1
|
Sun M, Han G, Yu H, Wang F. Kudiezi Injection Attenuates Apoptosis and Neuroinflammation for Ameliorating Angiogenesis via miR-21/PDCD4/NF-κB Axis in MCAO/Reperfusion Rats. JOURNAL OF ETHNOPHARMACOLOGY 2025:120066. [PMID: 40449697 DOI: 10.1016/j.jep.2025.120066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 05/24/2025] [Accepted: 05/27/2025] [Indexed: 06/03/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE miR-21 has multiple neuroprotective effects in central nervous system (CNS) ischemic damage. Kudiezi (KDZ), a traditional Chinese medication, has a longstanding application in cerebral ischemia treatment and has been clinically confirmed to be effective for ischemic injury. However, its specific effects on miR-21 regulation are yet unclear. AIM OF THE STUDY Investigate the fundamental mechanisms of the anti-inflammatory as well as antiapoptotic effects of KDZ in rats subjected to middle cerebral artery occlusion-reperfusion(MCAO/R). MATERIALS AND METHODS MCAO rats were developed, and the TTC staining and Garcia JH scoring methods were used to detect infarct size and analyze the neuroprotective effects of KDZ on this rat model. Real-time PCR and miRNA antagomir lateral ventricular injection were used to evaluate the change in miR-21 after the KDZ treatment. Furthermore, multicolor fluorescent staining, TUNEL staining, and microvessel density detection were conducted to analyze the effects of KDZ on MCAO rats, which included anti-inflammatory, neuroprotective, and apoptosis inhibition effects. Additionally, Western blotting was employed to determine the amounts of relevant inflammatory agents and proteins expressed. RESULTS KDZ injection improved the infarct area and neurological function scores in MCAO rats, aggravated the level of miR-21 expression, and regulated protein levels, including toll-like receptor 4, programmed death cell factor 4 (PDCD4), mitogen-activated protein kinase (MAPK) (p38, ERK), and nuclear factor kappa-B (NF-κB) as well. Besides, KDZ injection can regulate the amounts of inflammatory factors, angiopoietins, and brain water content and also increase the level of tight junction proteins. CONCLUSIONS KDZ injection has a neuroprotective effect by increasing the quantity of miR-21 of MCAO rats, preventing the PDCD4/MAPK/NF-κB signaling pathway from being activated, drastically lowering the level of pro-inflammatory proteins, and reversing the increase of apoptosis factors in the brain.
Collapse
Affiliation(s)
- Mengjie Sun
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, China.
| | - Guanghui Han
- Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, 100102, China.
| | - Hongni Yu
- Xiangya Hospital of Central South University, Changsha, Hunan 410008, China.
| | - Fengli Wang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, 100078, China.
| |
Collapse
|
2
|
Luo X, Niu JY, Chen HS. The potential value of traditional Chinese medicine monomers in cerebral ischemia-reperfusion injury: a network meta-analysis based on animal model. BMC Complement Med Ther 2025; 25:163. [PMID: 40325432 PMCID: PMC12051284 DOI: 10.1186/s12906-025-04899-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 04/25/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Cerebral ischemia-reperfusion injury (CIRI) is a complex pathological process, which can further aggravate the damage of ischemic tissues. Traditional Chinese medicine (TCM) monomers, bioactive compounds extracted from Chinese herbal medicines, have been demonstrated to have various protective effects against reperfusion injury. This network meta-analysis (NMA) aimed to investigate the optimal treatment strategy of TCM monomers for CIRI in animal models. METHODS Four databases including PubMed, Embase, Web of Science, and Cochrane were searched up to January 06, 2024. First, prospective registration was done at PROSPERO (ID: CRD42024496289), the quality of the included studies was evaluated with SYRCLE's risk of bias tool, and statistical analysis was conducted with Stata Version 18.0 and RStudio. RESULTS In total, 26 studies were included, involving 506 animals and 12 TCM monomers. The results of a meta-analysis demonstrated that, compared to the control group, puerarin, paeoniflorin, hydroxysafflor yellow A, sinomenine, and salvianolic acid significantly reduced mNSS scores. Furthermore, ginsenoside, scutellarin, and baicalein significantly reduced Longa scores. In addition, salvianolic acid treatment significantly decreased brain water content. Regarding infarct volume, bilobalide, baicalein and puerarin all demonstrated remarkable effects. The network meta-analysis suggested that paeoniflorin might be the most effective intervention in terms of mNSS score, with a surface under the cumulative ranking curve (SUCRA) value of 92.8%; Scutellarin might be the most effective intervention to reduce Longa score (SUCRA = 87.6%); And salvianolic acid might be the most effective intervention to reduce brain water content (SUCRA = 98.2%); For infarct volume specifically, bilobalide may be the most effective intervention (SUCRA = 95.5%). In our meta-regression, we found that dose and duration of treatment may contribute to heterogeneity among mNSS studies. CONCLUSION TCM monomers could provide a favorable neuroprotection on CIRI, with heterogeneous protective effects. Given the small number and the differences in quality of included studies, more high-quality, programmatic animal studies were needed to validate our findings. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Xin Luo
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, China
- Department of Neurology, General Hospital of Northern Theater Command, 83 Wen Hua Road, Shenyang, 110016, China
| | - Jing-Yuan Niu
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, China
- Department of Neurology, General Hospital of Northern Theater Command, 83 Wen Hua Road, Shenyang, 110016, China
| | - Hui-Sheng Chen
- Department of Neurology, General Hospital of Northern Theater Command, 83 Wen Hua Road, Shenyang, 110016, China.
| |
Collapse
|
3
|
Frolov A, Wadood A, Kelley BJ. Review of Supplements That Patients Commonly Report Using for Dementia. J Clin Med 2024; 13:7541. [PMID: 39768463 PMCID: PMC11727725 DOI: 10.3390/jcm13247541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/08/2024] [Accepted: 12/01/2024] [Indexed: 01/16/2025] Open
Abstract
Dietary supplements are readily available over the counter in the United States and are used by the majority of older adults to address a variety of concerns and conditions. Many older adults report using dietary supplements for cognitive health-either to address memory loss or dementia or in efforts to prevent cognitive decline. Our objective for this narrative review is to summarize the available efficacy and safety data for several supplements commonly reported in our clinic as being used for symptoms of dementia. Using a validated survey instrument, we conducted a survey of patients in our tertiary referral center memory clinic population to assess for the most commonly reported supplements for cognition. In our review, we compare the strength of published medical and scientific evidence to advertising or other lay press claims made about the nine most reported supplements with the aim of providing a representation of general trends in this industry. We found little or no scientific evidence available to support the use of any of these substances to ameliorate memory loss or other cognitive symptoms. Although most appear safe in the studies conducted to date, several authors have highlighted the lack of Food and Drug Administration oversight in the supplement industry, raising concerns over unknown or undeclared contaminants in these over-the-counter products. This review will better prepare clinicians to discuss these considerations with their patients who are considering the use of dietary supplements.
Collapse
Affiliation(s)
| | | | - Brendan J. Kelley
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (A.F.)
| |
Collapse
|
4
|
Ye J, Yin X, Xie S, Hua Q, Zhu J, Chen J, Zheng W, Cai L. Methacrylated hyaluronic acid/laponite photosensitive, sustained-release hydrogel loaded with bilobalide for enhancing random flap survival through mitigation of endoplasmic reticulum stress. Int J Biol Macromol 2024; 281:136277. [PMID: 39370062 DOI: 10.1016/j.ijbiomac.2024.136277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/04/2024] [Accepted: 10/02/2024] [Indexed: 10/08/2024]
Abstract
Random flaps are extensively utilized in plastic surgery due to their flexibility compared to traditional axial vascular system arrangements and their resemblance to injured skin in color, thickness, and texture. Despite these advantages, they are susceptible to ischemia-reperfusion injuries and subsequent necrosis post-transplantation. Bilobalide (BB), a sesquiterpene compound derived from Ginkgo biloba, exhibits notable antioxidant and anti-inflammatory properties and is commonly used to treat ischemiareperfusion injuries. However, its short half-life restricts its sustained efficacy in random flaps. In this study, we synthesized a multi-crosslinked, photosensitive methacryloyl hyaluronic acid(HAMA)/laponite(Lap)/bilobalide (BB) hydrogel. This dualcrosslinked hydrogel demonstrates superior mechanical properties and biocompatibility while providing a stable release of bilobalide. In vitro experiments showed that it significantly reduces edema, promotes angiogenesis, and enhances the survival of random flaps. Further network pharmacology analysis and recovery experiments suggested that the hydrogel's beneficial effects are mediated by the regulation of endoplasmic reticulum stress and specifically identified the regulation of the PERK/TXNIP/NLRP3 signaling pathway as crucial to its anti-inflammatory effects. Therefore, this HAMA/Lap/BB hydrogel promotes the survival of random flaps in rats by alleviating endoplasmic reticulum stress, providing a novel intervention strategy for the treatment of random flaps injuries.
Collapse
Affiliation(s)
- Jiangtian Ye
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China; The Second School of Medicine of Wenzhou Medical University, Wenzhou 325000, PR China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, PR China
| | - Xinghao Yin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China; The Second School of Medicine of Wenzhou Medical University, Wenzhou 325000, PR China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, PR China
| | - Shangjing Xie
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Qianqian Hua
- The First School of Medicine of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Jinrong Zhu
- The Second School of Medicine of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Jiawei Chen
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang Province, China
| | - Wenhao Zheng
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China; The Second School of Medicine of Wenzhou Medical University, Wenzhou 325000, PR China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, PR China
| | - Leyi Cai
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, PR China; The Second School of Medicine of Wenzhou Medical University, Wenzhou 325000, PR China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000, PR China.
| |
Collapse
|
5
|
Gulia S, Chandra P, Das A. Combating anoikis resistance: bioactive compounds transforming prostate cancer therapy. Anticancer Drugs 2024; 35:687-697. [PMID: 38743565 DOI: 10.1097/cad.0000000000001616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The study aims to discuss the challenges associated with treating prostate cancer (PCa), which is known for its complexity and drug resistance. It attempts to find differentially expressed genes (DEGs), such as those linked to anoikis resistance and circulating tumor cells, in PCa samples. This study involves analyzing the functional roles of these DEGs using gene enrichment analysis, and then screening of 102 bioactive compounds to identify a combination that can control the expression of the identified DEGs. In this study, 53 DEGs were identified from PCa samples including anoikis-resistant PCa cells and circulating tumor cells in PCa. Gene enrichment analysis with regards to functional enrichment of DEGs was performed. An inclusive screening process was carried out among 102 bioactive compounds to identify a combination capable of affecting and regulating the expression of selected DEGs. Eventually, gastrodin, nitidine chloride, chenodeoxycholic acid, and bilobalide were selected, as their combination demonstrated ability to modulate expression of 50 out of the 53 genes targeted. The subsequent analysis focused on investigating the biological pathways and processes influenced by this combination. The findings revealed a multifaceted and multidimensional approach to tumor regression. The combination of bioactive compounds exhibited effects on various genes including those related to production of inflammatory cytokines, cell proliferation, autophagy, apoptosis, angiogenesis, and metastasis. The current study has made a valuable contribution to the development of a combination of bioactive natural compounds that can significantly impede the development of treatment resistance in prostate tumor while countering the tumors' evasion of the immune system. The implications of this study are highly significant as it suggests the creation of an enhanced immunotherapeutic, natural therapeutic concoction with combinatorial potential.
Collapse
Affiliation(s)
- Shweta Gulia
- Department of Biotechnology, Delhi Technological University, Delhi, India
| | | | | |
Collapse
|
6
|
Ma J, Zhao Y, Cui Y, Lin H. Hypoxia Postconditioning Attenuates Hypoxia-Induced Inflammation and Endothelial Barrier Dysfunction. J Surg Res 2024; 301:413-422. [PMID: 39042975 DOI: 10.1016/j.jss.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 05/15/2024] [Accepted: 06/16/2024] [Indexed: 07/25/2024]
Abstract
INTRODUCTION In recent years, a number of studies have demonstrated that hypoxia reoxygenation (HR) induced by ischemia postconditioning (IPC) reduces endothelial barrier dysfunction and inflammation in various models. When HR occurs, the P38 mitogen-activated protein kinase (P38 MAPK) breaks down the endothelial barrier. But no study has clearly clarified the effect of hypoxia postconditioning (HPC) on P38 MAPK in human dermal microvascular endothelial cells. Therefore, we investigated the function of HPC on P38 MAPK during HR in vitro. METHODS Human dermal microvascular endothelial cells were cultured in a hypoxic incubator for 8 h. Then cells were reperfused for 12 h (reoxygenation) or postconditioned by 5 min of reoxygenation and 5 min of re-hypoxia 3 times followed by 11.5 h reoxygenation. SB203580 was used as an inhibitor of P38 MAPK. Cell counting kit-8 assay kits were employed to detect cell activity. The corresponding levels of IL-6, IL-8 and IL-1β were examined via Enzyme-Linked ImmunoSorbent Assay. The endothelial barrier was evaluated using fluorescein isothiocyanate-dextran leakage assay. Western blot was used to detect claudin-5, phosphorylation of P38 MAPK (P-P38 MAPK) and P38 MAPK expression. Claudin-5 localization was studied by immunofluorescence. RESULTS HR induced endothelial barrier hyperpermeability, elevated inflammation levels, and increased the P-P38 MAPK. But HPC reduced cell injury and maintained the integrity of the endothelial barrier while inhibiting P-P38 MAPK and increasing expression of claudin-5. HPC redistributed claudin-5 in a continuous and linear pattern on the cell membrane. CONCLUSIONS HPC protects against HR induced downregulation and redistribution of claudin-5 by inhibiting P-P38 MAPK.
Collapse
Affiliation(s)
- Jiaxing Ma
- Plastic and Reconstructive Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yinhua Zhao
- Plastic and Reconstructive Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yue Cui
- Plastic and Reconstructive Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Huang Lin
- Plastic and Reconstructive Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
7
|
Zhang K, Wang ZC, Sun H, Long H, Wang Y. Esculentoside H reduces the PANoptosis and protects the blood-brain barrier after cerebral ischemia/reperfusion through the TLE1/PI3K/AKT signaling pathway. Exp Neurol 2024; 379:114850. [PMID: 38857750 DOI: 10.1016/j.expneurol.2024.114850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/27/2024] [Accepted: 06/06/2024] [Indexed: 06/12/2024]
Abstract
AIMS Matrix metalloproteinases 9 (MMP9) plays a role in the destruction of blood-brain barrier (BBB) and cell death after cerebral ischemic/reperfusion (I/R). Esculentoside H (EH) is a saponin found in Phytolacca esculenta. It can block JNK1/2 and NF-κB signal mediated expression of MMP9. In this study, we determined whether EH can protect against cerebral I/R injury by inhibiting MMP9 and elucidated the underlying mechanism. MAIN METHODS Male SD rats were used to construct middle cerebral artery occlusion (MCAO) models. We determined the effect of EH on MMP9 inhibition, BBB destruction, neuronal death, PANoptosis, infarct volume, and the protective factor TLE1. Adeno-associated virus (AAV) infection was used to establish TLE1 gene overexpression and knockdown rats, which were used to determine the function. LY294002 was used to determine the role of PI3K/AKT signaling in TLE1 function. KEY FINDINGS After EH treatment, MMP9 expression, BBB destruction, neuronal death, and infarct volume decreased. We found that TLE1 expression decreased obviously after cerebral I/R. TLE1-overexpressing rats revealed distinct protective effects to cerebral I/R injury. After treatment with LY294002, the protective effect was inhibited. The curative effect of EH also decreased when TLE1 was knocked down. SIGNIFICANCE EH alleviates PANoptosis and protects BBB after cerebral I/R via the TLE1/PI3K/AKT signaling pathway. Our findings reveal a novel strategy and new target for treating cerebral I/R injury.
Collapse
Affiliation(s)
- Kuo Zhang
- Department of Urology, the Affiliated Lihuili Hospital, Ningbo University, Ningbo, China; Department of Urology, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo, China
| | - Zhi-Chao Wang
- Department of Urology, Ningbo Yinzhou No.2 Hospital, Ningbo, China
| | - Hongxue Sun
- Department of Neurology, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Huimin Long
- Department of Urology, the Affiliated Lihuili Hospital, Ningbo University, Ningbo, China; Department of Urology, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo, China.
| | - Yingju Wang
- Department of Emergency Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| |
Collapse
|
8
|
Liu Q, Wang J, Gu Z, Ouyang T, Gao H, Kan H, Yang Y. Comprehensive Exploration of the Neuroprotective Mechanisms of Ginkgo biloba Leaves in Treating Neurological Disorders. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:1053-1086. [PMID: 38904550 DOI: 10.1142/s0192415x24500435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Neurological disorders (NDs) are diseases that seriously affect the health of individuals worldwide, potentially leading to a significant reduction in the quality of life for patients and their families. Herbal medicines have been widely used in the treatment of NDs due to their multi-target and multi-pathway features. Ginkgo biloba leaves (GBLs), one of the most popular herbal medicines in the world, have been demonstrated to present therapeutic effects on NDs. However, the pharmacological mechanisms of GBLs in the treatment of neurological disorders have not been systematically summarized. This study aimed to summarize the molecular mechanism of GBLs in treating NDs from the cell models, animal models, and clinical trials of studies. Four databases, i.e., PubMed, Google Scholar, CNKI, and Web of Science were searched using the following keywords: "Ginkgo biloba", "Ginkgo biloba extract", "Ginkgo biloba leaves", "Ginkgo biloba leaves extract", "Neurological disorders", "Neurological diseases", and "Neurodegenerative diseases". All items meeting the inclusion criteria on the treatment of NDs with GBLs were extracted and summarized. Additionally, PRISMA 2020 was performed to independently evaluate the screening methods. Out of 1385 records in the database, 52 were screened in relation to the function of GBLs in the treatment of NDs; of these 52 records, 39 were preclinical trials and 13 were clinical studies. Analysis of pharmacological studies revealed that GBLs can improve memory, cognition, behavior, and psychopathology of NDs and that the most frequently associated GBLs are depression, followed by Alzheimer's disease, stroke, Huntington's disease, and Parkinson's disease. Additionally, the clinical studies of depression, AD, and stroke are the most common, and most of the remaining ND data are available from in vitro or in vivo animal studies. Moreover, the possible mechanisms of GBLs in treating NDs are mainly through free radical scavenging, anti-oxidant activity, anti-inflammatory response, mitochondrial protection, neurotransmitter regulation, and antagonism of PAF. This is the first paper to systematically and comprehensively investigate the pharmacological effects and neuroprotective mechanisms of GBLs in the treatment of NDs thus far. All findings contribute to a better understanding of the efficacy and complexity of GBLs in treating NDs, which is of great significance for the further clinical application of this herbal medicine.
Collapse
Affiliation(s)
- Qiwei Liu
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine Hefei, Anhui 230012, P. R. China
| | - Jinghui Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine Hefei, Anhui 230012, P. R. China
| | - Zongyun Gu
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine Hefei, Anhui 230012, P. R. China
| | - Ting Ouyang
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine Hefei, Anhui 230012, P. R. China
| | - Honglei Gao
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine Hefei, Anhui 230012, P. R. China
| | - Hongxing Kan
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine Hefei, Anhui 230012, P. R. China
- Anhui Computer Application Research Institute of Chinese Medicine, China Academy of Chinese Medical Sciences, Hefei, P. R. China
| | - Yinfeng Yang
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine Hefei, Anhui 230012, P. R. China
| |
Collapse
|
9
|
Zhu C, Wang D, Chang C, Liu A, Zhou J, Yang T, Jiang Y, Li X, Jiang W. Dexmedetomidine alleviates blood-brain barrier disruption in rats after cerebral ischemia-reperfusion by suppressing JNK and p38 MAPK signaling. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2024; 28:239-252. [PMID: 38682172 PMCID: PMC11058545 DOI: 10.4196/kjpp.2024.28.3.239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/17/2024] [Accepted: 01/18/2024] [Indexed: 05/01/2024]
Abstract
Dexmedetomidine displays multiple mechanisms of neuroprotection in ameliorating ischemic brain injury. In this study, we explored the beneficial effects of dexmedetomidine on blood-brain barrier (BBB) integrity and neuroinflammation in cerebral ischemia/reperfusion injury. Sprague-Dawley rats were subjected to middle cerebral artery occlusion (MCAO) for 1.5 h and reperfusion for 24 h to establish a rat model of cerebral ischemia/reperfusion injury. Dexmedetomidine (9 g/kg) was administered to rats 30 min after MCAO through intravenous injection, and SB203580 (a p38 MAPK inhibitor, 200 g/kg) was injected intraperitoneally 30 min before MCAO. Brain damages were evaluated by 2,3,5-triphenyltetrazolium chloride staining, hematoxylin-eosin staining, Nissl staining, and brain water content assessment. BBB permeability was examined by Evans blue staining. Expression levels of claudin-5, zonula occludens-1, occludin, and matrix metalloproteinase-9 (MMP-9) as well as M1/M2 phenotypes-associated markers were assessed using immunofluorescence, RT-qPCR, Western blotting, and gelatin zymography. Enzyme-linked immunosorbent assay was used to examine inflammatory cytokine levels. We found that dexmedetomidine or SB203580 attenuated infarct volume, brain edema, BBB permeability, and neuroinflammation, and promoted M2 microglial polarization after cerebral ischemia/reperfusion injury. Increased MMP-9 activity by ischemia/reperfusion injury was inhibited by dexmedetomidine or SB203580. Dexmedetomidine inhibited the activation of the ERK, JNK, and p38 MAPK pathways. Moreover, activation of JNK or p38 MAPK reversed the protective effects of dexmedetomidine against ischemic brain injury. Overall, dexmedetomidine ameliorated brain injury by alleviating BBB permeability and promoting M2 polarization in experimental cerebral ischemia/reperfusion injury model by inhibiting the activation of JNK and p38 MAPK pathways.
Collapse
Affiliation(s)
- Canmin Zhu
- Department of Neurology, The First Peopleʼs Hospital of Jiangxia District, Wuhan 430200, Hubei, China
| | - Dili Wang
- Department of Neurology, The First Peopleʼs Hospital of Jiangxia District, Wuhan 430200, Hubei, China
| | - Chang Chang
- Department of Neurology, The First Peopleʼs Hospital of Jiangxia District, Wuhan 430200, Hubei, China
| | - Aofei Liu
- Department of Medicine, Soochow University, Suzhou 215006, Jiangsu, China
- Department of Vascular Neurosurgery, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
| | - Ji Zhou
- Department of Medicine, Soochow University, Suzhou 215006, Jiangsu, China
- Department of Vascular Neurosurgery, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
| | - Ting Yang
- Department of Neurology, The First Peopleʼs Hospital of Jiangxia District, Wuhan 430200, Hubei, China
| | - Yuanfeng Jiang
- Department of Medicine, Soochow University, Suzhou 215006, Jiangsu, China
- Department of Vascular Neurosurgery, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
| | - Xia Li
- Department of Medicine, Soochow University, Suzhou 215006, Jiangsu, China
- Department of Vascular Neurosurgery, PLA Rocket Force Characteristic Medical Center, Beijing 100088, China
| | - Weijian Jiang
- Department of Medicine, Soochow University, Suzhou 215006, Jiangsu, China
| |
Collapse
|
10
|
Song W, Chen Z, Zhang M, Fu H, Wang X, Ma J, Zang X, Hu J, Ai F, Chen K. Bilobalide Prevents Apoptosis and Improves Cardiac Function in Myocardial Infarction. Mol Biotechnol 2024; 66:442-453. [PMID: 37199885 DOI: 10.1007/s12033-023-00753-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 04/11/2023] [Indexed: 05/19/2023]
Abstract
Myocardial infarction (MI) is an extremely severe cardiovascular disease, which ranks as the leading cause of sudden death worldwide. Studies have proved that cardiac injury following MI can cause cardiomyocyte apoptosis and myocardial fibrosis. Bilobalide (Bilo) from Ginkgo biloba leaves have been widely reported to possess excellent cardioprotective effects. However, concrete roles of Bilo in MI have not been investigated yet. We here designed both in vitro and in vivo experiments to explore the effects of Bilo on MI-induced cardiac injury and the underlying mechanisms of its action. We conducted in vitro experiments using oxygen-glucose deprivation (OGD)-treated H9c2 cells. Cell apoptosis in H9c2 cells was assessed by conducting flow cytometry assay and evaluating apoptosis-related proteins with western blotting. MI mouse model was established by performing left anterior descending artery (LAD) ligation. Cardiac function of MI mice was determined by assessing ejection fraction (EF), fractional shortening (FS), left ventricular end-systolic diameter (LVESD), and left ventricular end-diastolic diameter (LVEDD). Histological changes were analyzed, infarct size and myocardial fibrosis were measured by hematoxylin and eosin (H&E) and Masson staining in cardiac tissues from the mice. The apoptosis of cardiomyocytes in MI mice was assessed by TUNEL staining. Western blotting was applied to detect the effect of Bilo on c-Jun N-terminal kinase (JNK)/p38 mitogen-activated protein kinases (p38 MAPK) signaling both in vitro and in vivo. Bilo inhibited OGD-induced cell apoptosis and lactate dehydrogenase (LDH) release in H9c2 cells. The protein levels of p-JNK and p-p38 were significantly downregulated by Bilo treatment. SB20358 (inhibitor of p38) and SP600125 (inhibitor of JNK) suppressed OGD-induced cell apoptosis as Bilo did. In MI mouse model, Bilo improved the cardiac function and significantly reduced the infarct size and myocardial fibrosis. Bilo inhibited MI-induced cardiomyocytes apoptosis in mice. Bilo suppressed the protein levels of p-JNK and p-p38 in cardiac tissues from MI mice. Bilo alleviated OGD-induced cell apoptosis in H9c2 cells and suppressed MI-induced cardiomyocyte apoptosis and myocardial fibrosis in mice via the inactivation of JNK/p38 MAPK signaling pathways. Thus, Bilo may be an effective anti-MI agent.
Collapse
Affiliation(s)
- Weifeng Song
- Department of Cardiology, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou, 463599, China
| | - Zhen Chen
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, No. 26 Shengli Street, Jiang 'an District, Wuhan, 430014, China
| | - Meng Zhang
- Department of Ophthalmology, Shenzhen Baoan Women's and Children's Hospital, Shenzhen, 518102, China
| | - Haixia Fu
- Department of Cardiology, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou, 463599, China
| | - Xianqing Wang
- Department of Cardiology, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou, 463599, China
| | - Jifang Ma
- Department of Cardiology, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou, 463599, China
| | - Xiaobiao Zang
- Department of Cardiology, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou, 463599, China
| | - Juan Hu
- Department of Cardiology, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou, 463599, China
| | - Fen Ai
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, No. 26 Shengli Street, Jiang 'an District, Wuhan, 430014, China.
| | - Ke Chen
- Department of Cardiology, Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, No. 7 Weiwu Road, Jinshui District, Zhengzhou, 463599, China.
| |
Collapse
|
11
|
Mao S, Yao J, Zhang T, Zhang X, Tan W, Li C. Bilobalide attenuates lipopolysaccharide‑induced HepG2 cell injury by inhibiting TLR4‑NF‑κB signaling via the PI3K/Akt pathway. Exp Ther Med 2024; 27:24. [PMID: 38125341 PMCID: PMC10728898 DOI: 10.3892/etm.2023.12312] [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] [Received: 01/22/2021] [Accepted: 09/23/2021] [Indexed: 12/23/2023] Open
Abstract
Inflammation is involved in the pathological process underlying a number of liver diseases. Bilobalide (BB) is a natural compound from Ginkgo biloba leaves that was recently demonstrated to exert hepatoprotective effects by inhibiting oxidative stress in the liver cancer cell line HepG2. The anti-inflammatory activity of BB has been reported in recent studies. The major objective of the present study was to investigate whether BB could attenuate inflammation-associated cell damage. HepG2 cells were cultured with lipopolysaccharide (LPS) and BB, and cell damage was evaluated by measuring cell viability using MTT assay. The activity of the NF-κB signaling pathway was assessed by measuring the levels of IκBα, NF-κB p65, phosphorylated (p)-IκBα, p-p65, p65 DNA-binding activity and inflammatory cytokines IL-1β, IL-6 and TNF-α. A toll-like receptor (TLR)4 inhibitor (CLI-095) was used to detect the involvement of TLR4 in cell injury caused by LPS. In addition, the PI3K/Akt inhibitor LY294002 was applied to explore the involvement of the PI3K/Akt axis in mediating the effects of BB. The results demonstrated that LPS induced HepG2 cell injury. LPS also elevated the levels of p-IκBα, p-p65, p65 DNA-binding activity and inflammatory cytokines. However, CLI-095 significantly attenuated the LPS-induced cell damage and inhibited the activation of NF-κB signaling. BB also dose-dependently attenuated the LPS-induced cell damage, activation of NF-κB signaling and TLR4 overexpression. Furthermore, it was observed that LY294002 diminished the cytoprotective effects of BB on cell injury, TLR4 expression and NF-κB activation. These findings indicated that BB could attenuate LPS-induced inflammatory injury to HepG2 cells by regulating TLR4-NF-κB signaling.
Collapse
Affiliation(s)
- Shumei Mao
- Department of Pharmacology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Jinpeng Yao
- Department of Cardiology, Yantai Kaifaqu Hospital, Yantai, Shandong 264006, P.R. China
| | - Teng Zhang
- Department of Pharmacology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Xiang Zhang
- Department of Pharmacology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Wei Tan
- Department of Respiratory Medicine, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| | - Chengde Li
- Department of Clinical Pharmacy, Key Laboratory of Applied Pharmacology in Universities of Shandong, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
12
|
Gouda NA, Alshammari SO, Abourehab MAS, Alshammari QA, Elkamhawy A. Therapeutic potential of natural products in inflammation: underlying molecular mechanisms, clinical outcomes, technological advances, and future perspectives. Inflammopharmacology 2023; 31:2857-2883. [PMID: 37950803 DOI: 10.1007/s10787-023-01366-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 10/06/2023] [Indexed: 11/13/2023]
Abstract
Chronic inflammation is a common underlying factor in many major diseases, including heart disease, diabetes, cancer, and autoimmune disorders, and is responsible for up to 60% of all deaths worldwide. Metformin, statins, and corticosteroids, and NSAIDs (non-steroidal anti-inflammatory drugs) are often given as anti-inflammatory pharmaceuticals, however, often have even more debilitating side effects than the illness itself. The natural product-based therapy of inflammation-related diseases has no adverse effects and good beneficial results compared to substitute conventional anti-inflammatory medications. In this review article, we provide a concise overview of present pharmacological treatments, the pathophysiology of inflammation, and the signaling pathways that underlie it. In addition, we focus on the most promising natural products identified as potential anti-inflammatory therapeutic agents. Moreover, preclinical studies and clinical trials evaluating the efficacy of natural products as anti-inflammatory therapeutic agents and their pragmatic applications with promising outcomes are reviewed. In addition, the safety, side effects and technical barriers of natural products are discussed. Furthermore, we also summarized the latest technological advances in the discovery and scientific development of natural products-based medicine.
Collapse
Affiliation(s)
- Noha A Gouda
- College of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi, 10326, Republic of Korea
| | - Saud O Alshammari
- Department of Pharmacognosy and Alternative Medicine, Faculty of Pharmacy, Northern Border University, Rafha, 76321, Saudi Arabia
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, College of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | - Qamar A Alshammari
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Northern Border University, Rafha, 76321, Saudi Arabia
| | - Ahmed Elkamhawy
- College of Pharmacy, Dongguk University-Seoul, Goyang, Gyeonggi, 10326, Republic of Korea.
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| |
Collapse
|
13
|
Li S, Yang C, Wu Z, Chen Y, He X, Liu R, Ma W, Deng S, Li J, Liu Q, Wang Y, Zhang W. Suppressive effects of bilobalide on depression-like behaviors induced by chronic unpredictable mild stress in mice. Food Funct 2023; 14:8409-8419. [PMID: 37615035 DOI: 10.1039/d3fo02681g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Background: Depression is a psychiatric disorder with depressed mood and even suicide attempts as the main clinical symptoms, and its pathogenesis has not yet been fully elucidated. Brain derived neurotrophic factor (BDNF) plays an important role in the pathogenesis of depression. Purpose: The main aim of the present study was to evaluate the effectiveness and reveal the potential mechanisms of bilobalide (BB) intervention in alleviating depression-like behaviors by using chronic unpredictable mild stress (CUMS) mice via mediating the BDNF pathway. Methods: Behavioral assessments were carried out by using the sucrose preference test (SPT), tail suspension test (TST), and forced swimming test (FST). CUMS mice were randomly divided into 5 groups: CUMS + solvent, CUMS + BB low, CUMS + BB medium, CUMS + BB high and CUMS + fluoxetine. Total serum levels of tumor necrosis factor (TNF-α) and interleukin-6 (IL-6) were measured by ELISA. Expression of TNF-α, IL-6, AKT, GSK3β, β-catenin, Trk-B and BDNF in the mouse hippocampus was assessed by western blotting. Results: BB treatment reduced the levels of pro-inflammatory cytokines (IL-6 and TNF-α) and increased the protein expression of BDNF in the hippocampus region of the CUMS mice. Moreover, BB treatment enhanced the AKT/GSK3β/β-catenin signaling pathway which is downstream of the BDNF receptor Trk-B in the hippocampus of these mice. Conclusions: Overall, the experimental results indicated that BB reverses CUMS-induced depression-like behavior. BB exerts antidepressant-like effects by inhibiting neuroinflammation and enhancing the function of neurotrophic factors.
Collapse
Affiliation(s)
- Shengnan Li
- Engineering Research Center of Bio-Process of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230001, China.
| | - Chengying Yang
- Engineering Research Center of Bio-Process of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230001, China.
| | - Zeyu Wu
- Engineering Research Center of Bio-Process of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230001, China.
| | - Yuanli Chen
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei 230001, China
| | - Xiaoyu He
- Key Laboratory of Metabolism and Regulation for Major Diseases of Anhui Higher Education Institutes, Hefei University of Technology, Hefei 230001, China
| | - Rui Liu
- Engineering Research Center of Bio-Process of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230001, China.
| | - Wanru Ma
- Engineering Research Center of Bio-Process of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230001, China.
| | - Shaohuan Deng
- Engineering Research Center of Bio-Process of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230001, China.
| | - Jianwen Li
- Engineering Research Center of Bio-Process of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230001, China.
| | - Qingsong Liu
- Engineering Research Center of Bio-Process of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230001, China.
| | - Yunchun Wang
- Engineering Research Center of Bio-Process of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230001, China.
| | - Wencheng Zhang
- Engineering Research Center of Bio-Process of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei 230001, China.
| |
Collapse
|
14
|
Xie Q, Lu D, Yuan J, Ren M, Li Y, Wang J, Ma R, Wang J. l-borneol promotes neurovascular unit protection in the subacute phase of transient middle cerebral artery occlusion rats: p38-MAPK pathway activation, anti-inflammatory, and anti-apoptotic effect. Phytother Res 2023; 37:4166-4184. [PMID: 37310024 DOI: 10.1002/ptr.7878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 06/14/2023]
Abstract
Our previous study showed l-borneol reduced cerebral infarction in the acute stage after cerebral ischemia, but there is little about the study of subacute phase. We herein investigated the cerebral protective effects of l-borneol on neurovascular units (NVU) in the subacute phase after transient middle cerebral artery occlusion (t-MCAO). The t-MCAO model was prepared by the line embolus method. Zea Longa, mNss, HE, and TTC staining were used to evaluate the effect of l-borneol. We evaluated the mechanisms of l-borneol on inflammation, p38 MAPK pathway, and apoptosis, etc. through various technologies. l-borneol 0.2, 0.1, 0.05 g·kg-1 could significantly reduce cerebral infarction rate, alleviate the pathological injury, and inhibit inflammation reaction. l-borneol could also significantly increase brain blood supply, Nissl bodies, and the expression of GFAP. Additionally, l-borneol activated the p38 MAPK signaling pathway, inhibited cell apoptosis, and maintained BBB integrity. l-borneol had a neuroprotective effect, which was related to activating the p38 MAPK signaling pathway, inhibiting inflammatory response and apoptosis, and improving cerebral blood supply to protect BBB and stabilize and remodel NVU. The study will provide a reference for the use of l-borneol in the treatment of ischemic stroke in the subacute phase.
Collapse
Affiliation(s)
- Qian Xie
- State Key Laboratory of Southwestern Chinese Medicine Resource, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Foshan University, Foshan, China
| | - Danni Lu
- State Key Laboratory of Southwestern Chinese Medicine Resource, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianmei Yuan
- State Key Laboratory of Southwestern Chinese Medicine Resource, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mihong Ren
- State Key Laboratory of Southwestern Chinese Medicine Resource, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yong Li
- State Key Laboratory of Southwestern Chinese Medicine Resource, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiajun Wang
- State Key Laboratory of Southwestern Chinese Medicine Resource, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rong Ma
- State Key Laboratory of Southwestern Chinese Medicine Resource, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Foshan University, Foshan, China
- South China University of Technology, Guangzhou, China
| | - Jian Wang
- State Key Laboratory of Southwestern Chinese Medicine Resource, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
15
|
Moreira J, Machado M, Dias-Teixeira M, Ferraz R, Delerue-Matos C, Grosso C. The neuroprotective effect of traditional Chinese medicinal plants-A critical review. Acta Pharm Sin B 2023; 13:3208-3237. [PMID: 37655317 PMCID: PMC10465969 DOI: 10.1016/j.apsb.2023.06.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/23/2023] [Accepted: 04/03/2023] [Indexed: 09/02/2023] Open
Abstract
Neurodegenerative and neuropsychiatric diseases are increasingly affecting individuals' quality of life, thus increasing their cost to social and health systems. These diseases have overlapping mechanisms, such as oxidative stress, protein aggregation, neuroinflammation, neurotransmission impairment, mitochondrial dysfunction, and excitotoxicity. Currently, there is no cure for neurodegenerative diseases, and the available therapies have adverse effects and low efficacy. For neuropsychiatric disorders, such as depression, the current therapies are not adequate to one-third of the patients, the so-called treatment-resistant patients. So, searching for new treatments is fundamental. Medicinal plants appear as a strong alternative and complement towards new treatment protocols, as they have been used for health purposes for thousands of years. Thus, the main goal of this review is to revisit the neuroprotective potential of some of the most predominant medicinal plants (and one fungus) used in traditional Chinese medicine (TCM), focusing on their proven mechanisms of action and their chemical compositions, to give clues on how they can be useful against neurodegeneration progression.
Collapse
Affiliation(s)
- João Moreira
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Porto 4249-015, Portugal
| | - Mariana Machado
- Ciências Químicas e das Biomoléculas/CISA, Escola Superior de Saúde—Instituto Politécnico do Porto, Porto 4200-072, Portugal
| | - Mónica Dias-Teixeira
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Porto 4249-015, Portugal
- NICiTeS—Núcleo de Investigação em Ciências e Tecnologias da Saúde, Escola Superior de Saúde Ribeiro Sanches, Lisboa 1950-396, Portugal
| | - Ricardo Ferraz
- Ciências Químicas e das Biomoléculas/CISA, Escola Superior de Saúde—Instituto Politécnico do Porto, Porto 4200-072, Portugal
- REQUIMTE/LAQV, Departamento de Química e Bioquímica Faculdade de Ciências, Universidade do Porto, Porto 4169-007, Portugal
| | - Cristina Delerue-Matos
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Porto 4249-015, Portugal
| | - Clara Grosso
- REQUIMTE/LAQV, Instituto Superior de Engenharia do Porto, Instituto Politécnico do Porto, Porto 4249-015, Portugal
| |
Collapse
|
16
|
Malaník M, Čulenová M, Sychrová A, Skiba A, Skalicka-Woźniak K, Šmejkal K. Treating Epilepsy with Natural Products: Nonsense or Possibility? Pharmaceuticals (Basel) 2023; 16:1061. [PMID: 37630977 PMCID: PMC10459181 DOI: 10.3390/ph16081061] [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: 06/05/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
Epilepsy is a neurological disease characterized by recurrent seizures that can lead to uncontrollable muscle twitching, changes in sensitivity to sensory perceptions, and disorders of consciousness. Although modern medicine has effective antiepileptic drugs, the need for accessible and cost-effective medication is urgent, and products derived from plants could offer a solution. For this review, we have focused on natural compounds that have shown anticonvulsant activity in in vivo models of epilepsy at relevant doses. In some cases, the effects have been confirmed by clinical data. The results of our search are summarized in tables according to their molecular targets. We have critically evaluated the data we present, identified the most promising therapeutic candidates, and discussed these in the text. Their perspectives are supported by both pharmacokinetic properties and potential interactions. This review is intended to serve as a basis for future research into epilepsy and related disorders.
Collapse
Affiliation(s)
- Milan Malaník
- Department of Natural Drugs, Faculty of Pharmacy, Masaryk University, Palackého 1946/1, 61200 Brno, Czech Republic; (A.S.); (K.Š.)
| | - Marie Čulenová
- Department of Natural Drugs, Faculty of Pharmacy, Masaryk University, Palackého 1946/1, 61200 Brno, Czech Republic; (A.S.); (K.Š.)
| | - Alice Sychrová
- Department of Natural Drugs, Faculty of Pharmacy, Masaryk University, Palackého 1946/1, 61200 Brno, Czech Republic; (A.S.); (K.Š.)
| | - Adrianna Skiba
- Department of Natural Products Chemistry, Faculty of Pharmacy, Medical University of Lublin, 1 Chodzki Str., 20-093 Lublin, Poland; (A.S.); (K.S.-W.)
| | - Krystyna Skalicka-Woźniak
- Department of Natural Products Chemistry, Faculty of Pharmacy, Medical University of Lublin, 1 Chodzki Str., 20-093 Lublin, Poland; (A.S.); (K.S.-W.)
| | - Karel Šmejkal
- Department of Natural Drugs, Faculty of Pharmacy, Masaryk University, Palackého 1946/1, 61200 Brno, Czech Republic; (A.S.); (K.Š.)
| |
Collapse
|
17
|
Akanchise T, Angelova A. Ginkgo Biloba and Long COVID: In Vivo and In Vitro Models for the Evaluation of Nanotherapeutic Efficacy. Pharmaceutics 2023; 15:pharmaceutics15051562. [PMID: 37242804 DOI: 10.3390/pharmaceutics15051562] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 05/28/2023] Open
Abstract
Coronavirus infections are neuroinvasive and can provoke injury to the central nervous system (CNS) and long-term illness consequences. They may be associated with inflammatory processes due to cellular oxidative stress and an imbalanced antioxidant system. The ability of phytochemicals with antioxidant and anti-inflammatory activities, such as Ginkgo biloba, to alleviate neurological complications and brain tissue damage has attracted strong ongoing interest in the neurotherapeutic management of long COVID. Ginkgo biloba leaf extract (EGb) contains several bioactive ingredients, e.g., bilobalide, quercetin, ginkgolides A-C, kaempferol, isorhamnetin, and luteolin. They have various pharmacological and medicinal effects, including memory and cognitive improvement. Ginkgo biloba, through its anti-apoptotic, antioxidant, and anti-inflammatory activities, impacts cognitive function and other illness conditions like those in long COVID. While preclinical research on the antioxidant therapies for neuroprotection has shown promising results, clinical translation remains slow due to several challenges (e.g., low drug bioavailability, limited half-life, instability, restricted delivery to target tissues, and poor antioxidant capacity). This review emphasizes the advantages of nanotherapies using nanoparticle drug delivery approaches to overcome these challenges. Various experimental techniques shed light on the molecular mechanisms underlying the oxidative stress response in the nervous system and help comprehend the pathophysiology of the neurological sequelae of SARS-CoV-2 infection. To develop novel therapeutic agents and drug delivery systems, several methods for mimicking oxidative stress conditions have been used (e.g., lipid peroxidation products, mitochondrial respiratory chain inhibitors, and models of ischemic brain damage). We hypothesize the beneficial effects of EGb in the neurotherapeutic management of long-term COVID-19 symptoms, evaluated using either in vitro cellular or in vivo animal models of oxidative stress.
Collapse
Affiliation(s)
- Thelma Akanchise
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France
| | - Angelina Angelova
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, 91400 Orsay, France
| |
Collapse
|
18
|
Wei HP, Peng ZF, Shao KM, Zhang PH, Chen L, Hu JA, Chai H, Liu JM. cPKCγ Inhibits Caspase-9-Initiated Neuronal Apoptosis in an Ischemia Reperfusion Model In Vitro Through p38 MAPK-p90RSK-Bad Pathway. Neurochem Res 2023; 48:362-374. [PMID: 36152136 DOI: 10.1007/s11064-022-03747-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 08/03/2022] [Accepted: 08/30/2022] [Indexed: 02/08/2023]
Abstract
Strokes are one of the leading causes of death and disability in the world. Previously we have found that conventional protein kinase Cγ (cPKCγ) plays neuroprotective role in ischemic strokes. Further, we found that cPKCγ knockdown increased the level of cleaved (cl)-Caspase-3. However, the precise mechanisms underlying cPKCγ-mediated neuronal death remain unclear. To this end, a model incorporating 1 h oxygen-glucose deprivation/24 h reoxygenation (1 h OGD/24 h R) was established in cortical neurons. We found that cPKCγ knockdown remarkably increased neuronal death after OGD. We also found that cPKCγ knockdown increased the level of cl-Caspase-3 through the upstream initiators Capsases-9 (not Caspase-8/12) in OGD-treated neurons. Overexpression of cPKCγ could decrease neuronal death and cl-Caspase-3 and -9 levels. Moreover, cPKCγ knockdown further reduced the phosphorylation levels of p38 MAPK, p90RSK, and Bad. In addition, the protein levels of Bcl-2 and Bcl-xl were decreased after cPKCγ knockdown, whereas that of Bax was increased. In conclusion, our results suggest that cPKCγ partly alleviates ischemic injury through activating the p38 MAPK-p90RSK-Bad pathway and inhibiting Caspase-9 initiated apoptosis. This may have potential as a therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Hai-Ping Wei
- Department of Neurology, Lanzhou University Second Hospital, No. 82 Cuiyingmen Street, Chengguan District, Lanzhou, 730030, Gansu, China.
| | - Zhi-Feng Peng
- Department of Physiology, School of Medicine, Shanxi Datong University, Xingyun Street, Pingcheng District, Datong, 037009, Shanxi, China
| | - Kang-Mei Shao
- The Second Clinical Medical College, Lanzhou University Second Hospital, No. 82 Cuiyingmen Street, Chengguan District, Lanzhou, 730030, Gansu, China
| | - Pei-Hao Zhang
- The Second Clinical Medical College, Lanzhou University Second Hospital, No. 82 Cuiyingmen Street, Chengguan District, Lanzhou, 730030, Gansu, China
| | - Lei Chen
- The Second Clinical Medical College, Lanzhou University Second Hospital, No. 82 Cuiyingmen Street, Chengguan District, Lanzhou, 730030, Gansu, China
| | - Jin-An Hu
- The Second Clinical Medical College, Lanzhou University Second Hospital, No. 82 Cuiyingmen Street, Chengguan District, Lanzhou, 730030, Gansu, China
| | - Hui Chai
- The Second Clinical Medical College, Lanzhou University Second Hospital, No. 82 Cuiyingmen Street, Chengguan District, Lanzhou, 730030, Gansu, China
| | - Jin-Mei Liu
- The Second Clinical Medical College, Lanzhou University Second Hospital, No. 82 Cuiyingmen Street, Chengguan District, Lanzhou, 730030, Gansu, China
| |
Collapse
|
19
|
Liu A, Hu J, Yeh TS, Wang C, Tang J, Huang X, Chen B, Huangfu L, Yu W, Zhang L. Neuroprotective Strategies for Stroke by Natural Products: Advances and Perspectives. Curr Neuropharmacol 2023; 21:2283-2309. [PMID: 37458258 PMCID: PMC10556387 DOI: 10.2174/1570159x21666230717144752] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 09/09/2023] Open
Abstract
Cerebral ischemic stroke is a disease with high prevalence and incidence. Its management focuses on rapid reperfusion with intravenous thrombolysis and endovascular thrombectomy. Both therapeutic strategies reduce disability, but the therapy time window is short, and the risk of bleeding is high. Natural products (NPs) have played a key role in drug discovery, especially for cancer and infectious diseases. However, they have made little progress in clinical translation and pose challenges to the treatment of stroke. Recently, with the investigation of precise mechanisms in cerebral ischemic stroke and the technological development of NP-based drug discovery, NPs are addressing these challenges and opening up new opportunities in cerebral stroke. Thus, in this review, we first summarize the structure and function of diverse NPs, including flavonoids, phenols, terpenes, lactones, quinones, alkaloids, and glycosides. Then we propose the comprehensive neuroprotective mechanism of NPs in cerebral ischemic stroke, which involves complex cascade processes of oxidative stress, mitochondrial damage, apoptosis or ferroptosis-related cell death, inflammatory response, and disruption of the blood-brain barrier (BBB). Overall, we stress the neuroprotective effect of NPs and their mechanism on cerebral ischemic stroke for a better understanding of the advances and perspective in NPs application that may provide a rationale for the development of innovative therapeutic regimens in ischemic stroke.
Collapse
Affiliation(s)
- Aifen Liu
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Jingyan Hu
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Tzu-Shao Yeh
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong 226019, China
| | - Chengniu Wang
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Jilong Tang
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Xiaohong Huang
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Bin Chen
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Liexiang Huangfu
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Weili Yu
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
| | - Lei Zhang
- Institute of Interdisciplinary Integrative Medicine Research, School of Medicine, Nantong University, Nantong 226001, China
- Department of Pharmaceutical Botany, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
20
|
Obrenovich M, Singh SK, Li Y, Perry G, Siddiqui B, Haq W, Reddy VP. Natural Product Co-Metabolism and the Microbiota-Gut-Brain Axis in Age-Related Diseases. Life (Basel) 2022; 13:41. [PMID: 36675988 PMCID: PMC9865576 DOI: 10.3390/life13010041] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Complementary alternative medicine approaches are growing treatments of diseases to standard medicine practice. Many of these concepts are being adopted into standard practice and orthomolecular medicine. Age-related diseases, in particular neurodegenerative disorders, are particularly difficult to treat and a cure is likely a distant expectation for many of them. Shifting attention from pharmaceuticals to phytoceuticals and "bugs as drugs" represents a paradigm shift and novel approaches to intervention and management of age-related diseases and downstream effects of aging. Although they have their own unique pathologies, a growing body of evidence suggests Alzheimer's disease (AD) and vascular dementia (VaD) share common pathology and features. Moreover, normal metabolic processes contribute to detrimental aging and age-related diseases such as AD. Recognizing the role that the cerebral and cardiovascular pathways play in AD and age-related diseases represents a common denominator in their pathobiology. Understanding how prosaic foods and medications are co-metabolized with the gut microbiota (GMB) would advance personalized medicine and represents a paradigm shift in our view of human physiology and biochemistry. Extending that advance to include a new physiology for the advanced age-related diseases would provide new treatment targets for mild cognitive impairment, dementia, and neurodegeneration and may speed up medical advancements for these particularly devastating and debilitating diseases. Here, we explore selected foods and their derivatives and suggest new dementia treatment approaches for age-related diseases that focus on reexamining the role of the GMB.
Collapse
Affiliation(s)
- Mark Obrenovich
- Research Service, Department of Veteran's Affairs Medical Center, Cleveland, OH 44106, USA
- Department of Chemistry, Case Western Reserve University, Cleveland, OH 44106, USA
- The Gilgamesh Foundation for Medical Science and Research, Cleveland, OH 44116, USA
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
- Departments of Chemistry and Biological and Environmental Sciences, Cleveland State University, Cleveland, OH 44115, USA
| | - Sandeep Kumar Singh
- Indian Scientific Education and Technology (ISET) Foundation, Lucknow 226002, India
| | - Yi Li
- Department of Nutrition and Dietetics, Saint Louis University, Saint Louis, MO 63103, USA
| | - George Perry
- Department of Neuroscience Developmental and Regenerative Biology, University of Texas, San Antonio, TX 78249, USA
| | - Bushra Siddiqui
- School of Medicine, Northeast Ohio College of Medicine, Rootstown, OH 44272, USA
| | - Waqas Haq
- School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - V Prakash Reddy
- Department of Chemistry, Missouri University of Science and Technology, Rolla, MO 65409, USA
| |
Collapse
|
21
|
Xu B, Bai L, Chen L, Tong R, Feng Y, Shi J. Terpenoid natural products exert neuroprotection via the PI3K/Akt pathway. Front Pharmacol 2022; 13:1036506. [PMCID: PMC9606746 DOI: 10.3389/fphar.2022.1036506] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
PI3K/Akt, an essential signaling pathway widely present in cells, has been shown to be relevant to neurological disorders. As an important class of natural products, terpenoids exist in large numbers and have diverse backbones, so they have a great chance to be identified as neuroprotective agents. In this review, we described and summarized recent research for a range of terpenoid natural products associated with the PI3K/Akt pathway by classifying their basic chemical structures of the terpenes, identified by electronic searches on PubMed, Web of Science for research, and Google Scholar websites. Only articles published in English were included. Our discussion here concerned 16 natural terpenoids and their mechanisms of action, the associated diseases, and the methods of experimentation used. We also reviewed the discovery of their chemical structures and their derivatives, and some compounds have been concluded for their structure–activity relationships (SAR). As a result, terpenoids are excellent candidates for research as natural neuroprotective agents, and our content will provide a stepping stone for further research into these natural products. It may be possible for more terpenoids to serve as neuroprotective agents in the future.
Collapse
Affiliation(s)
- Bingyao Xu
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lan Bai
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- The State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Chen
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Rongsheng Tong
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- The State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Rongsheng Tong, ; Yibin Feng, ; Jianyou Shi,
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- *Correspondence: Rongsheng Tong, ; Yibin Feng, ; Jianyou Shi,
| | - Jianyou Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- The State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Rongsheng Tong, ; Yibin Feng, ; Jianyou Shi,
| |
Collapse
|
22
|
Hu Q, Luo K, Liu P, Mei Y. To discuss the mechanism of colchicine in the treatment of acute cerebral infarction based on network pharmacology. Medicine (Baltimore) 2022; 101:e30720. [PMID: 36197265 PMCID: PMC9509177 DOI: 10.1097/md.0000000000030720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
To explore the mechanism of action of colchicine in the treatment of acute cerebral infarction (ACI) based on network pharmacology. The Swiss Target Prediction Database and CTD database were used to predict the target information of colchicine. ACI-related targets were retrieved using the GeneCards database, and the target protein interaction network (PPI) and active ingredient-target network were obtained by combining Cytoscape 3.7.1 software and R language. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis and gene function analysis (GO) enrichment analysis were performed using R language to preliminarily explore the multiple pharmacological mechanisms of action of colchicine. There were 200 targets identified by network parameter analysis; 958 ACI targets were identified. Overlapping comparisons allowed the extraction of 143 overlapping targets, and the top 30 targets were screened according to the topological isomerization parameters. Component-target networks were constructed. A PPI of overlapping targets was established to identify key targets. In addition, Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis and GO functional enrichment analysis were performed to explore the multiple mechanisms of action of colchicine in the treatment of ACI. Colchicine treatment of ACI is characterized by multi-component, multi-target and multi-pathway, and can exert complex network regulation through the interaction between different targets, providing a new idea and new basis for further exploration of the mechanism of action of colchicine in the treatment of ACI.
Collapse
Affiliation(s)
- Qiaoxia Hu
- Department of Geriatrics, the Affiliated Hospital of Medical School of Ningbo University, Institute of Geriatrics of Ningbo University, Ningbo, China
- *Correspondence: Qiaoxia Hu, Department of Geriatrics, the Affiliated Hospital of Medical School of Ningbo University, Institute of Geriatrics of Ningbo University, 247 Renmin Road, Ningbo 315020, China (e-mail: )
| | - Kena Luo
- Department of Geriatrics, the Affiliated Hospital of Medical School of Ningbo University, Institute of Geriatrics of Ningbo University, Ningbo, China
| | - Puheng Liu
- Department of Geriatrics, the Affiliated Hospital of Medical School of Ningbo University, Institute of Geriatrics of Ningbo University, Ningbo, China
| | - Yifei Mei
- Department of Geriatrics, the Affiliated Hospital of Medical School of Ningbo University, Institute of Geriatrics of Ningbo University, Ningbo, China
| |
Collapse
|
23
|
Schepetkin IA, Chernysheva GA, Aliev OI, Kirpotina LN, Smol’yakova VI, Osipenko AN, Plotnikov MB, Kovrizhina AR, Khlebnikov AI, Plotnikov EV, Quinn MT. Neuroprotective Effects of the Lithium Salt of a Novel JNK Inhibitor in an Animal Model of Cerebral Ischemia–Reperfusion. Biomedicines 2022; 10:biomedicines10092119. [PMID: 36140222 PMCID: PMC9495587 DOI: 10.3390/biomedicines10092119] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/24/2022] [Accepted: 08/25/2022] [Indexed: 01/31/2023] Open
Abstract
The c-Jun N-terminal kinases (JNKs) regulate many physiological processes, including inflammatory responses, morphogenesis, cell proliferation, differentiation, survival, and cell death. Therefore, JNKs represent attractive targets for therapeutic intervention. In an effort to develop improved JNK inhibitors, we synthesized the lithium salt of 11H-indeno[1,2-b]quinoxaline-11-one oxime (IQ-1L) and evaluated its affinity for JNK and biological activity in vitro and in vivo. According to density functional theory (DFT) modeling, the Li+ ion stabilizes the six-membered ring with the 11H-indeno[1,2-b]quinoxaline-11-one (IQ-1) oximate better than Na+. Molecular docking showed that the Z isomer of the IQ-1 oximate should bind JNK1 and JNK3 better than (E)-IQ-1. Indeed, experimental analysis showed that IQ-1L exhibited higher JNK1-3 binding affinity in comparison with IQ-1S. IQ-1L also was a more effective inhibitor of lipopolysaccharide (LPS)-induced nuclear factor-κB/activating protein 1 (NF-κB/AP-1) transcriptional activity in THP-1Blue monocytes and was a potent inhibitor of proinflammatory cytokine production by MonoMac-6 monocytic cells. In addition, IQ-1L inhibited LPS-induced c-Jun phosphorylation in MonoMac-6 cells, directly confirming JNK inhibition. In a rat model of focal cerebral ischemia (FCI), intraperitoneal injections of 12 mg/kg IQ-1L led to significant neuroprotective effects, decreasing total neurological deficit scores by 28, 29, and 32% at 4, 24, and 48 h after FCI, respectively, and reducing infarct size by 52% at 48 h after FCI. The therapeutic efficacy of 12 mg/kg IQ-1L was comparable to that observed with 25 mg/kg of IQ-1S, indicating that complexation with Li+ improved efficacy of this compound. We conclude that IQ-1L is more effective than IQ-1S in treating cerebral ischemia injury and thus represents a promising anti-inflammatory compound.
Collapse
Affiliation(s)
- Igor A. Schepetkin
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA
| | - Galina A. Chernysheva
- Department of Pharmacology, Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk NRMC, 634028 Tomsk, Russia
| | - Oleg I. Aliev
- Department of Pharmacology, Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk NRMC, 634028 Tomsk, Russia
| | - Liliya N. Kirpotina
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA
| | - Vera I. Smol’yakova
- Department of Pharmacology, Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk NRMC, 634028 Tomsk, Russia
| | - Anton N. Osipenko
- Department of Pharmacology, Siberian State Medical University, 2 Moskovskiy tract, 634050 Tomsk, Russia
| | - Mark B. Plotnikov
- Department of Pharmacology, Goldberg Research Institute of Pharmacology and Regenerative Medicine, Tomsk NRMC, 634028 Tomsk, Russia
- Radiophysical Faculty, National Research Tomsk State University, 634050 Tomsk, Russia
| | | | | | - Evgenii V. Plotnikov
- Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Mark T. Quinn
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA
- Correspondence: ; Tel.: +1-406-994-4707; Fax: +1-406-994-4303
| |
Collapse
|
24
|
Maternal Inflammation Exaggerates Offspring Susceptibility to Cerebral Ischemia–Reperfusion Injury via the COX-2/PGD2/DP2 Pathway Activation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1571705. [PMID: 35437456 PMCID: PMC9013311 DOI: 10.1155/2022/1571705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 03/15/2022] [Accepted: 03/23/2022] [Indexed: 12/04/2022]
Abstract
The pathogenesis of cerebral ischemia–reperfusion (I/R) injury is complex and does not exhibit an effective strategy. Maternal inflammation represents one of the most important factors involved in the etiology of brain injury in newborns. We aimed to investigate the effect of maternal inflammation on offspring susceptibility to cerebral I/R injury and the mechanisms by which it exerts its effects. Pregnant SD rats were intraperitoneally injected with LPS (300 μg/kg/day) at gestational days 11, 14, and 18. Pups were subjected to MCAO/R on postnatal day 60. Primary neurons were obtained from postnatal day 0 SD rats and subjected to OGD/R. Neurological deficits, brain injury, neuronal viability, neuronal damage, and neuronal apoptosis were assessed. Oxidative stress and inflammation were evaluated, and the expression levels of COX-2/PGD2/DP pathway-related proteins and apoptotic proteins were detected. Maternal LPS exposure significantly increased the levels of oxidative stress and inflammation, significantly activated the COX-2/PGD2/DP2 pathway, and increased proapoptotic protein expression. However, maternal LPS exposure significantly decreased the antiapoptotic protein expression, which subsequently increased neurological deficits and cerebral I/R injury in offspring rats. The corresponding results were observed in primary neurons. Moreover, these effects of maternal LPS exposure were reversed by a COX-2 inhibitor and DP1 agonist but exacerbated by a DP2 agonist. In conclusion, maternal inflammatory exposure may increase offspring susceptibility to cerebral I/R injury. Moreover, the underlying mechanism might be related to the activation of the COX-2/PGD2/DP2 pathway. These findings provide a theoretical foundation for the development of therapeutic drugs for cerebral I/R injury.
Collapse
|
25
|
Ma T, Lv L, Yu Y, Jia L, Song X, Xu X, Li T, Sheng X, Wang H, Zhang J, Gao L. Bilobalide Exerts Anti-Inflammatory Effects on Chondrocytes Through the AMPK/SIRT1/mTOR Pathway to Attenuate ACLT-Induced Post-Traumatic Osteoarthritis in Rats. Front Pharmacol 2022; 13:783506. [PMID: 35281931 PMCID: PMC8905364 DOI: 10.3389/fphar.2022.783506] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 01/21/2022] [Indexed: 12/28/2022] Open
Abstract
Although osteoarthritis (OA) significantly affects the quality of life of the elderly, there is still no effective treatment strategy. The standardized Ginkgo biloba L. extract preparation has been shown to have a wide range of therapeutic effects. Bilobalide, a unique ingredient of Ginkgo biloba, has anti-inflammatory and antioxidant pharmacological properties, but its mechanism of action on OA remains unknown. In this study, we investigated the effects of bilobalide on the development of OA through in vivo and in vitro experiments, as well as its potential anti-inflammatory mechanisms. The in vitro experiments demonstrated that bilobalide significantly inhibited the production of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and matrix metalloproteinase 13 (MMP13) in ATDC5 chondrocytes induced by Interleukin-1β (IL-1β). At the molecular level, bilobalide induced chondrocyte autophagy by activating the AMPK/SIRT1/mTOR signaling pathway, which increased the expression of autophagy-related Atg genes, up-regulated the expression of LC3 protein, and reduced the expression of the p62 protein. In vivo, bilobalide exerted significant anti-inflammatory and anti-extracellular matrix (ECM) degradation effects in a rat model of post-traumatic OA (PTOA) induced by anterior cruciate ligament transection (ACLT). Bilobalide could relieve joint pain in PTOA rats, inhibit the expression of iNOS and COX-2 protein in cartilage via the AMPK/SIRT1/mTOR pathway, and reduce the level of ECM degradation biomarkers in serum. In conclusion, bilobalide exhibits vigorous anti-inflammatory activity, presenting it as an interesting potential therapeutic agent for OA.
Collapse
Affiliation(s)
- Tianwen Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory of Animals Disease Pathogenesis and Comparative Medicine, Harbin, China
| | - Liangyu Lv
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory of Animals Disease Pathogenesis and Comparative Medicine, Harbin, China
| | - Yue Yu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory of Animals Disease Pathogenesis and Comparative Medicine, Harbin, China
| | - Lina Jia
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory of Animals Disease Pathogenesis and Comparative Medicine, Harbin, China
| | - Xiaopeng Song
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory of Animals Disease Pathogenesis and Comparative Medicine, Harbin, China
| | - XinYu Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory of Animals Disease Pathogenesis and Comparative Medicine, Harbin, China
| | - Ting Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory of Animals Disease Pathogenesis and Comparative Medicine, Harbin, China
| | - Xuanbo Sheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory of Animals Disease Pathogenesis and Comparative Medicine, Harbin, China
| | - Haoran Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory of Animals Disease Pathogenesis and Comparative Medicine, Harbin, China
| | - Jiantao Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory of Animals Disease Pathogenesis and Comparative Medicine, Harbin, China
| | - Li Gao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.,Heilongjiang Key Laboratory of Animals Disease Pathogenesis and Comparative Medicine, Harbin, China
| |
Collapse
|
26
|
GSK-126 Protects CA1 Neurons from H3K27me3-Mediated Apoptosis in Cerebral Ischemia. Mol Neurobiol 2022; 59:2552-2562. [PMID: 35091962 PMCID: PMC9016005 DOI: 10.1007/s12035-021-02677-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 11/30/2021] [Indexed: 12/16/2022]
Abstract
Epigenetics, including histone modifications, play a significant role in central nervous system diseases, but the underlying mechanism remains to be elucidated. The aim of this study was to evaluate the role of H3K27me3 in regulating transcriptomic and pathogenic mechanisms following global ischemic stroke. Here, we found that in vivo ischemic/reperfusion (I/R) injury induced marked upregulation of H3K27me3 in the hippocampus. The administration of GSK-126 to rat brains decreased the levels of H3K27me3 in the hippocampus and reduced neuronal apoptosis after experimental stroke. Furthermore, ChIP-seq data demonstrated that the primary role of GSK-126 in the ischemic brain is to reduce H3K27me3 enrichment, mediating negative regulation of the execution phase of apoptosis and the MAPK signaling pathway. Further study suggested that the protective role of GSK-126 in ischemic rats was antagonized by U0126, an inhibitor of ERK1/2. Collectively, we demonstrated the potential of H3K27me3 as a novel stroke therapeutic target, and GSK-126 exerted a neuroprotective function in ischemic brain injury, which might be associated with activation of the MAPK/ERK pathway.
Collapse
|
27
|
Liu R, Song P, Gu X, Liang W, Sun W, Hua Q, Zhang Y, Qiu Z. Comprehensive Landscape of Immune Infiltration and Aberrant Pathway Activation in Ischemic Stroke. Front Immunol 2022; 12:766724. [PMID: 35140708 PMCID: PMC8818702 DOI: 10.3389/fimmu.2021.766724] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 12/16/2021] [Indexed: 11/24/2022] Open
Abstract
Ischemic stroke (IS) is a multifactorial disease caused by the interaction of multiple environmental and genetic risk factors, and it is the most common cause of disability. The immune microenvironment and inflammatory response participate in the whole process of IS occurrence and development. Therefore, the rational use of relevant markers or characteristic pathways in the immune microenvironment will become one of the important therapeutic strategies for the treatment of IS. We collected peripheral blood samples from 10 patients diagnosed with IS at the First Affiliated Hospital of Gannan Medical University and First Affiliated Hospital, Jinan" University, and from 10 normal people. The GSE16561 dataset was downloaded from the Gene Expression Omnibus (GEO) database. xCell, gene set enrichment analysis (GSEA), single-sample GSEA (ssGSEA) and immune-related gene analysis were used to evaluate the differences in the immune microenvironment and characteristic pathways between the IS and control groups of the two datasets. xCell analysis showed that the IS-24h group had significantly reduced central memory CD8+ T cell, effector memory CD8+ T cell, B cell and Th1 cell scores and significantly increased M1 macrophage and macrophage scores. GSEA showed that the IS-24h group had significantly increased inflammation-related pathway activity(myeloid leukocyte activation, positive regulation of tumor necrosis factor biosynthetic process, myeloid leukocyte migration and leukocyte chemotaxis), platelet-related pathway activity(platelet activation, signaling and aggregation; protein polymerization; platelet degranulation; cell-cell contact zone) and pathology-related pathway activity (ERBB signaling pathway, positive regulation of ERK1 and ERK2 cascade, vascular endothelial growth factor receptor signaling pathway, and regulation of MAP kinase activity). Immune-related signature analysis showed that the macrophage signature, antigen presentation-related signature, cytotoxicity-related signature, B cell-related signature and inflammation-related signature were significantly lower in the IS-24h group than in the control group. In this study, we found that there were significant differences in the immune microenvironment between the peripheral blood of IS patients and control patients, as shown by the IS group having significantly reduced CD8+ Tcm, CD8+ Tem, B cell and Th1 cell scores and significantly increased macrophage and M1 macrophage scores. Additionally, inflammation-related, pathological, and platelet-related pathway activities were significantly higher in the IS group than in the control group.
Collapse
Affiliation(s)
- Rongrong Liu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Department of Neurology, Ganzhou People’s Hospital, Ganzhou, China
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Clinical Neuroscience Institute, Jinan University, Guangzhou, China
| | - Pingping Song
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Clinical Neuroscience Institute, Jinan University, Guangzhou, China
| | - Xunhu Gu
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Weidong Liang
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Department of Anesthesiology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Wei Sun
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Clinical Neuroscience Institute, Jinan University, Guangzhou, China
| | - Qian Hua
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Yusheng Zhang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Clinical Neuroscience Institute, Jinan University, Guangzhou, China
- *Correspondence: Yusheng Zhang, ; Zhengang Qiu,
| | - Zhengang Qiu
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- *Correspondence: Yusheng Zhang, ; Zhengang Qiu,
| |
Collapse
|
28
|
Knockdown of circRNA-Memo1 Reduces Hypoxia/Reoxygenation Injury in Human Brain Endothelial Cells Through miRNA-17-5p/SOS1 Axis. Mol Neurobiol 2022; 59:2085-2097. [PMID: 35041140 DOI: 10.1007/s12035-022-02743-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 01/09/2022] [Indexed: 12/23/2022]
Abstract
Circ-Memo1 has been proved to be upregulated in ischemia-reperfusion induced acute injury of kidney tissues. However, the potential role of circ-Memo1 in cerebral hypoxia/reoxygenation (H/R) injury is still unclear.Blood samples were collected from 25 ischemic stroke patients and 25 healthy controls. To construct the H/R model, human brain microvascular endothelial cells (HBMVECs) were cultured under the hypoxic condition, followed by reoxygenation. Cell viability was analyzed by MTT assay. Flow cytometry was carried out to examine cell apoptosis. The level of malondialdehyde (MDA) and the activity of superoxide dismutase (SOD) were measured by MDA and SOD assay kits, respectively. The levels of TNF-α, IL-1β, and IL-6 were determined by enzyme-linked immunosorbent assay (ELISA). Dual-luciferase reporter gene detection was employed to verify the binding relationships between circ-Memo1, miR-17-5p, and SOS1.Circ-Memo1 and SOS1 expressions were increased, and miR-17-5p expression was reduced in ischemic stroke patients. Circ-Memo1 silencing promoted cell viability, inhibited the activation of ERK/NF-κB signaling pathway, reduced oxidative stress and inflammatory response, and inhibited cell apoptosis. Moreover, miR-17-5p functioned as the sponge of circ-Memo1, and SOS1 was identified as the target of miR-17-5p. The protective effect of circ-Memo1 knockdown on cell injury after H/R treatment was weakened by miR-17-5p inhibition.Knockdown of circ-Memo1 alleviated H/R injury of HBMVEC cells by regulating the miR-17-5p/SOS1 axis, indicating that circ-Memo1 might be a potential treatment target for cerebral H/R injury.
Collapse
|
29
|
Li R, Zhou Y, Zhang S, Li J, Zheng Y, Fan X. The natural (poly)phenols as modulators of microglia polarization via TLR4/NF-κB pathway exert anti-inflammatory activity in ischemic stroke. Eur J Pharmacol 2022; 914:174660. [PMID: 34863710 DOI: 10.1016/j.ejphar.2021.174660] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/04/2021] [Accepted: 11/29/2021] [Indexed: 12/24/2022]
Abstract
Increasing evidences suggest that inflammation plays a key role in the pathogenesis of stroke, a devastating disease second only to cardiac ischemia as a cause of death worldwide. Microglia are the first non-neuronal cells on the scene during the innate immune response to acute ischemic stroke. Microglia respond to acute brain injury by activating and developing classic M1-like (pro-inflammatory) or alternative M2-like (anti-inflammatory) phenotypes. M1 microglia produce pro-inflammatory cytokines to exacerbate neural death, astrocyte apoptosis, and blood brain barrier (BBB) disruption, while M2 microglia play the opposite role. NF-κB, a central regulator of the inflammatory response, was responsible for microglia M1 and M2 polarization. NF-κB p65 and p50 form a heterodimer to initiate a pro-inflammatory cytokine response, which enhances M1 activation and impair M2 response of microglia. TLR4, expressed on the surface of microglia, plays an important role in activating NF-κB, ultimately causing the M1 response of microglia. Therefore, modulation of microglial phenotypes via TLR4/NF-κB signaling pathway may be a promising therapeutic approach for ischemic stroke. Dietary (poly)phenols are present in various foods, which have shown promising protective effects on ischemic stroke. In vivo studies strongly suggest that many (poly)phenols have a pronounced impact on ischemic stroke, as demonstrated by lower neuroinflammation. Thus, this review focuses on the anti-inflammatory properties of dietary (poly)phenols and discusses their effects on the polarization of microglia through modulating TLR4/NF-κB signaling pathway in the ischemic stroke.
Collapse
Affiliation(s)
- Ruoqi Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yuan Zhou
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Shanshan Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jieying Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yingyi Zheng
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiang Fan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
30
|
Liu P, Yang X, Niu J, Hei C. Hyperglycemia aggravates ischemic brain damage via ERK1/2 activated cell autophagy and mitochondrial fission. Front Endocrinol (Lausanne) 2022; 13:928591. [PMID: 35992111 PMCID: PMC9388937 DOI: 10.3389/fendo.2022.928591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Hyperglycemia is one of the major risk factors for stroke and stroke recurrence, leading to aggravated neuronal damage after cerebral ischemia/reperfusion (I/R). ERK1/2 signaling pathway plays a vital role in cerebral ischemic injury. However, the role of the ERK1/2 pathway in hyperglycemia-aggravated ischemic brain damage is not clear. METHODS Streptozotocin (STZ; 50 mg/kg)-induced diabetes (blood glucose ≥12 mmol/L) or control groups in adult Sprague-Dawley rats were further subdivided into I/R (carotid artery/vein clamping), I/R + PD98059 (I/R plus ERK1/2 inhibitor), and Sham-operated groups (n = 10 each). Neurobehavioral status (Neurological behavior scores) and the volume of the cerebral infarction (TTC staining); brain mitochondrial potential (JCI ratio test) and cell apoptosis (TUNEL assay); RAS protein expression, phosphorylated/total ERK1/2 and Drp-1 (Dynamic-related protein 1) protein levels (Western blotting); mitochondrial fusion-related proteins mitofusin-1/2 (Mfn1/2), optic atrophy (OPA-1) and mitochondrial fission 1 (Fis1), and autophagy-associated proteins Beclin-1, LC3-I/II and P62 (Western blotting and immunohistochemistry) were analyzed. RESULTS The I/R + PD98059 group demonstrated better neurobehavior on the 1st (p < 0.05) and the 3rd day (p < 0.01) than the I/R group. Compared to the Sham group, cerebral ischemia/reperfusion brought about neuronal damage in the I/R group (p <0.01). However, treatment with PD98059 showed an improved situation with faster recovery of mitochondrial potential and less apoptosis of neuronal cells in the I/R + PD98059 group (p < 0.01). The I/R group had a higher-level expression of RAS and phosphorylated ERK1/2 and Drp-1 than the diabetes mellitus (DM) group (p < 0.01). The PD98059 treated group showed decreased expression of p-ERK1/2, p-Drp-1, Fis1, and Beclin-1, LC3-I/II and P62, but increased Mfn1/2 and OPA-1 than the I/R group (p < 0.01). CONCLUSION Hyperglycemia worsens cerebral ischemia/reperfusion-induced neuronal damage via ERK1/2 activated cell autophagy and mitochondrial fission.
Collapse
Affiliation(s)
- Ping Liu
- Department of Endocrinology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Xiao Yang
- Neuroscience Center, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Jianguo Niu
- Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, China
| | - Changchun Hei
- Ningxia Key Laboratory of Cerebrocranial Disease, Ningxia Medical University, Yinchuan, China
- Department of Human Anatomy, Histology and Embryology, Ningxia Medical University, Yinchuan, China
- *Correspondence: Changchun Hei,
| |
Collapse
|
31
|
Zhang X, Zhong W, Ma X, Zhang X, Chen H, Wang Z, Lou M. Ginkgolide With Intravenous Alteplase Thrombolysis in Acute Ischemic Stroke Improving Neurological Function: A Multicenter, Cluster-Randomized Trial (GIANT). Front Pharmacol 2021; 12:792136. [PMID: 34925044 PMCID: PMC8681856 DOI: 10.3389/fphar.2021.792136] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 11/04/2021] [Indexed: 12/25/2022] Open
Abstract
Background and Purpose: We aimed to investigate the effect of Ginkgolide® treatment on neurological function in patients receiving intravenous (IV) recombinant tissue plasminogen activator (rt-PA). Methods: This cluster randomized controlled trial included acute ischemic stroke patients in 24 centers randomized to intervention of intravenous Ginkgolide® or control group within the first 24 h after IV rt-PA therapy (IVT). Clinical outcome at 90 days was assessed with modified Rankin Scale (mRS) score and dichotomized into good outcome (0-2) and poor outcome (3-6). Hemorrhagic transformation represented the conversion of a bland infarction into an area of hemorrhage by computed tomography. Symptomatic intracerebral hemorrhage (sICH) was defined as cerebral hemorrhagic transformation in combination with clinical deterioration of National Institutes of Health Stroke Scale (NIHSS) score ≥4 points at 7-day or if the hemorrhage was likely to be the cause of the clinical deterioration. We performed logistic regression analysis and propensity score matching analysis to investigate the impact of Ginkgolide® treatment with IV rt-PA on good outcome, hemorrhagic transformation and sICH, respectively. Results: A total of 1113 patients were finally included and 513 (46.1%) were in the intervention group. Patients in the Ginkgolide® group were more likely to have good outcomes (78.6 vs. 66.7%, p < 0.01) and lower rate of sICH (0 vs. 2.72%, p < 0.01), compared with patients in the control group. The intra-cluster correlation coefficient (ICC) for good outcome at 90 days was 0.033. Binary logistic regression analysis revealed that treatment with Ginkgolide® was independently associated with 90-day mRS in patients with IV rt-PA therapy (OR 1.498; 95% CI 1.006-2.029, p = 0.009). After propensity score matching, conditional logistic regression showed intervention with Ginkgolide® was significantly associated with 90-day good outcome (OR 1.513; 95% CI 1.073-2.132, p = 0.018). No significant difference in hemorrhage transformation was seen between the 2 matched cohorts (OR 0.885; 95% CI 0.450-1.741, p = 0.724). Conclusion: Using Ginkgolide® within 24-hour after IV rt-PA is effective and safe and might be recommended in combination with rtPA therapy in acute ischemic stroke. Clinical Trial Registration: http://www.clinicaltrials.gov, identifier NCT03772847.
Collapse
Affiliation(s)
- Xuting Zhang
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Wansi Zhong
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Xiaodong Ma
- Department of Neurology, Haiyan People's Hospital, Jiaxing, China
| | - Xiaoling Zhang
- Department of Neurology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Hongfang Chen
- Department of Neurology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Zhimin Wang
- Department of Neurology, The First People's Hospital of Taizhou, Taizhou, China
| | - Min Lou
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| |
Collapse
|
32
|
Fan XX, Cao ZY, Liu MX, Liu WJ, Xu ZL, Tu PF, Wang ZZ, Cao L, Xiao W. Diterpene Ginkgolides Meglumine Injection inhibits apoptosis induced by optic nerve crush injury via modulating MAPKs signaling pathways in retinal ganglion cells. JOURNAL OF ETHNOPHARMACOLOGY 2021; 279:114371. [PMID: 34181957 DOI: 10.1016/j.jep.2021.114371] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/02/2021] [Accepted: 06/23/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Diterpene Ginkgolides Meglumine Injection (DGMI) is made of extracts from Ginkgo biloba L, including Ginkgolides A, B, and K and some other contents, and has been widely used as the treatment of cerebral ischemic stroke in clinic. It can be learned from the "Compendium of Materia Medica" that Ginkgo possesses the effect of "dispersing toxin". The ancient Chinese phrase "dispersing toxin" is now explained as elimination of inflammation and oxidative state in human body. And it led to the original ideas for today's anti-oxidation studies of Ginkgo in apoptosis induced by optic nerve crush injury. AIM OF THE STUDY To investigate the underlying molecular mechanism of the DGMI in retinal ganglion cells (RGCs) apoptosis. MATERIALS AND METHODS TUNEL staining was used to observe the anti-apoptotic effects of DGMI on the adult rat optic nerve injury (ONC) model, and flow cytometry and hoechst 33,342 staining were used to observe the anti-apoptotic effects of DGMI on the oxygen glucose deprivation (OGD) induced RGC-5 cells injury model. The regulation of apoptosis and MAPKs pathways were investigated with Immunohistochemistry and Western blotting. RESULTS This study demonstrated that DGMI is able to decrease the conduction time of F-VEP and ameliorate histological features induced by optic nerve crush injury in rats. Immunohistochemistry and TUNEL staining results indicated that DGMI can also inhibit cell apoptosis via modulating MAPKs signaling pathways. In addition, treatment with DGMI markedly improved the morphological structures and decreased the apoptotic index in RGC-5 cells. Mechanistically, DGMI could significantly inhibit cell apoptosis by inhibiting p38, JNK and Erk1/2 activation. CONCLUSION The study shows that DGMI and ginkgolides inhibit RGCs apoptosis by impeding the activation of MAPKs signaling pathways in vivo and in vitro. Therefore, the present study provided scientific evidence for the underlying mechanism of DGMI and ginkgolides on optic nerve crush injury.
Collapse
Affiliation(s)
- Xiao-Xue Fan
- Jiangsu Kanion Pharmaceutical Co.Ltd., Lianyungang, 222001, China; State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Lianyungang, 222001, China; Modern Chinese Medicine Innovation Cluster and Digital Pharmaceutical Technology Platform, Lianyungang, 222001, China
| | - Ze-Yu Cao
- Jiangsu Kanion Pharmaceutical Co.Ltd., Lianyungang, 222001, China; State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Lianyungang, 222001, China; Modern Chinese Medicine Innovation Cluster and Digital Pharmaceutical Technology Platform, Lianyungang, 222001, China
| | - Min-Xuan Liu
- Jiangsu Kanion Pharmaceutical Co.Ltd., Lianyungang, 222001, China; State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Lianyungang, 222001, China; Modern Chinese Medicine Innovation Cluster and Digital Pharmaceutical Technology Platform, Lianyungang, 222001, China
| | - Wen-Jun Liu
- Jiangsu Kanion Pharmaceutical Co.Ltd., Lianyungang, 222001, China; State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Lianyungang, 222001, China; Modern Chinese Medicine Innovation Cluster and Digital Pharmaceutical Technology Platform, Lianyungang, 222001, China
| | - Zhi-Liang Xu
- Jiangsu Kanion Pharmaceutical Co.Ltd., Lianyungang, 222001, China; State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Lianyungang, 222001, China; Modern Chinese Medicine Innovation Cluster and Digital Pharmaceutical Technology Platform, Lianyungang, 222001, China
| | - Peng-Fei Tu
- Jiangsu Kanion Pharmaceutical Co.Ltd., Lianyungang, 222001, China; Peking University, Beijing, 100871, China
| | - Zhen-Zhong Wang
- Jiangsu Kanion Pharmaceutical Co.Ltd., Lianyungang, 222001, China; State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Lianyungang, 222001, China; Modern Chinese Medicine Innovation Cluster and Digital Pharmaceutical Technology Platform, Lianyungang, 222001, China
| | - Liang Cao
- Jiangsu Kanion Pharmaceutical Co.Ltd., Lianyungang, 222001, China; State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Lianyungang, 222001, China; Modern Chinese Medicine Innovation Cluster and Digital Pharmaceutical Technology Platform, Lianyungang, 222001, China.
| | - Wei Xiao
- Jiangsu Kanion Pharmaceutical Co.Ltd., Lianyungang, 222001, China; State Key Laboratory of New-tech for Chinese Medicine Pharmaceutical Process, Lianyungang, 222001, China; Modern Chinese Medicine Innovation Cluster and Digital Pharmaceutical Technology Platform, Lianyungang, 222001, China.
| |
Collapse
|
33
|
Zhan L, Wang X, Zhang Y, Zhu G, Ding Y, Chen X, Jiang W, Wu S. Benazepril hydrochloride protects against doxorubicin cardiotoxicity by regulating the PI3K/Akt pathway. Exp Ther Med 2021; 22:1082. [PMID: 34447475 PMCID: PMC8355712 DOI: 10.3892/etm.2021.10516] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 07/08/2021] [Indexed: 02/05/2023] Open
Abstract
Doxorubicin (DOX) stimulates the generation of reactive oxygen species, thereby impairing mitochondrial functions. Angiotensin-converting enzyme inhibitors (ACEIs) have been identified to exhibit protective effects on cardiovascular diseases. The present study aimed to test the hypothesis that an ACEI benazepril hydrochloride (HCl) may protect against DOX-induced cardiotoxicity. The DOX injury model was established using rat embryonic cardiac myoblast cells (H9c2 cell line) treated with DOX in vitro. H9c2 cells were treated with benazepril-HCl, DOX or a mixture of DOX and benazepril-HCl to measure the activities of myocardial enzymes including lactate dehydrogenase (LDH), superoxide dismutase, catalase and glutathione peroxidase, in addition to the concentration of malondialdehyde in the culture medium. Cells without any treatment were used as a control. DOX treatment increased the levels of activity of myocardial enzymes in H9c2 cells compared with those in the untreated control cells. Additionally, co-treatment with benazepril-HCl significantly reduced the levels of apoptosis occurring due to DOX-mediated cellular damage. The mechanistic experiment revealed that pretreatment with benazepril-HCl counteracted the DOX-induced oxidative stress and suppressed the activation of apoptosis via the PI3K/Akt signaling pathway. By contrast, an Akt inhibitor (MK2206) inhibited the protective effects of benazepril-HCl against DOX-induced H9c2 cell injury, as revealed by increased LDH release in H9c2 cells. These results suggested that benazepril-HCl may potentially be administered as an adjuvant for DOX in long-term clinical use.
Collapse
Affiliation(s)
- Lan Zhan
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xiangxiu Wang
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yanjing Zhang
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Guonian Zhu
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yu Ding
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xuemei Chen
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Wei Jiang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Sisi Wu
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
34
|
Xu D, Kong T, Shao Z, Liu M, Zhang R, Zhang S, Kong Q, Chen J, Cheng B, Wang C. Orexin-A alleviates astrocytic apoptosis and inflammation via inhibiting OX1R-mediated NF-κB and MAPK signaling pathways in cerebral ischemia/reperfusion injury. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166230. [PMID: 34358627 DOI: 10.1016/j.bbadis.2021.166230] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/11/2021] [Accepted: 07/21/2021] [Indexed: 12/22/2022]
Abstract
Orexin-A (OXA) is a neuropeptide with neuroprotective effect by reducing cerebral ischemia/reperfusion injury (CIRI). Inflammation and apoptosis mediated by astrocyte activation are the key pathological mechanisms for CIRI. We thus attempted to confirm neuroprotective effects of OXA on astrocytic inflammation and apoptosis in CIRI and clarify the relative mechanisms. A middle cerebral artery occlusion and reperfusion (MCAO/R) rat model and U251 glioma cells model subjected to oxygen glucose deprivation and reperfusion (OGD/R) were established, with or without OXA treatment. Neurological deficit score was determined, and cerebral infarct volume was evaluated by 2,3,5-triphenyltetrazolium chloride (TTC) staining. Western Blot was used to detect the expressions of NF-κB p65, p-p65, p-ERK, p-p38, GFAP, OX1R, IL-1β, TNF-α, IL-6, iNOS, Bcl-2, Bax, CytC, cleaved caspase-9 and cleaved caspase-3 in vivo and in vitro. Pro-inflammatory cytokines in cell supernatant IL-1β, TNF-α and IL-6 were determined by ELISA. Hoechst 33342 staining was used to detect the apoptosis of astrocyte. Immunofluorescent staining was performed to assess the nuclear translocation of p65 and the expression of GFAP. The results showed that OXA significantly improved neurological deficit score and decreased the volume of infarct area in brain. OXA decreased inflammatory mediators, inhibited astrocyte activation and nuclear translocation of NF-κB and phosphorylation of NF-κB, MAPK/ERK and MAPK/p38. Besides, OXA suppressed apoptosis via upregulating the ratio of Bcl-2/Bax and downregulating cytochrome C, cleaved-caspase-9 and cleaved caspase-3. Overall, it was concluded that OXA exerts neuroprotective effect during CIRI through attenuating astrocytes apoptosis, astrocytes activation and pro-inflammatory cytokines production, by Inhibiting OX1R-mediated NF-κB, MAPK/ERK and MAPK/p38 signaling pathways. The progress in our study is helpful to elucidate the molecular mechanisms of OXA neuroprotection, which could lead to the development of new treatment strategies for ischemic stroke.
Collapse
Affiliation(s)
- Dandan Xu
- Neurobiology Key Laboratory of Jining Medical University, Jining 272067, China
| | | | - Ziqi Shao
- Neurobiology Key Laboratory of Jining Medical University, Jining 272067, China
| | - Minghui Liu
- Basic Medical Sciences, Jining Medical University, Jining 272067, China
| | - Rumin Zhang
- Neurobiology Key Laboratory of Jining Medical University, Jining 272067, China
| | - Shengnan Zhang
- Neurobiology Key Laboratory of Jining Medical University, Jining 272067, China
| | - Qingxia Kong
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining 272029, China
| | - Jing Chen
- Neurobiology Key Laboratory of Jining Medical University, Jining 272067, China; Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Baohua Cheng
- Neurobiology Key Laboratory of Jining Medical University, Jining 272067, China.
| | - Chunmei Wang
- Neurobiology Key Laboratory of Jining Medical University, Jining 272067, China.
| |
Collapse
|
35
|
Lu J, Xie L, Liu K, Zhang X, Wang X, Dai X, Liang Y, Cao Y, Li X. Bilobalide: A review of its pharmacology, pharmacokinetics, toxicity, and safety. Phytother Res 2021; 35:6114-6130. [PMID: 34342079 DOI: 10.1002/ptr.7220] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 06/25/2021] [Accepted: 07/03/2021] [Indexed: 12/18/2022]
Abstract
Bilobalide is a natural sesquiterpene trilactone from Ginkgo biloba leaves. It has good water solubility and is widely used in food and pharmaceutical fields. In the last decade, a plethora of studies on the pharmacological activities of bilobalide has been conducted and demonstrated that bilobalide possessed an extensive range of pharmacological activities such as neuroprotective, antioxidative, antiinflammatory, anti-ischemic, and cardiovascular protective activities. Pharmacokinetic studies indicated that bilobalide may have the characteristics of rapid absorption, good bioavailability, wide distribution, and slow elimination. This review aims to summarize the advances in pharmacological, pharmacokinetics, toxicity, and safety studies of bilobalide in the last decade with an emphasis on its neuroprotective and antiinflammatory activities, to provide researchers with the latest information and point out the limitations of relevant research at the current stage and the aspects that should be strengthened in future research.
Collapse
Affiliation(s)
- Jing Lu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Long Xie
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kai Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xuming Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xian Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaolin Dai
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Youdan Liang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi Cao
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofang Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
36
|
Zhu Y, Zhao H, Zhang W, Ma X, Liu Y. Dexmedetomidine attenuates neuronal injury induced by cerebral ischemia‑reperfusion by regulating miR‑199a. Mol Med Rep 2021; 24:574. [PMID: 34109426 PMCID: PMC8201450 DOI: 10.3892/mmr.2021.12213] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 04/21/2021] [Indexed: 12/30/2022] Open
Abstract
As is well known, dexmedetomidine (DEX) serves a neuroprotective role in cerebral ischemia‑reperfusion (CIR) injury, and microRNA (miR)‑199a has been re‑ported to be associated with IR injury. However, the association between DEX and miR‑199a in CIR injury remains unknown. Thus, the aim of the present study was to verify whether the neuroprotective effect of DEX on cerebral ischemia‑reperfusion rats is associated with miR‑199a. A rat model of CIR was established, and the modified neurological severity score (mNSS) was evaluated. The effect of DEX on the patholog‑ical structure of the cerebral cortex in CIR rats was observed by hematoxylin and eosin and Nissl staining. Reverse transcription‑quantitative PCR was used to analyze the expression levels of miR‑199a in brain tissue following intracerebroventricular injection of miR‑199a antagomir. The co‑expression of NeuN and microtubule‑associated proteins 1A/1B light chain 3B in the cerebral cortex was analyzed by immunofluorescence staining. Western blotting and immunohistochemistry were performed to analyze the expression of autophagy‑associated proteins in the brain tissue. DEX inhibited the expression of miR‑199a, decreased the mNSS and improved pathological damage to the cerebral cortex. DEX also inhibited autophagy and expression levels of associated proteins and decreased nerve cell injury. In conclusion, DEX inhibited expression of miR‑199a and improved neurocyte injury induced by CIR.
Collapse
Affiliation(s)
- Yulin Zhu
- Department of Anesthesiology, Yantaishan Hospital, Yantai, Shandong 264000, P.R. China
| | - Huatang Zhao
- Department of Anesthesiology, The Second Hospital of Liaocheng Affiliated to Shan-dong First Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Wenshan Zhang
- Department of Anesthesiology, Laixi People's Hospital, Qingdao, Shandong 266600, P.R. China
| | - Xingang Ma
- Department of Anesthesiology, Zibo Maternal and Child Health Hospital, Zibo, Shandong 255000, P.R. China
| | - Ye Liu
- Department of Anesthesiology, Zibo Maternal and Child Health Hospital, Zibo, Shandong 255000, P.R. China
| |
Collapse
|
37
|
Xie W, Wang X, Xiao T, Cao Y, Wu Y, Yang D, Zhang S. Protective Effects and Network Analysis of Ginsenoside Rb1 Against Cerebral Ischemia Injury: A Pharmacological Review. Front Pharmacol 2021; 12:604811. [PMID: 34276353 PMCID: PMC8283782 DOI: 10.3389/fphar.2021.604811] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 05/13/2021] [Indexed: 12/30/2022] Open
Abstract
Ischemic stroke is a leading cause of death and disability worldwide. Currently, only a limited number of drugs are available for treating ischemic stroke. Hence, studies aiming to explore and develop other potential strategies and agents for preventing and treating ischemic stroke are urgently needed. Ginseng Rb1 (GRb1), a saponin from natural active ingredients derived from traditional Chinese medicine (TCM), exerts neuroprotective effects on the central nervous system (CNS). We conducted this review to explore and summarize the protective effects and mechanisms of GRb1 on cerebral ischemic injury, providing a valuable reference and insights for developing new agents to treat ischemic stroke. Our summarized results indicate that GRb1 exerts significant neuroprotective effects on cerebral ischemic injury both in vivo and in vitro, and these network actions and underlying mechanisms are mediated by antioxidant, anti-inflammatory, and antiapoptotic activities and involve the inhibition of excitotoxicity and Ca2+ influx, preservation of blood–brain barrier (BBB) integrity, and maintenance of energy metabolism. These findings indicate the potential of GRb1 as a candidate drug for treating ischemic stroke. Further studies, in particular clinical trials, will be important to confirm its therapeutic value in a clinical setting.
Collapse
Affiliation(s)
- Weijie Xie
- Shanghai Mental Health Centre, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xinyue Wang
- Shanghai Mental Health Centre, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tianbao Xiao
- First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yibo Cao
- First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Yumei Wu
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Dongsheng Yang
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Song Zhang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
38
|
Targeting Common Signaling Pathways for the Treatment of Stroke and Alzheimer's: a Comprehensive Review. Neurotox Res 2021; 39:1589-1612. [PMID: 34169405 DOI: 10.1007/s12640-021-00381-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/11/2021] [Accepted: 05/24/2021] [Indexed: 12/30/2022]
Abstract
Neurodegenerative diseases such as stroke and Alzheimer's disease (AD) are two inter-related disorders that affect the neurons in the brain and central nervous system. Alzheimer's is a disease by undefined origin and causes. Stroke and its most common type, ischemic stroke (IS), occurs due to the blockade of cerebral blood vessels. As an important feature, both of disorders are associated with irreversible damages to the brain and nervous system. In this regard, finding common signaling pathways and the same molecular origin between these two diseases may be a promising way for their solution. On the basis of literature appraisal, the most common signaling cascades implicated in the pathogenesis of AD and stroke including notch, autophagy, inflammatory, and insulin signaling pathways were reviewed. Furthermore, current therapeutic strategies including natural and synthetic pharmaceuticals aiming modulation of respective signaling factors were scrutinized to ameliorate neural deficits in AD and stroke. Taken together, digging deeper in the common connections and signal targeting can be greatly helpful in understanding and unified treating of these disorders.
Collapse
|
39
|
Du J, Li W, Wang B. Long non-coding RNA TUG1 aggravates cerebral ischemia and reperfusion injury by sponging miR-493-3p/miR-410-3p. Open Med (Wars) 2021; 16:919-930. [PMID: 34222667 PMCID: PMC8231466 DOI: 10.1515/med-2021-0253] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 12/13/2022] Open
Abstract
Background Cerebral ischemia and reperfusion injury (CIRI) affects bodily function by causing irreversible damage to brain cells. The diverse pathophysiological course factors hinder the research work to go deeper. Long noncoding RNA taurine-upregulated gene 1 (TUG1) has been reported to be related to CIRI. This study explored the undefined regulatory pathway of TUG1 in CIRI. Methods Quantitative real-time polymerase chain reaction was applied to test the expression of TUG1, microRNA (miR)-493-3p and miR-410-3p. The viability and apoptosis of oxygen and glucose deprivation/reoxygen (OGD/R) model cells were evaluated by cell counting kit-8 and flow cytometry assay, respectively. The determination of inflammatory factors of interleukin-6, interleukin-1β and tumor necrosis factor-α was presented by enzyme-linked immunosorbent assay. The oxidative stress was performed by measuring the generation of malondialdehyde, reactive oxygen species and the activity of superoxide dismutase. Cytotoxicity was presented by measuring the generation of lactate dehydrogenase. Western blot assay was devoted to assessing the level of apoptosis-related factors (cleaved-caspase-3 and cleaved-caspase-9) and the protein level of c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (p38 MAPK) pathway-related factors in neuro-2a cells treated by OGD/R. Besides, online database starBase was applied to predict the potential binding sites of TUG1 to miR-493-3p and miR-410-3p, which was further confirmed by the dual-luciferase reporter system. Results The expression of TUG1 was upregulated, while miR-493-3p or miR-410-3p was downregulated in the serum of CIRI and OGD/R model cells. Meanwhile, knockdown of TUG1 eliminated the suppression in proliferation, the promotion in apoptosis, inflammation and oxidative stress, as well as the cytotoxicity in OGD/R model cells. Interestingly, the inhibition of miR-493-3p or miR-410-3p allayed the above effects. In addition, TUG1 harbored miR-493-3p or miR-410-3p and negatively regulated their expression. Finally, the TUG1 activated JNK and p38 MAPK pathways by sponging miR-493-3p/miR-410-3p. Conclusion TUG1 motivated the development of CIRI by sponging miR-493-3p/miR-410-3p to activate JNK and p38 pathways. The novel role of TUG1 in CIRI may contribute to the advancement of CIRI treatment.
Collapse
Affiliation(s)
- Jinlong Du
- Department of Critical Care Medicine, Jingzhou Central Hospital, The Second Clinical Medical College of Yangtze University, Jingzhou, Hubei, 434020, China
| | - Wenjing Li
- Department of Ultrasound, Jingzhou Central Hospital, The Second Clinical Medical College of Yangtze University, Jingzhou, Hubei, 434020, China
| | - Bing Wang
- Department of Critical Care Medicine, Jingzhou Central Hospital, The Second Clinical Medical College of Yangtze University, Jingzhou, Hubei, 434020, China
| |
Collapse
|
40
|
Xu H, Wang E, Chen F, Xiao J, Wang M. Neuroprotective Phytochemicals in Experimental Ischemic Stroke: Mechanisms and Potential Clinical Applications. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6687386. [PMID: 34007405 PMCID: PMC8102108 DOI: 10.1155/2021/6687386] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/10/2021] [Accepted: 03/29/2021] [Indexed: 02/06/2023]
Abstract
Ischemic stroke is a challenging disease with high mortality and disability rates, causing a great economic and social burden worldwide. During ischemic stroke, ionic imbalance and excitotoxicity, oxidative stress, and inflammation are developed in a relatively certain order, which then activate the cell death pathways directly or indirectly via the promotion of organelle dysfunction. Neuroprotection, a therapy that is aimed at inhibiting this damaging cascade, is therefore an important therapeutic strategy for ischemic stroke. Notably, phytochemicals showed great neuroprotective potential in preclinical research via various strategies including modulation of calcium levels and antiexcitotoxicity, antioxidation, anti-inflammation and BBB protection, mitochondrial protection and antiapoptosis, autophagy/mitophagy regulation, and regulation of neurotrophin release. In this review, we summarize the research works that report the neuroprotective activity of phytochemicals in the past 10 years and discuss the neuroprotective mechanisms and potential clinical applications of 148 phytochemicals that belong to the categories of flavonoids, stilbenoids, other phenols, terpenoids, and alkaloids. Among them, scutellarin, pinocembrin, puerarin, hydroxysafflor yellow A, salvianolic acids, rosmarinic acid, borneol, bilobalide, ginkgolides, ginsenoside Rd, and vinpocetine show great potential in clinical ischemic stroke treatment. This review will serve as a powerful reference for the screening of phytochemicals with potential clinical applications in ischemic stroke or the synthesis of new neuroprotective agents that take phytochemicals as leading compounds.
Collapse
Affiliation(s)
- Hui Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen 508060, China
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | | | - Feng Chen
- Institute for Advanced Study, Shenzhen University, Shenzhen 508060, China
| | - Jianbo Xiao
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China
| | - Mingfu Wang
- Institute for Advanced Study, Shenzhen University, Shenzhen 508060, China
- School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| |
Collapse
|
41
|
Bilobalide Enhances AMPK Activity to Improve Liver Injury and Metabolic Disorders in STZ-Induced Diabetes in Immature Rats via Regulating HMGB1/TLR4/NF- κB Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8835408. [PMID: 33959665 PMCID: PMC8075671 DOI: 10.1155/2021/8835408] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 02/27/2021] [Accepted: 03/25/2021] [Indexed: 12/19/2022]
Abstract
This study was aimed at examining the effect and underlying mechanisms of bilobalide (BB) on hepatic injury in streptozotocin- (STZ-) induced diabetes mellitus (DM) in immature rats. Immature rats (one day old) were randomly divided into five groups: group I, control nondiabetic rats; group II, STZ-induced, untreated diabetic rats; groups III/IV/V, STZ-induced and BB-treated diabetic rats, which were intraperitoneally injected with BB (2.5 mg/kg, 5 mg/kg, or 10 mg/kg) after 3 days followed by STZ treatment. We observed that BB improved the histopathological changes and maintained normal glucose metabolism, blood lipid, and liver function indicators, such as fasting blood glucose, obesity index, HbA1c, HOMA-IR, fast serum insulin, adiponectin, total cholesterol (TC), triglyceride (TG), high-density lipoprotein (HDL), low-density lipoprotein (LDL), aspartate transaminase (AST), and alanine transaminase (ALT) in STZ-induced DM in immature rats by a biochemical analyzer or ELISA. Meanwhile, Western blot analysis showed that in STZ-induced DM immature rats, BB decreased the expression of apoptosis-related proteins Bax, cleaved caspase-3, and cleaved caspase-9 while enhancing the Bcl-2 expression; BB downregulated the expression of ACC related to fat anabolism, while upregulating the expression of CPT-1 related to fat catabolism. Strikingly, treatment with BB significantly increased the expression of AMPKα1 as well as inhibited HMGB1, TLR4, and p-P65 expression in hepatic tissues of immature DM rats. AMPK inhibitor (compound C, CC) cotreated with BB undermined the protective effect of BB on the liver injury. The results of the present study suggested BB may have a significant role in alleviating liver damage in the STZ-induced immature DM rats.
Collapse
|
42
|
Du H, He Y, Pan Y, Zhao M, Li Z, Wang Y, Yang J, Wan H. Danhong Injection Attenuates Cerebral Ischemia-Reperfusion Injury in Rats Through the Suppression of the Neuroinflammation. Front Pharmacol 2021; 12:561237. [PMID: 33927611 PMCID: PMC8076794 DOI: 10.3389/fphar.2021.561237] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 03/12/2021] [Indexed: 11/17/2022] Open
Abstract
Neuroinflammation is one of the major causes of damage of the central nervous system (CNS) and plays a vital role in the pathogenesis of cerebral ischemia, which can result in long-term disability and neuronal death. Danhong injection (DHI), a traditional Chinese medicine injection, has been applied to the clinical treatment of cerebral stoke for many years. In this study, we investigated the protective effects of DHI on cerebral ischemia-reperfusion injury (CIRI) in rats and explored its potential anti-neuroinflammatory properties. CIRI in adult male SD rats was induced by middle cerebral artery occlusion (MCAO) for 1 h and reperfusion for 24 h. Results showed that DHI (0.5, 1, and 2 ml/kg) dose-dependently improved the neurological deficits and alleviated cerebral infarct volume and histopathological damage of the cerebral cortex caused by CIRI. Moreover, DHI (0.5, 1, and 2 ml/kg) inhibited the mRNA expressions of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), intercellular cell adhesion molecule-1 (ICAM-1), cyclooxygenase-2 (COX-2), and inducible nitric oxide synthase (iNOS) in ischemic brains, downregulated TNF-α, IL-1β, and monocyte chemotactic protein-1 (MCP-1) levels in serum, and reduced the neutrophil infiltration (myeloperoxidase, MPO) in ischemic brains, in a dose-dependent manner. Immunohistochemical staining results also revealed that DHI dose-dependently diminished the protein expressions of ICAM-1 and COX-2, and suppressed the activation of microglia (ionized calcium-binding adapter molecule 1, Iba-1) and astrocyte (glial fibrillary acidic protein, GFAP) in the cerebral cortex. Western blot analysis showed that DHI significantly downregulated the phosphorylation levels of the proteins in nuclear factor κB (NF-κB) and mitogen-activated protein kinas (MAPK) signaling pathways in ischemic brains. These results indicate that DHI exerts anti-neuroinflammatory effects against CIRI, which contribute to the amelioration of CNS damage.
Collapse
Affiliation(s)
- Haixia Du
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu He
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuanjiang Pan
- Department of Chemistry, Zhejiang University, Hangzhou, China
| | - Mengdi Zhao
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhiwei Li
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu Wang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiehong Yang
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Haitong Wan
- College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China.,College of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
43
|
Protective Effect of Triphala against Oxidative Stress-Induced Neurotoxicity. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6674988. [PMID: 33898626 PMCID: PMC8052154 DOI: 10.1155/2021/6674988] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 03/04/2021] [Accepted: 03/27/2021] [Indexed: 11/17/2022]
Abstract
Background Oxidative stress is implicated in the progression of many neurological diseases, which could be induced by various chemicals, such as hydrogen peroxide (H2O2) and acrylamide. Triphala is a well-recognized Ayurvedic medicine that possesses different therapeutic properties (e.g., antihistamine, antioxidant, anticancer, anti-inflammatory, antibacterial, and anticariogenic effects). However, little information is available regarding the neuroprotective effect of Triphala on oxidative stress. Materials and Methods An in vitro H2O2-induced SH-SY5Y cell model and an in vivo acrylamide-induced zebrafish model were established. Cell viability, apoptosis, and proliferation were examined by MTT assay, ELISA, and flow cytometric analysis, respectively. The molecular mechanism underlying the antioxidant activity of Triphala against H2O2 was investigated dose dependently by Western blotting. The in vivo neuroprotective effect of Triphala on acrylamide-induced oxidative injury in Danio rerio was determined using immunofluorescence staining. Results The results indicated that Triphala plays a neuroprotective role against H2O2 toxicity in inhibiting cell apoptosis and promoting cell proliferation. Furthermore, Triphala pretreatment suppressed the phosphorylation of the mitogen-activated protein kinase (MARK) signal pathway (p-Erk1/2, p-JNK1/2, and p-p38), whereas it restored the activities of antioxidant enzymes (superoxide dismutase 1 (SOD1) and catalase) in the H2O2-treated SH-SY5Y cells. Consistently, similar protective effects of Triphala were observed in declining neuroapoptosis and scavenging free radicals in the zebrafish central neural system, possessing a critical neuroprotective property against acrylamide-induced oxidative stress. Conclusion In summary, Triphala is a promising neuroprotective agent against oxidative stress in SH-SY5Y cells and zebrafishes with significant antiapoptosis and antioxidant activities.
Collapse
|
44
|
Xu D, Kong T, Cheng B, Zhang R, Yang C, Chen J, Wang C. Orexin-A alleviates cerebral ischemia-reperfusion injury by inhibiting endoplasmic reticulum stress-mediated apoptosis. Mol Med Rep 2021; 23:266. [PMID: 33576468 PMCID: PMC7893697 DOI: 10.3892/mmr.2021.11905] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 01/08/2021] [Indexed: 12/12/2022] Open
Abstract
Orexin‑A (OXA) protects neurons against cerebral ischemia‑reperfusion injury (CIRI). Endoplasmic reticulum stress (ERS) induces apoptosis after CIRI by activating caspase‑12 and the CHOP pathway. The present study aimed to determine whether OXA mitigates CIRI by inhibiting ERS‑induced neuronal apoptosis. A model of CIRI was established, in which rats were subjected to middle cerebral artery occlusion with ischemic intervention for 2 h, followed by reperfusion for 24 h. Neurological deficit examination and 2,3,5‑triphenyltetrazolium chloride staining were performed to assess the level of CIRI and neuroprotection by OXA. Expression levels of ERS‑related proteins and cleaved caspase‑3 were measured via western blotting, while the rate of neuronal apoptosis in the cortex was determined using a TUNEL assay. OXA treatment decreased the infarct volume of rats after CIRI and attenuated neuron apoptosis. Furthermore, administration of OXA decreased the expression levels of GRP78, phosphorylated (p)‑PERK, p‑eukaryotic initiation factor‑2α, p‑inositol requiring enzyme 1α, p‑JNK, cleaved caspase‑12, CHOP and cleaved caspase‑3, all of which were induced by CIRI. Collectively, these findings suggested that OXA attenuated CIRI by inhibiting ERS‑mediated apoptosis, thus clarifying the mechanism underlying its neuroprotective effect and providing a novel therapeutic direction for the treatment of CIRI.
Collapse
Affiliation(s)
- Dandan Xu
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Tingting Kong
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Baohua Cheng
- Neurobiology Key Laboratory of Jining Medical University in Colleges of Shandong, Jining, Shandong 272067, P.R. China
| | - Rumin Zhang
- Neurobiology Key Laboratory of Jining Medical University in Colleges of Shandong, Jining, Shandong 272067, P.R. China
| | - Chunqing Yang
- Neurobiology Key Laboratory of Jining Medical University in Colleges of Shandong, Jining, Shandong 272067, P.R. China
| | - Jing Chen
- Neurobiology Key Laboratory of Jining Medical University in Colleges of Shandong, Jining, Shandong 272067, P.R. China
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Chunmei Wang
- Neurobiology Key Laboratory of Jining Medical University in Colleges of Shandong, Jining, Shandong 272067, P.R. China
| |
Collapse
|
45
|
Ko IG, Jin JJ, Hwang L, Kim SH, Kim CJ, Jeon JW, Chung JY, Han JH. Adenosine A2A receptor agonist polydeoxyribonucleotide ameliorates short-term memory impairment by suppressing cerebral ischemia-induced inflammation via MAPK pathway. PLoS One 2021; 16:e0248689. [PMID: 33735236 PMCID: PMC7971468 DOI: 10.1371/journal.pone.0248689] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 03/03/2021] [Indexed: 12/12/2022] Open
Abstract
Cerebral ischemia causes tissue death owing to occlusion of the cerebral blood vessels, and cerebral ischemia activates mitogen-activated protein kinase (MAPK) and induces secretion of pro-inflammatory cytokines. Adenosine A2A receptor agonist, polydeoxyribonucleotide (PDRN), suppresses the secretion of pro-inflammatory cytokines and exhibits anti-inflammatory effect. In the current study, the therapeutic effect of PDRN on cerebral ischemia was evaluated using gerbils. For the induction of cerebral ischemia, the common carotid arteries were exposed, and then aneurysm clips were used to occlude the common carotid arteries bilaterally for 7 minutes. In the PDRN-treated groups, the gerbils were injected intraperitoneally with 0.3 mL of saline containing 8 mg/kg PDRN, per a day for 7 days following cerebral ischemia induction. In order to confirm the participation of the adenosine A2A receptor in the effects mediated by PDRN, 8 mg/kg 7-dimethyl-1-propargylxanthine (DMPX), adenosine A2A receptor antagonist, was treated with PDRN. In the current study, induction of ischemia enhanced the levels of pro-inflammatory cytokines and increased phosphorylation of MAPK signaling factors in the hippocampus and basolateral amygdala. However, treatment with PDRN ameliorated short-term memory impairment by suppressing the production of pro-inflammatory cytokines and inactivation of MAPK signaling factors in cerebral ischemia. Furthermore, PDRN treatment enhanced the concentration of cyclic adenosine-3,5'-monophosphate (cAMP) as well as phosphorylation of cAMP response element-binding protein (p-CREB). Co-treatment of DMPX and PDRN attenuated the therapeutic effect of PDRN on cerebral ischemia. Based on these findings, PDRN may be developed as the primary treatment in cerebral ischemia.
Collapse
Affiliation(s)
- Il-Gyu Ko
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Jun-Jang Jin
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Lakkyong Hwang
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Sang-Hoon Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Chang-Ju Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Jung Won Jeon
- Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Jun-Young Chung
- Department of Anesthesiology and Pain Medicine, Kyung Hee University Hospital at Gangdong, College of Medicine, Kyung Hee University, Seoul, Korea
| | - Jin Hee Han
- Department of Anesthesiology and Pain Medicine, Kyung Hee Medical Center, College of Medicine, Kyung Hee University, Seoul, Korea
- * E-mail:
| |
Collapse
|
46
|
Li C, Liu S, Aerqin Q, Shen D, Wu X, Liu K. The therapeutic effects of ginkgolides in Guillain-Barré syndrome and experimental autoimmune neuritis. J Clin Neurosci 2021; 87:44-49. [PMID: 33863532 DOI: 10.1016/j.jocn.2021.02.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 12/14/2020] [Accepted: 02/15/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Guillain-Barré syndrome (GBS) is an acquired immune-mediated inflammatory peripheral neuropathy. The immune regulation of ginkgolides have been revealed in recent years. We herein investigate the potential therapeutic effects of ginkgolides both on GBS and its animal model, experimental autoimmune neuritis (EAN). METHODS EAN in C57BL/6 mice induced by subcutaneous injection with peripheral nerve myelin P0 protein peptide 180-199 (P0 peptide) were treated with ginkgolides at three different doses. GBS patients were randomly divided into two groups, the experimental group and the control group. The experimental group were treated with ginkgolides as soon as diagnosed. RESULTS Our data indicated that ginkgolides administration daily ameliorated the score of EAN and delayed the peak of disease in EAN mice. Ginkgolides also down-regulated the proportions of T helper (Th) 17 cells in EAN spleens. Furthermore, we also found that administration of ginkgolides significantly decreased the levels of interferon (IFN)-γ and interleukin-12 (IL)-12 in GBS patients. CONCLUSIONS Our results suggested that ginkgolides ameliorated the clinical score of EAN through down-regulating the proportions of Th 17 cells. Ginkgolides also suppressed inflammation response by decreasing pro-inflammatory cytokines IFN-γ and IL-12, suggesting ginkgolides had potential therapeutic effects on GBS patients and EAN in the future.
Collapse
Affiliation(s)
- Chunrong Li
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Shan Liu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Qiaolifan Aerqin
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Donghui Shen
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Xiujuan Wu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China
| | - Kangding Liu
- Neuroscience Center, Department of Neurology, The First Hospital of Jilin University, Jilin University, Changchun, China.
| |
Collapse
|
47
|
Saad MAEL, Fahmy MIM, Al-Shorbagy M, Assaf N, Hegazy AAEA, El-Yamany MF. Nateglinide Exerts Neuroprotective Effects via Downregulation of HIF-1α/TIM-3 Inflammatory Pathway and Promotion of Caveolin-1 Expression in the Rat's Hippocampus Subjected to Focal Cerebral Ischemia/Reperfusion Injury. Inflammation 2021; 43:401-416. [PMID: 31863220 DOI: 10.1007/s10753-019-01154-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ischemic stroke is a major cause of death and motor disabilities all over the world. It is a muti-factorial disorder associated with inflammatory, apoptotic, and oxidative responses. Nateglinide (NAT), an insulinotropic agent used for the treatment of type 2 diabetes mellitus, recently showed potential anti-inflammatory and anti-apoptotic effects. The aim of our study was to elucidate the unique neuroprotective role of NAT in the middle cerebral artery occlusion (MCAO)-induced stroke in rats. Fifty-six male rats were divided to 4 groups (n = 14 in each group): the sham-operated group, sham receiving NAT (50 mg/kg/day, p.o) group, ischemia/reperfusion (IR) group, and IR receiving NAT group (50 mg/kg/day, p.o). MCAO caused potent deficits in motor and behavioral functions of the rats. Significant increase in inflammatory and apoptotic biomarkers has been observed in rats' hippocampi. Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) pathway was significantly stimulated causing activation of series inflammatory biomarkers ending up neuro-inflammatory milieu. Pretreatment with NAT preserved rats' normal behavioral and motor functions. Moreover, NAT opposed the expression of hypoxia-inducible factor-1α (HIF-1α) resulting in downregulation of more inflammatory mediators namely, NF-κB, tumor necrosis factor-β (TNF-β), and the anti-survival gene PMAIP-1. NAT stimulated caveolin-1 (Cav-1) which prevented expression of oxidative biomarkers, nitric oxide (NO), and myeloperoxidase (MPO) and hamper the activation of apoptotic biomarker caspase-3. In conclusion, our work postulated that NAT exhibited its neuroprotective effects in rats with ischemic stroke via attenuation of different unique oxidative, apoptotic, and inflammatory pathways.
Collapse
Affiliation(s)
- Muhammad Abd El-Latif Saad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Giza, Egypt.,School of Pharmacy, NewGiza University, Giza, Egypt
| | - Mohamed Ibrahim Mohamed Fahmy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Drug Technology, Heliopolis University for Sustainable Development, Cairo, Egypt.
| | - Muhammad Al-Shorbagy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Giza, Egypt.,School of Pharmacy, NewGiza University, Giza, Egypt
| | - Naglaa Assaf
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Misr University for Science and Technology (MUST), Giza, Egypt
| | | | | |
Collapse
|
48
|
Zhang B, Shen J, Zhong Z, Zhang L. PKM2 Aggravates Cerebral Ischemia Reperfusion-Induced Neuroinflammation via TLR4/MyD88/TRAF6 Signaling Pathway. Neuroimmunomodulation 2021; 28:29-37. [PMID: 33744886 DOI: 10.1159/000509710] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 06/24/2020] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES Cerebral ischemia-reperfusion (I/R) injury is the leading cause of ischemic stroke. Pyruvate Kinase isozymes M2 (PKM2), as a critical glycolytic enzyme during glycolysis, is involved in neuronal apoptosis in rats with hypoxic-ischemic encephalopathy. This study focused on functional investigation and potential molecular mechanism toward PKM2 in cerebral I/R injury. METHODS Cerebral I/R injury model was established by middle cerebral artery occlusion (MCAO) in vivo or oxygen-glucose deprivation and reoxygenation (OGD/R) in vitro. qRT-PCR and Western blot were used to detect the expression of PKM2 in I/R injury models. The effects of PKM2 on I/R injury were determined via triphenyl tetrazolium chloride staining and evaluation of neurological deficits. Cell Counting Kit-8 was employed to detect cell viability, and ELISA was conducted to detect pro-inflammatory cytokines. The underlying mechanism involved in regulation of PKM2 on I/R injury was investigated via ELISA and Western blot. RESULTS PKM2 was upregulated after cerebral I/R injury. Knockdown of PKM2 alleviated MCAO-induced infarction and neurological dysfunction. Moreover, PKM2 knockdown also alleviated OGD/R-induced neuronal cell injury and inflammatory response. Mechanistically, PKM2 knockdown-induced neuroprotection was accompanied by inhibition of high-mobility group box 1 (HMGB1), reflected by inactivation of TLR4/MyD88 (myeloid differentiation factor 88)/TRAF6 (TNF receptor-associated factor 6) signaling pathway. CONCLUSIONS Knockdown of PKM2 attenuated cerebral I/R injury through HMGB1-mediated TLR4/MyD88/TRAF6 expression change, providing a potential target for cerebral I/R injury treatment.
Collapse
Affiliation(s)
- Baocheng Zhang
- Department of Intensive Care Unit, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Jie Shen
- Department of Intensive Care Unit, Jinshan Hospital Affiliated to Fudan University, Shanghai, China,
| | - Zhiyue Zhong
- Department of Intensive Care Unit, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Lin Zhang
- Department of Intensive Care Unit, Jinshan Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
49
|
Xian XH, Gao JX, Qi J, Fan SJ, Zhang M, Li WB. Activation of p38 MAPK participates in the sulbactam-induced cerebral ischemic tolerance mediated by glial glutamate transporter-1 upregulation in rats. Sci Rep 2020; 10:20601. [PMID: 33244020 PMCID: PMC7692545 DOI: 10.1038/s41598-020-77583-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 11/12/2020] [Indexed: 12/13/2022] Open
Abstract
Our previous studies have shown that sulbactam can play a neuroprotection role in hippocampal neurons by upregulating the expression and function of glial glutamate transporter-1 (GLT-1) during ischemic insult. Here, using rat global cerebral ischemia model, we studied in vivo the role of p38 mitogen-activated protein kinases (MAPK) in the sulbactam-induced GLT-1 upregulation and neuroprotection against ischemia. The hippocampal CA1 field was selected as observing target. The expressions of phosphorylated-p38 MAPK and GLT-1 were assayed with western blot analysis and immunohistochemistry. The condition of delayed neuronal death (DND) was assayed with neuropathological evaluation under thionin staining. It was shown that administration of sulbactam protected CA1 hippocampal neurons against ischemic insult accompanied with significantly upregulation in the expressions of phosphorylated-p38 MAPK and GLT-1. The time course analysis showed that sulbactam activated p38 MAPK before the GLT-1 upregulation in either normal or global cerebral ischemic rats. Furthermore, inhibiting p38 MAPK activation by SB203580 blocked the GLT-1 upregulation and neuroprotection induced by sulbactam. The above results suggested that p38 MAPK, at least partly, participated in the sulbactam-induced brain tolerance to ischemia mediated by GLT-1 upregulation in rats.
Collapse
Affiliation(s)
- Xiao-Hui Xian
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China
| | - Jun-Xia Gao
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China
| | - Jie Qi
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China
| | - Shu-Juan Fan
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China
| | - Min Zhang
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China. .,Neuroscience Research Center of Hebei Medical University, Shijiazhuang, People's Republic of China.
| | - Wen-Bin Li
- Department of Pathophysiology, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, People's Republic of China. .,Neuroscience Research Center of Hebei Medical University, Shijiazhuang, People's Republic of China.
| |
Collapse
|
50
|
Identification of the in vivo relevant dissolution media for the three active components in EGb 761 tablet for better correlation with their pharmacokinetics in healthy subjects. Eur J Pharm Sci 2020; 154:105515. [DOI: 10.1016/j.ejps.2020.105515] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/22/2020] [Accepted: 08/13/2020] [Indexed: 11/22/2022]
|