1
|
Ye H, Wu L, Liu YM, Zhang JX, Hu HT, Dong ML, Ren J. Wogonin attenuates septic cardiomyopathy by suppressing ALOX15-mediated ferroptosis. Acta Pharmacol Sin 2025:10.1038/s41401-025-01547-1. [PMID: 40205009 DOI: 10.1038/s41401-025-01547-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 03/17/2025] [Indexed: 04/11/2025]
Abstract
Septic cardiomyopathy (SCM), a severe complication in sepsis, significantly increases the mortality of septic patients. Ferroptosis, an iron-regulated cell death, has been implicated in the development of SCM. Wogonin, a flavonoid from the root of the skullcap, exhibits anti-inflammatory, anti-allergic, and anti-apoptotic activities. In this study, we investigated the effects of wogonin on SCM and associated cardiomyocyte ferroptosis. Cecal ligation and puncture (CLP) surgery was performed in mice to establish a SCM model. Wogonin (20, 40 and 60 mg·kg-1, i.p.) was administered 2 h prior to CLP surgery. We showed that wogonin pretreatment dose-dependently mitigated CLP-induced cardiac dysfunction, myocardial damage, and deranged cardiomyocyte contractility. Furthermore, wogonin pretreatment ameliorated cardiac inflammation, oxidative stress, and mitochondrial dysfunction in CLP-challenged mice. We demonstrated that wogonin exerted the cardioprotective effects through suppressing cardiomyocyte ferroptosis both in vivo and in vitro. We revealed that wogonin directly bound to and inhibited ALOX15 (arachidonic acid 15-lipoxygenase), a lipoxygenase that governed the oxidation of polyunsaturated fatty acids to initiate ferroptosis. Pharmacological inhibition of ALOX15 using a specific inhibitor ML351 (10 mg·kg-1·d-1, i.p. for 7 days prior to CLP surgery) markedly diminished cardiac abnormalities and cardiomyocyte ferroptosis in CLP-challenged mice. In LPS-challenged HL-1 cardiomyocytes, overexpression of ALOX15 or supplement of its downstream metabolite 15-HpETE (1 μM) diminished the anti-ferroptotic effects of wogonin. Our results demonstrate that wogonin protects against SCM through inhibition of ALOX15-meditated ferroptosis.
Collapse
Affiliation(s)
- Hua Ye
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Department of Burns & Plastic and Wound Repair, Ganzhou People's Hospital, Ganzhou, 341000, China
| | - Lin Wu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China
| | - Yan-Mei Liu
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Jun-Xia Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research; Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences; Haihe Laboratory of Cell Ecosystem, Beijing, 100191, China
| | - Huan-Tao Hu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Mao-Long Dong
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
- Department of Burns, Tongren Hospital of Wuhan University & Wuhan Third Hospital, Wuhan, 430060, China.
| | - Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, 200032, China.
- National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| |
Collapse
|
2
|
Yuan M, Yi S, Wang X, Han G, Wei P, Lv Z, Gui B, Chen X, Wang Y, Zhu L. Promoted Translocation of Perfluorooctanoic Acid across the Blood-Retinal Barrier due to its Inhibition of Tight Junction Assembly by Antagonizing LPAR1. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:4807-4819. [PMID: 40038073 DOI: 10.1021/acs.est.4c12051] [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: 03/06/2025]
Abstract
Eye health is becoming a significant public health concern, and a recent epidemiological investigation suggested that perfluorooctanoic acid (PFOA), a so-called forever chemical, was correlated with decreased human visual acuity; however, it remains unknown whether PFOA can pass through the blood-retinal barrier (BRB) to cause visual toxicity. In this study, the mice received a 28-day subchronic oral exposure to PFOA. The results of spatial mass spectrometry imaging indicated that the eye-enriched PFOA dispersed into the subretina primarily through the outer BRB (oBRB), which subsequently resulted in significantly increased apoptosis and decreased thickness of multiple oBRB-associated layers. BRB integrity and function were compromised due to decreased expression of the tight junction (TJ). Mechanistically, PFOA outcompeted lysophosphatidic acid to bind strongly with lysophosphatidic acid receptor 1 (LPAR1) in its antagonism, abolishing its ability to stimulate the TJ assembly-related signaling pathway. This subsequently attenuated phosphorylation of the myosin light chain, rendering insufficient contraction of the actomyosin cytoskeleton, leading to decreased TJ assembly and BRB leakage. This, in turn, facilitated PFOA translocation across the BRB and accumulation within the subretinal space. Our findings suggest that oBRB is particularly vulnerable to PFOA, which targets directly LPAR1 to disable its function of maintaining TJ assembly cascades, leading to adverse visual effects.
Collapse
Affiliation(s)
- Meng Yuan
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Shujun Yi
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Xiaoyan Wang
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Guoge Han
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Nankai University Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University, Tianjin 300020, China
| | - Pinghui Wei
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Nankai University Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University, Tianjin 300020, China
| | - Zixuan Lv
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Bingxin Gui
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| | - Xuan Chen
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Nankai University Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University, Tianjin 300020, China
| | - Yan Wang
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Nankai University Eye Institute, Nankai University Affiliated Eye Hospital, Nankai University, Tianjin 300020, China
| | - Lingyan Zhu
- Key Laboratory of Pollution Processes and Environmental Criteria of Ministry of Education, Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin 300350, China
| |
Collapse
|
3
|
Vissenaekens H, Criel H, Grootaert C, Raes K, Smagghe G, Van Camp J. Flavonoids and cellular stress: a complex interplay affecting human health. Crit Rev Food Sci Nutr 2021; 62:8535-8566. [PMID: 34098806 DOI: 10.1080/10408398.2021.1929822] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Flavonoid consumption has beneficial effects on human health, however, clinical evidence remains often inconclusive due to high interindividual variability. Although this high interindividual variability has been consistently observed in flavonoid research, the potential underlying reasons are still poorly studied. Especially the knowledge on the impact of health status on flavonoid responsiveness is limited and merits more investigation. Here, we aim to highlight the bidirectional interplay between flavonoids and cellular stress. First, the state-of-the-art concerning inflammatory stress and mitochondrial dysfunction is reviewed and a comprehensive overview of recent in vitro studies investigating the impact of flavonoids on cellular stress, induced by tumor necrosis factor α, lipopolysaccharide and mitochondrial stressors, is given. Second, we critically discuss the influence of cellular stress on flavonoid uptake, accumulation, metabolism and cell responses, which has, to our knowledge, never been extensively reviewed before. Next, we advocate the innovative insight that stratification of the general population based on health status can reveal subpopulations that benefit more from flavonoid consumption. Finally, suggestions are given for the development of future cell models that simulate the physiological micro-environment, including interindividual variability, since more mechanistic research is needed to establish scientific-based personalized food recommendations for specific subpopulations.
Collapse
Affiliation(s)
- Hanne Vissenaekens
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium.,Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Hanne Criel
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Charlotte Grootaert
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Katleen Raes
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Guy Smagghe
- Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - John Van Camp
- Department of Food Technology, Safety and Health, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| |
Collapse
|
4
|
Li C, Zhang Y, Liu R, Mai Y. Anagliptin Protected against Hypoxia/Reperfusion-Induced Brain Vascular Endothelial Permeability by Increasing ZO-1. ACS OMEGA 2021; 6:7771-7777. [PMID: 33778288 PMCID: PMC7992143 DOI: 10.1021/acsomega.1c00242] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/22/2021] [Indexed: 05/17/2023]
Abstract
BACKGROUND AND PURPOSE Cerebral ischemia-reperfusion injury is commonly induced during the treatment of ischemic stroke and is reported to be related to the blood-brain barrier destruction and brain vascular endothelial cell dysfunction. Anagliptin is a novel antidiabetic agent recently reported to protect neurons from oxidative stress. In the present study, we aim to investigate the protective property of anagliptin against oxygen-glucose deprivation and reperfusion (OGD/R)-induced injury on endothelial cells and clarify the potential underlying mechanism. METHODS OGD/R modeling was established on bEnd.3 brain endothelial cells. Cell viability was detected using the MTT assay, and the mitochondrial reactive oxygen species (ROS) level was measured using the mitoses red staining assay. The endothelial monolayer permeability was determined using an FITC-dextran permeation assay. The expression levels of NOX-4 and ZO-1 were evaluated using qRT-PCR and Western blot assays. The expressions of MLC-2, p-MLC-2, and myosin light chain kinase (MLCK) were determined using Western blot. RESULTS First, the decreased cell viability, upregulated NOX-4, and elevated mitochondrial ROS level in the endothelial cells induced by OGD/R were reversed by treatment with anagliptin. Second, the enlarged endothelial permeability and the decreased expression level of ZO-1 in the endothelial cells induced by OGD/R were alleviated by anagliptin. Third, the downregulation of ZO-1 and enlarged brain endothelial monolayer permeability induced by OGD/R were ameliorated by an MLCK inhibitor, ML-7. Lastly, the elevated expressions of MLCK and p-MLC-2 induced by OGD/R were suppressed by anagliptin. CONCLUSION Anagliptin protected against hypoxia/reperfusion-induced brain vascular endothelial permeability by increasing the expression ZO-1, mediated by inhibition of the MLCK/MLC-2 signaling pathway.
Collapse
Affiliation(s)
- Chuo Li
- Department
of Neurology, Guangzhou Eighth People’s
Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510440, China
- . Phone/Fax: +86-020-36473145
| | - Yusheng Zhang
- Department
of Neurology and Stroke Center, The First
Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, China
| | - Rongrong Liu
- Department
of Neurology and Stroke Center, The First
Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, China
| | - Yuzhen Mai
- Department
of Neurology, Guangzhou Eighth People’s
Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510440, China
| |
Collapse
|
5
|
Chan YH, Harith HH, Israf DA, Tham CL. Differential Regulation of LPS-Mediated VE-Cadherin Disruption in Human Endothelial Cells and the Underlying Signaling Pathways: A Mini Review. Front Cell Dev Biol 2020; 7:280. [PMID: 31970155 PMCID: PMC6955238 DOI: 10.3389/fcell.2019.00280] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/31/2019] [Indexed: 12/18/2022] Open
Abstract
Endothelial cells lining the inner vascular wall form a monolayer that contributes to the selective permeability of endothelial barrier. This selective permeability is mainly regulated by an endothelium-specific adherens junctional protein, known as vascular endothelial-cadherin (VE-cadherin). In endothelial cells, the adherens junction comprises of VE-cadherin and its associated adhesion molecules such as p120, α-catenin, and β-catenin, in which α-catenin links cytoplasmic tails of VE-cadherin to actin cytoskeleton through β-catenin. Proinflammatory stimuli such as lipopolysaccharide (LPS) are capable of attenuating vascular integrity through the disruption of VE-cadherin adhesion in endothelial cells. To date, numerous studies demonstrated the disruption of adherens junction as a result of phosphorylation-mediated VE-cadherin disruption. However, the outcomes from these studies were inconsistent and non-conclusive as different cell fractions were used to examine the effect of LPS on the disruption of VE-cadherin. By using Western Blot, some studies utilized total protein lysate and reported decreased protein expression while some studies reported unchanged expression. Other studies which used membrane and cytosolic fractions of protein extract demonstrated decreased and increased VE-cadherin expression, respectively. Despite the irregularities, the results of immunofluorescence staining are consistent with the formation of intercellular gap. Besides that, the overall underlying disruptive mechanisms of VE-cadherin remain largely unknown. Therefore, this mini review will focus on different experiment approaches in terms of cell fractions used in different human endothelial cell studies, and relate these differences to the results obtained in Western blot and immunofluorescence staining in order to give some insights into the overall differential regulatory mechanisms of LPS-mediated VE-cadherin disruption and address the discrepancy in VE-cadherin expression.
Collapse
Affiliation(s)
- Yee Han Chan
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Hanis Hazeera Harith
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Daud Ahmad Israf
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Chau Ling Tham
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
6
|
Hu P, Zhao F, Wang J, Zhu W. Lactoferrin attenuates lipopolysaccharide-stimulated inflammatory responses and barrier impairment through the modulation of NF-κB/MAPK/Nrf2 pathways in IPEC-J2 cells. Food Funct 2020; 11:8516-8526. [DOI: 10.1039/d0fo01570a] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Lactoferrin attenuated LPS-induced inflammatory responsesviainhibiting NF-κB/MAPK pathways in IPEC-J2 cells.
Collapse
Affiliation(s)
- Ping Hu
- National Center for International Research on Animal Gut Nutrition
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health
- Laboratory of Gastrointestinal Microbiology
- National Experimental Teaching Demonstration Center of Animal Science
- College of Animal Science and Technology
| | - Fangzhou Zhao
- National Center for International Research on Animal Gut Nutrition
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health
- Laboratory of Gastrointestinal Microbiology
- National Experimental Teaching Demonstration Center of Animal Science
- College of Animal Science and Technology
| | - Jing Wang
- National Center for International Research on Animal Gut Nutrition
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health
- Laboratory of Gastrointestinal Microbiology
- National Experimental Teaching Demonstration Center of Animal Science
- College of Animal Science and Technology
| | - Weiyun Zhu
- National Center for International Research on Animal Gut Nutrition
- Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health
- Laboratory of Gastrointestinal Microbiology
- National Experimental Teaching Demonstration Center of Animal Science
- College of Animal Science and Technology
| |
Collapse
|
7
|
Huang C, Zhang Z, Wang L, Liu J, Gong X, Zhang C. ML-7 attenuates airway inflammation and remodeling via inhibiting the secretion of Th2 cytokines in mice model of asthma. Mol Med Rep 2018; 17:6293-6300. [PMID: 29512725 PMCID: PMC5928606 DOI: 10.3892/mmr.2018.8683] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/01/2017] [Indexed: 01/05/2023] Open
Abstract
Previous studies have indicated that smooth muscle myosin light chain kinase (MLCK) has a prominent role in the regulation of smooth muscle contraction, which tends to be upregulated in asthma. In recent years, numerous studies have reported that MLCK is intimately connected with the immunoregulatory mechanism of T cells. The imbalance of T helper type 1 cells (Th1)/Th2 constitutes the immune-associated pathological basis of chronic asthma. Th2-associated cytokines, including interleukin-4, −5, −13, −25 and −33, are involved in airway inflammation, hyperresponsiveness and remodeling, which leads to a progressive decline in lung function. The purpose of the present study was to verify whether inhibition of bronchial MLCK attenuated the expression Th2-associated cytokines in asthmatic mice, including the above-mentioned ones. Female BALB/c mice were used to establish an ovalbumin (OVA)-induced model of asthma, of which one group was treated with the MLCK inhibitor (5-iodonaphthalene-1-sulfonyl) homopiperazine (ML-7). The inhibitor of MLCK, ML-7 attenuated airway inflammation and remodeling by reducing inflammatory cell infiltration and the secretion of Th2 cytokines in mice model of asthma, which may represent a promising therapeutic strategy for asthma.
Collapse
Affiliation(s)
- Chuanjun Huang
- Department of Respiratory Diseases, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250013, P.R. China
| | - Zewen Zhang
- Department of Medical Imaging and Nuclear Medicine, Shandong University, Jinan, Shandong 250014, P.R. China
| | - Liuxin Wang
- Department of Respiratory Medicine, The First People's Hospital of Jining, Jining, Shandong 272011, P.R. China
| | - Ju Liu
- Department of Medical Research Center, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| | - Xiaodan Gong
- Department of Respiratory Diseases, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| | - Caiqing Zhang
- Department of Respiratory Diseases, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, P.R. China
| |
Collapse
|
8
|
Bourque C, Zhang Y, Fu M, Racine M, Greasley A, Pei Y, Wu L, Wang R, Yang G. H 2S protects lipopolysaccharide-induced inflammation by blocking NFκB transactivation in endothelial cells. Toxicol Appl Pharmacol 2017; 338:20-29. [PMID: 29128401 DOI: 10.1016/j.taap.2017.11.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 10/23/2017] [Accepted: 11/07/2017] [Indexed: 01/26/2023]
Abstract
Hydrogen sulfide (H2S) is a novel gasotransmitter and acts as a multifunctional regulator in various cellular functions. Past studies have demonstrated a significant role of H2S and its generating enzyme cystathionine gamma-lyase (CSE) in the cardiovascular system. Lipopolysaccharide (LPS), a major pathogenic factor, is known to initiate the inflammatory immune response. The cross talk between LPS-induced inflammation and the CSE/H2S system in vascular cells has not yet been elucidated in detail. Here we showed that LPS decreased CSE mRNA and protein expression in human endothelial cells and blocked H2S production in mouse aorta tissues. Transfection of the cells with TLR4-specific siRNA knockdown TLR4 mRNA expression and abolished the inhibitory role of LPS on CSE expression. Higher dose of LPS (100μg/ml) decreased cell viability, which was reversed by exogenously applied H2S at physiologically relevant concentration (30μM). Lower dose of LPS (10μg/ml) had no effect on cell viability, but significantly induced inflammation gene expressions and cytokines secretion and stimulated cell hyper-permeability. H2S treatment prevented LPS-induced inflammation and hyper-permeability. Lower VE-cadherin expression in LPS-incubated cells would contribute to cell hyper-permeability, which was reversed by H2S co-incubation. In addition, H2S treatment blocked LPS-induced NFκB transactivation. We further validated that LPS-induced hyper-permeability was reversed by CSE overexpression but further deteriorated by CRISPR/Cas9-mediated knockout of CSE. In vivo, deficiency of CSE sensitized the mice to LPS-induced inflammation in vascular tissues. Take together, these data suggest that CSE/H2S system protects LPS-induced inflammation and cell hyper-permeability by blocking NFκB transactivation.
Collapse
Affiliation(s)
- Caitlyn Bourque
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Canada; Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada
| | - Yanjie Zhang
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada; School of Life Science, Shanxi University, Taiyuan, China
| | - Ming Fu
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada; School of Human Kinetics, Laurentian University, Sudbury, Canada; Department of Biology, Laurentian University, Sudbury, Canada; Health Science North Research Institute, Sudbury, Canada
| | - Mélanie Racine
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Canada; Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada
| | - Adam Greasley
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Canada; Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada
| | - Yanxi Pei
- School of Life Science, Shanxi University, Taiyuan, China
| | - Lingyun Wu
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada; School of Human Kinetics, Laurentian University, Sudbury, Canada; Health Science North Research Institute, Sudbury, Canada
| | - Rui Wang
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada; Department of Biology, Laurentian University, Sudbury, Canada
| | - Guangdong Yang
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Canada; Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada.
| |
Collapse
|
9
|
Xiong Y, Wang C, Shi L, Wang L, Zhou Z, Chen D, Wang J, Guo H. Myosin Light Chain Kinase: A Potential Target for Treatment of Inflammatory Diseases. Front Pharmacol 2017; 8:292. [PMID: 28588494 PMCID: PMC5440522 DOI: 10.3389/fphar.2017.00292] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 05/08/2017] [Indexed: 01/30/2023] Open
Abstract
Myosin light chain kinase (MLCK) induces contraction of the perijunctional apical actomyosin ring in response to phosphorylation of the myosin light chain. Abnormal expression of MLCK has been observed in respiratory diseases, pancreatitis, cardiovascular diseases, cancer, and inflammatory bowel disease. The signaling pathways involved in MLCK activation and triggering of endothelial barrier dysfunction are discussed in this review. The pharmacological effects of regulating MLCK expression by inhibitors such as ML-9, ML-7, microbial products, naturally occurring products, and microRNAs are also discussed. The influence of MLCK in inflammatory diseases starts with endothelial barrier dysfunction. The effectiveness of anti-MLCK treatment may depend on alleviation of that primary pathological mechanism. This review summarizes evidence for the potential benefits of anti-MLCK agents in the treatment of inflammatory disease and the importance of avoiding treatment-related side effects, as MLCK is widely expressed in many different tissues.
Collapse
Affiliation(s)
- Yongjian Xiong
- Central Laboratory, The First Affiliated Hospital, Dalian Medical UniversityDalian, China
| | - Chenou Wang
- Laboratory Animal Center, Dalian Medical UniversityDalian, China
| | - Liqiang Shi
- Laboratory Animal Center, Dalian Medical UniversityDalian, China
| | - Liang Wang
- Laboratory Animal Center, Dalian Medical UniversityDalian, China
| | - Zijuan Zhou
- Laboratory Animal Center, Dalian Medical UniversityDalian, China
| | - Dapeng Chen
- Laboratory Animal Center, Dalian Medical UniversityDalian, China
| | - Jingyu Wang
- Laboratory Animal Center, Dalian Medical UniversityDalian, China
| | - Huishu Guo
- Central Laboratory, The First Affiliated Hospital, Dalian Medical UniversityDalian, China
| |
Collapse
|
10
|
Key Molecular Mechanisms of Chaiqinchengqi Decoction in Alleviating the Pulmonary Albumin Leakage Caused by Endotoxemia in Severe Acute Pancreatitis Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 2016:3265368. [PMID: 27413385 PMCID: PMC4930819 DOI: 10.1155/2016/3265368] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 05/02/2016] [Accepted: 05/16/2016] [Indexed: 02/05/2023]
Abstract
To reveal the key molecular mechanisms of Chaiqinchengqi decoction (CQCQD) in alleviating the pulmonary albumin leakage caused by endotoxemia in severe acute pancreatitis (SAP) rats. Rats models of SAP endotoxemia-induced acute lung injury were established, the studies in vivo provided the important evidences that the therapy of CQCQD significantly ameliorated the increases in plasma levels of lipopolysaccharide (LPS), sCd14, and Lbp, the elevation of serum amylase level, the enhancements of systemic and pulmonary albumin leakage, and the depravation of airways indicators, thus improving respiratory dysfunction and also pancreatic and pulmonary histopathological changes. According to the analyses of rats pulmonary tissue microarray and protein-protein interaction network, c-Fos, c-Src, and p85α were predicted as the target proteins for CQCQD in alleviating pulmonary albumin leakage. To confirm these predictions, human umbilical vein endothelial cells were employed in in vitro studies, which provide the evidences that (1) LPS-induced paracellular leakage and proinflammatory cytokines release were suppressed by pretreatment with inhibitors of c-Src (PP1) or PI3K (LY294002) or by transfection with siRNAs of c-Fos; (2) fortunately, CQCQD imitated the actions of these selective inhibitions agents to inhibit LPS-induced high expressions of p-Src, p-p85α, and c-Fos, therefore attenuating paracellular leakage and proinflammatory cytokines release.
Collapse
|