1
|
Zhang X, Hu F, Li J, Chen L, Mao YF, Li QB, Nie CY, Lin C, Xiao J. IGF-1 inhibits inflammation and accelerates angiogenesis via Ras/PI3K/IKK/NF-κB signaling pathways to promote wound healing. Eur J Pharm Sci 2024; 200:106847. [PMID: 38972611 DOI: 10.1016/j.ejps.2024.106847] [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: 03/12/2024] [Revised: 06/13/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
Exogenous insulin-like growth factor-1 (IGF-1) has been reported to promote wound healing through regulation of vascular endothelial cells (VECs). Despite the existing studies of IGF-1 on VEC and its role in angiogenesis, the mechanisms regarding anti-inflammatory and angiogenetic effects of IGF-1 remain unclear. In this study, we investigated the wound-healing process and the related signaling pathway of IGF-1 using an inflammation model induced by IFN-γ. The results demonstrated that IGF-1 can increase cell proliferation, suppress inflammation in VECs, and promote angiogenesis. In vivo studies further confirmed that IGF-1 can reduce inflammation, enhance vascular regeneration, and improve re-epithelialization and collagen deposition in acute wounds. Importantly, the Ras/PI3K/IKK/NF-κB signaling pathways was identified as the mechanisms through which IGF-1 exerts its anti-inflammatory and pro-angiogenic effects. These findings contribute to the understanding of IGF-1's role in wound healing and may have implications for the development of new wound treatment approaches.
Collapse
Affiliation(s)
- Xin Zhang
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China; Department of Burn, the First Affiliated Hospital of Wenzhou Medical University, Nan Bai Xiang, Wenzhou, Zhejiang 325000, China
| | - Fei Hu
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Jie Li
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Lin Chen
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Yu-Fei Mao
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China
| | - Qiu-Bo Li
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China; Department of Burn, the First Affiliated Hospital of Wenzhou Medical University, Nan Bai Xiang, Wenzhou, Zhejiang 325000, China
| | - Chen-Yao Nie
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China.
| | - Cai Lin
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China; Department of Burn, the First Affiliated Hospital of Wenzhou Medical University, Nan Bai Xiang, Wenzhou, Zhejiang 325000, China.
| | - Jian Xiao
- School of Pharmaceutical Sciences, Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, China; Department of Burn, the First Affiliated Hospital of Wenzhou Medical University, Nan Bai Xiang, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
2
|
Qi M, Su X, Li Z, Huang H, Wang J, Lin N, Kong X. Bibliometric analysis of research progress on tetramethylpyrazine and its effects on ischemia-reperfusion injury. Pharmacol Ther 2024; 259:108656. [PMID: 38735486 DOI: 10.1016/j.pharmthera.2024.108656] [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: 12/18/2023] [Revised: 04/22/2024] [Accepted: 05/07/2024] [Indexed: 05/14/2024]
Abstract
In recent decades, natural products have attracted worldwide attention and become one of the most important resources for pharmacological industries and medical sciences to identify novel drug candidates for disease treatment. Tetramethylpyrazine (TMP) is an alkaloid extracted from Ligusticum chuanxiong Hort., which has shown great therapeutic potential in cardiovascular and cerebrovascular diseases, liver and renal injury, as well as cancer. In this review, we analyzed 1270 papers published on the Web of Science Core Collection from 2002 to 2022 and found that TMP exerted significant protective effects on ischemia-reperfusion (I/R) injury that is the cause of pathological damages in a variety of conditions, such as ischemic stroke, myocardial infarction, acute kidney injury, and liver transplantation. TMP is limited in clinical applications to some extent due to its rapid metabolism, a short biological half-life and poor bioavailability. Obviously, the structural modification, administration methods and dosage forms of TMP need to be further investigated in order to improve its bioavailability. This review summarizes the clinical applications of TMP, elucidates its potential mechanisms in protecting I/R injury, provides strategies to improve bioavailability, which presents a comprehensive understanding of the important compound. Hopefully, the information and knowledge from this review can help researchers and physicians to better improve the applications of TMP in the clinic.
Collapse
Affiliation(s)
- Mingzhu Qi
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xiaohui Su
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Zhuohang Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Helan Huang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jingbo Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Na Lin
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Xiangying Kong
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
3
|
Chen Y, Cheng Q, Zeng S, Lv S. Potential analgesic effect of Foshousan oil-loaded chitosan-alginate nanoparticles on the treatment of migraine. Front Pharmacol 2023; 14:1190920. [PMID: 37680717 PMCID: PMC10482050 DOI: 10.3389/fphar.2023.1190920] [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: 03/21/2023] [Accepted: 08/07/2023] [Indexed: 09/09/2023] Open
Abstract
Background: Migraine is a common neurovascular disorder with typical throbbing and unilateral headaches, causing a considerable healthcare burden on the global economy. This research aims to prepare chitosan-alginate (CS-AL) nanoparticles (NPs) containing Foshousan oil (FSSO) and investigate its potential therapeutic effects on the treatment of migraine. Methods: FSSO-loaded CS-AL NPs were prepared by using the single emulsion solvent evaporation method. Lipopolysaccharide (LPS)-stimulated BV-2 cells and nitroglycerin (NTG)-induced migraine mice were further used to explore anti-migraine activities and potential mechanisms of this botanical drug. Results: FSSO-loaded CS-AL NPs (212.1 ± 5.2 nm, 45.1 ± 6.2 mV) had a well-defined spherical shape with prolonged drug release and good storage within 4 weeks. FSSO and FSSO-loaded CS-AL NPs (5, 10, and 15 μg/mL) showed anti-inflammatory activities in LPS-treated BV-2 cells via reducing the levels of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), and nitric oxide (NO), but elevating interleukin-10 (IL-10) expressions. Moreover, FSSO-loaded CS-AL NPs (52 and 104 mg/kg) raised pain thresholds against the hot stimulus and decreased acetic acid-induced writhing frequency and foot-licking duration in NTG-induced migraine mice. Compared with the model group, calcitonin gene-related peptide (CGRP) and NO levels were downregulated, but 5-hydroxytryptamine (5-HT) and endothelin (ET) levels were upregulated along with rebalanced ET/NO ratio, and vasomotor dysfunction was alleviated by promoting cerebral blood flow (CBF) in the FSSO-loaded CS-AL NPs (104 mg/kg) group. Conclusion: FSSO-loaded CS-AL NPs could attenuate migraine via inhibiting neuroinflammation in LPS-stimulated BV-2 cells and regulating vasoactive substances in NTG-induced migraine mice. These findings suggest that the FSS formula may be exploited as new phytotherapy for treating migraine.
Collapse
Affiliation(s)
- Yulong Chen
- College of Medicine and Health Science, Wuhan Polytechnic University, Wuhan, China
| | - Qingzhou Cheng
- College of Medicine and Health Science, Wuhan Polytechnic University, Wuhan, China
| | - Shan Zeng
- School of Mathematics and Computer Science, Wuhan Polytechnic University, Wuhan, China
| | - Site Lv
- School of Mathematics and Computer Science, Wuhan Polytechnic University, Wuhan, China
| |
Collapse
|
4
|
Zhao Y, Ma C, Qiu Q, Huang X, Qiaolongbatu X, Qu H, Wu J, Fan G, Wu Z. Exploring the components and mechanisms of Shen-qi-wang-mo granule in the treatment of retinal vein occlusion by UPLC-Triple TOF MS/MS and network pharmacology. Sci Rep 2023; 13:5330. [PMID: 37005436 PMCID: PMC10066998 DOI: 10.1038/s41598-023-32472-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/28/2023] [Indexed: 04/04/2023] Open
Abstract
This study aimed to explore the substance basis and mechanisms of Shen-qi-wang-mo Granule (SQWMG), a traditional Chinese medicine prescription that had been clinically utilized to treat retinal vein occlusion (RVO) for 38 years. Components in SQWMG were analyzed by UPLC-Triple-TOF/MS and a total of 63 components were identified with ganoderic acids (GA) being the largest proportion. Potential targets of active components were retrieved from SwissTargetPrediction. RVO-related targets were acquired from related disease databases. Core targets of SQWMG against RVO were acquired by overlapping the above targets. The 66 components (including 5 isomers) and 169 targets were obtained and concluded into a component-target network. Together with biological enrichment analysis of targets, it revealed the crucial role of the "PI3K-Akt signaling pathway", "MAPK signaling pathway" and their downstream factor iNOS and TNF-α. The 20 key targets of SQWMG in treating RVO were acquired from the network and pathway analysis. The effects of SQWMG on targets and pathways were validated by molecular docking based on AutoDock Vina and qPCR experiment. The molecular docking showed great affinity for these components and targets, especially on ganoderic acids (GA) and alisols (AS), which were both triterpenoids and qPCR exhibited remarkably reduced inflammatory factor gene expression through regulation of these two pathways. Finally, the key components were also identified from rat serum after treatment of SQWMG.
Collapse
Affiliation(s)
- Yi Zhao
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Cui Ma
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Qinghua Qiu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Xucong Huang
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
- School of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Xijier Qiaolongbatu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Han Qu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, People's Republic of China
| | - Jiaqi Wu
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China
| | - Guorong Fan
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China.
| | - Zhenghua Wu
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China.
| |
Collapse
|
5
|
Liu Y, Yang G, Cui W, Zhang Y, Liang X. Regulatory mechanisms of tetramethylpyrazine on central nervous system diseases: A review. Front Pharmacol 2022; 13:948600. [PMID: 36133805 PMCID: PMC9483103 DOI: 10.3389/fphar.2022.948600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Central nervous system (CNS) diseases can lead to motor, sensory, speech, cognitive dysfunction, and sometimes even death. These diseases are recognized to cause a substantial socio-economic impact on a global scale. Tetramethylpyrazine (TMP) is one of the main active ingredients extracted from the Chinese herbal medicine Ligusticum striatum DC. (Chuan Xiong). Many in vivo and in vitro studies have demonstrated that TMP has a certain role in the treatment of CNS diseases through inhibiting calcium ion overload and glutamate excitotoxicity, anti-oxidative/nitrification stress, mitigating inflammatory response, anti-apoptosis, protecting the integrity of the blood-brain barrier (BBB) and facilitating synaptic plasticity. In this review, we summarize the roles and mechanisms of action of TMP on ischemic cerebrovascular disease, spinal cord injury, Parkinson’s disease, Alzheimer’s disease, cognitive impairments, migraine, and depression. Our review will provide new insights into the clinical applications of TMP and the development of novel therapeutics.
Collapse
Affiliation(s)
- Yue Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guang Yang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Wenqiang Cui
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yunling Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yunling Zhang, ; Xiao Liang,
| | - Xiao Liang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Yunling Zhang, ; Xiao Liang,
| |
Collapse
|
6
|
Luxen M, van Meurs M, Molema G. Unlocking the Untapped Potential of Endothelial Kinase and Phosphatase Involvement in Sepsis for Drug Treatment Design. Front Immunol 2022; 13:867625. [PMID: 35634305 PMCID: PMC9136877 DOI: 10.3389/fimmu.2022.867625] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/28/2022] [Indexed: 11/29/2022] Open
Abstract
Sepsis is a devastating clinical condition that can lead to multiple organ failure and death. Despite advancements in our understanding of molecular mechanisms underlying sepsis and sepsis-associated multiple organ failure, no effective therapeutic treatment to directly counteract it has yet been established. The endothelium is considered to play an important role in sepsis. This review highlights a number of signal transduction pathways involved in endothelial inflammatory activation and dysregulated endothelial barrier function in response to sepsis conditions. Within these pathways – NF-κB, Rac1/RhoA GTPases, AP-1, APC/S1P, Angpt/Tie2, and VEGF/VEGFR2 – we focus on the role of kinases and phosphatases as potential druggable targets for therapeutic intervention. Animal studies and clinical trials that have been conducted for this purpose are discussed, highlighting reasons why they might not have resulted in the expected outcomes, and which lessons can be learned from this. Lastly, opportunities and challenges that sepsis and sepsis-associated multiple organ failure research are currently facing are presented, including recommendations on improved experimental design to increase the translational power of preclinical research to the clinic.
Collapse
Affiliation(s)
- Matthijs Luxen
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- *Correspondence: Matthijs Luxen,
| | - Matijs van Meurs
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Grietje Molema
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
7
|
Unfractionated heparin attenuates endothelial barrier dysfunction via the phosphatidylinositol-3 kinase/serine/threonine kinase/nuclear factor kappa-B pathway. Chin Med J (Engl) 2020; 133:1815-1823. [PMID: 32649510 PMCID: PMC7470014 DOI: 10.1097/cm9.0000000000000905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background Vascular endothelial dysfunction is considered a key pathophysiologic process for the development of acute lung injury. In this study, we aimed at investigating the effects of unfractionated heparin (UFH) on the lipopolysaccharide (LPS)-induced changes of vascular endothelial-cadherin (VE-cadherin) and the potential underlying mechanisms. Methods Male C57BL/6 J mice were randomized into three groups: vehicle, LPS, and LPS + UFH groups. Intraperitoneal injection of 30 mg/kg LPS was used to induce sepsis. Mice in the LPS + UFH group received subcutaneous injection of 8 U UFH 0.5 h before LPS injection. The lung tissue of the mice was collected for assessing lung injury by measuring the lung wet/dry (W/D) weight ratio and observing histological changes. Human pulmonary microvascular endothelial cells (HPMECs) were cultured and used to analyze the effects of UFH on LPS- or tumor necrosis factor-alpha (TNF-α)-induced vascular hyperpermeability, membrane expression of VE-cadherin, p120-catenin, and phosphorylated myosin light chain (p-MLC), and F-actin remodeling, and on the LPS-induced activation of the phosphatidylinositol-3 kinase (PI3K)/serine/threonine kinase (Akt)/nuclear factor kappa-B (NF-κB) signaling pathway. Results In vivo, UFH pretreatment significantly attenuated LPS-induced pulmonary histopathological changes (neutrophil infiltration and erythrocyte effusion, alveolus pulmonis collapse, and thicker septum), decreased the lung W/D, and increased protein concentration (LPS vs. LPS + UFH: 0.57 ± 0.04 vs. 0.32 ± 0.04 mg/mL, P = 0.0092), total cell count (LPS vs. LPS + UFH: 9.57 ± 1.23 vs. 3.65 ± 0.78 × 105/mL, P = 0.0155), polymorphonuclear neutrophil percentage (LPS vs. LPS + UFH: 88.05% ± 2.88% vs. 22.20% ± 3.92%, P = 0.0002), and TNF-α (460.33 ± 23.48 vs. 189.33 ± 14.19 pg/mL, P = 0.0006) in the bronchoalveolar lavage fluid. In vitro, UFH pre-treatment prevented the LPS-induced decrease in the membrane expression of VE-cadherin (LPS vs. LPS + UFH: 0.368 ± 0.044 vs. 0.716 ± 0.064, P = 0.0114) and p120-catenin (LPS vs. LPS + UFH: 0.208 ± 0.018 vs. 0.924 ± 0.092, P = 0.0016), and the LPS-induced increase in the expression of p-MLC (LPS vs. LPS + UFH: 0.972 ± 0.092 vs. 0.293 ± 0.025, P = 0.0021). Furthermore, UFH attenuated LPS- and TNF-α-induced hyperpermeability of HPMECs (LPS vs. LPS + UFH: 8.90 ± 0.66 vs. 15.84 ± 1.09 Ω·cm2, P = 0.0056; TNF-α vs. TNF-α + UFH: 11.28 ± 0.64 vs. 18.15 ± 0.98 Ω·cm2, P = 0.0042) and F-actin remodeling (LPS vs. LPS + UFH: 56.25 ± 1.51 vs. 39.70 ± 1.98, P = 0.0027; TNF-α vs. TNF-α + UFH: 55.42 ± 1.42 vs. 36.51 ± 1.20, P = 0.0005) in vitro. Additionally, UFH decreased the phosphorylation of Akt (LPS vs. LPS + UFH: 0.977 ± 0.081 vs. 0.466 ± 0.035, P = 0.0045) and I kappa B Kinase (IKK) (LPS vs. LPS + UFH: 1.023 ± 0.070 vs. 0.578 ± 0.044, P = 0.0060), and the nuclear translocation of NF-κB (LPS vs. LPS + UFH: 1.003 ± 0.077 vs. 0.503 ± 0.065, P = 0.0078) in HPMECs, which was similar to the effect of the PI3K inhibitor, wortmannin. Conclusions The protective effect of UFH against LPS-induced pulmonary endothelial barrier dysfunction involves VE-cadherin stabilization and PI3K/Akt/NF-κB signaling.
Collapse
|
8
|
Sun Y, Liu J, Zhang X, Li X, Zhou B, Lv Z. Mechanisms involved in inflammatory pulmonary fibrosis induced by lunar dust simulant in rats. ENVIRONMENTAL TOXICOLOGY 2019; 34:131-140. [PMID: 30496634 DOI: 10.1002/tox.22665] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/17/2018] [Accepted: 09/23/2018] [Indexed: 06/09/2023]
Abstract
Lunar dust is one of the biggest risk factors in the future manned exploration mission. Much is not known about the pulmonary toxicity of lunar dust. The aim of this study was to evaluate the lung inflammation and oxidative stress induced by subacute exposure to lunar dust stimulant (LDS) in rats. Wistar rats were intratracheally administered LDS, twice a week for 3 weeks. Inflammatory cell counting and cytokine assays using bronchoalveolar lavage fluid (BALF) were performed. Lung tissues were processed for histopathological examination and immunohistochemical staining. Biomarkers of oxidative stress and genes and proteins related to inflammation and fibrosis in lung tissue were also determined. The neutrophil count in the BALF of LDS-exposed groups was higher than that in controls (P < .05). LDS caused a significant increase in some of biochemical indicators and proinflammatory factors levels in BALF compared with control group. The normal balance between oxidation and antioxidation was broken by LDS. Pathological characteristics of lung tissue and immunohistochemical results for α-smooth muscle actin (α-SMA) indicated that inflammatory response was an extremely important passage to pulmonary fibrosis. Real-time PCR analysis showed elevated levels of nitric oxide synthase (NOS) and nicotinamide adenine dinucleotide phosphate oxidase (NOX) mRNA in the lungs (P < .05). Western blotting results were consistent with immunohistochemistry and qPCR results. These results indicate that inhalation of lunar dust may cause inflammatory pulmonary fibrosis. NOX4 may be a key potential therapeutic target for inflammatory injury and fibrosis in the lung.
Collapse
Affiliation(s)
- Yan Sun
- College of Basic Medical Sciences, Shenyang Medical College, Shenyang, China
- Lunar and Planetary Science Laboratory, MUST-Partner Laboratory of Key Laboratory of Lunar and Deep Space Exploration, CAS, Macau, China
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China
| | - Jinguo Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, China
| | - Xiaoping Zhang
- Lunar and Planetary Science Laboratory, MUST-Partner Laboratory of Key Laboratory of Lunar and Deep Space Exploration, CAS, Macau, China
- Lunar and Planetary Science Laboratory, Macau University of Science and Technology, Macau, China
| | - Xiongyao Li
- Center for Lunar and Planetary Sciences, Institute of Geochemistry, Chinese Academy of Sciences, Guiyang, China
| | - Baichu Zhou
- College of Basic Medical Sciences, Shenyang Medical College, Shenyang, China
| | - Zengjing Lv
- College of Basic Medical Sciences, Shenyang Medical College, Shenyang, China
| |
Collapse
|
9
|
Zhou P, Xie W, Luo Y, Lu S, Dai Z, Wang R, Sun G, Sun X. Protective Effects of Total Saponins of Aralia elata (Miq.) on Endothelial Cell Injury Induced by TNF-α via Modulation of the PI3K/Akt and NF-κB Signalling Pathways. Int J Mol Sci 2018; 20:ijms20010036. [PMID: 30577658 PMCID: PMC6337668 DOI: 10.3390/ijms20010036] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 12/11/2018] [Accepted: 12/18/2018] [Indexed: 12/19/2022] Open
Abstract
Atherosclerosis is an arterial disease associated with inflammation. Hence, the discovery of novel therapeutic agents for suppressing inflammatory responses is urgent and vital for the treatment of atherosclerosis in cardiovascular diseases. The total saponins of Aralia elata (Miq.) Seem. (TAS) are the main components extracted from the Chinese traditional herb Longya Aralia chinensis L., a folk medicine used in Asian countries for treating numerous diseases, enhancing energy and boosting immunity. However, the protective effects of TAS against inflammation-triggered vascular endothelial dysfunction, a critical early event during the course of atherosclerosis, and the potential mechanisms of this protection have been not demonstrated. Accordingly, the aim of this study was to investigate the anti-inflammatory and anti-apoptotic effects and the protective mechanisms of TAS, and show how TAS ameliorates human umbilical vein endothelial cell (HUVEC) damage caused by tumour necrosis factor-α (TNF-α). The results indicated that TAS exerted cytoprotective effects by inhibiting TNF-α-triggered HUVEC apoptosis, mitochondrial membrane potential depolarisation, and the regulation of inflammatory factors (IL-6, MCP-1, and VCAM-1) while suppressing NF-κB transcription. Furthermore, this phenomenon was related to activation of the phosphoinositide 3-kinase (PI3K)/Akt signalling pathway. Blocking the Akt pathway with LY294002, a PI3K inhibitor, reversed the cytoprotective effect of TAS against TNF-α-induced endothelial cell death. Moreover, LY294002 partially abolished the effects of TAS on the upregulation of the Bcl-2 family of proteins and the downregulation of Bax protein expression. In conclusion, the results of our study suggest that TAS suppresses the inflammation and apoptosis of HUVECs induced by TNF-α and that PI3K/Akt signalling plays a key role in promoting cell survival and anti-inflammatory reactions during this process.
Collapse
Affiliation(s)
- Ping Zhou
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing 100193, China.
| | - Weijie Xie
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing 100193, China.
| | - Yun Luo
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing 100193, China.
| | - Shan Lu
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing 100193, China.
| | - Ziru Dai
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing 100193, China.
| | - Ruiying Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing 100193, China.
| | - Guibo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing 100193, China.
| | - Xiaobo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100193, China.
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing 100193, China.
- Key Laboratory of Efficacy Evaluation of Chinese Medicine against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Beijing 100193, China.
- Zhongguancun Open Laboratory of the Research and Development of Natural Medicine and Health Products, Beijing 100193, China.
| |
Collapse
|
10
|
Ishikawa C, Senba M, Mori N. Anti-adult T‑cell leukemia/lymphoma activity of cerdulatinib, a dual SYK/JAK kinase inhibitor. Int J Oncol 2018; 53:1681-1690. [PMID: 30066853 DOI: 10.3892/ijo.2018.4513] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 07/03/2018] [Indexed: 11/06/2022] Open
Abstract
Adult T‑cell leukemia/lymphoma (ATLL) constitutes an aggressive malignancy caused by human T‑cell leukemia virus type 1 (HTLV‑1) that is resistant to available chemotherapeutics. The constitutive activation of Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling is an important feature of ATLL, and spleen tyrosine kinase (SYK) is overexpressed in HTLV‑1-transformed T‑cell lines. In this study, we evaluated the effects of SYK- (PRT060318) or JAK- (JAK inhibitor 1) selective inhibitors and the dual SYK/JAK inhibitor, cerdulatinib, on the viability of HTLV‑1-transformed and ATLL-derived T‑cell lines. Cell proliferation, viability, cell cycle, apoptosis and intracellular signaling cascades were analyzed by the water-soluble tetrazolium-8 assay, flow cytometry and western blot analysis. HTLV‑1-infected T‑cell lines were sensitive to both SYK-selective and pan-JAK inhibitors, whereas cerdulatinib more potently suppressed cell proliferation and reduced cell viability than either of these agents alone. By contrast, the cytotoxic effects of cerdulatinib on uninfected T‑cell lines and peripheral blood mononuclear cells from a healthy donor were less pronounced. Cerdulatinib induced cell cycle arrest in the G2/M phase, which was associated with a decreased cyclin-dependent kinase 1 and cyclin B1, and an increased p21 and p27 expression. Hoechst staining revealed chromatin condensation and nuclear fragmentation in the cells treated with cerdulatinib, and an increased fraction of apoptotic APO2.7-stained cells was detected by flow cytometry. This corresponded to the activation of caspase-8, -9 and -3, and decreased levels of the anti-apoptotic factors, Bcl-xL, survivin, X-linked inhibitor of apoptosis (XIAP) and c‑FLIP. The cerdulatinib-induced decrease in cell viability was partly reversed by the caspase inhibitor, z‑VAD‑FMK. These anti-ATLL effects were associated with the suppression of SYK and JAK/STAT signaling, along with that of the downstream factors, AKT, ERK, activator protein‑1 and nuclear factor-κB. Finally, oral dosing with cerdulatinib lowered the tumor burden in a murine model of ATLL. Thus, our findings indicate that the simultaneous inhibition of therapeutically relevant targets, such as SYK and JAK is a more effective approach than single-agent therapy for the treatment of ATLL.
Collapse
Affiliation(s)
- Chie Ishikawa
- Department of Microbiology and Oncology, Graduate School of Medicine, University of The Ryukyus, Nishihara, Okinawa 903-0215, Japan
| | - Masachika Senba
- Department of Pathology, Institute of Tropical Medicine, Nagasaki University, Nagasaki 852-8523, Japan
| | - Naoki Mori
- Department of Microbiology and Oncology, Graduate School of Medicine, University of The Ryukyus, Nishihara, Okinawa 903-0215, Japan
| |
Collapse
|
11
|
Shan CS, Xu QQ, Shi YH, Wang Y, He ZX, Zheng GQ. Chuanxiong Formulae for Migraine: A Systematic Review and Meta-Analysis of High-Quality Randomized Controlled Trials. Front Pharmacol 2018; 9:589. [PMID: 30013473 PMCID: PMC6036270 DOI: 10.3389/fphar.2018.00589] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 05/16/2018] [Indexed: 12/25/2022] Open
Abstract
Objective: Migraine is a complex, prevalent and disabling neurological disorder characterized by recurrent episodes of headache without ideal treatment. We aim to assess the current available evidence of herbal Chuanxiong (Ligusticum chuanxiong Hort. root) formulae for the treatment of migraine according to the high-quality randomized controlled trials (RCTs). Methods: English and Chinese electronic databases were searched from their inceptions until March 2017. The methodological quality of included study was assessed by the Cochrane Collaboration risk of bias tool. RCTs with Cochrane risk of bias (RoB) score ≥4 were included in the analyses. Meta-analysis was conducted using RevMan 5.3 software. Publication bias was assessed by funnel plot analysis and Egger's test. Results: Nineteen RCTs with 1832 participants were identified. The studies investigated the Chuanxiong formulae vs. placebo (n = 5), Chuanxiong formulae vs. conventional pharmacotherapy (CP) (n = 13 with 15 comparisons), and Chuanxiong formulae plus CP vs. CP (n = 1). Meta-analysis indicated that Chuanxiong formulae could reduce frequency, duration, days and pain severity of migraine and improve the total clinical efficacy rate (P < 0.05). Adverse event monitoring was reported in 16 out of 19 studies and occurrence rate of adverse event was low. Conclusion: The findings of present study indicated that Chuanxiong formulae exerted the symptom reliefs of for migraine.
Collapse
Affiliation(s)
- Chun-Shuo Shan
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qing-Qing Xu
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yi-Hua Shi
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yong Wang
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhang-Xin He
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guo-Qing Zheng
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
12
|
Hou M, Tang Q, Xue Q, Zhang X, Liu Y, Yang S, Chen L, Xu X. Pharmacodynamic action and mechanism of Du Liang soft capsule, a traditional Chinese medicine capsule, on treating nitroglycerin-induced migraine. JOURNAL OF ETHNOPHARMACOLOGY 2017; 195:231-237. [PMID: 27866934 DOI: 10.1016/j.jep.2016.11.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 10/31/2016] [Accepted: 11/11/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Du Liang soft capsule (DL) is a traditional Chinese medicine for treating migraines; it is made from two Chinese herbs, including LigusticumstriatumDC., root; Angelica dahurica (Hoffm.) Benth. & Hook.f. ex Franch. & Sav., root. AIM OF THE STUDY In the present study, we aimed to elucidate the pharmacodynamic action of DL and its mechanism in an animal model of migraines induced by glyceryl trinitrate (GTN). MATERIALS AND METHODS Sixty rats were randomly divided into six groups, including a normal control group, model control group, positive group (Sumatriptan 0.006gkg-1), and three DL groups (0.44, 1.31 and 3.93gkg-1). All rats were intragastrically treated with the corresponding treatment for 7 consecutive days, and they were subcutaneously injected with GTN (10mgkg-1) 30min after the last treatment, except in the normal control group. After model establishment, the behaviors of all rats, including head scratching, cage climbing, and the development of red ears were observed continuously by digital camera every 30min for 3h. Four hours after GTN treatment, all rats were anaesthetized and the blood and tissue samples were collected. Plasma calcitonin gene related to peptide (CGRP) and endothelin (ET) levels were measured using the radioimmunoassay method, and serum NO was determined by the colorimetric method. Afterwards, the brainstem tissues were dissected and washed with physiological saline, and divided evenly into two parts. One part was used to test the monoamine levels, including levels of 5-hydroxytryptamine (5-HT), norepinephrine (NE) and dopamine (DA), by the fluorometric method, and the other part was used to determine the nuclear factor kappaB (NF-κB) p65, nuclear c-fos, inducible nitric oxide synthase (iNOS), interleukin (IL)-1β (IL-1β), and cyclooxygenase-2 (COX-2) levels by Western blot analysis. RESULTS In the pharmacodynamic action assay, DL (1.31 and 3.93gkg-1) greatly improved the abnormal behaviors of migraine rats, including head scratching and cage climbing, and the development of red ears. In the mechanism assay, compared with the control group, the plasma CGRP and serum NO levels and the brainstem 5-HT, NE and DA levels in the DL administration groups were significantly decreased; and the plasma ET levels were remarkably increased. Moreover, down-regulation of NF-κB p65, c-fos and pro-inflammatory cytokines, including iNOS, IL-1β and COX-2 in the brainstem in the DL administration groups were observed by Western blot analysis. CONCLUSIONS The above results suggested that DL has a therapeutic effect on migraines, and its mechanism may be related to adjusting the level of neurotransmitters and vasoactive substances, consequently relieving neurogenic inflammation.
Collapse
Affiliation(s)
- Min Hou
- College of Pharmaceutical Sciences & College of Traditional Chinese Medicine, Southwest University, Chongqing 400715, PR China; Department of pharmacy, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, PR China
| | - Qing Tang
- College of Pharmaceutical Sciences & College of Traditional Chinese Medicine, Southwest University, Chongqing 400715, PR China
| | - Qiang Xue
- College of Pharmaceutical Sciences & College of Traditional Chinese Medicine, Southwest University, Chongqing 400715, PR China
| | - Xiaona Zhang
- College of Pharmaceutical Sciences & College of Traditional Chinese Medicine, Southwest University, Chongqing 400715, PR China
| | - Yang Liu
- College of Pharmaceutical Sciences & College of Traditional Chinese Medicine, Southwest University, Chongqing 400715, PR China
| | - Sheng Yang
- College of Pharmaceutical Sciences & College of Traditional Chinese Medicine, Southwest University, Chongqing 400715, PR China
| | - Liechun Chen
- Chongqing Pharscin Pharmaceutical Group Co., Ltd, Chongqing 401121, PR China
| | - Xiaoyu Xu
- College of Pharmaceutical Sciences & College of Traditional Chinese Medicine, Southwest University, Chongqing 400715, PR China.
| |
Collapse
|
13
|
Jia Y, Wang Z, Zang A, Jiao S, Chen S, Fu Y. Tetramethylpyrazine inhibits tumor growth of lung cancer through disrupting angiogenesis via BMP/Smad/Id-1 signaling. Int J Oncol 2016; 48:2079-86. [PMID: 26984046 DOI: 10.3892/ijo.2016.3443] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 02/24/2016] [Indexed: 11/05/2022] Open
Abstract
The underlying mechanisms of inhibitory effects induced by tetramethylpyrazine (TMP) on angiogenesis and tumor growth of lung cancer were investigated. In vitro cell proliferation, migration, and tube formation of human microvascular endothelial cells (HMEC-1) were evaluated by a 3-(4,5-dimethylthiazol-2-yl)-2,5-dephenyltetrazolium bromide (MTT), wound healing, Transwell, and Matrigel assays. The expression of BMP/Smad/Id-1 signals was detected by RT-PCR and western blotting. In an A549 xenograft tumor model, TMP (40 and 80 mg/kg/day) was intraperitoneally injected into mice. The expressions of CD31, phosphorylated Smad1/5/8, and Id-1 were measured by immunohistochemistry. We demonstrated that TMP inhibited proliferation, migration, and capillary tube formation of HMEC-1 in a dose- and time-dependent manner. Furthermore, treatment of HMEC-1 cells with TMP (0.4 mg/ml) significantly upregulated BMP2 expression and downregulated BMPRIA, BMPRII, phosphorylated Smad1/5/8, and Id-1 expression. In addition, administrations of TMP remarkably inhibited tumor growth of A549 xenograft in nude mice. The CD31, phosphorylated Smad1/5/8, and Id-1 expression were significantly inhibited in TMP-treated xenograft tumors compared with the vehicle. In conclusion, our results indicated that TMP suppressed angiogenesis and tumor growth of lung cancer via blocking the BMP/Smad/Id-1 signaling.
Collapse
Affiliation(s)
- Youchao Jia
- Department of Medical Oncology, General Hospital of Chinese PLA, Beijing 100853, P.R. China
| | - Zhigang Wang
- Department of Medical Oncology, Baoding Hengxing Hospital of Traditional Chinese and Western Medicine, Baoding 071000, P.R. China
| | - Aimin Zang
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding 071000, P.R. China
| | - Shunchang Jiao
- Department of Medical Oncology, General Hospital of Chinese PLA, Beijing 100853, P.R. China
| | - Sumei Chen
- Department of Medical Oncology, General Hospital of Chinese PLA, Beijing 100853, P.R. China
| | - Yan Fu
- Department of Medical Oncology, General Hospital of Chinese PLA, Beijing 100853, P.R. China
| |
Collapse
|
14
|
Zhang S, Shao SY, Song XY, Xia CY, Yang YN, Zhang PC, Chen NH. Protective effects of Forsythia suspense extract with antioxidant and anti-inflammatory properties in a model of rotenone induced neurotoxicity. Neurotoxicology 2016; 52:72-83. [DOI: 10.1016/j.neuro.2015.09.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 09/20/2015] [Accepted: 09/21/2015] [Indexed: 12/21/2022]
|
15
|
Tetramethylpyrazine Protects Retinal Capillary Endothelial Cells (TR-iBRB2) against IL-1β-Induced Nitrative/Oxidative Stress. Int J Mol Sci 2015; 16:21775-90. [PMID: 26370989 PMCID: PMC4613279 DOI: 10.3390/ijms160921775] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 08/19/2015] [Accepted: 08/31/2015] [Indexed: 12/22/2022] Open
Abstract
Blood-retinal barrier (BRB) breakdown is one of the primary causes of diabetic retinopathy (DR). The pro-inflammatory factor interleukin-1β (IL-1β) was reported to be involved in the induction of BRB breakdown during the pathogenesis of DR. In the present study, we investigated the protective effect of tetramethylpyrazine (TMP), a major active component of the traditional herb Ligusticum chuanxiong, on IL-1β-induced cell death of the rat retinal capillary endothelial TR-iBRB2 cells. Our results showed that IL-1β-induced cell dysfunction in TR-iBRB2 cells via inducing nitrative/oxidative stress; however, such effect was attenuated with the pre-treatment of TMP. The cellular protective effect of TMP was likely to be mediated through the inhibition of inducible nitric oxide synthase (iNOS) expression and leukostasis as well as suppression of reactive oxygen species (ROS) generation, mitochondrial dysfunction and MAPKs activation. These findings significantly contribute to a better understanding of the protective effect of TMP in DR and form the basis of the therapeutic development of TMP in treating such disease in the future.
Collapse
|
16
|
Liu Y, Zhao G, Lin J, Li C, Li Q, Che C, Wang Q, Hu L. The role of Syk signaling in antifungal innate immunity of human corneal epithelial cells. BMC Ophthalmol 2015; 15:55. [PMID: 26036769 PMCID: PMC4451931 DOI: 10.1186/s12886-015-0041-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 05/18/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Fungal keratitis is a kind of intractable and sight-threatening diseases. Spleen-tyrosine kinase (Syk) is a non-receptor tyrosine kinase, which plays an important role in the signaling pathway of the receptors. In the current study, we investigate the expression and function of Syk in human corneal epithelial cells with Aspergillus fumigatus (A. fumigatus) infection. METHODS Cultured telomerase-immortalized human corneal epithelial cells (THCEs) were treated with A. fumigatus hyphae with or without treatment of Syk inhibitors. Activation of Syk and the role of Syk in regulating inflammatory cytokines and chemokines expression were evaluated. The mRNA expression was determined by real time PCR, and protein activation was measured by western blotting. RESULTS Syk protein was detected in THCEs, and its activation was enhanced after treatment of A. fumigatus hyphae. Expression of inflammatory cytokines (IL-1β and IL-6) and chemokines (IL-8 and CXCL1) mRNA were significantly increased after stimulation of A. fumigatus hyphae in THCEs. Activation of Syk and expression of IL-1β, IL-6, IL-8 and CXCL1 by A. fumigatus hyphae were blocked by Syk inhibitors. CONCLUSION These findings demonstrate that normal human corneal epithelial cells produce Syk, and Syk activation plays an important role in regulating A. fumigatus hyphae-induced inflammatory responses in THCEs.
Collapse
Affiliation(s)
- Ying Liu
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China. .,Department of Ophthalmology, the Affiliated Hospital of Xuzhou Medical College, Xuzhou, 221000, Jiangsu Province, China.
| | - Guiqiu Zhao
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China.
| | - Jing Lin
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China.
| | - Cui Li
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China.
| | - Qing Li
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China.
| | - Chengye Che
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China.
| | - Qian Wang
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China.
| | - Liting Hu
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong Province, China.
| |
Collapse
|
17
|
Gao HJ, Liu PF, Li PW, Huang ZY, Yu FB, Lei T, Chen Y, Cheng Y, Mu QC, Huang HY. Ligustrazine monomer against cerebral ischemia/reperfusion injury. Neural Regen Res 2015; 10:832-40. [PMID: 26109963 PMCID: PMC4468780 DOI: 10.4103/1673-5374.156991] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2015] [Indexed: 12/13/2022] Open
Abstract
Ligustrazine (2,3,5,6-tetramethylpyrazine) is a major active ingredient of the Szechwan lovage rhizome and is extensively used in treatment of ischemic cerebrovascular disease. The mechanism of action of ligustrazine use against ischemic cerebrovascular diseases remains unclear at present. This study summarizes its protective effect, the optimum time window of administration, and the most effective mode of administration for clinical treatment of cerebral ischemia/reperfusion injury. We examine the effects of ligustrazine on suppressing excitatory amino acid release, promoting migration, differentiation and proliferation of endogenous neural stem cells. We also looked at its effects on angiogenesis and how it inhibits thrombosis, the inflammatory response, and apoptosis after cerebral ischemia. We consider that ligustrazine gives noticeable protection from cerebral ischemia/reperfusion injury. The time window of ligustrazine administration is limited. The protective effect and time window of a series of derivative monomers of ligustrazine such as 2-[(1,1-dimethylethyl)oxidoimino]methyl]-3,5,6-trimethylpyrazine, CXC137 and CXC195 after cerebral ischemia were better than ligustrazine.
Collapse
Affiliation(s)
- Hai-Jun Gao
- Department of Neurosurgery, First Bethune Hospital of Jilin University, Changchun, Jilin Province, China ; Department of Neurosurgery, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, China
| | - Peng-Fei Liu
- Department of Neurosurgery, First Bethune Hospital of Jilin University, Changchun, Jilin Province, China
| | - Pei-Wen Li
- Department of Neurosurgery, First Bethune Hospital of Jilin University, Changchun, Jilin Province, China
| | - Zhuo-Yan Huang
- Clinical Medical College of Beihua University, Jilin, Jilin Province, China
| | - Feng-Bo Yu
- School of Pharmacy, Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, China
| | - Ting Lei
- Department of Neurosurgery, First Bethune Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yong Chen
- Department of Neurosurgery, First Bethune Hospital of Jilin University, Changchun, Jilin Province, China
| | - Ye Cheng
- Department of Neurosurgery, First Bethune Hospital of Jilin University, Changchun, Jilin Province, China
| | - Qing-Chun Mu
- Department of Neurosurgery, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, China
| | - Hai-Yan Huang
- Department of Neurosurgery, First Bethune Hospital of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
18
|
Theodorakis NG, Wang YN, Korshunov VA, Maluccio MA, Skill NJ. Thalidomide ameliorates portal hypertension via nitric oxide synthase independent reduced systolic blood pressure. World J Gastroenterol 2015; 21:4126-4135. [PMID: 25892862 PMCID: PMC4394073 DOI: 10.3748/wjg.v21.i14.4126] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 07/30/2014] [Accepted: 10/15/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: Portal hypertension is a common complication of liver cirrhosis and significantly increases mortality and morbidity. Previous reports have suggested that the compound thalidomide attenuates portal hypertension (PHT). However, the mechanism for this action is not fully elucidated. One hypothesis is that thalidomide destabilizes tumor necrosis factor α (TNFα) mRNA and therefore diminishes TNFα induction of nitric oxide synthase (NOS) and the production of nitric oxide (NO). To examine this hypothesis, we utilized the murine partial portal vein ligation (PVL) PHT model in combination with endothelial or inducible NOS isoform gene knockout mice.
METHODS: Wild type, inducible nitric oxide synthase (iNOS)-/- and endothelial nitric oxide synthase (eNOS)-/- mice received either PVL or sham surgery and were given either thalidomide or vehicle. Serum nitrate (total nitrate, NOx) was measured daily for 7 d as a surrogate of NO synthesis. Serum TNFα level was quantified by enzyme-linked immunosorbent assay. TNFα mRNA was quantified in liver and aorta tissue by reverse transcription-polymerase chain reaction. PHT was determined by recording splenic pulp pressure (SPP) and abdominal aortic flow after 0-7 d. Response to thalidomide was determined by measurement of SPP and mean arterial pressure (MAP).
RESULTS: SPP, abdominal aortic flow (Qao) and plasma NOx were increased in wild type and iNOS-/- PVL mice when compared to sham operated control mice. In contrast, SPP, Qao and plasma NOx were not increased in eNOS-/- PVL mice when compared to sham controls. Serum TNFα level in both sham and PVL mice was below the detection limit of the commercial ELISA used. Therefore, the effect of thalidomide on serum TNFα levels was undetermined in wild type, eNOS-/- or iNOS-/- mice. Thalidomide acutely increased plasma NOx in wild type and eNOS-/- mice but not iNOS-/- mice. Moreover, thalidomide temporarily (0-90 min) decreased mean arterial pressure, SPP and Qao in wild type, eNOS-/- and iNOS-/- PVL mice, after which time levels returned to the respective baseline.
CONCLUSION: Thalidomide does not reduce portal pressure in the murine PVL model by modulation of NO biosynthesis. Rather, thalidomide reduces PHT by decreasing MAP by an undetermined mechanism.
Collapse
|
19
|
Effects of incretin agonists on endothelial nitric oxide synthase expression and nitric oxide synthesis in human coronary artery endothelial cells exposed to TNFα and glycated albumin. Pharmacol Rep 2015; 67:69-77. [PMID: 25560578 DOI: 10.1016/j.pharep.2014.08.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 08/08/2014] [Accepted: 08/08/2014] [Indexed: 12/25/2022]
|
20
|
Li J, Yang X, Yang J, Lai L. Study on the interaction between ligustrazine and 12‐tungstophosphoric acid using resonance Rayleigh scattering and resonance nonlinear scattering spectra, and its analytical applications. LUMINESCENCE 2014; 30:643-8. [DOI: 10.1002/bio.2799] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 08/28/2014] [Accepted: 09/15/2014] [Indexed: 11/12/2022]
Affiliation(s)
- Junbo Li
- Pharmaceutical DepartmentChangzhi Medical College Changzhi 046000 China
| | - Xiaoli Yang
- Pharmaceutical DepartmentChangzhi Medical College Changzhi 046000 China
| | - Jinxiang Yang
- Pharmaceutical DepartmentChangzhi Medical College Changzhi 046000 China
| | - Lina Lai
- Pharmaceutical DepartmentChangzhi Medical College Changzhi 046000 China
| |
Collapse
|