1
|
Qin H, Su L, Zhou B, Yang P, Zhu YL, Liang D. Remote Ischemic Postconditioning Improve Cerebral Ischemia-Reperfusion Injury Induced Cognitive Dysfunction through Suppressing Mitochondrial Apoptosis in Hippocampus via TK/BK/B2R-Mediated PI3K/AKT. Mol Neurobiol 2025:10.1007/s12035-025-04864-y. [PMID: 40229456 DOI: 10.1007/s12035-025-04864-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 03/17/2025] [Indexed: 04/16/2025]
Abstract
Remote ischemic postconditioning (RIPostC) is known to improve motor function recovery in animal models, but its efficacy in alleviating cognitive impairment caused by ischemic stroke remains unclear. This study aims to investigate the beneficial role of RIPostC in recovering cognitive impairment induced by cerebral ischemia-reperfusion injury (CIRI). Building upon our previous research findings, we proved that the TK/BK/B2R pathway is crucial for understanding the crosstalk between cognitive impairment and RIPostC. Additionally, in vitro experiments were conducted using the oxygen glucose deprivation/re-oxygenation (OGD/r) HT-22 cell model, which revealed that the mechanism by which RIPostC suppressed mitochondrial apoptosis was mainly through the activation of the B2R/PI3K/AKT signaling pathway, thereby protecting neurons in the ischemic hippocampus from ischemic damage. To investigate the effect of RIPostC on cognitive function recovery following ischemic stroke, we established a rat model using left middle cerebral artery occlusion reperfusion (MCAO/r). 48 h after MCAO/r, rats were subjected to 3 circles of RIPostC therapy daily for 12 consecutive days. HOE140 was used to antagonize the bradykinin 2 receptor (B2R). Cognitive function was assessed using a modified neurological severity score, the Morris water maze, and the novel object recognition test. Local infarct volume in the hippocampus was measured through MRI scanning. The apoptosis rate of hippocampal neurons was quantified using TUNEL staining. Protein expression levels of kallikrein (TK) and mitochondrial apoptosis-related proteins, Cyt c, Bcl-2, Bax, cleaved caspase-3, and cleaved caspase-9, were detected in ischemic hippocampal tissue using Western blot (WB). The expression of bradykinin (BK) in serum and the ischemic penumbra was measured using an enzyme-linked immunosorbent (ELISA) assay. In the cell experiments, the HT-22 cell line and OGD/r model were used to simulate in vitro hippocampal ischemia. WB was performed to detect the expression of apoptosis-related proteins and PI3K/AKT pathway proteins. The apoptosis rate of HT-22 cells was detected using Annexin-V/PI flow cytometry and a cell viability kit. JC-1 staining and reactive oxygen species staining were used to evaluate mitochondrial condition. The PI3K/AKT pathway was inhibited using LY294002. RIPostC significantly upregulated the concentrations of TK and BK in the ischemic hippocampus. Behavioral function tests demonstrated that daily RIPostC therapy for 12 days significantly promoted cognitive function recovery in MCAO/r rats. Through MRI analysis, we found that RIPostC therapy effectively reduced the infarct volume in the hippocampus. Additionally, TUNEL staining and WB results of apoptosis-related proteins showed that RIPostC therapy significantly reduced apoptosis of hippocampal neurons. However, the therapeutic effect of RIPostC was reversed by the B2R antagonist HOE14, indicating that the TK/BK/B2R pathway mediated the neuroprotective effect of RIPostC. Cell experiments further confirmed that BK/B2R significantly attenuated mitochondrial apoptosis induced by ischemia-hypoxia injury in HT-22 cells. In vivo and in vitro results from WB demonstrated that the BK/B2R pathway activated the PI3K/AKT signaling pathway. Finally, the PI3K inhibitor LY294002 reversed the anti-apoptotic effect induced by BK/B2R. RIPostC therapy effectively inhibited mitochondrial apoptosis of hippocampal neurons and significantly alleviated cognitive dysfunction associated with CIRI by regulating the TK/BK/B2R-medated PI3K/AKT pathway. In conclusion, RIPostC represents a promising therapeutic strategy for combating cognitive dysfunction by inhibiting cell apoptosis in hippocampus. Moreover, our results suggest that RIPostC may have a broader protective effect against apoptosis in other ischemia-reperfusion-related diseases.
Collapse
Affiliation(s)
- Haocheng Qin
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Lu Su
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Bao Zhou
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Pengkun Yang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Wulumuqi Road, Jing'an District, Shanghai, 200040, China
| | - Yu-Lian Zhu
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Wulumuqi Road, Jing'an District, Shanghai, 200040, China.
| | - Dan Liang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, No. 12 Wulumuqi Road, Jing'an District, Shanghai, 200040, China.
| |
Collapse
|
2
|
Zhao Z, Xie L, Shi J, Liu T, Wang S, Huang J, Wu D, Zhang X. Neuroprotective Effect of Zishen Huoxue Decoction treatment on Vascular Dementia by activating PINK1/Parkin mediated Mitophagy in the Hippocampal CA1 Region. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117172. [PMID: 37709106 DOI: 10.1016/j.jep.2023.117172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 09/08/2023] [Accepted: 09/10/2023] [Indexed: 09/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zishen Huoxue Decoction (ZSHXD) is a Traditional Chinese Medicine (TCM) prescription for the treatment of vascular dementia (VD). Although the clinical effects of ZSHXD have been demonstrated, the molecular mechanisms underlying the neuroprotective effects of ZSHXD remain unclear. AIM OF THE STUDY To explore whether the neuroprotective effect of Zishen Huoxue Decoction (ZSHXD) treatment is associated with the PINK1/Parkin pathway-mediated mitophagy in hippocampal CA1 region of 2-VO model rats. MATERIALS AND METHODS Seventy-two male SD rats were randomly divided into the sham group, model group, Donepezil (0.45 mg/kg) group, ZSHXD low dose group (8.9 g/kg), ZSHXD medium dose group (17.8 g/kg), and ZSHXD high dose group (35.6 g/kg). Two-vessel occlusion (2-VO) rat model is established to evaluate the therapeutic effect of ZSHXD pretreatment. Hematoxylin-eosin (HE) staining is conducted to detect the morphological changes of neurons and the number of normal neurons in the hippocampal CA1 region. Then, the mitochondrial function and structure were reflected by the mitochondrial membrane potential (MMP) levels and transmission electron microscopy (TEM). Meanwhile, the expression of mitophagy related proteins mediated by PINK1/Parkin was detected by western blot (WB). After that, malondialdehyde (MDA) and superoxide dismutase (SOD) levels were measured by Elisa. At last, the apoptosis-related proteins Caspase-3、Bax、Bcl-2 were measured by WB. RESULTS The results depict that ZSHXD has dose-dependently improved the cognitive function in 2-VO model rats. It has also been showed that ZSHXD can alleviate neuron damage, rescue the mitochondrial structural injury and dysfunction in hippocampal CA1 region. Besides, ZSHXD has increased the activity of SOD and decreased the activity of MDA. In addition, ZSHXD can inhibit apoptosis with Caspase-3, Bax decreasing and Bcl-2 increasing. Specially, the protection of ZSHXD showed in 2-VO model rats is along with the upregulation of PINK1, Parkin and LC3-Ⅱ/Ⅰ, and downregulation of p62 in the hippocampal CA1 region. CONCLUSIONS This study reveals that ZSHXD protects the 2-VO model rats from ischemic injury by activating the PINK1/Parkin-mediated mitophagy in the hippocampal CA1 region.
Collapse
Affiliation(s)
- Ziting Zhao
- Hunan University of Chinese Medicine, Changsha, 410208, Hunan Province, China
| | - Le Xie
- Hunan Hospital of Integrated Traditional Chinese and Western Medicine, Changsha, 410006, Hunan Province, China
| | - Jiayi Shi
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410218, Hunan Province, China
| | - Tonghe Liu
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410218, Hunan Province, China
| | - Shiliang Wang
- Hunan Hospital of Integrated Traditional Chinese and Western Medicine, Changsha, 410006, Hunan Province, China
| | - Jianhua Huang
- Hunan Academy of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410006, Hunan Province, China
| | - Dahua Wu
- Hunan Hospital of Integrated Traditional Chinese and Western Medicine, Changsha, 410006, Hunan Province, China.
| | - Xiuli Zhang
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410218, Hunan Province, China.
| |
Collapse
|
3
|
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
|
4
|
Zhao H, Wang M, Huang X, Wu X, Xiao H, Jin F, Lv J, Cheng J, Zhao Y, Zhang C. Wasp venom from Vespa magnifica acts as a neuroprotective agent to alleviate neuronal damage after stroke in rats. PHARMACEUTICAL BIOLOGY 2022; 60:334-346. [PMID: 35171059 PMCID: PMC8863380 DOI: 10.1080/13880209.2022.2032207] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 11/24/2021] [Accepted: 01/17/2022] [Indexed: 06/14/2023]
Abstract
CONTEXT Acute ischaemic stroke (AIS) is a major cause of disability and death, which is a serious threat to human health and life. Wasp venom extracted from Vespa magnifica Smith (Vespidae) could treat major neurological disorders. OBJECTIVE This study investigated the effects of wasp venom on AIS in rats. MATERIAL AND METHODS We used a transient middle cerebral artery occlusion (MCAO) model in Sprague-Dawley rats (260-280 g, n = 8-15) with a sham operation group being treated as negative control. MCAO rats were treated with wasp venom (0.05, 0.2 and 0.6 mg/kg, i.p.) using intraperitoneal injection. After treatment 48 h, behavioural tests, cortical blood flow (CBF), TTC staining, H&E staining, Nissl staining, TUNEL assay, immunohistochemistry (IHC) and ELISA were employed to investigate neuroprotective effects of wasp venom. RESULTS Compared with the MCAO group, wasp venom (0.6 mg/kg) improved neurological impairment, accelerated CBF recovery (205.6 ± 52.92 versus 216.7 ± 34.56), reduced infarct volume (337.1 ± 113.2 versus 140.7 ± 98.03) as well as BBB permeability as evidenced by changes in claudin-5 and AQP4. In addition, function recovery of stroke by wasp venom treatment was associated with a decrease in TNF-α, IL-1β, IL-6 and inhibition activated microglia as well as apoptosis. Simultaneously, the wasp venom regulated the angiogenesis factors VEGF and b-FGF in the brain. CONCLUSIONS Wasp venom exhibited a potential neuroprotective effect for AIS. In the future, we will focus on determining whether the observed actions were due to a single compound or the interaction of multiple components of the venom.
Collapse
Affiliation(s)
- Hairong Zhao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, PR China
- School of Medicine, Xiamen University, Xiamen, PR China
| | - Mei Wang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, PR China
| | - Xi Huang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, PR China
| | - Xiumei Wu
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, PR China
- National-Local Joint Engineering Research Center of Entomoceutics, Dali, PR China
| | - Huai Xiao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, PR China
- National-Local Joint Engineering Research Center of Entomoceutics, Dali, PR China
| | - Fanmao Jin
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, PR China
| | - Jiaming Lv
- School of Medicine, Xiamen University, Xiamen, PR China
| | - Jidong Cheng
- School of Medicine, Xiamen University, Xiamen, PR China
| | - Yu Zhao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, PR China
- National-Local Joint Engineering Research Center of Entomoceutics, Dali, PR China
| | - Chenggui Zhang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, PR China
- National-Local Joint Engineering Research Center of Entomoceutics, Dali, PR China
| |
Collapse
|
5
|
Lin SR, Lin QM, Lin YJ, Qian X, Wang XP, Gong Z, Chen F, Song B. Bradykinin postconditioning protects rat hippocampal neurons after restoration of spontaneous circulation following cardiac arrest via activation of the AMPK/mTOR signaling pathway. Neural Regen Res 2022; 17:2232-2237. [PMID: 35259843 PMCID: PMC9083139 DOI: 10.4103/1673-5374.337049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Bradykinin (BK) is an active component of the kallikrein-kinin system that has been shown to have cardioprotective and neuroprotective effects. We previously showed that BK postconditioning strongly protects rat hippocampal neurons upon restoration of spontaneous circulation (ROSC) after cardiac arrest. However, the precise mechanism underlying this process remains poorly understood. In this study, we treated a rat model of ROSC after cardiac arrest (induced by asphyxiation) with 150 μg/kg BK via intraperitoneal injection 48 hours after ROSC following cardiac arrest. We found that BK postconditioning effectively promoted the recovery of rat neurological function after ROSC following cardiac arrest, increased the amount of autophagosomes in the hippocampal tissue, inhibited neuronal cell apoptosis, up-regulated the expression of autophagy-related proteins LC3 and NBR1 and down-regulated p62, inhibited the expression of the brain injury marker S100β and apoptosis-related protein caspase-3, and affected the expression of adenosine monophosphate-activated protein kinase/mechanistic target of rapamycin pathway-related proteins. Adenosine monophosphate-activated protein kinase inhibitor compound C clearly inhibited BK-mediated activation of autophagy in rats after ROSC following cardiac arrest, which aggravated the injury caused by ROSC. The mechanistic target of rapamycin inhibitor rapamycin enhanced the protective effects of BK by stimulating autophagy. Our findings suggest that BK postconditioning protects against injury caused by ROSC through activating the adenosine monophosphate-activated protein kinase/mechanistic target of the rapamycin pathway.
Collapse
Affiliation(s)
- Shi-Rong Lin
- Provincial College of Clinical Medicine, Fujian Medical University; Department of Emergency, Fujian Provincial Hospital South Branch; Department of Emergency, Fujian Provincial Hospital; Fujian Emergency Medical Center; Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, Fujian Province, China
| | - Qing-Ming Lin
- Provincial College of Clinical Medicine, Fujian Medical University; Department of Emergency, Fujian Provincial Hospital; Fujian Emergency Medical Center; Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, Fujian Province, China
| | - Yu-Jia Lin
- Provincial College of Clinical Medicine, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Xin Qian
- Provincial College of Clinical Medicine, Fujian Medical University; Department of Emergency, Fujian Provincial Hospital; Fujian Emergency Medical Center; Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, Fujian Province, China
| | - Xiao-Ping Wang
- Provincial College of Clinical Medicine, Fujian Medical University; Department of Emergency, Fujian Provincial Hospital; Fujian Emergency Medical Center; Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, Fujian Province, China
| | - Zheng Gong
- Provincial College of Clinical Medicine, Fujian Medical University; Department of Emergency, Fujian Provincial Hospital; Fujian Emergency Medical Center; Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, Fujian Province, China
| | - Feng Chen
- Provincial College of Clinical Medicine, Fujian Medical University; Department of Emergency, Fujian Provincial Hospital; Fujian Emergency Medical Center; Fujian Provincial Key Laboratory of Emergency Medicine, Fuzhou, Fujian Province, China
| | - Bin Song
- Department of Human Anatomy, School of Basic Medical Sciences, Fujian Medical University; Key Laboratory of Brain Aging and Neurodegenerative Diseases of Fujian Province; Laboratory of Clinical Applied Anatomy, Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
6
|
Vespakinin-M, a natural peptide from Vespa magnifica, promotes functional recovery in stroke mice. Commun Biol 2022; 5:74. [PMID: 35058552 PMCID: PMC8776894 DOI: 10.1038/s42003-022-03024-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 12/28/2021] [Indexed: 02/06/2023] Open
Abstract
Acute ischemic stroke triggers complex systemic pathological responses for which the exploration of drug resources remains a challenge. Wasp venom extracted from Vespa magnifica (Smith, 1852) is most commonly used to treat rheumatoid arthritis as well as neurological disorders. Vespakinin-M (VK), a natural peptide from wasp venom, has remained largely unexplored for stroke. Herein, we first confirmed the structure, stability, toxicity and distribution of VK as well as its penetration into the blood–brain barrier. VK (150 and 300 µg/kg, i.p.) was administered to improve stroke constructed by middle cerebral artery occlusion in mice. Our results indicate that VK promote functional recovery in mice after ischemia stroke, including an improvement of neurological impairment, reduction of infarct volume, maintenance of blood-brain barrier integrity, and an obstruction of the inflammatory response and oxidative stress. In addition, VK treatment led to reduced neuroinflammation and apoptosis associated with the activation of PI3K–AKT and inhibition of IκBα–NF-κB signaling pathways. Simultaneously, we confirmed that VK can combine with bradykinin receptor 2 (B2R) as detected by molecular docking, the B2R antagonist HOE140 could counteract the neuro-protective effects of VK on stroke in mice. Overall, targeting the VK–B2R interaction can be considered as a practical strategy for stroke therapy. Zhao et al establish the structure, stability, toxicity and distribution of vespakinin-M (VK) as well as its penetration into the blood–brain barrier in mice. They go on to show that VK promotes functional recovery in mice after ischemia stroke and shed light on the potential underlying mechanisms, which together indicates the potential therapeutic value of targeting VK in stroke therapy.
Collapse
|
7
|
Saklani P, Khan H, Gupta S, Kaur A, Singh TG. Neuropeptides: Potential neuroprotective agents in ischemic injury. Life Sci 2022; 288:120186. [PMID: 34852271 DOI: 10.1016/j.lfs.2021.120186] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 12/24/2022]
Abstract
AIM Ischemic damage to the brain is linked to an increased rate of morbidity and mortality worldwide. In certain parts of the world, it remains a leading cause of mortality and the primary cause of long-term impairment. Ischemic injury is exacerbated when particular neuropeptides are removed, or their function in the brain is blocked, whereas supplying such neuropeptides lowers ischemic harm. Here, we have discussed the role of neuropeptides in ischemic injury. MATERIALS & METHODS Numerous neuropeptides had their overexpression following cerebral ischemia. Neuropeptides such as NPY, CGRP, CART, SP, BK, PACAP, oxytocin, nociception, neurotensin and opioid peptides act as transmitters, documented in several "in vivo" and "in vitro" studies. Neuropeptides provide neuroprotection by activating the survival pathways or inhibiting the death pathways, i.e., MAPK, BDNF, Nitric Oxide, PI3k/Akt and NF-κB. KEY FINDINGS Neuropeptides have numerous beneficial effects in ischemic models, including antiapoptotic, anti-inflammatory, and antioxidant actions that provide a powerful protective impact in neurons when combined. These innovative therapeutic substances have the potential to treat ischemia injury due to their pleiotropic modes of action. SIGNIFICANCE This review emphasizes the neuroprotective role of neuropeptides in ischemic injury via modulation of various signalling pathways i.e., MAPK, BDNF, Nitric Oxide, PI3k/Akt and NF-κB.
Collapse
Affiliation(s)
- Priyanka Saklani
- Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| | - Saurabh Gupta
- Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, 140401, Punjab, India
| | | |
Collapse
|
8
|
Burda J, Burda R. Ischemic tolerance - blessing or curse. Physiol Res 2021; 70:661-670. [PMID: 34505532 DOI: 10.33549/physiolres.934644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Application of knowledge about ischemic tolerance to clinic requires the solid understanding of mechanism of creation of this phenomenon. This review summarizes research that has been carried out in many laboratories over a long period of time, but the main focus will be on own experimental research. The main emphasis is devoted to the possibility of preparing full tolerance in the donor's body and its transfer to the patient in the form of activated blood plasma. Such plasma could be administered as soon as the patient is transported to the hospital and would take effect immediately after administration to the patient's bloodstream. One chapter is also devoted to anticonditioning, i.e. the possibility of preventing the activation of tolerance. Anticonditioning could be used to treat oncologic patients. We expect that this method could increase effectiveness of cancer treatment. Cross-tolerance with a wide range of diverse stressors gives us the courage to assume that activated plasma can significantly help with a wide range of pathological events.
Collapse
Affiliation(s)
- J Burda
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Košice, Slovak Republic. Faculty of Medicine, Clinic of Trauma Surgery, P. J. Šafárik University, Košice, Slovak Republic.
| | | |
Collapse
|
9
|
Xia M, Ruan Z, Chen B, Wang Y, Zhou Z, Ren S, Wu L, Tang N. Wuzang Wenyang Huayu decoction regulates differentially expressed transcripts in the rats' hippocampus after cerebral hypoperfusion. J Cell Mol Med 2020; 24:294-303. [PMID: 31705584 PMCID: PMC6933406 DOI: 10.1111/jcmm.14723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 08/14/2019] [Accepted: 08/27/2019] [Indexed: 11/30/2022] Open
Abstract
The modified Wenyang Huayu decoction has been widely used to treat vascular dementia in China for thousands of years. We have previously proved that a modified version, Wuzang Wenyang Huayu decoction has the potential to be a more effective clinical treatment that can attenuate cerebral ischaemic injury. However, the global transcript profile and signalling conduction pathways regulated by this recipe remains unclear. This study established a two-vessel occlusion rat model by bilateral common carotid artery occlusion. Two groups of rats were intragastrically treated Wuzang Wenyang Huayu 2.5 g/kg vs or Piracetam 0.15 g/kg for 2 weeks. Learning and memory abilities were measured with Morris water maze. Neuronal plasticity was observed by HE staining. Differentially expressed transcripts of rat hippocampus were analysed by transcriptomics with Illumina HiSeq2500 platform. Results showed that Wuzang Wenyang Huayu decoction significantly alleviated learning, memory deficits, coordination dysfunction and prevented hippocampus cellular injury; Results further revealed the increased gene expression in KEGG metabolic pathways (MT-ND2. MT-ND3, MT-ND4, MT-ND4L, MT-ND5 and MT-ATP8) and genes involved in signal transduction, carcinogenesis, immune system, endocrine system, nervous system etc (Results further revealed differential expression of genes involved in various systems, including MT-ND2) Our discovery is likely to provide new insights to molecular mechanisms of Wuzang Wenyang Huayu regarding hippocampal transcripts in a murine vascular dementia model.
Collapse
Affiliation(s)
- Meng Xia
- School of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory for Foundational Research of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Ziyun Ruan
- School of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Ben Chen
- Guangxi Key Laboratory for Foundational Research of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Yunqiao Wang
- School of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, China
- Guangxi Key Laboratory for Foundational Research of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
| | - Zengzi Zhou
- The 3rd Xiangya Hospital, Central South University, Changsha, China
| | - Shiding Ren
- Guangxi Key Laboratory for Foundational Research of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- The First Affiliated Hospital to Guangxi University of Chinese Medicine, Nanning, China
| | - Lin Wu
- Guangxi Key Laboratory for Foundational Research of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- The First Affiliated Hospital to Guangxi University of Chinese Medicine, Nanning, China
| | - Nong Tang
- Guangxi Key Laboratory for Foundational Research of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, China
- The First Affiliated Hospital to Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
10
|
Danková M, Domoráková I, Fagová Z, Stebnický M, Kunová A, Mechírová E. Bradykinin and noradrenaline preconditioning influences level of antioxidant enzymes SOD, CuZn-SOD, Mn-SOD and catalase in the white matter of spinal cord in rabbits after ischemia/reperfusion. Eur J Histochem 2019; 63. [PMID: 31631645 PMCID: PMC6802454 DOI: 10.4081/ejh.2019.3045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 09/28/2019] [Indexed: 11/23/2022] Open
Abstract
The aim of present work is to assess the effects of bradykinin (Br) or noradrenaline (Nor) preconditioning to the levels of antioxidant enzymes: superoxide dismutase (SOD), copper, zinc superoxide dismutase (CuZn-SOD), manganese superoxide dismutase (Mn-SOD) and catalase in ischemia/reperfusion (I/R) model in the rabbit spinal cord white matter as well as effect on glial fibrillary acidic protein (GFAP) and ubiquitin immunoreaction in glial cells. Rabbits were preconditioned by intraperitoneal single dose of Br or Nor 48 h prior to 20 min of ischemia followed by 24 or 48 h of reperfusion. White matter of L3-L6 spinal cord segments was used for comparison of antioxidant enzyme levels in sham control, ischemic groups and four preconditioned groups. The total SOD level in the Br or Nor preconditioned groups after 48 h of reperfusion was increased vs Br or Nor preconditioned groups after 24 h of reperfusion. The comparison among the ischemic group vs Br preconditioned (P<0.05), and Nor preconditioned (P<0.001) groups after 48 h of reperfusion, showed statistically significant decrease of Mn-SOD activity. Tissue catalase level activity was significantly decreased in the Br preconditioned group after 48 h of reperfusion (P<0.05) and Nor preconditioned groups after 24 h of reperfusion (P<0.001) and also after 48 h of reperfusion (P<0.001), in comparison to ischemic group after 48 h of reperfusion. Significantly decreased tissue catalase activity (P<0.05) in both Nor preconditioned groups after 24 or 48 h of reperfusion was measured vs Br preconditioned group after 48 h of reperfusion. According to our results, in the white matter, activation of stress proteins in glial cells, as well as antioxidant enzymes levels, were influenced by pharmacological preconditioning followed by 20 min of ischemia and 24 or 48 h of reperfusion. These changes contribute to ischemic tolerance acquisition and tissue protection from oxidative stress during reperfusion period.
Collapse
Affiliation(s)
- Marianna Danková
- Department of Histology and Embryology, Pavol Jozef Šafárik University, Košice.
| | | | | | | | | | | |
Collapse
|
11
|
Fu D, Liu H, Liu H, Yao J. Effects of D‑Ala2, D‑Leu5‑Enkephalin pre‑ and post‑conditioning in a rabbit model of spinal cord ischemia and reperfusion injury. Mol Med Rep 2019; 20:4811-4820. [PMID: 31638217 PMCID: PMC6854538 DOI: 10.3892/mmr.2019.10729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 06/20/2019] [Indexed: 11/15/2022] Open
Abstract
It has recently been revealed that during the aorta-clamped period, D-Ala2, D-Leu5-Enkephalin (DADLE) infusion can protect the spinal cord against ischemia and reperfusion (I/R) injury. However, the protective effects of DADLE administration prior to ischemia or at the time of early reperfusion have not yet been investigated. Drug pre- or post-conditioning can serve as a more valuable clinical strategy. Therefore, the present study was designed to investigate the neuroprotective effect of DADLE infusion at different time intervals in order to determine the optimum time point for ischemic spinal cord protection. A total of 40 New Zealand white rabbits were randomly divided into 5 groups: Sham-operated (Sham), normal saline pre-conditioning (NS), DADLE per-conditioning (Dper), DADLE pre-conditioning (Dpre) and DADLE post-conditioning (Dpost). All animals were subjected to spinal cord ischemia for 30 min followed by 48 h reperfusion. Hind limb motor functions were assessed according to the Tarlov criterion when the animals regained consciousness, 6, 24 and 48 h after reperfusion. Histological analysis and the number of viable α-motor neurons were also used to assess the extent of spinal cord injury. Compared with the NS group, the Tarlov scores and the number of normal neurons were significantly higher in the Dper group (P<0.05), which were consistent with the results of a previous study. In addition, the paraplegia rate and loss of normal motor neurons were lower in the DADLE per- and post-conditioning groups compared with the DADLE pre-conditioning; however, these were not statistically significant. DADLE 0.05 mg/kg administration at three time points all mitigated normal motor neuron injury in the anterior horn and decreased the paraplegia rates in rabbits. The therapeutic benefits appeared best in the post-conditioning group with DADLE, and worst in the pre-conditioning group.
Collapse
Affiliation(s)
- Danyun Fu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai 200080, P.R. China
| | - Haitong Liu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai 200080, P.R. China
| | - Hua Liu
- Department of Anesthesiology, Shanghai General Hospital, Shanghai 200080, P.R. China
| | - Junyan Yao
- Department of Anesthesiology, Shanghai General Hospital, Shanghai 200080, P.R. China
| |
Collapse
|
12
|
Fagová Z, Domoráková I, Danková M, Mechírová E, Kunová A, Stebnický M. Ubiquitin and endogenous antioxidant enzymes participate in neuroprotection of the rabbit spinal cord after ischemia and bradykinin postconditioning. Acta Histochem 2019; 121:732-741. [PMID: 31270014 DOI: 10.1016/j.acthis.2019.06.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 06/05/2019] [Accepted: 06/25/2019] [Indexed: 11/25/2022]
Abstract
The aim of this study was to investigate neuroprotective effect of bradykinin postconditioning on the rabbit spinal cord after 20 min of ischemia and 3 days of reperfusion. Bradykinin was administered by single i.p. application at 1, 6, 12 or 24 h after ischemia. Assessment of neurological function of hind limbs (Tarlov score) was estimated. Quantitative analysis was evaluated by Fluoro Jade B method, NeuN and ubiquitin immunohistochemistry in anterior horn neurons of the spinal cord. Histomorphologically distribution of ubiquitin and endogenous antioxidant enzymes (SOD1, SOD2, catalase) immunoreaction was described. Bradykinin postconditioning showed decreased number of degenerated neurons, increased number of surviving neurons and increase in number of ubiquitin positive neurons in all bradykinin postconditioned groups versus ischemia/reperfusion group. According to our results bradykinin postconditioning applied 24 h after ischemia significantly decreased (p < 0.001) number of degenerated neurons versus ischemia/reperfusion group. The least effective time window for bradykinin postconditioning was at 12 h after ischemia. Tarlov score was significantly improved (p < 0.05) in groups with bradykinin postconditioning applied 1, 6 or 24 h after ischemia versus ischemia/reperfusion group. Tarlov score in group with bradykinin application 12 h after ischemia was significantly decreased (p < 0.05) versus sham control group. Neuronal immunoreaction of ubiquitin, SOD1, SOD2 and catalase influenced by bradykinin postconditioning was dependent on neuronal survival or degeneration. In conclusion, bradykinin postconditioning showed protective effect on neurons in anterior horns of the rabbit spinal cord and improved motor function of hind limbs.
Collapse
|
13
|
Grewal AK, Singh N, Singh TG. Neuroprotective effect of pharmacological postconditioning on cerebral ischaemia-reperfusion-induced injury in mice. ACTA ACUST UNITED AC 2019; 71:956-970. [PMID: 30809806 DOI: 10.1111/jphp.13073] [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: 09/18/2018] [Accepted: 01/01/2019] [Indexed: 12/31/2022]
Abstract
OBJECTIVES To investigate the mechanism of neuroprotection rendered via pharmacological postconditioning in cerebral ischaemia-reperfusion-induced injury in mice. METHODS Pharmacological postconditioning is strategy which either involves hindering deleterious pathway or inducing modest stress level which triggers intracellular defence pathway to sustain more vigorous insult leading to conditioning. Hence, in current research we explored the potentiality of CGS21680 (0.5 mg/kg; i.p), an adenosine A2 A receptor agonist and PTEN inhibitor, SF1670 (3 mg/kg; i.p.) to trigger postconditioning after inducing cerebral global ischaemia (17 min) and reperfusion (24 h)-induced injury via occlusion of both carotid arteries. Mice were also given treatment with LY294002 (1.5 mg/kg; i.p.), a PI3K inhibitor and adenosine A2 A receptor antagonist, Istradefylline (2 mg/kg; i.p.), to establish the precise mechanism of postconditioning. Various biochemical and behavioural parameters were assessed to examine the effect of pharmacological postconditioning. KEY FINDINGS Pharmacological postconditioning induced with CGS21680 and SF1670 attenuated the infarction along with improved behavioural and biochemical parameters in comparison with ischaemia-reperfusion control group. The outcome of postconditioning with CGS21680 and SF1670 was significantly reversed by LY294002 and Istradefylline, respectively. CONCLUSIONS The neuroprotective effects of CGS21680 and SF1670 postconditioning on cerebral ischaemia-reperfusion injury may be due to PI3K/Akt pathway activation.
Collapse
Affiliation(s)
- Amarjot Kaur Grewal
- Department of Pharmacology, Chitkara college of Pharmacy, Chitkara University, Patiala, India
| | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | - Thakur Gurjeet Singh
- Department of Pharmacology, Chitkara college of Pharmacy, Chitkara University, Patiala, India
| |
Collapse
|
14
|
Wang YH, Yang YL, Cheng X, Zhang J, Li W, Du GH. Xiao-Xu-Ming decoction extract regulates differentially expressed proteins in the hippocampus after chronic cerebral hypoperfusion. Neural Regen Res 2019; 14:470-479. [PMID: 30539815 PMCID: PMC6334616 DOI: 10.4103/1673-5374.245471] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Xiao-Xu-Ming decoction has been widely used to treat stroke and sequelae of stroke. We have previously shown that the active fractions of Xiao-Xu-Ming decoction attenuate cerebral ischemic injury. However, the global protein profile and signaling conduction pathways regulated by Xiao-Xu-Ming decoction are still unclear. This study established a two-vessel occlusion rat model by bilateral common carotid artery occlusion. Rats were intragastrically administered 50 or 150 mg/kg Xiao-Xu-Ming decoction for 4 consecutive weeks. Learning and memory abilities were measured with Morris water maze. Motor ability was detected with prehensile test. Coordination ability was examined using the inclined screen test. Neuronal plasticity was observed by immunofluorescent staining. Differentially expressed proteins of rat hippocampus were analyzed by label-free quantitative proteomics. Real time-polymerase chain reaction and western blot assay were used to identify the changes in proteins. Results showed that Xiao-Xu-Ming decoction dramatically alleviated learning and memory deficits, and motor and coordination dysfunction, and increased the expression of microtubule-associated protein 2. Xiao-Xu-Ming decoction extract remarkably decreased 13 upregulated proteins and increased 39 downregulated proteins. The regulated proteins were mainly involved in oxidation reduction process, intracellular signaling cascade process, and protein catabolic process. The signaling pathways were mainly involved in ubiquitin mediated proteolysis and the phosphatidylinositol signaling system. Furthermore, there was an interaction among Rab2a, Ptpn1, Ppm1e, Cdk18, Gorasp2, Eps15, Capza2, Syngap1 and Mt-nd1. Protein analyses confirmed the changes in expression of MT-ND1. The current findings provide new insights into the molecular mechanisms of Xiao-Xu-Ming decoction extract’s effects on chronic cerebral hypoperfusion.
Collapse
Affiliation(s)
- Yue-Hua Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines; Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ying-Lin Yang
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiao Cheng
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jun Zhang
- Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wan Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines; Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Guan-Hua Du
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines; Beijing Key Laboratory of Drug Target Identification and Drug Screening, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
15
|
Nokkari A, Abou-El-Hassan H, Mechref Y, Mondello S, Kindy MS, Jaffa AA, Kobeissy F. Implication of the Kallikrein-Kinin system in neurological disorders: Quest for potential biomarkers and mechanisms. Prog Neurobiol 2018; 165-167:26-50. [PMID: 29355711 PMCID: PMC6026079 DOI: 10.1016/j.pneurobio.2018.01.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 01/15/2018] [Indexed: 01/06/2023]
Abstract
Neurological disorders represent major health concerns in terms of comorbidity and mortality worldwide. Despite a tremendous increase in our understanding of the pathophysiological processes involved in disease progression and prevention, the accumulated knowledge so far resulted in relatively moderate translational benefits in terms of therapeutic interventions and enhanced clinical outcomes. Aiming at specific neural molecular pathways, different strategies have been geared to target the development and progression of such disorders. The kallikrein-kinin system (KKS) is among the most delineated candidate systems due to its ubiquitous roles mediating several of the pathophysiological features of these neurological disorders as well as being implicated in regulating various brain functions. Several experimental KKS models revealed that the inhibition or stimulation of the two receptors of the KKS system (B1R and B2R) can exhibit neuroprotective and/or adverse pathological outcomes. This updated review provides background details of the KKS components and their functions in different neurological disorders including temporal lobe epilepsy, traumatic brain injury, stroke, spinal cord injury, Alzheimer's disease, multiple sclerosis and glioma. Finally, this work will highlight the putative roles of the KKS components as potential neurotherapeutic targets and provide future perspectives on the possibility of translating these findings into potential clinical biomarkers in neurological disease.
Collapse
Affiliation(s)
- Amaly Nokkari
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon
| | - Hadi Abou-El-Hassan
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, USA
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Mark S Kindy
- Department of Pharmaceutical Science, College of Pharmacy, University of South Florida, Tampa, FL, USA; James A. Haley VA Medical Center, Tampa, FL, USA
| | - Ayad A Jaffa
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon; Department of Medicine, Medical University of South, Charleston, SC, USA.
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon; Center for Neuroproteomics & Biomarkers Research, Department of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
16
|
Zhang K, Yan J, Wang L, Tian X, Zhang T, Guo L, Li B, Wang W, Liu X. The Pyk2/MCU pathway in the rat middle cerebral artery occlusion model of ischemic stroke. Neurosci Res 2018; 131:52-62. [DOI: 10.1016/j.neures.2017.09.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 08/02/2017] [Accepted: 09/06/2017] [Indexed: 12/16/2022]
|
17
|
Neuroprotection of bradykinin/bradykinin B2 receptor system in cerebral ischemia. Biomed Pharmacother 2017; 94:1057-1063. [DOI: 10.1016/j.biopha.2017.08.042] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 08/07/2017] [Accepted: 08/07/2017] [Indexed: 12/17/2022] Open
|
18
|
Wu D, Wang J, Wang H, Ji A, Li Y. Protective roles of bioactive peptides during ischemia-reperfusion injury: From bench to bedside. Life Sci 2017; 180:83-92. [PMID: 28527782 DOI: 10.1016/j.lfs.2017.05.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 05/09/2017] [Accepted: 05/10/2017] [Indexed: 12/14/2022]
Abstract
Ischemia-reperfusion (I/R) is a well-known pathological condition which may lead to disability and mortality. I/R injury remains an unresolved and complicated situation in a number of clinical conditions, such as cardiac arrest with successful reanimation, as well as ischemic events in brain and heart. Peptides have many attractive advantages which make them suitable candidate drugs in treating I/R injury, such as low toxicity and immunogenicity, good solubility property, distinct tissue distribution pattern, and favorable pharmacokinetic profile. An increasing number of studies indicate that peptides could protect against I/R injury in many different organs and tissues. Peptides also face several therapeutic challenges that limit their clinical application. In this review, we present the mechanisms of action of peptides in reducing I/R injury, as well as further discuss modification strategies to improve the functional properties of bioactive peptides.
Collapse
Affiliation(s)
- Dongdong Wu
- Henan University School of Basic Medical Sciences, Kaifeng 475004, Henan, China; Institute of Environmental Medicine, Henan University, Kaifeng 475004, Henan, China
| | - Jun Wang
- Henan University School of Basic Medical Sciences, Kaifeng 475004, Henan, China; Institute of Environmental Medicine, Henan University, Kaifeng 475004, Henan, China
| | - Honggang Wang
- Henan University School of Basic Medical Sciences, Kaifeng 475004, Henan, China; Institute of Environmental Medicine, Henan University, Kaifeng 475004, Henan, China
| | - Ailing Ji
- Henan University School of Basic Medical Sciences, Kaifeng 475004, Henan, China; Institute of Environmental Medicine, Henan University, Kaifeng 475004, Henan, China.
| | - Yanzhang Li
- Henan University School of Basic Medical Sciences, Kaifeng 475004, Henan, China; Institute of Environmental Medicine, Henan University, Kaifeng 475004, Henan, China.
| |
Collapse
|
19
|
Nemethova M, Talian I, Danielisova V, Tkacikova S, Bonova P, Bober P, Matiasova M, Sabo J, Burda J. Delayed bradykinin postconditioning modulates intrinsic neuroprotective enzyme expression in the rat CA1 region after cerebral ischemia: a proteomic study. Metab Brain Dis 2016; 31:1391-1403. [PMID: 27393013 DOI: 10.1007/s11011-016-9859-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 06/14/2016] [Indexed: 10/21/2022]
Abstract
Pyramidal cells in the CA1 brain region exhibit an ischemic tolerance after delayed postconditioning; therefore, this approach seems to be a promising neuroprotective procedure in cerebral postischemic injury improvement. However, little is known about the effect of postconditioning on protein expression patterns in the brain, especially in the affected hippocampal neurons after global cerebral ischemia. This study is focused on the examination of the ischemia-vulnerable CA1 neuronal layer and on the acquisition of protection from delayed neuronal death after ischemia. Ischemic-reperfusion injury was induced in Wistar rats and bradykinin was applied 2 days after the ischemic insult in an attempt to overcome delayed cell death. Analysis of complex peptide CA1 samples was performed by automated two dimensional liquid chromatography (2D-LC) fractionation coupled to tandem matrix assisted laser desorption/ionization time-of-flight (MALDI TOF/TOF) mass spectrometry instrumentation. We devoted our attention to differences in protein expression mapping in ischemic injured CA1 neurons in comparison with equally affected neurons, but with bradykinin application. Proteomic analysis identified several proteins occurring only after postconditioning and control, which could have a potentially neuroprotective influence on ischemic injured neurons. Among them, the prominent position occupies a regulator of glutamate level aspartate transaminase AATC, a scavenger of glutamate in brain neuroprotection after ischemia-reperfusion. We identified this enzyme in controls and after postconditioning, but AATC presence was not detected in the ischemic injured CA1 region. This finding was confirmed by two-dimensional differential electrophoresis followed by MALDI-TOF/TOF MS identification. Results suggest that bradykinin as delayed postconditioning may be associated with modulation of protein expression after ischemic injury and thus this procedure can be involved in neuroprotective metabolic pathways.
Collapse
Affiliation(s)
| | - Ivan Talian
- Department of Medical and Clinical Biophysics, Faculty of Medicine, P. J. Safarik University, Kosice, Slovakia
| | | | - Sona Tkacikova
- Department of Medical and Clinical Biophysics, Faculty of Medicine, P. J. Safarik University, Kosice, Slovakia
| | - Petra Bonova
- Institute of Neurobiology, SAS, Kosice, Slovakia
| | - Peter Bober
- Department of Medical and Clinical Biophysics, Faculty of Medicine, P. J. Safarik University, Kosice, Slovakia
| | | | - Jan Sabo
- Department of Medical and Clinical Biophysics, Faculty of Medicine, P. J. Safarik University, Kosice, Slovakia
| | - Jozef Burda
- Institute of Neurobiology, SAS, Kosice, Slovakia
| |
Collapse
|
20
|
Jin Z, Wu J, Yan LJ. Chemical Conditioning as an Approach to Ischemic Stroke Tolerance: Mitochondria as the Target. Int J Mol Sci 2016; 17:351. [PMID: 27005615 PMCID: PMC4813212 DOI: 10.3390/ijms17030351] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 02/26/2016] [Accepted: 03/04/2016] [Indexed: 12/17/2022] Open
Abstract
It is well established that the brain can be prepared to resist or tolerate ischemic stroke injury, and mitochondrion is a major target for this tolerance. The preparation of ischemic stroke tolerance can be achieved by three major approaches: ischemic conditioning, hypoxic conditioning and chemical conditioning. In each conditioning approach, there are often two strategies that can be used to achieve the conditioning effects, namely preconditioning (Pre-C) and postconditioning (Post-C). In this review, we focus on chemical conditioning of mitochondrial proteins as targets for neuroprotection against ischemic stroke injury. Mitochondrial targets covered include complexes I, II, IV, the ATP-sensitive potassium channel (mitoKATP), adenine dinucleotide translocase (ANT) and the mitochondrial permeability transition pore (mPTP). While numerous mitochondrial proteins have not been evaluated in the context of chemical conditioning and ischemic stroke tolerance, the paradigms and approaches reviewed in this article should provide general guidelines on testing those mitochondrial components that have not been investigated. A deep understanding of mitochondria as the target of chemical conditioning for ischemic stroke tolerance should provide valuable insights into strategies for fighting ischemic stroke, a leading cause of death in the world.
Collapse
Affiliation(s)
- Zhen Jin
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA.
| | - Jinzi Wu
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA.
| | - Liang-Jun Yan
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX 76107, USA.
| |
Collapse
|
21
|
dos Anjos LC, Gomes FMM, do Couto LL, Mourão CA, Moreira KG, Silva LP, Mortari MR. Anxiolytic activity and evaluation of potentially adverse effects of a bradykinin-related peptide isolated from a social wasp venom. Life Sci 2016; 149:153-9. [PMID: 26898126 DOI: 10.1016/j.lfs.2016.02.063] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 02/12/2016] [Accepted: 02/16/2016] [Indexed: 01/04/2023]
Abstract
Anxiety disorders are major health problems in terms of costs stemming from sick leave, disabilities, healthcare and premature mortality. Despite the availability of classic anxiolytics, some anxiety disorders are still resistant to treatment, with higher rates of adverse effects. In this respect, several toxins isolated from arthropod venoms are useful in identifying new compounds to treat neurological disorders, particularly pathological anxiety. Thus, the aims of this study were to identify and characterize an anxiolytic peptide isolated from the venom of the social wasp Polybia paulista. The peptide was identified as Polisteskinin R, with nominal molecular mass [M+H](+)=1301Da and primary structure consisting of Ala-Arg-Arg-Pro-Pro-Gly-Phe-Thr-Pro-Phe-Arg-OH. The anxiolytic effect was tested using the elevated plus maze test. Moreover, adverse effects on the spontaneous behavior and motor coordination of animals were assessed using the open field and rotarod tests. Polisteskinin R induced a dose-dependent anxiolytic effect. Animals treated with the peptide and diazepam spent significantly more time into the open arms when compared to the groups treated with the vehicle and pentylenetetrazole. No significant differences in spontaneous behavior or motor coordination were observed between the groups, showing that the peptide was well tolerated. The interaction by agonists in both known BK receptors induces a variability of physiological effects; Polisteskinin R can act on these receptors, inducing modulatory activity and thus, attenuating anxiety behaviors. The results of this study demonstrated that the compound Polisteskinin R exerted potent anxiolytic effects and its analogues are promising candidates for experimental pharmacology.
Collapse
Affiliation(s)
- Lilian Carneiro dos Anjos
- Neuropharmacology Laboratory, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, DF, Brazil; Graduate Program of Animal Biology, University of Brasília, Brasília, DF, Brazil
| | - Flávia Maria Medeiros Gomes
- Neuropharmacology Laboratory, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, DF, Brazil; Graduate Program of Animal Biology, University of Brasília, Brasília, DF, Brazil
| | - Lucianna Lopes do Couto
- Neuropharmacology Laboratory, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, DF, Brazil
| | - Cecília Alves Mourão
- Neuropharmacology Laboratory, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, DF, Brazil
| | | | - Luciano Paulino Silva
- Graduate Program of Animal Biology, University of Brasília, Brasília, DF, Brazil; Laboratory of Mass Spectrometry and Laboratory of Nanobiotechnology, Embrapa Genetic Resources and Biotechnology, Brasília, DF, Brazil
| | - Márcia Renata Mortari
- Neuropharmacology Laboratory, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, DF, Brazil; Graduate Program of Animal Biology, University of Brasília, Brasília, DF, Brazil.
| |
Collapse
|
22
|
Sharma R, Randhawa PK, Singh N, Jaggi AS. Bradykinin in ischemic conditioning-induced tissue protection: Evidences and possible mechanisms. Eur J Pharmacol 2015; 768:58-70. [DOI: 10.1016/j.ejphar.2015.10.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 10/15/2015] [Accepted: 10/15/2015] [Indexed: 01/02/2023]
|
23
|
Pharmacological Alternatives for the Treatment of Neurodegenerative Disorders: Wasp and Bee Venoms and Their Components as New Neuroactive Tools. Toxins (Basel) 2015; 7:3179-209. [PMID: 26295258 PMCID: PMC4549745 DOI: 10.3390/toxins7083179] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 08/01/2015] [Accepted: 08/05/2015] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative diseases are relentlessly progressive, severely impacting affected patients, families and society as a whole. Increased life expectancy has made these diseases more common worldwide. Unfortunately, available drugs have insufficient therapeutic effects on many subtypes of these intractable diseases, and adverse effects hamper continued treatment. Wasp and bee venoms and their components are potential means of managing or reducing these effects and provide new alternatives for the control of neurodegenerative diseases. These venoms and their components are well-known and irrefutable sources of neuroprotectors or neuromodulators. In this respect, the present study reviews our current understanding of the mechanisms of action and future prospects regarding the use of new drugs derived from wasp and bee venom in the treatment of major neurodegenerative disorders, including Alzheimer’s Disease, Parkinson’s Disease, Epilepsy, Multiple Sclerosis and Amyotrophic Lateral Sclerosis.
Collapse
|
24
|
Leandra C, Tasca CI, Boeck CR. The Role of NMDA Receptors in the Development of Brain Resistance through Pre- and Postconditioning. Aging Dis 2014; 5:430-41. [PMID: 25489494 DOI: 10.14336/ad.2014.0500430] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 01/30/2014] [Accepted: 02/09/2014] [Indexed: 11/01/2022] Open
Abstract
Brain tolerance or resistance can be achieved by interventions before and after injury through potential toxic agents used in low stimulus or dose. For brain diseases, the neuroprotection paradigm desires an attenuation of the resulting motor, cognitive, emotional, or memory deficits following the insult. Preconditioning is a well-established experimental and clinical translational strategy with great beneficial effects, but limited applications. NMDA receptors have been reported as protagonists in the adjacent cellular mechanisms contributing to the development of brain tolerance. Postconditioning has recently emerged as a new neuroprotective strategy, which has shown interesting results when applied immediately, i.e. several hours to days, after a stroke event. Investigations using chemical postconditioning are still incipient, but nevertheless represent an interesting and promising clinical strategy. In the present review pre- and postconditioning are discussed as neuroprotective paradigms and the focus of our attention lies on the participation of NMDA receptors proteins in the processes related to neuroprotection.
Collapse
Affiliation(s)
| | - Carla Inês Tasca
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina-UFSC, Campus Trindade, 88040-900, Florianópolis, SC, Brazil
| | - Carina Rodrigues Boeck
- Laboratório de Biologia Celular e Molecular, Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM), Programa de Pós-graduação Ciências da Saúde, Universidade do Extremo Sul Catarinense-UNESC, Criciúma, 88806-000, SC, Brazil
| |
Collapse
|
25
|
Biological networks in ischemic tolerance - rethinking the approach to clinical conditioning. Transl Stroke Res 2014; 4:114-29. [PMID: 24223074 DOI: 10.1007/s12975-012-0244-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The adaptive response (conditioning) to environmental stressors evokes evolutionarily conserved programs in uni- and multicellular organisms that result in increased fitness and resistance to stressor induced injury. Although the concept of conditioning has been around for a while, its translation into clinical therapies targeting neurovascular diseases has only recently begun. The slow pace of clinical adoption might be partially explained by our poor understanding of underpinning mechanisms and of the complex responses of the organism to the stressor. At the 2(nd) Translational Preconditioning Meeting participants engaged in an intense discussion addressing whether the time has come to more aggressively implement clinical conditioning protocols in the treatment of cerebrovascular diseases or whether it would be better to wait until preclinical data would help to minimize clinical empiricism. This review addresses the complex involvement of biological networks in establishing ischemic tolerance at the organism level using two clinically promising conditioning modalities, namely remote ischemic preconditioning, and per- or post-conditioning, as examples.
Collapse
|
26
|
Danielisova V, Gottlieb M, Bonova P, Nemethova M, Burda J. Bradykinin postconditioning ameliorates focal cerebral ischemia in the rat. Neurochem Int 2014; 72:22-9. [DOI: 10.1016/j.neuint.2014.04.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 03/20/2014] [Accepted: 04/08/2014] [Indexed: 01/10/2023]
|
27
|
Bradykinin preconditioning affects the number of degenerated neurons and the level of antioxidant enzymes in spinal cord ischemia in rabbits. Acta Histochem 2014; 116:252-7. [PMID: 23981244 DOI: 10.1016/j.acthis.2013.07.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Revised: 07/22/2013] [Accepted: 07/23/2013] [Indexed: 12/16/2022]
Abstract
Bradykinin preconditioning has been used for acquisition of tolerance after spinal cord ischemia. Rabbits were preconditioned intraperitoneally with bradykinin 48 h prior to 20 min of abdominal aorta ligation followed by 24 and 48 h of reperfusion. The activities of SOD and catalase were measured and Fluoro Jade B (FJB)-positive degenerated neurons were evaluated. The outcomes of Tarlov scoring system used to assess neurological functions showed significant improvement in bradykinin groups compared to the ischemic group. The number of FJB-positive degenerated neurons was decreased in ventral horns of both bradykinin groups. Significantly decreased activities of total SOD and mitochondrial Mn-SOD were also detected in both bradykinin groups versus ischemic group while CuZn-SOD and catalase activities were significantly decreased only in the bradykinin group after 24h of reperfusion versus ischemic group. These findings suggest that one of the possibilities of the neuroprotective effect of delayed bradykinin preconditioning against spinal cord ischemic injury could be realized by mitochondrial protection and decreased synthesis of Mn-SOD as well as by promotion of neuronal survival.
Collapse
|
28
|
Zhang J, Lu W, Lei Q, Tao X, You H, Xie P. Salvianolate increases heat shock protein expression in a cerebral ischemia-reperfusion injury model. Neural Regen Res 2013; 8:2327-2335. [PMID: 25206542 PMCID: PMC4146039 DOI: 10.3969/j.issn.1673-5374.2013.25.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2013] [Accepted: 07/25/2013] [Indexed: 11/18/2022] Open
Abstract
Stroke remains a worldwide health problem. Salvianolate exerts a protective effect in various mi-crocirculatory disturbance-related diseases, but studies of the mechanisms underlying its protective action have mainly focused on the myocardium, whereas little research has been carried out in brain tissue following ischemia-reperfusion. We assessed the neuroprotective effects of salvianolate in a rat model of cerebral ischemia-reperfusion injury induced using the suture method. At onset and 24 and 48 hours after reperfusion, rats were intraperitoneally injected with salvianolate (18 mg/kg) or saline. Neurological deficit scores at 72 hours showed that the neurological functions of rats that had received salvianolate were significantly better than those of the rats that had received saline. 2,3,5-Triphenyltetrazolium chloride was used to stain cerebral tissue to determine the extent of the infarct area. A significantly smaller infarct area and a significantly lower number of apoptotic cells were observed after treatment with salvianolate compared with the saline treatment. Expression of heat shock protein 22 and phosphorylated protein kinase B in ischemic brain tissue was significantly greater in rats treated with salvianolate compared with rats treated with saline. Our findings suggest that salvianolate provides neuroprotective effects against cerebral ischemia-reperfusion injury by upregulating heat shock protein 22 and phosphorylated protein kinase B expression.
Collapse
Affiliation(s)
- Jinnan Zhang
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Wei Lu
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Qiang Lei
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Xi Tao
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Hong You
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Pinghui Xie
- Department of Neurology, Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
| |
Collapse
|
29
|
Vinten-Johansen J, Shi W. The science and clinical translation of remote postconditioning. J Cardiovasc Med (Hagerstown) 2013; 14:206-13. [PMID: 23412366 DOI: 10.2459/jcm.0b013e32835cecc6] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The treatment of reperfusion injury requires measures beyond timely reperfusion. Conventional postconditioning (PostC) of ischemic tissues offers a strategy to reduce reperfusion injury, but its adoption is challenged by requiring access and imposing additional ischemia to the ischemic organ. Generating protective signals by PostC in a tissue remote from the target organ such as the limb, i.e. remote PostC (rPostC), may present an alternative approach to exerting endogenous tissue protection. Because rPostC is only recently reported, the fundamental biology of rPostC is not well understood, and studies to date are largely observational. rPostC has been observed to reduce ischemia-reperfusion injury experimentally in heart, kidney, brain and skeletal muscle in multiple species, including rat, rabbit and pig. Both necrosis and apoptosis are reduced. As in remote ischemic preconditioning, rPostC requires a transfer or communication of protective factors or signals through humoral and/or neural pathways. Triggers of target organ protection include G-protein-coupled receptor ligands, metabolites of ischemia, or small thermolabile molecules. Some evidence suggests that reperfusion injury salvage kinases may be involved in rPostC, in agreement with both preconditioning and conventional PostC. Clinical studies investigating improvements in clinical outcomes or biomarkers with rPostC are encouraging.
Collapse
Affiliation(s)
- Jakob Vinten-Johansen
- Division of Cardiothoracic Surgery, Cardiothoracic Research Laboratory, Carlyle Fraser Heart Center of Emory University Hospital Midtown, Emory University School of Medicine, Atlanta, Georgia 30308-2225, USA.
| | | |
Collapse
|
30
|
Torres-Rivera W, Pérez D, Park KY, Carrasco M, Platt MO, Eterović VA, Ferchmin PA, Ulrich H, Martins AH. Kinin-B2 receptor exerted neuroprotection after diisopropylfluorophosphate-induced neuronal damage. Neuroscience 2013; 247:273-9. [PMID: 23735753 DOI: 10.1016/j.neuroscience.2013.05.054] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 05/23/2013] [Accepted: 05/24/2013] [Indexed: 02/06/2023]
Abstract
The kinin-B2 receptor (B2BKR) activated by its endogenous ligand bradykinin participates in various metabolic processes including the control of arterial pressure and inflammation. Recently, functions for this receptor in brain development and protection against glutamate-provoked excitotoxicity have been proposed. Here, we report neuroprotective properties for bradykinin against organophosphate poisoning using acute hippocampal slices as an in vitro model. Following slice perfusion for 10min with diisopropylfluorophosphate (DFP) to initiate the noxious stimulus, responses of pyramidal neurons upon an electric impulse were reduced to less than 30% of control amplitudes. Effects on synaptic-elicited population spikes were reverted when preparations had been exposed to bradykinin 30min after challenging with DFP. Accordingly, bradykinin-induced population spike recovery was abolished by HOE-140, a B2BKR antagonist. However, the kinin-B1 receptor (B1BKR) agonist Lys-des-Arg(9)-bradykinin, inducing the phosphorylation of mitogen-activated protein kinase (MEK/MAPK) and cell death, abolished bradykinin-mediated neuroprotection, an effect, which was reverted by the ERK inhibitor PD98059. In agreement with pivotal B1BKR functions in this process, antagonism of endogenous B1BKR activity alone was enough for restoring population spike activity. On the other hand pralidoxime, an oxime, reactivating acetylcholinesterase (AChE) after organophosphate poisoning, induced population spike recovery after DFP exposure in the presence of bradykinin and Lys-des-Arg(9)-bradykinin. Lys-des-Arg(9)-bradykinin did not revert protection exerted by pralidoxime, however when instead bradykinin and Ly-des-Arg(9)-bradykinin were superfused together, recovery of population spikes diminished. These findings again confirm the neuroprotective feature of bradykinin, which is, diminished by its endogenous metabolites, stimulating the B1BKR, providing a novel understanding of the physiological roles of these receptors.
Collapse
Affiliation(s)
- W Torres-Rivera
- Department of Biochemistry, Universidad Central del Caribe, Bayamón, PR 00956, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Remifentanil postconditioning improves global cerebral ischemia-induced spatial learning and memory deficit in rats via inhibition of neuronal apoptosis through the PI3K signaling pathway. Neurol Sci 2013; 34:1955-62. [DOI: 10.1007/s10072-013-1419-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 03/20/2013] [Indexed: 02/06/2023]
|
32
|
Bonova P, Burda J, Danielisova V, Nemethova M, Gottlieb M. Delayed post-conditioning reduces post-ischemic glutamate level and improves protein synthesis in brain. Neurochem Int 2013; 62:854-60. [PMID: 23454191 DOI: 10.1016/j.neuint.2013.02.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 01/23/2013] [Accepted: 02/03/2013] [Indexed: 11/16/2022]
Abstract
In the clinic delayed post-conditioning would represent an attractive strategy for the survival of vulnerable neurons after an ischemic event. In this paper we studied the impact of ischemia and delayed post-conditioning on blood and brain tissue concentrations of glutamate and protein synthesis. We designed two groups of animals for analysis of brain tissues and blood after global ischemia and post-conditioning, and one for analysis of blood glutamate after transient focal ischemia. Our results showed elevated blood glutamate in two models of transient brain ischemia and decreases in blood glutamate to control in the first 20min of post-conditioning recirculation followed by a consecutive drop of about 20.5% on the first day. Similarly, we recorded reduced protein synthesis in hippocampus and cortex 2 and 3days after ischemia. However, increased glutamate was registered only in the hippocampus. Post-conditioning improves protein synthesis in CA1 and dentate gyrus and, surprisingly, leads to 50% reduction in glutamate in whole hippocampus and cortex. In conclusion, ischemia leads to meaningful elevation of blood and tissue glutamate. Post-conditioning activates mechanisms resulting in rapid elimination of glutamate from brain tissue and/or in the circulatory system that could otherwise impede brain-to-blood glutamate efflux mechanisms. Moreover, post-conditioning induces protein synthesis renewing in ischemia affected tissues that could also contribute to elimination of excitotoxicity. In addition, the potential of glutamate for monitoring the progress of ischemia and efficacy of therapy was shown.
Collapse
Affiliation(s)
- Petra Bonova
- Institute of Neurobiology, Slovak Academy of Sciences, Kosice, Slovak Republic.
| | | | | | | | | |
Collapse
|
33
|
Bonova P, Burda J, Danielisova V, Nemethova M, Gottlieb M. Development of a pattern in biochemical parameters in the core and penumbra during infarct evolution after transient MCAO in rats. Neurochem Int 2013; 62:8-14. [DOI: 10.1016/j.neuint.2012.10.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 10/04/2012] [Accepted: 10/30/2012] [Indexed: 10/27/2022]
|
34
|
Albert-Weißenberger C, Sirén AL, Kleinschnitz C. Ischemic stroke and traumatic brain injury: the role of the kallikrein-kinin system. Prog Neurobiol 2012; 101-102:65-82. [PMID: 23274649 DOI: 10.1016/j.pneurobio.2012.11.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 11/15/2012] [Accepted: 11/20/2012] [Indexed: 12/16/2022]
Abstract
Acute ischemic stroke and traumatic brain injury are a major cause of mortality and morbidity. Due to the paucity of therapies, there is a pressing clinical demand for new treatment options. Successful therapeutic strategies for these conditions must target multiple pathophysiological mechanisms occurring at different stages of brain injury. In this respect, the kallikrein-kinin system is an ideal target linking key pathological hallmarks of ischemic and traumatic brain damage such as edema formation, inflammation, and thrombosis. In particular, the kinin receptors, plasma kallikrein, and coagulation factor XIIa are highly attractive candidates for pharmacological development, as kinin receptor antagonists or inhibitors of plasma kallikrein and coagulation factor XIIa are neuroprotective in animal models of stroke and traumatic brain injury. Nevertheless, conflicting preclinical evaluation as well as limited and inconclusive data from clinical trials suggest caution when transferring observations made in animals into the human situation. This review summarizes current evidence on the pathological significance of the kallikrein-kinin system during ischemic and traumatic brain damage, with a particular focus on experimental data derived from animal models. Experimental findings are also compared with human data if available, and potential therapeutic implications are discussed.
Collapse
|
35
|
Inhibition of autophagy contributes to ischemic postconditioning-induced neuroprotection against focal cerebral ischemia in rats. PLoS One 2012; 7:e46092. [PMID: 23029398 PMCID: PMC3461004 DOI: 10.1371/journal.pone.0046092] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 08/27/2012] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Ischemic postconditioning (IPOC), or relief of ischemia in a stuttered manner, has emerged as an innovative treatment strategy to reduce programmed cell death, attenuate ischemic injuries, and improve neurological outcomes. However, the mechanisms involved have not been completely elucidated. Recent studies indicate that autophagy is a type of programmed cell death that plays elusive roles in controlling neuronal damage and metabolic homeostasis. This study aims to determine the role of autophagy in IPOC-induced neuroprotection against focal cerebral ischemia in rats. METHODOLOGY/PRINCIPAL FINDINGS A focal cerebral ischemic model with permanent middle cerebral artery (MCA) occlusion plus transient common carotid artery (CCA) occlusion was established. The autophagosomes and the expressions of LC3/Beclin 1/p62 were evaluated for their contribution to the activation of autophagy. We found that autophagy was markedly induced with the upregulation of LC3/Beclin 1 and downregulation of p62 in the penumbra at various time intervals following ischemia. IPOC, performed at the onset of reperfusion, reduced infarct size, mitigated brain edema, inhibited the induction of LC3/Beclin 1 and reversed the reduction of p62 simultaneously. Rapamycin, an inducer of autophagy, partially reversed all the aforementioned effects induced by IPOC. Conversely, autophagy inhibitor 3-methyladenine (3-MA) attenuated the ischemic insults, inhibited the activation of autophagy, and elevated the expression of anti-apoptotic protein Bcl-2, to an extent comparable to IPOC. CONCLUSIONS/SIGNIFICANCE The present study suggests that inhibition of the autophagic pathway plays a key role in IPOC-induced neuroprotection against focal cerebral ischemia. Thus, pharmacological inhibition of autophagy may provide a novel therapeutic strategy for the treatment of stroke.
Collapse
|
36
|
Danielisova V, Burda J, Nemethova M, Gottlieb M, Burda R. An Effective Combination of Two Different Methods of Postconditioning. Neurochem Res 2012; 37:2085-91. [DOI: 10.1007/s11064-012-0829-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 05/02/2012] [Accepted: 06/08/2012] [Indexed: 10/28/2022]
|
37
|
Rehni AK, Singh TG. Involvement of CCR-2 chemokine receptor activation in ischemic preconditioning and postconditioning of brain in mice. Cytokine 2012; 60:83-9. [PMID: 22704692 DOI: 10.1016/j.cyto.2012.05.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2011] [Revised: 04/24/2012] [Accepted: 05/08/2012] [Indexed: 11/25/2022]
Abstract
The present study has been designed to investigate the potential role of CCR-2 chemokine receptor in ischemic preconditioning as well as postconditioning induced reversal of ischemia-reperfusion injury in mouse brain. Bilateral carotid artery occlusion of 17 min followed by reperfusion for 24h was employed in present study to produce ischemia and reperfusion induced cerebral injury in mice. Cerebral infarct size was measured using triphenyltetrazolium chloride staining. Memory was evaluated using elevated plus-maze test and Morris water maze test. Rota rod test was employed to assess motor incoordination. Bilateral carotid artery occlusion followed by reperfusion produced cerebral infarction and impaired memory and motor co-ordination. Three preceding episodes of bilateral carotid artery occlusion for 1 min and reperfusion of 1 min were employed to elicit ischemic preconditioning of brain, while three episodes of bilateral carotid artery occlusion for 10s and reperfusion of 10s immediately after the completion of were employed to elicit ischemic postconditioning of brain. Both prior ischemic preconditioning as well as ischemic postconditioning immediately after global cerebral ischemia prevented markedly ischemia-reperfusion-induced cerebral injury as measured in terms of infarct size, loss of memory and motor coordination. RS 102895, a selective CCR-2 chemokine receptor antagonist, attenuated the neuroprotective effect of both the ischemic preconditioning as well as postconditioning. It is concluded that the neuroprotective effect of both ischemic preconditioning as well as ischemic postconditioning may involve the activation of CCR-2 chemokine receptors.
Collapse
Affiliation(s)
- Ashish K Rehni
- Chitkara College of Pharmacy, Chandigarh-Patiala National Highway, Rajpura, 140 401 Patiala, Punjab, India
| | | |
Collapse
|
38
|
Kinin-B2 receptor mediated neuroprotection after NMDA excitotoxicity is reversed in the presence of kinin-B1 receptor agonists. PLoS One 2012; 7:e30755. [PMID: 22348022 PMCID: PMC3277507 DOI: 10.1371/journal.pone.0030755] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Accepted: 12/21/2011] [Indexed: 01/21/2023] Open
Abstract
Background Kinins, with bradykinin and des-Arg9-bradykinin being the most important ones, are pro-inflammatory peptides released after tissue injury including stroke. Although the actions of bradykinin are in general well characterized; it remains controversial whether the effects of bradykinin are beneficial or not. Kinin-B2 receptor activation participates in various physiological processes including hypotension, neurotransmission and neuronal differentiation. The bradykinin metabolite des-Arg9-bradykinin as well as Lys-des-Arg9-bradykinin activates the kinin-B1 receptor known to be expressed under inflammatory conditions. We have investigated the effects of kinin-B1 and B2 receptor activation on N-methyl-D-aspartate (NMDA)-induced excitotoxicity measured as decreased capacity to produce synaptically evoked population spikes in the CA1 area of rat hippocampal slices. Principal Findings Bradykinin at 10 nM and 1 µM concentrations triggered a neuroprotective cascade via kinin-B2 receptor activation which conferred protection against NMDA-induced excitotoxicity. Recovery of population spikes induced by 10 nM bradykinin was completely abolished when the peptide was co-applied with the selective kinin-B2 receptor antagonist HOE-140. Kinin-B2 receptor activation promoted survival of hippocampal neurons via phosphatidylinositol 3-kinase, while MEK/MAPK signaling was not involved in protection against NMDA-evoked excitotoxic effects. However, 100 nM Lys-des-Arg9-bradykinin, a potent kinin-B1 receptor agonist, reversed bradykinin-induced population spike recovery. The inhibition of population spikes recovery was reversed by PD98059, showing that MEK/MAPK was involved in the induction of apoptosis mediated by the B1 receptor. Conclusions Bradykinin exerted protection against NMDA-induced excitotoxicity which is reversed in the presence of a kinin-B1 receptor agonist. As bradykinin is converted to the kinin-B1 receptor metabolite des-Arg9-bradykinin by carboxypeptidases, present in different areas including in brain, our results provide a mechanism for the neuroprotective effect in vitro despite of the deleterious effect observed in vivo.
Collapse
|
39
|
Dutra RC, Leite DFP, Bento AF, Manjavachi MN, Patrício ES, Figueiredo CP, Pesquero JB, Calixto JB. The role of kinin receptors in preventing neuroinflammation and its clinical severity during experimental autoimmune encephalomyelitis in mice. PLoS One 2011; 6:e27875. [PMID: 22132157 PMCID: PMC3222659 DOI: 10.1371/journal.pone.0027875] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Accepted: 10/27/2011] [Indexed: 11/19/2022] Open
Abstract
Background Multiple sclerosis (MS) is a demyelinating and neuroinflammatory disease of the human central nervous system (CNS). The expression of kinins is increased in MS patients, but the underlying mechanisms by which the kinin receptor regulates MS development have not been elucidated. Methodology/Principal Findings Experimental autoimmune encephalomyelitis (EAE) was induced in female C57BL/6 mice by immunization with MOG35–55 peptide emulsified in complete Freund's adjuvant and injected with pertussis toxin on day 0 and day 2. Here, we report that blockade of the B1R in the induction phase of EAE markedly suppressed its progression by interfering with the onset of the immune response. Furthermore, B1R antagonist suppressed the production/expression of antigen-specific TH1 and TH17 cytokines and transcription factors, both in the periphery and in the CNS. In the chronic phase of EAE, the blockade of B1R consistently impaired the clinical progression of EAE. Conversely, administration of the B1R agonist in the acute phase of EAE suppressed disease progression and inhibited the increase in permeability of the blood-brain barrier (BBB) and any further CNS inflammation. Of note, blockade of the B2R only showed a moderate impact on all of the studied parameters of EAE progression. Conclusions/Significance Our results strongly suggest that kinin receptors, mainly the B1R subtype, play a dual role in EAE progression depending on the phase of treatment through the lymphocytes and glial cell-dependent pathways.
Collapse
MESH Headings
- Animals
- Bradykinin Receptor Antagonists
- CD4-Positive T-Lymphocytes/immunology
- Chronic Disease
- Cytokines/biosynthesis
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/complications
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Gene Deletion
- Humans
- Inflammation/complications
- Inflammation/pathology
- Inflammation/prevention & control
- Lymphoid Tissue/immunology
- Lymphoid Tissue/pathology
- Mice
- Mice, Inbred C57BL
- Models, Biological
- Myelin Sheath/metabolism
- Receptors, Bradykinin/metabolism
- Spinal Cord/metabolism
- Spinal Cord/pathology
- Th1 Cells/immunology
- Th17 Cells/immunology
Collapse
Affiliation(s)
- Rafael C. Dutra
- Department of Pharmacology, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Daniela F. P. Leite
- Department of Pharmacology, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Allisson F. Bento
- Department of Pharmacology, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Marianne N. Manjavachi
- Department of Pharmacology, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Eliziane S. Patrício
- Department of Pharmacology, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Cláudia P. Figueiredo
- Department of Pharmacology, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - João B. Pesquero
- Department of Biophysics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - João B. Calixto
- Department of Pharmacology, Centre of Biological Sciences, Universidade Federal de Santa Catarina, Florianópolis, Santa Catarina, Brazil
- * E-mail:
| |
Collapse
|
40
|
Delayed ischemic postconditioning protects hippocampal CA1 neurons by preserving mitochondrial integrity via Akt/GSK3β signaling. Neurochem Int 2011; 59:749-58. [PMID: 21867737 DOI: 10.1016/j.neuint.2011.08.008] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Revised: 08/05/2011] [Accepted: 08/08/2011] [Indexed: 11/20/2022]
Abstract
Delayed ischemic postconditioning (Post C), which involves a brief ischemia followed by reperfusion 2 days after 8-10min global cerebral ischemia (GCI), has been shown to exert a remarkable protection of the vulnerable hippocampal CA1 region of the brain and attenuation of behavioral deficits, although the mechanisms remain poorly understood. The purpose of the current study was to explore the effect of Post C upon mitochondrial integrity, cytochrome c release and Bax translocation as a potential key mechanism for Post C protection of the critical hippocampal CA1 region neurons. The results of the study revealed that ischemic Post C (3min) administered 2 days after 8-min GCI exerted a robust preservation from GCI injury, as evidenced by the increase of NeuN-positive and the decrease of TUNEL-positive cells, as well as morphological features of mitochondrial integrity in the hippocampal CA1 region. We also found that Post C significantly blocked inner mitochondrial membrane potential depolarization, as shown by JC-1 staining, and attenuates cytochrome c release and Bax translocation induced by GCI. Pre-treatment of the PI3K inhibitor LY294002, 20min prior to Post C, significantly attenuated Post C-induced elevation of p-Akt and p-GSK3β, as well as prevented Post C enhancement of mitochondrial integrity and Post C neuroprotection. The results suggest that phosphorylation of Akt and subsequent inactivation of GSK3β signaling is critical in mediating Post C beneficial effects upon mitochondrial integrity, function and neuroprotection following GCI injury.
Collapse
|
41
|
Nagy D, Kocsis K, Fuzik J, Marosi M, Kis Z, Teichberg VI, Toldi J, Farkas T. Kainate postconditioning restores LTP in ischemic hippocampal CA1: onset-dependent second pathophysiological stress. Neuropharmacology 2011; 61:1026-32. [PMID: 21781978 DOI: 10.1016/j.neuropharm.2011.07.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 06/14/2011] [Accepted: 07/06/2011] [Indexed: 11/29/2022]
Abstract
Postconditioning can be induced by a broad range of stimuli within minutes to days after an ischemic cerebral insult. A special form is elicited by pharmacological intervention called second pathophysiological stress. The present study aimed to evaluate the effects of low-dose (5 mg/kg) kainate postconditioning with onsets 0, 24 and 48 h after the ischemic insult on the hippocampal synaptic plasticity in a 2-vessel occlusion model in rat. The hippocampal function was tested by LTP measurements of Schaffer collateral-CA1 pyramidal cell synapses in acute slices and the changes in density of Golgi-Cox-stained apical dendritic spines. Postconditioning 0 and 24 h after ischemia was not protective, whereas 48-h-onset postconditioning resulted in the reappearance of a normal spine density (>100,000 spines) 3 days after ischemia, in parallel with the long-term restoration of the damaged LTP function. Similar, but somewhat less effects were observed after 10 days. Our data clearly demonstrate the onset dependence of postconditioning elicited by a subconvulsant dose of kainate treatment in global ischemia, with restoration of the structural plasticity and hippocampal function.
Collapse
Affiliation(s)
- Dávid Nagy
- Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Wang S, Duan Y, Su D, Li W, Tan J, Yang D, Wang W, Zhao Z, Wang X. Delta opioid peptide [D-Ala2, D-Leu5] enkephalin (DADLE) triggers postconditioning against transient forebrain ischemia. Eur J Pharmacol 2011; 658:140-4. [PMID: 21349267 DOI: 10.1016/j.ejphar.2011.02.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 01/14/2011] [Accepted: 02/06/2011] [Indexed: 11/25/2022]
Abstract
Preconditioning with selective delta opioid peptide [d-Ala2, d-Leu5] enkephalin (DADLE) provides ischemic tolerance following transient forebrain ischemia in rats. However, whether DADLE postconditioning retains its neuroprotective efficacy and the underlying molecular mechanism in ischemic brain is largely unknown. We investigated DADLE postconditioning protection of hippocampal CA1 neurons against transient forebrain ischemia. 6 days after being implanted with cannula at the right lateral ventricle, rats underwent 10 min of forebrain ischemia by four vessel occlusion. Hippocampal CA1 neuronal survival and degeneration were measured in the hippocampi of rats at 3 days after ischemia. The behavioral and cognitive improvements of DADLE treatment in rats were also evaluated on days 5-9 using open-field and Morris water maze tests. The results showed that DADLE at doses of 0.25 and 2.5 nmol, but not 25 nmol, could significantly protect CA1 neurons against ischemia/reperfusion injury. Co-administration with the delta-opioid receptor antagonist naltrindole or pretreatment with the Akt antagonist LY294002 completely abolished the DADLE postconditioning effect. Furthermore, DADLE postconditioning exhibited cognitive benefits in rats with transient forebrain ischemia. The study thus suggested a therapeutic opportunity of postconditioning neuroprotection by DADLE and also provided important information in understanding the mechanism of DADLE action in the ischemic brain.
Collapse
Affiliation(s)
- Shuyan Wang
- Department of Anesthesiology, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Peng Y, Feng SF, Wang Q, Wang HN, Hou WG, Xiong L, Luo ZJ, Tan QR. Hyperbaric oxygen preconditioning ameliorates anxiety-like behavior and cognitive impairments via upregulation of thioredoxin reductases in stressed rats. Prog Neuropsychopharmacol Biol Psychiatry 2010; 34:1018-25. [PMID: 20493230 DOI: 10.1016/j.pnpbp.2010.05.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Revised: 05/12/2010] [Accepted: 05/12/2010] [Indexed: 11/18/2022]
Abstract
The present study examined the protective effect of hyperbaric oxygen preconditioning (HBO-PC) and the role of thioredoxin reductase (TrxR) in a post-traumatic stress disorder (PTSD)-induced rat model by using single prolonged stress (SPS). Rats were randomly divided into Sham, HBO, SPS and HBO+SPS groups. HBO-PC was conducted by exposing rats to 100% oxygen at 2.5atm absolute for 1h each day for 5 consecutive days. SPS was performed 24h after the last HBO-PC conditioning event. At 1h, 6h, 12h, 24h and 72h after SPS, TrxR mRNA expression was analyzed in the hippocampus; Nissl and TUNEL staining were performed at 72h after SPS. The results indicated that HBO-PC was able to significantly preserve viable neurons in the CA1 subfield of hippocampus following SPS exposure, as evidenced by reduced amounts of CA1 neuronal apoptosis. Furthermore, HBO-PC upregulate the expression of TrxR-1 and TrxR-2 mRNA in the hippocampus at 6h and 12h after SPS exposure and ameliorated anxiety-like behavior and cognitive impairments normally induced by SPS. Taken together, these findings suggest that HBO-PC is beneficial for the improvement of anxiety-like behavior and cognitive impairments induced by SPS exposure, and this effect might be associated with inhibition of neuronal apoptosis via upregulation of TrxR in stressed rats.
Collapse
Affiliation(s)
- Ye Peng
- Department of Orthopaedics, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Feng S, Wang Q, Wang H, Peng Y, Wang L, Lu Y, Shi T, Xiong L. Electroacupuncture pretreatment ameliorates hypergravity-induced impairment of learning and memory and apoptosis of hippocampal neurons in rats. Neurosci Lett 2010; 478:150-5. [PMID: 20457216 DOI: 10.1016/j.neulet.2010.05.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Revised: 04/06/2010] [Accepted: 05/04/2010] [Indexed: 01/14/2023]
Abstract
High-sustained positive acceleration (+Gz) exposures might lead to impairment in cognitive function. Our previous studies have shown that electroacupuncture (EA) pretreatment can attenuate transient focal cerebral ischemic injury in the rats. In this study we aimed to investigate whether EA pretreatment could ameliorate the impairment of learning and memory induced by a sustained +Gz exposure. Using the centrifuge model, rats of experimental groups were exposed to +10 Gz for 5 min. Morris water maze was used for assessing the cognitive ability, and the apoptotic hippocampal CA1 pyramidal neuronal cells were evaluated by caspase-3 activity and TUNEL staining. Our results showed that +Gz exposure significantly caused pyramidal neuronal damage, increased neuronal apoptosis and caspase-3 activity in hippocampal CA1 region, as well as resulted in an impairment of spatial learning and memory, as compared to the sham group animals. Furthermore, the EA pretreatment significantly attenuated the neuronal apoptosis, preserved neuronal morphology and inhibited the caspase-3 activity in hippocampal CA1 region resulted from +Gz exposure. The EA pretreatment also ameliorated the learning and memory function in rats exposed to +Gz. These findings indicate that EA pretreatment provides a novel method to prevent the cognitive damage caused by +Gz, which could significantly protect neuronal damage and impairment of learning and memory.
Collapse
Affiliation(s)
- Shufang Feng
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | | | | | | | | | | | | | | |
Collapse
|