and limit ischemia-reperfusion injury. A possible strategy is ischemic conditioning, which consists of inducing ischemia – either in the ischemic organ or in another body site [i.e., remote ischemic conditioning (RIC), e.g., by inflating a cuff around the patient's arm or leg]. The effects of ischemic conditioning have been studied, alone or in combination with revascularization techniques. Based on the timing (before, during, or after ischemia), RIC is classified as pre-, per-/peri-, or post-conditioning, respectively. In this review, we first highlight some pathophysiological and clinical similarities and differences between cardiac and cerebral ischemia. We report evidence that RIC reduces circulating biomarkers of myocardial necrosis, infarct size, and edema, although this effect appears not to translate into a better prognosis. We then review cutting-edge applications of RIC for the treatment of ischemic stroke. We also highlight that, although RIC is a safe procedure that can easily be implemented in hospital and pre-hospital settings, its efficacy in patients with ischemic stroke remains to be proven. We then discuss possible methodological issues of previous studies. We finish by highlighting some perspectives for future research, aimed at increasing the efficacy of ischemic conditioning for improving tissue protection and clinical outcomes, and stratifying myocardial infarction and brain ischemia patients to enhance treatment feasibility.
Collapse
Affiliation(s)
- Luigi F Saccaro
- Neurology and Stroke Care Unit, Versailles Hospital, Le Chesnay, France.,Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Alberto Aimo
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.,Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Michele Emdin
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.,Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Fernando Pico
- Neurology and Stroke Care Unit, Versailles Hospital, Le Chesnay, France.,Neurology Department, Versailles Saint-Quentin-en-Yvelines and Paris Saclay University, Versailles, France
| |
Collapse
|
4
|
Bartels RHMA. A new dimension in degenerative cervical myelopathy. Lancet Neurol 2020; 20:82-83. [PMID: 33357511 DOI: 10.1016/s1474-4422(20)30454-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 11/23/2020] [Indexed: 10/22/2022]
Affiliation(s)
- Ronald H M A Bartels
- Department of Neurosurgery, Radboud University Medical Center, 6525 GA, Nijmegen, Netherlands.
| |
Collapse
|
5
|
Xu D, Li F, Xue G, Hou K, Fang W, Li Y. Effect of Wnt signaling pathway on neurogenesis after cerebral ischemia and its therapeutic potential. Brain Res Bull 2020; 164:1-13. [PMID: 32763283 DOI: 10.1016/j.brainresbull.2020.07.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 12/08/2019] [Accepted: 07/08/2020] [Indexed: 02/07/2023]
Abstract
Neurogenesis process in the chronic phase of ischemic stroke has become the focus of research on stroke treatment recently, mainly through the activation of related pathways to increase the differentiation of neural stem cells (NSCs) in the brain sub-ventricular zone (SVZ) and subgranular zone (SGZ) of hippocampal dentate gyrus (DG) areas into neurons, promoting neurogenesis. While there is still debate about the longevity of active adult neurogenesis in humans, the SVZ and SGZ have the capacity to upregulate neurogenesis in response to cerebral ischemia, which opens discussion about potential treatment strategies to harness this neuronal regenerative response. Wnt signaling pathway is one of the most important approaches potentially targeting on neurogenesis after cerebral ischemia, appropriate activation of which in NSCs may help to improve the sequelae of cerebral ischemia. Various therapeutic approaches are explored on preclinical stage to target endogenous neurogenesis induced by Wnt signaling after stroke onset. This article describes the composition of Wnt signaling pathway and the process of neurogenesis after cerebral ischemia, and emphatically introduces the recent studies on the mechanisms of this pathway for post-stroke neurogenesis and the therapeutic possibility of activating the pathway to improve neurogenesis after stroke.
Collapse
Affiliation(s)
- Dan Xu
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Fengyang Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Gou Xue
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Kai Hou
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Weirong Fang
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| | - Yunman Li
- State Key Laboratory of Natural Medicines, Department of Physiology, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
6
|
TGF-β3/Smad3 Contributes to Isoflurane Postconditioning Against Cerebral Ischemia–Reperfusion Injury by Upregulating MEF2C. Cell Mol Neurobiol 2020; 40:1353-1365. [DOI: 10.1007/s10571-020-00822-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 02/27/2020] [Indexed: 10/24/2022]
|
7
|
Zhao L, Liao Q, Zhang Y, Tan S, Li S, Ke T. Ischemic Postconditioning Mitigates Retinopathy in Tree Shrews with Diabetic Cerebral Ischemia. J Diabetes Res 2020; 2020:6286571. [PMID: 32104713 PMCID: PMC7037873 DOI: 10.1155/2020/6286571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 01/04/2020] [Accepted: 01/17/2020] [Indexed: 12/25/2022] Open
Abstract
Ischemic postconditioning (PC) is proved to efficiently protect diabetic patients with acute myocardial infarction from ischemia-reperfusion injury. We aimed to explore the protective roles of ischemic PC on diabetic retinopathy in tree shrews with diabetic cerebral ischemia. A diabetic tree shrew model was established through high-fat diet feeding combined with streptozotocin (STZ) injection, while cortical thrombotic cerebral ischemia was induced photochemically. Tree shrews were divided into the normal control group, sham operation group, diabetes mellitus group, diabetes mellitus+cerebral ischemia group, and diabetes mellitus+cerebral ischemia+PC group (in which the tree shrews with diabetic cerebral ischemia were treated with ischemic PC). H&E staining was used to examine the pathological changes in the retina, and immunohistochemistry was performed to determine the retinal expression of VEGF (vascular endothelial growth factor). The modeling resulted in 77% tree shrews with diabetes. Ischemic PC reduced the blood glucose levels in the tree shrews with diabetic cerebral ischemia. Tree shrews with diabetes had thinned retina with disordered structures, and these pathological changes were aggravated after cerebral ischemia. The retinopathy was alleviated after ischemic PC. Retina expression of VEGF was mainly distributed in the ganglion cell layer in tree shrews. Diabetes and cerebral ischemia increased retinal VEGF expression in a step-wise manner, while additional ischemic PC reduced retinal VEGF expression. Therefore, ischemic PC effectively alleviates retinopathy in tree shrews with diabetic cerebral ischemia, and this effect is associated with reduced retinal VEGF expression.
Collapse
Affiliation(s)
- Ling Zhao
- Department of Endocrinology, The Second Affiliated Hospital of Kunming Medical University, Yunnan 650101, China
| | - Qiwei Liao
- Department of Cardiology, The Yan-an Affiliated Hospital of Kunming Medical University, Yunnan 650051, China
| | - Yueting Zhang
- Department of Endocrinology, The Second Affiliated Hospital of Kunming Medical University, Yunnan 650101, China
| | - Shufen Tan
- Department of Gynecologic Oncology, The Third Affiliated Hospital of Kunming Medical University, Yunnan 650101, China
| | - Shuqing Li
- Department of Pathophysiology, Kunming Medical University, Yunnan 650050, China
| | - Tingyu Ke
- Department of Endocrinology, The Second Affiliated Hospital of Kunming Medical University, Yunnan 650101, China
| |
Collapse
|
8
|
Cheng X, Yang YL, Li WH, Liu M, Wang YH, Du GH. Cerebral ischemia-reperfusion aggravated cerebral infarction injury and possible differential genes identified by RNA-Seq in rats. Brain Res Bull 2019; 156:33-42. [PMID: 31877338 DOI: 10.1016/j.brainresbull.2019.12.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 11/12/2019] [Accepted: 12/19/2019] [Indexed: 10/25/2022]
Abstract
Numerous studies have shown that local excessive inflammatory response in brain tissue was an important pathogenesis of secondary injury following cerebral ischemia-reperfusion (I/R). However, the inflammatory-related targets and pathways after cerebral I/R injury are still unclear. This study was to investigate possible targets and mechanisms after cerebral I/R injury. Rats were subjected to transient or permanent middle cerebral artery occlusion (MCAO). Neurological deficit scores test was used to evaluate neurological function. Cerebral infarction was evaluated by MRI, TTC staining and Nissl staining. Microglia activation was detected by immunofluorescence using Iba-1 antibody. Inflammatory factors were detected by ELISA assay. RNA-sequencing transcriptome analysis was processed and the differential genes were verified by real-time quantitative PCR (qPCR) and western blotting. The results showed that neurological function of rats in I/R group was more severe than that in I group on the 7th after cerebral I/R. Therefore, the differences between cerebral ischemia and cerebral I/R for 7 days were studied in further study. The results showed that the levels of pro-inflammatory factors in I/R group were higher and the levels of anti-inflammatory factors were lower than those in I group. KEGG pathway and gene network enrichment analysis revealed that some common differential up- and down-regulated genes were involved in most of significant pathways. These common differential up-regulated genes belonged to TLR4/MYD88 inflammatory signaling pathway and common differential down-regulated genes belonged to HRAS/RAF1 neurotrophic signaling pathway. Interestingly, according to the genetic interaction analysis of string database, these up-regulated differential genes might promote the development of inflammation, while the down-regulated differential genes might inhibit the development of inflammation. Furthermore, qPCR and WB results verified that these pro-inflammatory genes in the I/R group were higher than those in the I group, while possible anti-inflammatory genes in the I/R group were lower than those in the I group. It is concluded that TLR4/MYD88 inflammatory signaling pathway and HRAS/RAF1 neurotrophic signaling pathway may play different roles after cerebral I or I/R and may be therapeutic targets for stroke recovery.
Collapse
Affiliation(s)
- Xiao Cheng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Target Identification and New Drug Sreeening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ying-Lin Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Target Identification and New Drug Sreeening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Wei-Han Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Target Identification and New Drug Sreeening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Man Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Target Identification and New Drug Sreeening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Yue-Hua Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Target Identification and New Drug Sreeening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Guan-Hua Du
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; Beijing Key Laboratory of Drug Target Identification and New Drug Sreeening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
9
|
Wang Y, Pan WY, Ge JS, Wang XD, Chen W, Luo X, Wang YL. A review of the relationship between long noncoding RNA and post-stroke injury repair. J Int Med Res 2019; 47:4619-4624. [PMID: 31526155 PMCID: PMC6833389 DOI: 10.1177/0300060519867493] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 07/12/2019] [Indexed: 11/15/2022] Open
Abstract
Stroke is a cerebrovascular circulation disorder with sudden onset, which causes disorder of ion balance, inflammation, and acidosis, and that in turn induces ischemia-reperfusion injury, influencing the prognosis of stroke patients. Long noncoding RNAs (lncRNAs) are regulatory sequences involved at the transcriptional, post-transcriptional, and epigenetic levels, have high specific expression in the central nervous system, and effectively regulate the development of the central nervous system and progression of diseases. Stroke induces changes in the expression of many lncRNAs. Therefore, lncRNAs play an important role in the complex pathological process of stroke. Exploring lncRNA could facilitate a comprehensive understanding of the pathological mechanism of stroke and the post-injury molecular regulatory network. However, there are few reports on the role of lncRNA in the pathological development of stroke. In the present review, we discuss the association of lncRNA with post-stroke injury repair.
Collapse
Affiliation(s)
- Yao Wang
- Department of Rehabilitation Medicine, Shenzhen Dapeng New District Nan’ao People’s Hospital, Shenzhen, China
| | - Wei-Yi Pan
- Department of Rehabilitation Medicine, Shenzhen Dapeng New District Nan’ao People’s Hospital, Shenzhen, China
| | - Jun-Sheng Ge
- Department of Rehabilitation Medicine, Shenzhen Dapeng New District Nan’ao People’s Hospital, Shenzhen, China
| | - Xiao-Dong Wang
- Department of Rehabilitation, Shenzhen Second People’s Hospital, The First Affiliated Hospital, Shenzhen University School of Medicine, Shenzhen, China
| | - Wei Chen
- Kerry Rehabilitation Medicine Research Institute, Shenzhen, Guangdong, China
| | - Xun Luo
- Kerry Rehabilitation Medicine Research Institute, Shenzhen, Guangdong, China
| | - Yu-Long Wang
- Department of Rehabilitation, Shenzhen Second People’s Hospital, The First Affiliated Hospital, Shenzhen University School of Medicine, Shenzhen, China
| |
Collapse
|
10
|
SOD2 Mediates Curcumin-Induced Protection against Oxygen-Glucose Deprivation/Reoxygenation Injury in HT22 Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:2160642. [PMID: 31662771 PMCID: PMC6791267 DOI: 10.1155/2019/2160642] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 08/24/2019] [Accepted: 09/10/2019] [Indexed: 02/07/2023]
Abstract
Curcumin (Cur) induces neuroprotection against brain ischemic injury; however, the mechanism is still obscure. The aim of this study is to explore the potential neuroprotective mechanism of curcumin against oxygen-glucose deprivation/reoxygenation (OGD/R) injury in HT22 cells and investigate whether type-2 superoxide dismutase (SOD2) is involved in the curcumin-induced protection. In the present study, HT22 neuronal cells were treated with 3 h OGD plus 24 h reoxygenation to mimic ischemia/reperfusion injury. Compared with the normal cultured control group, OGD/R treatment reduced cell viability and SOD2 expression, decreased mitochondrial membrane potential (MMP) and mitochondrial complex I activity, damaged cell morphology, and increased lactic dehydrogenase (LDH) release, cell apoptosis, intracellular reactive oxygen species (ROS), and mitochondrial superoxide (P < 0.05). Meanwhile, coadministration of 100 ng/ml curcumin reduced the cell injury and apoptosis, inhibited intracellular ROS and mitochondrial superoxide accumulation, and ameliorated intracellular SOD2, cell morphology, MMP, and mitochondrial complex I activity. Downregulating the SOD2 expression by using siRNA, however, significantly reversed the curcumin-induced cytoprotection (P < 0.05). These findings indicated that curcumin induces protection against OGD/R injury in HT22 cells, and SOD2 protein may mediate the protection.
Collapse
|
11
|
Han M, Hu L, Chen Y. Rutaecarpine may improve neuronal injury, inhibits apoptosis, inflammation and oxidative stress by regulating the expression of ERK1/2 and Nrf2/HO-1 pathway in rats with cerebral ischemia-reperfusion injury. Drug Des Devel Ther 2019; 13:2923-2931. [PMID: 31692511 PMCID: PMC6708397 DOI: 10.2147/dddt.s216156] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 07/31/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Cerebral ischemia-reperfusion (CI/R) injury is a more serious brain injury caused by the recovery of blood supply after cerebral ischemia for a certain period of time. Rutaecarpine (Rut) is an alkaloid isolated from Evodia officinalis with various biological activities. Previous studies have shown that Rut has a certain protective effect on ischemic brain injury, but the specific molecular mechanism is still unknown. METHODS In this study, a rat model of CI/R was established to explore the effects and potential molecular mechanisms of Rut on CI/R injury in rats. RESULTS The results showed that Rut alleviated neuronal injury induced by CI/R in a dose-dependent manner. Besides, Rut inhibited neuronal apoptosis by inhibiting the activation of caspase 3 and the expression of Bax. In addition, Rut alleviated the inflammatory response and oxidative stress caused by CI/R through inhibiting the production of pro-inflammatory factors (IL-6 and IL-1β), lactate dehydrogenase (LDH), malondialdehyde (MDA) and ROS, and increased the levels of anti-inflammatory factors (IL-4 and IL-10) and superoxide dismutase (SOD). Biochemically, Western blot analyses showed that Rut inhibited the phosphorylation of ERK1/2 and promoted the expression of nuclear factor-erythroid 2 related factor 2 (Nrf2) pathway-related proteins (Nrf2, heme oxygenase 1 (HO-1) and NAD (P) H-quinone oxidoreductase 1) in a dose-dependent manner. These results show that Rut may alleviate brain injury induced by CI/R by regulating the expression of ERK1/2 and the activation of Nrf2/HO-1 pathway. CONCLUSION In conclusion, these results suggest that Rut may be used as an effective therapeutic agent for damage caused by CI/R.
Collapse
Affiliation(s)
- Meiyu Han
- Department of Internal Medicine, The Second People’s Hospital of Dongying City, Dongying City, Shandong Province257335, People’s Republic of China
| | - Lin Hu
- Department of Critical Care Medicine ICU, Zoucheng People’s Hospital, Zoucheng, Shandong Province273500, People’s Republic of China
| | - Yang Chen
- Department of Internal Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Pudong, Shanghai201399, People’s Republic of China
| |
Collapse
|
12
|
Roles Played by the Na +/Ca 2+ Exchanger and Hypothermia in the Prevention of Ischemia-Induced Carrier-Mediated Efflux of Catecholamines into the Extracellular Space: Implications for Stroke Therapy. Neurochem Res 2019; 45:16-33. [PMID: 31346893 PMCID: PMC6942591 DOI: 10.1007/s11064-019-02842-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 05/30/2019] [Accepted: 07/04/2019] [Indexed: 12/11/2022]
Abstract
The release of [3H]dopamine ([3H]DA) and [3H]noradrenaline ([3H]NA) in acutely perfused rat striatal and cortical slice preparations was measured at 37 °C and 17 °C under ischemic conditions. The ischemia was simulated by the removal of oxygen and glucose from the Krebs solution. At 37 °C, resting release rates in response to ischemia were increased; in contrast, at 17 °C, resting release rates were significantly reduced, or resting release was completely prevented. The removal of extracellular Ca2+ further increased the release rates of [3H]DA and [3H]NA induced by ischemic conditions. This finding indicated that the Na+/Ca2+ exchanger (NCX), working in reverse in the absence of extracellular Ca2+, fails to trigger the influx of Ca2+ in exchange for Na+ and fails to counteract ischemia by further increasing the intracellular Na+ concentration ([Na+]i). KB-R7943, an inhibitor of NCX, significantly reduced the cytoplasmic resting release rate of catecholamines under ischemic conditions and under conditions where Ca2+ was removed. Hypothermia inhibited the excessive release of [3H]DA in response to ischemia, even in the absence of Ca2+. These findings further indicate that the NCX plays an important role in maintaining a high [Na+]i, a condition that may lead to the reversal of monoamine transporter functions; this effect consequently leads to the excessive cytoplasmic tonic release of monoamines and the reversal of the NCX. Using HPLC combined with scintillation spectrometry, hypothermia, which enhances the stimulation-evoked release of DA, was found to inhibit the efflux of toxic DA metabolites, such as 3,4-dihydroxyphenylacetaldehyde (DOPAL). In slices prepared from human cortical brain tissue removed during elective neurosurgery, the uptake and release values for [3H]NA did not differ from those measured at 37 °C in slices that were previously maintained under hypoxic conditions at 8 °C for 20 h. This result indicates that hypothermia preserves the functions of the transport and release mechanisms, even under hypoxic conditions. Oxidative stress (H2O2), a mediator of ischemic brain injury enhanced the striatal resting release of [3H]DA and its toxic metabolites (DOPAL, quinone). The study supports our earlier findings that during ischemia transmitters are released from the cytoplasm. In addition, the major findings of this study that hypothermia of brain slice preparations prevents the extracellular calcium concentration ([Ca2+]o)-independent non-vesicular transmitter release induced by ischemic insults, inhibiting Na+/Cl−-dependent membrane transport of monoamines and their toxic metabolites into the extracellular space, where they can exert toxic effects.
Collapse
|
13
|
Liu SM, Cao XM, Qu XH, Cai W, Hu F, Cao WF, Wu LF, Wu XM. Effects of remote ischemic preconditioning on astrocyte proliferation and glial scars after cerebral infarction. EUR J INFLAMM 2019. [DOI: 10.1177/2058739219846325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The aim of this study was to investigate whether remote ischemic preconditioning (RIPC) can promote neurological function recovery after middle cerebral artery occlusion (MCAO) in rats and its possible mechanism. A total of 32 Sprague Dawley (SD) rats were randomly divided into RIPC group (n = 16) and MCAO group (n = 16). In the RIPC group, 1 h before induction of MCAO, the rats received bilateral femoral artery ischemic preconditioning (10 min/time), followed by 10 min of relaxation, and a total of three cycles were carried out. Then, the MCAO-2h model was established. In the MCAO group, the MCAO-2h model was established at 1 h after the separation of bilateral femoral arteries. The modified neurological severity score (mNSS) was assessed. At postmodeling day 7, triphenyltetrazolium chloride (TTC) staining and immunohistochemistry were conducted, and neurological function recovery, infarct size, and the expression levels of glial fibrillary acidic protein (GFAP), synaptophysin (SYN), and neurite outgrowth inhibitor A (Nogo-A) were observed. At postmodeling day 7, the difference in mNSS was statistically significant ( P < 0.05). Infarct size was significantly smaller in the RIPC group than in the MCAO group ( P < 0.05). The number of GFAP+ cells was significantly lesser in the RIPC group than in the MCAO group ( P < 0.05). The difference in thickness of the glial scar was not statistically significant ( P = 0.091). At postmodeling day 7, the expression level of SYN integrated optical density (IOD) was significantly higher in the RIPC group than in the MCAO group ( P < 0.05). The number of Nogo-A+ cells was significantly lesser in the RIPC group than in the MCAO group ( P < 0.05). At day 7 after MCAO, RIPC can promote neurological function recovery in rats and reduce infarct size. The mechanism may be that after 7 days, RIPC reduces GFAP expression, inhibits the trend of glial scar formation and Nogo-A expression, and increases SYN expression.
Collapse
Affiliation(s)
- Shi-Min Liu
- Department of Neurology, People’s Hospital of Jiangxi Province, Nanchang, China
| | - Xian-Min Cao
- Jiangxi University of Finance and Economics, Nanchang, China
| | - Xin-Hui Qu
- Department of Neurology, People’s Hospital of Jiangxi Province, Nanchang, China
| | - Wen Cai
- Department of Neurology, People’s Hospital of Jiangxi Province, Nanchang, China
| | - Fan Hu
- Department of Neurology, People’s Hospital of Jiangxi Province, Nanchang, China
| | - Wen-Feng Cao
- Department of Neurology, People’s Hospital of Jiangxi Province, Nanchang, China
| | - Lin-Feng Wu
- Department of Neurology, People’s Hospital of Jiangxi Province, Nanchang, China
| | - Xiao-Mu Wu
- Department of Neurology, People’s Hospital of Jiangxi Province, Nanchang, China
| |
Collapse
|
14
|
Zhang N, Zhu H, Han S, Sui L, Li J. cPKCγ alleviates ischemic injury through modulating synapsin Ia/b phosphorylation in neurons of mice. Brain Res Bull 2018; 142:156-162. [PMID: 30016727 DOI: 10.1016/j.brainresbull.2018.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 07/02/2018] [Accepted: 07/12/2018] [Indexed: 01/13/2023]
Abstract
Conventional protein kinase C (cPKC)γ and synapsin Ia/b have been implicated in the development of ischemic stroke, but their relationships and functions are unclear. In the present study, the oxygen-glucose deprivation (OGD)-induced ischemic insult in primary cultured cortical neurons in vitro and middle cerebral artery occlusion (MCAO)-induced ischemic stroke model in vivo were used to elucidate the function of cPKCγ and its modulation on synapsin Ia/b phosphorylation in ischemic stroke. We found that cPKCγ knockout significantly increased the infarct volume of mice after 1 h MCAO/72 h reperfusion by using triphenyltetrazolium chloride (TTC) staining. In the primarily cultured cortical neurons, cPKCγ knockout also aggravated the OGD-induced cell death and morphological damage of neurites, while cPKCγ restoration could alleviate the ischemic injury. Among the five phosphorylation sites of synapsin Ia/b, only the phosphorylation levels of Ser549 and 553 could be modulated by cPKCγ in neurons following 0.5 h OGD/24 h reoxygenation. In addition, we found that cPKCγ and synapsin Ia/b could be reciprocally co-immunoprecipitated in the cerebral cortex of MCAO mice. Taken together, we proposed that cPKCγ alleviates ischemic injury through modulating Ser549/553- synapsin Ia/b phosphorylation in neurons of mice.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Human Anatomy, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Chinese Medical Association Publishing House, Beijing 100710, PR China
| | - Hongyi Zhu
- Department of Neurobiology and Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, PR China
| | - Song Han
- Department of Neurobiology and Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, PR China
| | - Leiming Sui
- Core Facility Center, Capital Medical University, Beijing 100069, PR China
| | - Junfa Li
- Department of Neurobiology and Center of Stroke, Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, PR China.
| |
Collapse
|
15
|
Cabezas-Llobet N, Camprubí S, García B, Alberch J, Xifró X. Human alpha 1-antitrypsin protects neurons and glial cells against oxygen and glucose deprivation through inhibition of interleukins expression. Biochim Biophys Acta Gen Subj 2018; 1862:1852-1861. [PMID: 29857082 DOI: 10.1016/j.bbagen.2018.05.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 05/09/2018] [Accepted: 05/23/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Death due to cerebral stroke afflicts a large number of neuronal populations, including glial cells depending on the brain region affected. Drugs with a wide cellular range of protection are needed to develop effective therapies for stroke. Human alpha 1-antitrypsin (hAAT) is a serine proteinase inhibitor with potent anti-inflammatory, anti-apoptotic and immunoregulatory activities. This study aimed to test whether hAAT can protect different kind of neurons and glial cells after the oxygen and glucose deprivation (OGD). METHODS Addition of hAAT to mouse neuronal cortical, hippocampal and striatal cultures, as well as glial cultures, was performed 30 min after OGD induction and cell viability was assessed 24 h later. The expression of different apoptotic markers and several inflammatory parameters were assessed by immunoblotting and RT-PCR. RESULTS hAAT had a concentration-dependent survival effect in all neuronal cultures exposed to OGD, with a maximal effect at 1-2 mg/mL. The addition of hAAT at 1 mg/mL reduced the OGD-mediated necrotic and apoptotic death in all neuronal cultures. This neuroprotective activity of hAAT was associated with a decrease of cleaved caspase-3 and an increase of MAP2 levels. It was also associated with a reduction of pro-inflammatory cytokines protein levels and expression, increase of IL-10 protein levels and decrease of nuclear localization of nuclear factor-kappaB. Similar to neurons, addition of hAAT protected astrocytes and oligodendrocytes against OGD-induced cell death. CONCLUSIONS Human AAT protects neuronal and glial cells against OGD through interaction with cytokines. GENERAL SIGNIFICANCE Human AAT could be a good therapeutic neuroprotective candidate to treat ischemic stroke.
Collapse
Affiliation(s)
- Núria Cabezas-Llobet
- New Therapeutic Targets Group (TargetsLab), Departament de Ciències Mèdiques, Facultat de Medicina, Universitat de Girona, E-17071 Girona, Spain
| | | | | | - Jordi Alberch
- Departament de Biomedicina, Institut de Neurociències, Facultat de Medicina, Universitat de Barcelona, E-08036 Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), E-08036 Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain
| | - Xavier Xifró
- New Therapeutic Targets Group (TargetsLab), Departament de Ciències Mèdiques, Facultat de Medicina, Universitat de Girona, E-17071 Girona, Spain; Departament de Biomedicina, Institut de Neurociències, Facultat de Medicina, Universitat de Barcelona, E-08036 Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), E-08036 Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Spain.
| |
Collapse
|
16
|
Cheng X, Zhao H, Yan F, Tao Z, Wang R, Han Z, Li G, Luo Y, Ji X. Limb remote ischemic post-conditioning mitigates brain recovery in a mouse model of ischemic stroke by regulating reactive astrocytic plasticity. Brain Res 2018; 1686:94-100. [PMID: 29462606 DOI: 10.1016/j.brainres.2018.02.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Revised: 02/13/2018] [Accepted: 02/14/2018] [Indexed: 12/31/2022]
Abstract
Maladaptive alterations of astrocytic plasticity may cause brain edema in the acute stage of stroke and glial scar formation in the recovery stage. The present study was designed to investigate the potential regulation of limb remote ischemic post-conditioning (RIPC) on astrocytic plasticity in experimental cerebral ischemia-reperfusion injury. Cerebral ischemia was induced by transient middle cerebral artery occlusion (tMCAO) for 1 h in C57BL/6 mice, who were treated with RIPC immediately after reperfusion. The results showed that RIPC decreased hemispheric swelling, infarct volume and brain atrophy, and increased neurological function recovery and survival rates of ischemic mice at 3 and 14 d after cerebral ischemia-reperfusion, respectively. Moreover, the proportion of astrocyte subtypes was adjusted by RIPC treatment, demonstrated by decreased expression of the fibrous type (glial fibrillary acidic protein, GFAP) and increased expression of the protoplasmic type (glutamine synthetase, GS) in the ipsilateral side of the mouse brain at 14 d after cerebral ischemia-reperfusion. RIPC treatment adjusted the proportion of GFAP subtypes by downregulating the protein level of GFAPα, as well as upregulating the GFAPδ/GFAPα ratio in the ipsilateral side at 3 and 14 d after reperfusion. Notably, RIPC inhibited the phosphorylation of signal transducer and activators of transcriptions 3 (p-STAT3) in the ipsilateral side at 3 and 14 d after cerebral ischemia-reperfusion. Taken together, the results show that RIPC treatment could regulate reactive astrocytic plasticity and inhibition of STAT3 phosphorylation to promote neurological function recovery following ischemic stroke.
Collapse
Affiliation(s)
- Xue Cheng
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China; The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Haiping Zhao
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center, Beijing, China
| | - Feng Yan
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center, Beijing, China
| | - Zhen Tao
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center, Beijing, China
| | - Rongliang Wang
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center, Beijing, China
| | - Ziping Han
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center, Beijing, China
| | - Guangwen Li
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center, Beijing, China
| | - Yumin Luo
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center, Beijing, China; Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China
| | - Xunming Ji
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing, China; Beijing Geriatric Medical Research Center, Beijing, China; Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Beijing, China.
| |
Collapse
|