1
|
Fu ZP, Ying YG, Wang RY, Wang YQ. Aged gut microbiota promotes arrhythmia susceptibility via oxidative stress. iScience 2024; 27:110888. [PMID: 39381749 PMCID: PMC11460473 DOI: 10.1016/j.isci.2024.110888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/24/2024] [Accepted: 09/03/2024] [Indexed: 10/10/2024] Open
Abstract
Arrhythmias and sudden cardiac death (SCD) impose a significant burden. Their prevalence rises with age and is linked to gut dysbiosis. Our study aimed to determine whether aged gut microbiota affects arrhythmogenesis. Here, we demonstrated that arrhythmia susceptibility in aged mice could be transmitted to young mice using fecal microbiota transplantation (FMT). Mechanistically, increased intestinal reactive oxygen species (ROS) in aged mice reduced ion channel protein expression and promoted arrhythmias. Gut microbiota depletion by an antibiotic cocktail reduced ROS and arrhythmia in aged mice. Interestingly, oxidative stress in heart induced by hydrogen peroxide (H2O2) increased arrhythmia. Moreover, aged gut microbiota could induce oxidative stress in young mice colon by gut microbiota metabolites transplantation. Vitexin could reduce aging and arrhythmia through OLA1-Nrf2 signaling pathway. Overall, our study demonstrated that the gut microbiota of aged mice reduced cardiac ion channel protein expression through systemic oxidative stress, thereby increased the risk of arrhythmias.
Collapse
Affiliation(s)
- Zhi-ping Fu
- Collage of Pharmacology, North China University of Science and Technology, Tangshan 063200, China
| | - Yi-ge Ying
- Collage of Pharmacology, North China University of Science and Technology, Tangshan 063200, China
| | - Rui-yao Wang
- Collage of Pharmacology, North China University of Science and Technology, Tangshan 063200, China
| | - Yu-qing Wang
- Collage of Pharmacology, North China University of Science and Technology, Tangshan 063200, China
| |
Collapse
|
2
|
Gao F, Zhao Y, Zhang B, Xiao C, Sun Z, Gao Y, Dou X. Orientin alleviates ox-LDL-induced oxidative stress, inflammation and apoptosis in human vascular endothelial cells by regulating Sestrin 1 (SESN1)-mediated autophagy. J Mol Histol 2024; 55:109-120. [PMID: 38165567 DOI: 10.1007/s10735-023-10176-z] [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: 07/21/2023] [Accepted: 11/04/2023] [Indexed: 01/04/2024]
Abstract
Endothelial cells are a crucial component of the vessel-tissue wall and exert an important role in atherosclerosis (AS). To explore the role of Orientin in AS, human vascular endothelial cells (HUVECs) were induced by oxidized low-density lipoprotein (ox-LDL) to simulate the vascular endothelial injury during AS. Cell viability was detected by CCK-8 assay. Oxidative stress and inflammation related markers were measured using kits, RT-qPCR or western blot. Besides, cell apoptosis was assessed with TUNEL staining and cell autophagy was evaluated by LC3 immunofluorescent staining. Additionally, western blot was utilized to evaluate the expression of Sestrin 1 (SESN1) and proteins in AMPK/mTOR signaling. Afterwards, SESN1 was silenced to determine the expression of autophagy-related proteins. The further application of autophagy inhibitor 3-methyladenine (3-MA) was used to clarify the regulatory mechanism of Orientin on autophagy. Results showed that the decreased viability of HUVECs caused by ox-LDL induction was elevated by Orientin. Oxidative stress and inflammation were also attenuated after Orientin addition in HUVECs under ox-LDL condition. Moreover, Orientin suppressed apoptosis and induced autophagy of HUVECs stimulated by ox-LDL, accompanied by enhanced level of phospho (p)-AMPK and declined level of p-mTOR. Interestingly, SESN1 level was elevated by Orientin, and SESN1 depletion alleviated autophagy and reduced p-AMPK expression but enhanced p-mTOR expression. The further experiments indicated that SESN1 silencing or 3-MA addition reversed the inhibitory effects of Orientin on the oxidative stress, inflammation and apoptosis of HUVECs. Collectively, Orientin could induce autophagy by activating SESN1 expression, thereby regulating AMPK/mTOR signaling in ox-LDL-induced HUVECs.
Collapse
Affiliation(s)
- Feng Gao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China.
| | - Yongcheng Zhao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| | - Bin Zhang
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| | - Chunwei Xiao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| | - Zhanfa Sun
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| | - Yuan Gao
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| | - Xueyong Dou
- Department of Cardiovascular Surgery, Xuzhou Cancer Hospital, No. 131, Huancheng Road, Xuzhou, 221005, Jiangsu, People's Republic of China
| |
Collapse
|
3
|
Kundu A, Ghosh P, Bishayi B. Vitexin along with verapamil downregulates efflux pump P-glycoprotein in macrophages and potentiate M1 to M2 switching via TLR4-NF-κB-TNFR2 pathway in lipopolysaccharide treated mice. Immunobiology 2024; 229:152767. [PMID: 38103391 DOI: 10.1016/j.imbio.2023.152767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/22/2023] [Accepted: 11/29/2023] [Indexed: 12/19/2023]
Abstract
The lipopolysaccharide, a microbial toxin, is one of the major causative agents of sepsis. P-gp expression and its functions are altered during inflammation. LPS has been known to impair the functions of P-gp, an efflux transporter. But the effect of LPS on P-gp expression in murine peritoneal macrophages is poorly understood. Molecular docking studies reveal that vitexin is a potent substrate and verapamil a potent inhibitor of P-gp. In the present experimental study, the curative potential of vitexin as a fruit component and verapamil treated as a control inhibitor of P-gp was examined in a murine LPS sepsis model. The effects of vitexin and verapamil on P-gp expression in macrophages correlating with changes in macrophage polarization and associated functional responses during LPS induced sepsis were studied. Peritoneal macrophages of LPS (10 mg/kg body weight) challenged mice exhibited elevated levels of H2O2, superoxide, and NO in parallel with lower antioxidant activity. LPS treatment increased P-gp expression through increased TLR4/expression. However, LPS challenged mice treated with vitexin (5 mg/kg body weight) + verapamil (5 mg/kg body weight) showed higher anti-oxidant enzyme activity (SOD, CAT and GRx) resulting in reduced oxidative stress. This combination treatment also elevated TNFR2, concomitant with down-regulation of TLR4, NF-κB and P-gp expression in murine peritoneal macrophages, resulting in a switch from M1 to M2 polarisation of macrophages and reduced inflammatory responses. In conclusion, combined vitexin and verapamil treatment could be used as a promising therapy to regulate P-gp expression and protection against LPS mediated sepsis and inflammatory damages.
Collapse
Affiliation(s)
- Ayantika Kundu
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta 700009, West Bengal, India
| | - Pratiti Ghosh
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta 700009, West Bengal, India.
| | - Biswadev Bishayi
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, 92 APC Road, Calcutta 700009, West Bengal, India.
| |
Collapse
|
4
|
Mustapha M, Mat Taib CN. Beneficial Role of Vitexin in Parkinson's Disease. Malays J Med Sci 2023; 30:8-25. [PMID: 37102042 PMCID: PMC10125247 DOI: 10.21315/mjms2023.30.2.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/05/2021] [Indexed: 04/28/2023] Open
Abstract
Today, Parkinson's disease (PD) is the foremost neurological disorder all across the globe. In the quest for a novel therapeutic agent for PD with a multimodal mechanism of action and relatively better safety profile, natural flavonoids are now receiving greater attention as a potential source of neuroprotection. Vitexin have been shown to exhibit diverse biological benefits in various disease conditions, including PD. It exerts its anti-oxidative property in PD patients by either directly scavenging reactive oxygen species (ROS) or by upregulating the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and enhancing the activities of antioxidant enzymes. Also, vitexin activates the ERK1/1 and phosphatidyl inositol-3 kinase/Akt (PI3K/Akt) pro-survival signalling pathway, which upregulates the release of anti-apoptotic proteins and downregulates the expression of pro-apoptotic proteins. It could be antagonistic to protein misfolding and aggregation. Studies have shown that it can also act as an inhibitor of monoamine oxidase B (MAO-B) enzyme, thereby increasing striatal dopamine levels, and hence, restoring the behavioural deficit in experimental PD models. Such promising pharmacological potential of vitexin could be a game-changer in devising novel therapeutic strategies against PD. This review discusses the chemistry, properties, sources, bioavailability and safety profile of vitexin. The possible molecular mechanisms underlying the neuroprotective action of vitexin in the pathogenesis of PD alongside its therapeutic potential is also discussed.
Collapse
Affiliation(s)
- Musa Mustapha
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
- Department of Human Anatomy, Faculty of Basic Medical Sciences, College of Medical Sciences, Ahmadu Bello University, Zaria, Nigeria
| | - Che Norma Mat Taib
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| |
Collapse
|
5
|
Nyamweya B, Rukshala D, Fernando N, de Silva R, Premawansa S, Handunnetti S. Cardioprotective Effects of Vitex negundo: A Review of Bioactive Extracts and Compounds. J Evid Based Integr Med 2023; 28:2515690X231176622. [PMID: 37279951 DOI: 10.1177/2515690x231176622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023] Open
Abstract
There has been accumulating interest in the application of medicinal plants as alternative medicine to treat various diseases and/or to develop modern medicines. Vitex negundo is one of such medicinal plants that has been of interest to many researchers and has been of use in traditional medicine. V. negundo is found in Sri Lanka, Madagascar, Malaysia, India, China, The Philippines and East Africa. Therapeutic properties of V. negundo have previously been reviewed. Different parts, preparations and bioactive components of V. negundo possess potential protective and therapeutic effects against cardiovascular disease and related conditions as demonstrated in previous studies. We review the present state of scientific knowledge on the potential use of V. negundo and some of its bioactive components in protecting against cardiovascular diseases and related pathologies. Previous studies in animal and non-animal experimental models, although limited in number and vary in design, seem to support the cardioprotective effect of V. negundo and some of its active components. However, there is need for further preclinical and clinical studies to validate the use of V. negundo and its active constituents in protection and treatment of cardiovascular diseases. Additionally, since only a few V. negundo compounds have been evaluated, specific cardioprotective effects or mechanisms and possible side effects of other V. negundo compounds need to be extensively evaluated.
Collapse
Affiliation(s)
- Boniface Nyamweya
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo 03, Sri Lanka
| | - Dilani Rukshala
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo 03, Sri Lanka
| | - Narmada Fernando
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo 03, Sri Lanka
| | - Rajiva de Silva
- Department of Immunology, Medical Research Institute, Colombo 08, Sri Lanka
| | - Sunil Premawansa
- Departments of Zoology and Environment Sciences, University of Colombo, Colombo 03, Sri Lanka
| | - Shiroma Handunnetti
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo 03, Sri Lanka
| |
Collapse
|
6
|
Hu ZY, Yang ZB, Zhang R, Luo XJ, Peng J. The Protective Effect of Vitexin Compound B-1 on Rat Cerebral I/R Injury through a Mechanism Involving Modulation of miR-92b/NOX4 Pathway. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:137-147. [PMID: 35331124 DOI: 10.2174/1871527321666220324115848] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/29/2022] [Accepted: 01/29/2022] [Indexed: 01/01/2023]
Abstract
BACKGROUND Recent studies have uncovered that vitexin compound B-1 (VB-1) can protect neurons against hypoxia/reoxygenation (H/R)-induced oxidative injury through suppressing NOX4 expression. OBJECTIVE The aims of this study are to investigate whether VB-1 can protect the rat brain against ischemia/ reperfusion (I/R) injury and whether its effect on NOX4 expression is related to modulation of certain miRNAs expression. METHODS Rats were subjected to 2 h of cerebral ischemia followed by 24 h of reperfusion to establish an I/R injury model, which showed an increase in neurological deficit score and infarct volume concomitant with an upregulation of NOX4 expression, increase in NOX activity, and downregulation of miR-92b. RESULTS Administration of VB-1 reduced I/R cerebral injury accompanied by a reverse in NOX4 and miR-92b expression. Similar results were achieved in a neuron H/R injury model. Next, we evaluated the association of miR-92b with NOX4 by its mimics in the H/R model. H/R treatment increased neurons apoptosis concomitant with an upregulation of NOX4 and NOX activity while downregulation of miR-92b. All these effects were reversed in the presence of miR-92b mimics, confirming the function of miR-92b in suppressing NOX4 expression. CONCLUSION We conclude the protective effect of VB-1 against rat cerebral I/R injury through a mechanism involving modulation of miR-92b/NOX4 pathway.
Collapse
Affiliation(s)
- Zhong-Yang Hu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
- Department of Neurology, The Third Xiangya Hospital of Central South University, Changsha 410013, China
| | - Zhong-Bao Yang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Ruxu Zhang
- Department of Neurology, The Third Xiangya Hospital of Central South University, Changsha 410013, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha 410013, China
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| |
Collapse
|
7
|
CHENG X, ZHAO C, JIN Z, HU J, ZHANG Z, ZHANG C. Natural products: potential therapeutic agents for atherosclerosis. Chin J Nat Med 2022; 20:830-845. [DOI: 10.1016/s1875-5364(22)60219-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Indexed: 11/24/2022]
|
8
|
Lin GS, Zhang MY, Wu LN, Lin QY. Vitexin Glucolone Reinforces Radiosensitivity of Non-Small Cell Lung Cancer via Transforming Growth Factor Kinase 1/Adenylate Activated Protein Kinase Signaling Pathway. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Purpose: To discuss effects of vitexin glucolone (VG) to radiosensitivity of NSCLC (Non-small cell lung cancer) cell lines (A549 and H1299). Methods: Treating A549 and H1299 cells by VG with or without X-radiation. Cell viability was calculated by CCK8. Apoptosis rate
was measured by flow cytometry and Western blot to expressions of protein. Subsequently, cells were transferred with TAK1 siRNA, cell viability and apoptosis were measured. Results: VG decreased the viability of Huh7 cells and inhibited effects of VG was more strengthen than radiotherapy
in concentrations of 20 μmol/L and 40 μmol/L. Meanwhile, VG sensitized HCC exposed to radiation therapy to apoptosis as demonstrated by increased Bax/Bcl-2 ratio. In addition, VG enhanced the promotive effects of X-radiation on the expressions of TAK1, AMPKα1
and PPARγ. Furthermore, silence the expression of TAK1 partly reversed the effects of VG on HCC and radiosensitivity of NSCLC. Conclusion: VG enhances radiosensitivity of NSCLC via TAK1/AMPK pathway.
Collapse
Affiliation(s)
- Guo Sheng Lin
- Pulmonary and Critical Care Medicine, Affiliated Hospital of Putian University, Putian, 351100, China
| | - Mei Yi Zhang
- Pulmonary and Critical Care Medicine, Affiliated Hospital of Putian University, Putian, 351100, China
| | - Liang Ning Wu
- Pulmonary and Critical Care Medicine, Affiliated Hospital of Putian University, Putian, 351100, China
| | - Qun Ying Lin
- Pulmonary and Critical Care Medicine, Affiliated Hospital of Putian University, Putian, 351100, China
| |
Collapse
|
9
|
Vitexin Mitigates Staphylococcus aureus-Induced Mastitis via Regulation of ROS/ER Stress/NF- κB/MAPK Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7977433. [PMID: 35795861 PMCID: PMC9252844 DOI: 10.1155/2022/7977433] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/02/2022] [Accepted: 06/14/2022] [Indexed: 11/18/2022]
Abstract
Mastitis, caused by a variety of pathogenic microorganisms, seriously threatens the safety and economic benefits of the dairy industry. Vitexin, a flavone glucoside found in many plant species, has been widely reported to have antioxidant, anti-inflammatory, antiviral, anticancer, neuroprotective, and cardioprotective effects. However, few studies have explored the effect of vitexin on mastitis. This study is aimed at exploring whether the antioxidant and anti-inflammatory functions of vitexin can improve Staphylococcus aureus-induced mastitis and its possible molecular mechanism. The expression profiles of S. aureus-infected bovine mammary epithelial cells and gland tissues from the GEO data set (GSE94056 and GSE139612) were analyzed and found that DEGs were mainly involved in immune signaling pathways, apoptosis, and ER stress through GO and KEGG enrichment. Vitexin blocked the production of ROS and increased the activity of antioxidant enzymes (SOD, GSH-PX, and CAT) via activation of PPARγ in vivo and in vitro. In addition, vitexin reduced the production of inflammatory cytokines (TNF-α, IL-1β, and IL-6) and inhibited apoptosis in MAC-T cells and mouse mammary tissues infected with Staphylococcus aureus. Moreover, vitexin decreased the expression of PDI, Ero1-Lα, p-IRE1α, PERK, p-eIF2α, and CHOP protein but increased BiP in both mammary gland cells and tissues challenged by S. aureus. Western blot results also found that the phosphorylation levels of JNK, ERK, p38, and p65 were reduced in vitexin-treated tissues and cells. Vitexin inhibited the production of ROS through promoting PPARγ, increased the activity of antioxidant enzymes, and reduced inflammatory cytokines and apoptosis by alleviating ER stress and inactivation MAPKs and NF-κB signaling pathway. Vitexin maybe have great potential to be a preventive and therapeutic agent for mastitis.
Collapse
|
10
|
Zhou Q, Xu H, Zhao Y, Liu B, Cheng KW, Chen F, Wang M. 6-C-(E-Phenylethenyl)-naringenin, a Styryl Flavonoid, Inhibits Advanced Glycation End Product-Induced Inflammation by Upregulation of Nrf2. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:3842-3851. [PMID: 35297642 DOI: 10.1021/acs.jafc.2c00163] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Styryl flavonoids can be formed during the thermal processing of meats and flavonoid-enriched foods, showing high potentials in the prevention of different diseases. In this study, the protective effects of several styryl flavonoids against advanced glycation end product (AGE)-induced inflammation were evaluated, with 6-C-(E-phenylethenyl)-naringenin (6-PN) showing the strongest activity among them. The results indicated that 6-PN significantly ameliorated AGE-induced damages in human umbilical vein endothelial cells, including inhibition of pro-inflammatory cytokines and reactive oxygen species (ROS) production through downregulating the protein levels of the receptor for AGEs (RAGE) and NADPH oxidase. Notably, 6-PN possessed a much higher bioavailability than its parental compound, naringenin. Furthermore, 6-PN also promoted the activation of the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway that was suppressed by AGEs, and the anti-inflammatory effects of 6-PN disappeared when the cells were treated with ML385, a Nrf2 inhibitor. Hence, 6-PN might inhibit AGE-induced inflammation by the RAGE/ROS/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Qian Zhou
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, P. R. China
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, P. R. China
| | - Hui Xu
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, P. R. China
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, P. R. China
| | - Yueliang Zhao
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, P. R. China
| | - Bin Liu
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, P. R. China
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, P. R. China
| | - Ka-Wing Cheng
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, P. R. China
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, P. R. China
| | - Feng Chen
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, P. R. China
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, P. R. China
| | - Mingfu Wang
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, P. R. China
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, P. R. China
| |
Collapse
|
11
|
Zhang L, Chen D, Tu Y, Sang T, Pan T, Lin H, Cai C, Jin X, Wu F, Xu L, Chen Y. Vitexin attenuates autoimmune hepatitis in mouse induced by syngeneic liver cytosolic proteins via activation of AMPK/AKT/GSK-3β/Nrf2 pathway. Eur J Pharmacol 2022; 917:174720. [PMID: 34953801 DOI: 10.1016/j.ejphar.2021.174720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 12/04/2021] [Accepted: 12/20/2021] [Indexed: 12/18/2022]
Abstract
Autoimmune hepatitis (AIH) is a chronic progressive liver disease that currently does not have a successful therapeutic option. Vitexin, a bioflavonoid isolated from various medicinal plants, possesses a variety of activities; however, whether vitexin protects against AIH remains unclear. Therefore, the current study aims to investigate the hepatoprotective effects and mechanism of action of vitexin in both an experimental autoimmune hepatitis (EAH) mouse model and in D-galactosamine/lipopolysaccharide (D-GalN/LPS)-induced hepatocyte injury. Syngeneic liver antigen S100 was used to establish EAH. Vitexin treatment significantly decreased the infiltration of inflammatory and CD4+ T cells in the liver, reduced ALT and AST levels in the serum and attenuated hepatic injury due to oxidative stress. Moreover, vitexin mitigated the upregulation of Bax and cleaved caspase-3 and the downregulation of Bcl-2 in the livers of AIH mice. These regulations were accompanied by not only increased phosphorylation of AMPK, AKT and GSK-3β but also activation of Nrf2. Furthermore, vitexin inhibited apoptosis and the overexpression of inflammatory cytokines in D-GalN/LPS-treated AML12 cells. In addition, vitexin enhanced the phosphorylation of AMPK, AKT and GSK-3β. When AML12 cells were treated with an inhibitor of AMPK/AKT or specific siRNA targeting Nrf2, vitexin did not further induce the activation of Nrf2/HO-1. A molecular docking study confirmed that vitexin could interact with AMPK through hydrogen bonding interactions. In conclusion, vitexin ameliorated hepatic injury in EAH mice through activation of the AMPK/AKT/GSK-3β pathway and upregulation of the Nrf2 gene.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325006, China
| | - Dazhi Chen
- Department of Gastroenterology, The First Hospital of Peking University, BeiJing, 100032, China
| | - Yulu Tu
- Department of Gastroenterology, Ningbo Hangzhou Bay Hospital, Ningbo, 315336, Zhejiang, China
| | - Tiantian Sang
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, 325006, China
| | - Tongtong Pan
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325006, China
| | - Hongwei Lin
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325006, China
| | - Chao Cai
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325006, China
| | - Xiaozhi Jin
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325006, China
| | - Faling Wu
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325006, China
| | - Lanman Xu
- Department of Infectious Diseases and Liver Diseases, Ningbo Medical Center Lihuili Hospital, Affiliated Hospital of Ningbo University, Ningbo, 315040, Zhejiang, China.
| | - Yongping Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, Wenzhou Key Laboratory of Hepatology, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325006, China.
| |
Collapse
|
12
|
Shikonin Alleviates Endothelial Cell Injury Induced by ox-LDL via AMPK/Nrf2/HO-1 Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:5881321. [PMID: 34912465 PMCID: PMC8668324 DOI: 10.1155/2021/5881321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 11/07/2021] [Accepted: 11/09/2021] [Indexed: 11/18/2022]
Abstract
The present study aimed to explore the effects of shikonin (SKN) on the damage of human venous endothelial cells (HUVECs) induced by ox-LDL and the underlying molecular mechanism. The HUVECs were randomly divided into six groups: control, ox-LDL, SKN + ox-LDL, SKN + ox-LDL + compound C, SKN + ox-LDL + si-Nrf2, and SKN + ox-LDL + si-HO-1. The MTT method was used to detect cell viability, flow cytometry was used to detect cell apoptosis and reactive oxygen species (ROS) levels, and Western blot was used to detect protein levels. Compared to the control group, the cell viability of the ox-LDL group decreased, the apoptosis rate increased, the level of cleaved caspase-3 was upregulated, and the level of Bcl-2 protein was downregulated. The level of TNF-α, IL-1β, IL-6, vascular cell adhesion molecule-1 (VCAM1), intercellular adhesion molecule-1 (ICAM1), and E-selectin (E-sel) was increased, ROS levels increased, and superoxide dismutase (SOD) level decreased. Moreover, the protein levels of p-AMPK, Nrf2, and HO-1 were decreased. Compared to the ox-LDL group, SKN treatment improves cell viability, alleviates cell apoptosis and oxidative stress injury, and upregulates the protein levels of p-AMPK, Nrf2, and HO-1. Compound C, si-Nrf2, and si-HO-1 administration inhibits the AMPK/Nrf2/HO-1 signaling pathway, increases ROS generation, and inhibits the antagonistic effect of SKN on ox-LDL-induced HUVECs damage. In summary, SKN suppressed ox-LDL-induced ROS production and improved cell viability and cell apoptosis via the AMPK/Nrf2/HO-1 pathway.
Collapse
|
13
|
Vitexin inhibits APEX1 to counteract the flow-induced endothelial inflammation. Proc Natl Acad Sci U S A 2021; 118:2115158118. [PMID: 34810252 DOI: 10.1073/pnas.2115158118] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2021] [Indexed: 12/18/2022] Open
Abstract
Vascular endothelial cells are exposed to shear stresses with disturbed vs. laminar flow patterns, which lead to proinflammatory vs. antiinflammatory phenotypes, respectively. Effective treatment against endothelial inflammation and the consequent atherogenesis requires the identification of new therapeutic molecules and the development of drugs targeting these molecules. Using Connectivity Map, we have identified vitexin, a natural flavonoid, as a compound that evokes the gene-expression changes caused by pulsatile shear, which mimics laminar flow with a clear direction, vs. oscillatory shear (OS), which mimics disturbed flow without a clear direction. Treatment with vitexin suppressed the endothelial inflammation induced by OS or tumor necrosis factor-α. Administration of vitexin to mice subjected to carotid partial ligation blocked the disturbed flow-induced endothelial inflammation and neointimal formation. In hyperlipidemic mice, treatment with vitexin ameliorated atherosclerosis. Using SuperPred, we predicted that apurinic/apyrimidinic endonuclease1 (APEX1) may directly interact with vitexin, and we experimentally verified their physical interactions. OS induced APEX1 nuclear translocation, which was inhibited by vitexin. OS promoted the binding of acetyltransferase p300 to APEX1, leading to its acetylation and nuclear translocation. Functionally, knocking down APEX1 with siRNA reversed the OS-induced proinflammatory phenotype, suggesting that APEX1 promotes inflammation by orchestrating the NF-κB pathway. Animal experiments with the partial ligation model indicated that overexpression of APEX1 negated the action of vitexin against endothelial inflammation, and that endothelial-specific deletion of APEX1 ameliorated atherogenesis. We thus propose targeting APEX1 with vitexin as a potential therapeutic strategy to alleviate atherosclerosis.
Collapse
|
14
|
Zhou H, Jiang F, Leng Y. Propofol Ameliorates ox-LDL-Induced Endothelial Damage Through Enhancing Autophagy via PI3K/Akt/m-TOR Pathway: A Novel Therapeutic Strategy in Atherosclerosis. Front Mol Biosci 2021; 8:695336. [PMID: 34250023 PMCID: PMC8267008 DOI: 10.3389/fmolb.2021.695336] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 05/19/2021] [Indexed: 11/18/2022] Open
Abstract
Objective: Atherosclerosis (AS) represents a common age-associated disease, which may be accelerated by oxidized low-density lipoprotein (ox-LDL)-induced endothelial cell injury. This study aimed to investigate the effects of Propofol on ox-LDL-induced endothelial damage and the underlying molecular mechanisms. Methods: Human umbilical vein endothelial cells (HUVECs) were exposed to ox-LDL to induce endothelial damage. HUVECs were pretreated with 0, 5, 25 and 100°μM Propofol, followed by exposure to 100°μg/ml ox-LDL for 24°h. Cell viability was assessed by cell counting kit-8 (CCK-8) assay. The expression of autophagy- and apoptosis-related proteins was detected via western blot. Autophagosome was investigated under a transmission electron microscope. After co-treatment with autophagy inhibitor Bafilomycin A1 or si-Beclin-1, cell apoptosis was detected by flow cytometry. Furthermore, under cotreatment with PI3K activator 740Y-P, PI3K/Akt/m-TOR pathway- and autophagy-related proteins were examined by western blot. Results: With a concentration-dependent manner, Propofol promoted the viability of HUVECs exposed to ox-LDL, and increased LC3-II/I ratio and Beclin-1 expression, and decreased P62 expression. The formation of autophagosome was enhanced by Propofol. Furthermore, Propofol treatment elevated Bcl-2/Bax ratio and lowered Caspase-3 expression. Bafilomycin A1 or si-Beclin-1 distinctly ameliorated the inhibitory effects of Propofol on apoptosis in ox-LDL-exposed HUVECs. Moreover, Propofol lowered the activation of PI3K/Akt/m-TOR pathway in HUVECs under exposure to ox-LDL. However, its inhibitory effects were weakened by 740Y-P. Conclusion: Collectively, this study revealed that Propofol could ameliorate ox-LDL-induced endothelial damage through enhancing autophagy via PI3K/Akt/m-TOR pathway, which might offer a novel therapeutic strategy in AS.
Collapse
Affiliation(s)
- Hongyi Zhou
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China.,Department of Anesthesiology, Tongzhou Maternal and Child Health Hospital of Beijing, Beijing, China
| | - Fan Jiang
- Department of General Medicine, Beijing Luhe Hospital, Capital Medical University, Beijin, China
| | - Yufang Leng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China.,Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
15
|
Impaired Autophagy Induced by oxLDL/ β2GPI/anti- β2GPI Complex through PI3K/AKT/mTOR and eNOS Signaling Pathways Contributes to Endothelial Cell Dysfunction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6662225. [PMID: 34221236 PMCID: PMC8219424 DOI: 10.1155/2021/6662225] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/22/2021] [Accepted: 05/22/2021] [Indexed: 12/12/2022]
Abstract
Endothelial cell dysfunction plays a fundamental role in the pathogenesis of atherosclerosis (AS), and endothelial autophagy has protective effects on the development of AS. Our previous study had shown that oxidized low-density lipoprotein/β2-glycoprotein I/anti-β2-glycoprotein I antibody (oxLDL/β2GPI/anti-β2GPI) complex could promote the expressions of inflammatory cytokines and enhance the adhesion of leukocytes to endothelial cells. In the present study, we aimed to assess the effects of oxLDL/β2GPI/anti-β2GPI complex on endothelial autophagy and explore the associated potential mechanisms. Human umbilical vein endothelial cells (HUVECs) and mouse brain endothelial cell line (bEnd.3) were used as models of the vascular endothelial cells. Autophagy was evaluated by examining the expressions of autophagic proteins using western blotting analysis, autophagosome accumulation using transmission electron microscopy, and RFP-GFP-LC3 adenoviral transfection and autophagic flux using lysosome inhibitor chloroquine. The expressions of phospho-PI3K, phospho-AKT, phospho-mTOR, and phospho-eNOS were determined by western blotting analysis. 3-Methyladenine (3-MA) and rapamycin were used to determine the role of autophagy in oxLDL/β2GPI/anti-β2GPI complex-induced endothelial cell dysfunction. We showed that oxLDL/β2GPI/anti-β2GPI complex suppressed the autophagy, evidenced by an increase in p62 protein, a decrease in LC3-II and Beclin1, and a reduction of autophagosome generation in endothelial cells. Moreover, inhibition of autophagy was associated with PI3K/AKT/mTOR and eNOS signaling pathways. Rapamycin attenuated oxLDL/β2GPI/anti-β2GPI complex-induced endothelial inflammation, oxidative stress, and apoptosis, whereas 3-MA alone induced the endothelial injury. Our results suggested that oxLDL/β2GPI/anti-β2GPI complex inhibited endothelial autophagy via PI3K/AKT/mTOR and eNOS signaling pathways and further contributed to endothelial cell dysfunction. Collectively, our findings provided a novel mechanism for vascular endothelial injury in AS patients with an antiphospholipid syndrome (APS) background.
Collapse
|
16
|
Zhang Q, Fan Z, Xue W, Sun F, Zhu H, Huang D, Wang Z, Dong L. Vitexin regulates Epac and NLRP3 and ameliorates chronic cerebral hypoperfusion injury. Can J Physiol Pharmacol 2021; 99:1079-1087. [PMID: 33915055 DOI: 10.1139/cjpp-2021-0034] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Chronic cerebral hypoperfusion (CCH), as a critical factor of chronic cerebrovascular diseases, has greatly influenced the health of patients with vascular dementia. Vitexin, a flavone C-glycoside (apigenin-8-C-β-D-glucopyranoside) that belongs to the flavone subclass of flavonoids, has been shown to possess antioxidant and anti-ischemic properties; however, the putative protective effects of vitexin on the CCH need further investigation. In the current study, the role of vitexin and its underlying mechanism were investigated with permanent bilateral common carotid artery occlusion (2VO) in rats as well as mouse hippocampal neuronal (HT22) cells with oxygen and glucose deprivation/reoxygenation (OGD/R) injury model. The results demonstrated that vitexin improved cognitive dysfunction as well as alleviated pathological neuronal damage in hematoxylin plus eosin (HE) and TUNEL results. The decreased levels of exchange protein directly activated by cAMP 1 (Epac1), Epac2, Ras-associated protein 1 (Rap1), and phospho-extracellular signal-regulated kinase (p-ERK) were reversed by vitexin in rats with CCH. Furthermore, this study indicated that vitexin alleviated CCH-induced inflammation injuries by reducing the expression of NOD-like receptor 3 (NLRP3), caspase-1, interleukin 1β (IL-1β), IL-6, and cleaved caspase-3. In vitro, vitexin increased the expression of Epac1 and Epac2, decreased the activation of the NLRP3-mediated inflammation, and improved cell viability. Taken together, our findings suggest that vitexin can reduce the degree of the progressing pathological damage in the cortex and hippocampus and inhibit further deterioration of cognitive function in rats with CCH. Epac and NLRP3 can be regulated by vitexin in vivo and in vitro, which provides enlightenment for the protection of CCH injury.
Collapse
Affiliation(s)
- Qilong Zhang
- Department of Pharmacology, Key Laboratory of Anti-Inflammatory and Immunopharmacology of Ministry of Education, Key Laboratory of Chinese Medicine Research and Development of State Administration of Traditional Chinese Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Zhijia Fan
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Wei Xue
- Department of Pharmacology, Key Laboratory of Anti-Inflammatory and Immunopharmacology of Ministry of Education, Key Laboratory of Chinese Medicine Research and Development of State Administration of Traditional Chinese Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Fanfan Sun
- Department of Pharmacology, Key Laboratory of Anti-Inflammatory and Immunopharmacology of Ministry of Education, Key Laboratory of Chinese Medicine Research and Development of State Administration of Traditional Chinese Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Huaqing Zhu
- Laboratory of Molecular Biology and Department of Biochemistry, Anhui Medical University, Hefei, Anhui, China
| | - Dake Huang
- Synthetic Laboratory of School of Basic Medicine Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Zhicheng Wang
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Liuyi Dong
- Department of Pharmacology, Key Laboratory of Anti-Inflammatory and Immunopharmacology of Ministry of Education, Key Laboratory of Chinese Medicine Research and Development of State Administration of Traditional Chinese Medicine, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
17
|
Abdulai IL, Kwofie SK, Gbewonyo WS, Boison D, Puplampu JB, Adinortey MB. Multitargeted Effects of Vitexin and Isovitexin on Diabetes Mellitus and Its Complications. ScientificWorldJournal 2021; 2021:6641128. [PMID: 33935599 PMCID: PMC8055414 DOI: 10.1155/2021/6641128] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 03/19/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Till date, there is no known antidote to cure diabetes mellitus despite the discovery and development of diverse pharmacotherapeutic agents many years ago. Technological advancement in natural product chemistry has led to the isolation of analogs of vitexin and isovitexin found in diverse bioresources. These compounds have been extensively studied to explore their pharmacological relevance in diabetes mellitus. Aim of the Study. The present review was to compile results from in vitro and in vivo studies performed with vitexin and isovitexin derivatives relating to diabetes mellitus and its complications. A systematic online literature query was executed to collect all relevant articles published up to March 2020. RESULTS In this piece, we have collected data and presented it in a one-stop document to support the multitargeted mechanistic actions of vitexin and isovitexin in controlling diabetes mellitus and its complications. CONCLUSION Data collected hint that vitexin and isovitexin work by targeting diverse pathophysiological and metabolic pathways and molecular drug points involved in the clinical manifestations of diabetes mellitus. This is expected to provide a deeper understanding of its actions and also serve as a catapult for clinical trials and application research.
Collapse
Affiliation(s)
- Ibrahim Luru Abdulai
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, P.O. Box LG 54, Legon, Accra, Ghana
| | - Samuel Kojo Kwofie
- West African Centre for Cell Biology of Infectious Pathogens, College of Basic and Applied Sciences, University of Ghana, P.O. Box LG 54, Legon, Accra, Ghana
- Department of Biomedical Engineering, School of Engineering Sciences, College of Basic and Applied Sciences, University of Ghana, P.O. Box LG77, Legon, Accra, Ghana
| | - Winfred Seth Gbewonyo
- Department of Biochemistry, Cell and Molecular Biology, School of Biological Sciences, University of Ghana, Legon, Accra, Ghana
| | - Daniel Boison
- Department of Biochemistry, School of Biological Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Joshua Buer Puplampu
- Department of Biochemistry, School of Biological Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Michael Buenor Adinortey
- Department of Biochemistry, School of Biological Sciences, University of Cape Coast, Cape Coast, Ghana
| |
Collapse
|
18
|
Role of Flavonoids in The Interactions among Obesity, Inflammation, and Autophagy. Pharmaceuticals (Basel) 2020; 13:ph13110342. [PMID: 33114725 PMCID: PMC7692407 DOI: 10.3390/ph13110342] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/19/2022] Open
Abstract
Nowadays, obesity is considered as one of the main concerns for public health worldwide, since it encompasses up to 39% of overweight and 13% obese (WHO) adults. It develops because of the imbalance in the energy intake/expenditure ratio, which leads to excess nutrients and results in dysfunction of adipose tissue. The hypertrophy of adipocytes and the nutrients excess trigger the induction of inflammatory signaling through various pathways, among others, an increase in the expression of pro-inflammatory adipocytokines, and stress of the endoplasmic reticulum (ER). A better understanding of obesity and preventing its complications are beneficial for obese patients on two facets: treating obesity, and treating and preventing the pathologies associated with it. Hitherto, therapeutic itineraries in most cases are based on lifestyle modifications, bariatric surgery, and pharmacotherapy despite none of them have achieved optimal results. Therefore, diet can play an important role in the prevention of adiposity, as well as the associated disorders. Recent results have shown that flavonoids intake have an essential role in protecting against oxidative damage phenomena, and presents biochemical and pharmacological functions beneficial to human health. This review summarizes the current knowledge of the anti-inflammatory actions and autophagic flux of natural flavonoids, and their molecular mechanisms for preventing and/or treating obesity.
Collapse
|
19
|
Wang Y, Rijal B, Xu M, Li Z, An Y, Zhang F, Lu C. Renal denervation improves vascular endothelial dysfunction by inducing autophagy via AMPK/mTOR signaling activation in a rat model of type 2 diabetes mellitus with insulin resistance. Acta Diabetol 2020; 57:1227-1243. [PMID: 32488498 DOI: 10.1007/s00592-020-01532-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 03/31/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Recent clinical and animal studies have shown that renal denervation (RDN) improves insulin sensitivity and endothelial dysfunction. However, the specific mechanism remains incompletely understood. The purpose of this study is to investigate the effects of RDN on endothelial dysfunction of type 2 diabetes mellitus (T2DM) rat models with insulin resistance and to explore the underlying molecular mechanisms. METHODS Male Sprague-Dawley rats were fed with or without high-fat diet allocated in different groups, combined with low-dose streptozotocin which induces a rat model to develop T2DM with insulin resistance. RDN was conducted 1 week after the rat models fully developed T2DM. The animals were sub-divided into four groups randomly: control group (CON, n = 6), diabetic group (T2DM, n = 6), diabetic with sham surgery group (Sham, n = 6) and diabetic with RDN group (RDN, n = 6). Rats in all groups were studied at baseline, both preoperatively and 4 weeks after RDN, respectively. Western blot was used to detect the expression of angiotensin-converting enzyme 2 (ACE2) protein and the expression of autophagy-related proteins Beclin1, LC3 and p62 and autophagy signaling pathway AMPK/mTOR proteins and apoptosis-related protein caspase-3 in the aorta endothelial cells. In addition, the effects of ACE2 on autophagy of human umbilical vein insulin resistance endothelial cell culture in vitro were also studied. RESULTS RDN decreased plasma and renal tissue norepinephrine levels. The Von Willebrand factor level was also decreased, while the plasma level of nitric oxide (NO) was significantly increased after RDN. Compared with the T2DM group and the Sham group, the endothelium-dependent and endothelium-independent diastolic function of the RDN group was improved significantly, the expression of Beclin1, LC3, ACE2 and eNOS proteins was higher, and the level of p62 protein was decreased. Furthermore, we found that RDN can activate the expression of p-AMPK and inhibit the expression of p-mTOR. In cell culture experiment, ACE2 activated p-AMPK and inhibited p-mTOR, thus promoting autophagy. CONCLUSIONS RDN may not only increase the expression of ACE2 in the vascular endothelium, but also can via ACE2 activate p-AMPK and inhibit p-mTOR, thus promoting autophagy and improving endothelial dysfunction.
Collapse
MESH Headings
- AMP-Activated Protein Kinases/metabolism
- Animals
- Autophagy/physiology
- Denervation/methods
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Experimental/surgery
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/physiopathology
- Diabetes Mellitus, Type 2/surgery
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Insulin Resistance
- Kidney/innervation
- Kidney/metabolism
- Kidney/surgery
- Male
- Rats
- Rats, Sprague-Dawley
- Signal Transduction/physiology
- TOR Serine-Threonine Kinases/metabolism
Collapse
Affiliation(s)
- Yong Wang
- First Central Clinical College of Tianjin Medical University, Tianjin, China
- Department of Cardiology, The First People's Hospital of Shangqiu, Shangqiu, Henan, China
| | - Bikash Rijal
- First Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Mengping Xu
- Department of Cardiology, Tianjin First Central Hospital, Tianjin, China
| | - Zhuqing Li
- School of Medicine, Nankai University, Tianjin, China
| | - Yunan An
- First Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Feng Zhang
- Department of Cardiology, Tianjin First Central Hospital, Tianjin, China
| | - Chengzhi Lu
- Department of Cardiology, Tianjin First Central Hospital, Tianjin, China.
| |
Collapse
|
20
|
Ganesan K, Ramkumar KM, Xu B. Vitexin restores pancreatic β-cell function and insulin signaling through Nrf2 and NF-κB signaling pathways. Eur J Pharmacol 2020; 888:173606. [PMID: 32980348 DOI: 10.1016/j.ejphar.2020.173606] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/20/2020] [Accepted: 09/23/2020] [Indexed: 12/22/2022]
Abstract
Chronic hyperglycemia induces pancreatic β-cell dysfunction through several cell signaling pathways. The β-cell loss by apoptosis appears to play a crucial role in the onset and progression of diabetes. This study was aimed to investigate the role of vitexin against high glucose-induced β-cells apoptosis and the underlying mechanisms involved therein. INS-1 cells were pretreated with vitexin (20 and 40 μM) followed by high glucose (33 mM) exposure and the cytotoxicity was assessed by MTT. The effect of vitexin on nuclear factor erythroid 2-related factor 2 (Nrf2) and NF-kB signaling molecules have been studied. Vitexin-mediated stimulation of Nrf2 was assessed. Vitexin protected the cells against high glucose toxicity in a concentration-dependent manner. Vitexin improved insulin signaling as analyzed by the levels of functional proteins in the insulin pathways, viz., insulin receptor (IR), insulin receptor substrate (IRS)-1 and IRS-2, glucose transporter -2, and glucose-stimulated insulin secretion. Vitexin improved the high glucose-induced nuclear transcription factor system by suppressing Rel A, Rel B, P50/p105, and IκB expression resulting in decreased cell apoptosis, further confirmed by the reduction in the percentage of Annexin-V positive cells. Our data suggest that vitexin improves insulin secretion by activating key proteins, including NF-κB and Nrf2 in β-cells regulating apoptosis.
Collapse
Affiliation(s)
- Kumar Ganesan
- Food Science and Technology Program, Beijing Normal University-Hong Kong Baptist University United International College, Zhuhai, 519087, China; Laboratory and Clinical Research Institute for Pain, Department of Anaesthesiology, The University of Hong Kong, Hong Kong, China.
| | - Kunka Mohanram Ramkumar
- Life Science Division, SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur, Tamilnadu, 603 203, India.
| | - Baojun Xu
- Food Science and Technology Program, Beijing Normal University-Hong Kong Baptist University United International College, Zhuhai, 519087, China.
| |
Collapse
|
21
|
Boniface PK, Elizabeth FI. Flavones as a Privileged Scaffold in Drug Discovery: Current Developments. Curr Org Synth 2020; 16:968-1001. [PMID: 31984880 DOI: 10.2174/1570179416666190719125730] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/03/2019] [Accepted: 04/27/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Flavones are one of the main subclasses of flavonoids with diverse pharmacological properties. They have been reported to possess antimalarial, antimicrobial, anti-tuberculosis, anti-allergic, antioxidant, anti-inflammatory activities, among others. OBJECTIVE The present review summarizes the recent information on the pharmacological properties of naturally occurring and synthetic flavones. METHODS Scientific publications referring to natural and synthetic flavones in relation to their biological activities were hand-searched in databases such as SciFinder, PubMed (National Library of Medicine), Science Direct, Wiley, ACS, SciELO, Springer, among others. RESULTS As per the literature, seventy-five natural flavones were predicted as active compounds with reference to their IC50 (<20 µg/mL) in in vitro studies. Also, synthetic flavones were found active against several diseases. CONCLUSION As per the literature, flavones are important sources for the potential treatment of multifactorial diseases. However, efforts toward the development of flavone-based therapeutic agents are still needed. The appearance of new catalysts and chemical transformations is expected to provide avenues for the synthesis of unexplored flavones, leading to the discovery of flavones with new properties and biological activities.
Collapse
Affiliation(s)
- Pone K Boniface
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Ferreira I Elizabeth
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
22
|
Babaei F, Moafizad A, Darvishvand Z, Mirzababaei M, Hosseinzadeh H, Nassiri‐Asl M. Review of the effects of vitexin in oxidative stress-related diseases. Food Sci Nutr 2020; 8:2569-2580. [PMID: 32566174 PMCID: PMC7300089 DOI: 10.1002/fsn3.1567] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/18/2020] [Accepted: 03/22/2020] [Indexed: 12/12/2022] Open
Abstract
Vitexin is an apigenin flavone glycoside found in food and medicinal plants. It has a variety of pharmacological effects, including antioxidant, anti-inflammatory, anticancer, antinociceptive, and neuroprotective effects. This review study summarizes all the protective effects of vitexin as an antioxidant against reactive oxygen species, lipid peroxidation, and other oxidative damages in a variety of oxidative stress-related diseases, including seizure, memory impairment, cerebral ischemia, neurotoxicity, myocardial and respiratory injury, and metabolic dysfunction, with possible molecular and cellular mechanisms. This review describes any activation or inhibition of the signaling pathways that depend on the antioxidant activity of vitexin. More basic research is needed on the antioxidative effects of vitexin in vivo, and carrying out clinical trials for the treatment of oxidative stress-related diseases is also recommended.
Collapse
Affiliation(s)
- Fatemeh Babaei
- Department of Clinical BiochemistrySchool of MedicineStudent Research CommitteeShahid Beheshti University of Medical SciencesTehranIran
| | | | | | - Mohammadreza Mirzababaei
- Department of Clinical BiochemistrySchool of MedicineKermanshah University of Medical SciencesKermanshahIran
| | - Hossein Hosseinzadeh
- Pharmaceutical Research CenterPharmaceutical Technology InstituteMashhad University of Medical SciencesMashhadIran
- Department of Pharmacodynamic and ToxicologySchool of PharmacyPharmaceutical Technology InstituteMashhad University of Medical SciencesMashhadIran
| | - Marjan Nassiri‐Asl
- Department of Pharmacology and Neurobiology Research CenterSchool of MedicineShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
23
|
Absorption, metabolism, and bioactivity of vitexin: recent advances in understanding the efficacy of an important nutraceutical. Crit Rev Food Sci Nutr 2020; 61:1049-1064. [PMID: 32292045 DOI: 10.1080/10408398.2020.1753165] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
vitexin, an apigenin-8-C-glucoside, is widely present in numerous edible and medicinal plants. vitexin possesses a variety of bioactive properties, including antioxidation, anti-inflammation, anti-cancer, neuron-protection, and cardio-protection. Other beneficial health effects, such as fat reduction, glucose metabolism, and hepatoprotection, have also been reported in recent studies. This review briefly discusses the absorption and metabolism of vitexin, as well as its influence on gut microbiota. Recent advances in understanding the pharmacological and biological effects of vitexin are then reviewed. Improved knowledge of the absorption, metabolism, bioactivity, and molecular targets of vitexin is crucial for the better utilization of this emerging nutraceutical as a chemopreventive and chemotherapeutic agent.
Collapse
|
24
|
Mohd Ariff A, Abu Bakar NA, Abd Muid S, Omar E, Ismail NH, Ali AM, Mohd Kasim NA, Mohd Nawawi H. Ficus deltoidea suppresses endothelial activation, inflammation, monocytes adhesion and oxidative stress via NF-κB and eNOS pathways in stimulated human coronary artery endothelial cells. BMC Complement Med Ther 2020; 20:56. [PMID: 32066426 PMCID: PMC7076839 DOI: 10.1186/s12906-020-2844-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 02/06/2020] [Indexed: 12/25/2022] Open
Abstract
Background Ficus deltoidea (FD) has been shown to have antidiabetic, anti-inflammatory, antinociceptive and antioxidant properties. However, its effects on key events in the pathogenesis of atherosclerosis are unknown. Aim To investigate the endothelial activation, inflammation, monocyte-endothelial cell binding and oxidative stress effects of four FD varieties. Methods Human coronary artery endothelial cells (HCAEC) were incubated with different concentrations of aqueous ethanolic extracts of FD var. trengganuensis (FDT), var. kunstleri (FDK), var. deltoidea (FDD) and var. intermedia (FDI), together with LPS. Protein and gene expression of vascular cell adhesion molecule-1 (VCAM-1), intercellular cell adhesion molecule-1 (ICAM-1), endothelial-leukocyte adhesion molecule-1 (E-selectin), interleukin-6 (IL-6), Nuclear factor-κB (NF-κB) p50 and p65 and endothelial nitric oxide synthase (eNOS) were measured using ELISA and QuantiGene plex, respectively. Adhesion of monocyte to HCAEC and formation of reactive oxygen species (ROS) were detected by Rose Bengal staining and 2′-7′-dichlorofluorescein diacetate (DCFH-DA) assay. Results FDK exhibited the highest inhibition of biomarkers in relation to endothelial activation and inflammation, second in reducing monocyte binding (17.3%) compared to other varieties. FDK (25.6%) was also the most potent at decreasing ROS production. Conclusion FD has anti-atherogenic effects, possibly mediated by NF-κB and eNOS pathways; with FDK being the most potent variety. It is potentially beneficial in mitigating atherogenesis.
Collapse
Affiliation(s)
- Amirah Mohd Ariff
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, 47000, Sungai Buloh, Selangor, Malaysia
| | - Nurul Ain Abu Bakar
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, 47000, Sungai Buloh, Selangor, Malaysia
| | - Suhaila Abd Muid
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, 47000, Sungai Buloh, Selangor, Malaysia.,Faculty of Medicine, Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Sungai Buloh, Selangor, Malaysia
| | - Effat Omar
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, 47000, Sungai Buloh, Selangor, Malaysia.,Faculty of Medicine, Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Sungai Buloh, Selangor, Malaysia
| | - Nor Hadiani Ismail
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi MARA (UiTM), Puncak Alam Campus, 42300, Bandar Puncak Alam, Selangor, Malaysia
| | - Abdul Manaf Ali
- Faculty of Bioresources and Food Industry, Universiti Sultan Zainal Abidin (UniSZA), 20300, Kuala Terengganu, Terengganu, Malaysia
| | - Noor Alicezah Mohd Kasim
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, 47000, Sungai Buloh, Selangor, Malaysia.,Faculty of Medicine, Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Sungai Buloh, Selangor, Malaysia
| | - Hapizah Mohd Nawawi
- Institute of Pathology, Laboratory and Forensic Medicine (I-PPerForM), Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, 47000, Sungai Buloh, Selangor, Malaysia. .,Faculty of Medicine, Universiti Teknologi MARA (UiTM), Sungai Buloh Campus, Sungai Buloh, Selangor, Malaysia.
| |
Collapse
|
25
|
Galectin-3 Is a Potential Mediator for Atherosclerosis. J Immunol Res 2020; 2020:5284728. [PMID: 32149158 PMCID: PMC7042544 DOI: 10.1155/2020/5284728] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/13/2020] [Accepted: 02/04/2020] [Indexed: 12/22/2022] Open
Abstract
Atherosclerosis is a multifactorial chronic inflammatory arterial disease forming the pathological basis of many cardiovascular diseases such as coronary heart disease, heart failure, and stroke. Numerous studies have implicated inflammation as a key player in the initiation and progression of atherosclerosis. Galectin-3 (Gal-3) is a 30 kDa β-galactose, highly conserved and widely distributed intracellularly and extracellularly. Gal-3 has been demonstrated in recent years to be a novel inflammatory factor participating in the process of intravascular inflammation, lipid endocytosis, macrophage activation, cellular proliferation, monocyte chemotaxis, and cell adhesion. This review focuses on the role of Gal-3 in atherosclerosis and the mechanism involved and several classical Gal-3 agonists and antagonists in the current studies.
Collapse
|
26
|
Ginsenoside Rb1 Alleviates Oxidative Low-Density Lipoprotein–Induced Vascular Endothelium Senescence via the SIRT1/Beclin-1/Autophagy Axis. J Cardiovasc Pharmacol 2020; 75:155-167. [DOI: 10.1097/fjc.0000000000000775] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
27
|
da Silva LGS, Morelli AP, Pavan ICB, Tavares MR, Pestana NF, Rostagno MA, Simabuco FM, Bezerra RMN. Protective effects of beet (Beta vulgaris) leaves extract against oxidative stress in endothelial cells in vitro. Phytother Res 2020; 34:1385-1396. [PMID: 31989717 DOI: 10.1002/ptr.6612] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 12/04/2019] [Accepted: 12/31/2019] [Indexed: 12/22/2022]
Abstract
Beetroot is an herb used worldwide as a food product, raw material for food industry, ethanol production and source of food coloring. Beet leaves are an unconventional food with antioxidant properties, which might neutralize reactive oxygen species (ROS) induced by oxidized Low-Density Lipoprotein (LDL) present in dyslipidemias. This study aimed to elucidate the effects of beet leaves on the suppression of LDL oxidative processes. Beet leaves extract was produced, characterized, and tested for its antioxidant capacity using endothelial cells in vitro. A model of human umbilical vein endothelial cells was used in various tests, including viability assay, molecular analysis of antioxidant genes, ROS labeling, and macrophage adhesion assay. The extract improved the antioxidative protection of endothelial cells against different agents including oxidized LDL-cholesterol and H2 O2 . It acted on ROS directly due to its high content of natural antioxidants, but also due to the activation and improvement of cellular defenses such as Superoxide dismutase 1, Superoxide dismutase 2, and catalase. The inhibition of LDL-mediated oxidative effects on endothelial cells may turn this unconventional food a functional food with great potential for phytotherapy of atherosclerosis as an adjuvant for medicinal treatments.
Collapse
Affiliation(s)
- Luiz Guilherme Salvino da Silva
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Ana Paula Morelli
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Isadora Carolina Betim Pavan
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Mariana Rosolen Tavares
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Nathalie Fortes Pestana
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Mauricio Ariel Rostagno
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Fernando Moreira Simabuco
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Rosângela Maria Neves Bezerra
- Multidisciplinary Laboratory of Food and Health (LabMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| |
Collapse
|
28
|
Sundar UM, Ugusman A, Chua HK, Latip J, Aminuddin A. Piper sarmentosum Promotes Endothelial Nitric Oxide Production by Reducing Asymmetric Dimethylarginine in Tumor Necrosis Factor-α-Induced Human Umbilical Vein Endothelial Cells. Front Pharmacol 2019; 10:1033. [PMID: 31607906 PMCID: PMC6758593 DOI: 10.3389/fphar.2019.01033] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 08/14/2019] [Indexed: 12/11/2022] Open
Abstract
Asymmetric dimethylarginine (ADMA) is an endogenous inhibitor of endothelial nitric oxide synthase (eNOS). ADMA is degraded by dimethylarginine dimethylaminohydrolase (DDAH). Elevated levels of ADMA lead to reduction in nitric oxide (NO) production, which is linked to endothelial dysfunction and atherosclerosis. Piper sarmentosum is an herb that has shown stimulation on endothelial NO production by increasing both expression and activity of eNOS. Thus, this study determined whether the positive effect of P. sarmentosum on NO production is related to its modulation on the DDAH-ADMA pathway in cultured human umbilical vein endothelial cells (HUVEC) exposed to tumor necrosis factor-α (TNF-α). HUVEC were divided into four groups: control, treatment with 250 µg/ml of aqueous extract of P. sarmentosum leaves (AEPS), treatment with 30 ng/ml of TNF-α, and concomitant treatment with AEPS and TNF-α for 24 h. After treatments, HUVEC were collected to measure DDAH1 messenger RNA (mRNA) expression using quantitative real-time polymerase chain reaction. DDAH1 protein level was measured using enzyme-linked immunosorbent assay (ELISA), and DDAH enzyme activity was measured using colorimetric assay. ADMA concentration was measured using ELISA, and NO level was measured using Griess assay. Compared to control, TNF-α-treated HUVEC showed reduction in DDAH1 mRNA expression (P < 0.05), DDAH1 protein level (P < 0.01), and DDAH activity (P < 0.05). Treatment with AEPS successfully increased DDAH1 mRNA expression (P < 0.05), DDAH1 protein level (P < 0.01), and DDAH activity (P < 0.05) in TNF-α-treated HUVEC. Treatment with TNF-α caused an increase in ADMA level (P < 0.01) and a decrease in endothelial NO production (P < 0.001). Whereas treatment with AEPS was able to reduce ADMA level (P < 0.01) and restore NO (P < 0.001) in TNF-α-treated HUVEC. The results suggested that AEPS promotes endothelial NO production by stimulating DDAH activity and thus reducing ADMA level in TNF-α-treated HUVEC.
Collapse
Affiliation(s)
- Uma Mahgesswary Sundar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Azizah Ugusman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Hui Kien Chua
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Jalifah Latip
- Department of Pharmaceutical Chemistry, School of Chemical Sciences & Food Technology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Malaysia
| | - Amilia Aminuddin
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| |
Collapse
|
29
|
Zhang X, Han X, Zhang P, Zhou T, Chen Y, Jin J, Ma X. Morin attenuates oxidized low-density lipoprotein-mediated injury by inducing autophagy via activating AMPK signalling in HUVECs. Clin Exp Pharmacol Physiol 2019; 46:1053-1060. [PMID: 31407376 DOI: 10.1111/1440-1681.13160] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 07/25/2019] [Accepted: 08/06/2019] [Indexed: 01/07/2023]
Abstract
Endothelial dysfunction is a precursor of cardiovascular disease, and oxidized low-density lipoprotein (ox-LDL) has been implicated in the development of atherosclerosis by directly targeting endothelial cells. Morin, a natural flavonol, has been shown to protect endothelial cells from dysfunction. The present study was designed to evaluate the effect of morin on ox-LDL-induced injury and to investigate the underlying molecular mechanisms in human umbilical vein endothelial cells (HUVECs). The results showed that morin alleviated ox-LDL-induced endothelial injury and promoted the viability of HUVECs exposed to ox-LDL. Morin significantly inhibited the oxidative stress induced by ox-LDL by inhibiting the production of reactive oxygen species and malondialdehyde, and downregulating the level of superoxide dismutase. Moreover, morin markedly attenuated the overexpressed mRNA levels of the inflammatory factors interleukin (IL)-1β, IL-6, and the adhesion molecules ICAM-1 and VCAM-1 induced by exposure to ox-LDL. We found that morin attenuated ox-LDL-induced injury in HUVECs by inducing autophagy. The protective effects of morin against ox-LDL-induced injury were dramatically reversed by chloroquine phosphate (CQ) treatment. Furthermore, morin up-regulated the expression of p-AMPK and down-regulated the level of p-mTOR in HUVECs exposed to ox-LDL, and this was significantly reversed by the AMPK inhibitor Compound C (CC). Taken together, our results demonstrated that morin attenuates ox-LDL-mediated injury by inducing autophagy via activating AMPK signalling in HUVECs.
Collapse
Affiliation(s)
- Xiaodong Zhang
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, China
| | - Xiping Han
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, China
| | - Peng Zhang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Tingting Zhou
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yun Chen
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, China
| | - Jian Jin
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, China
| | - Xin Ma
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| |
Collapse
|
30
|
Inamdar S, Joshi A, Malik S, Boppana R, Ghaskadbi S. Vitexin alleviates non-alcoholic fatty liver disease by activating AMPK in high fat diet fed mice. Biochem Biophys Res Commun 2019; 519:106-112. [PMID: 31472955 DOI: 10.1016/j.bbrc.2019.08.139] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 08/25/2019] [Indexed: 01/14/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a most common liver disorder characterized by accumulation of fat in the liver and currently there is no approved treatment for it. Obesity and diabetes being leading cause of NAFLD, compounds having anti-obesity activity and potential to reduce insulin resistance are considered suitable candidate for NAFLD treatment. In this study, we checked effect of vitexin, a naturally occurring flavonoid, on high fat diet (HFD) induced NAFLD in C57BL/6J mice. In presence of vitexin, significant reduction in body and liver weight, triglyceride and cholesterol content in serum and liver was observed. Serum Alanine aminotransferase (ALT) and Aspartate aminotransferase (AST) levels were reduced significantly by vitexin which were elevated in HFD group whereas serum lipase activity remained unchanged. Vitexin suppressed de novo lipogenesis by downregulating expression of Peroxisome proliferator-activated receptor γ (PPARγ), CCAAT/enhancer-binding protein-α (C/EBP-α), sterol regulatory element-binding protein-1c (SREBP-1c), Fatty acid synthase (FAS) and Acetyl-CoA Carboxylase (ACC). Additionally, it also enhanced fatty acid oxidation and lipolysis by upregulating Peroxisome proliferator-activated receptor α (PPAR-α), carnitine palmitoyltransferase-1a (CPT-1a) and Adipose triglyceride lipase (ATGL). Inhibition of lipogenesis and activation of lipolysis and fatty acid oxidation by vitexin was found to be mediated by activation of AMP-activated protein kinase (AMPK). Vitexin also improved insulin signalling by activating insulin receptor substrate-1 (IRS-1) and its downstream target AKT. AMPK activation of vitexin was possibly through binding of vitexin to leptin receptor (LepR) which was confirmed by molecular docking studies and by observed enhanced expression of LepR. Thus, we propose that vitexin alleviates NAFLD by activating AMPK possibly by binding to LepR.
Collapse
Affiliation(s)
- Shrirang Inamdar
- Department of Zoology, Savitribai Phule Pune University, Pune, 411007, India
| | - Ankita Joshi
- Department of Zoology, Savitribai Phule Pune University, Pune, 411007, India
| | - Sajad Malik
- Department of Zoology, Savitribai Phule Pune University, Pune, 411007, India
| | | | - Saroj Ghaskadbi
- Department of Zoology, Savitribai Phule Pune University, Pune, 411007, India.
| |
Collapse
|
31
|
Meng N, Gong Y, Mu X, Wang YH, Su L, Jiang CS, Zhang H. Novel Role of Heterogeneous Nuclear Ribonucleoprotein E1 in Regulation of Apoptosis and Autophagy by a Triazole Derivative in Vascular Endothelial Cells. Int J Biol Sci 2019; 15:1299-1309. [PMID: 31223288 PMCID: PMC6567801 DOI: 10.7150/ijbs.32677] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 04/04/2019] [Indexed: 11/08/2022] Open
Abstract
Vascular endothelial cell (VEC) apoptosis and autophagy play an important role in the maintenance of vascular homeostasis. However, the association of molecular mechanisms between vascular endothelial cell apoptosis and autophagy has not been clarified. Here, we identified a novel triazole derivative, JL014, which could inhibit human umbilical vein vascular endothelial cell (HUVEC) apoptosis induced by deprivation of serum and fibroblast growth factor 2 and maintain HUVEC survival by promoting autophagy. Importantly, JL014 increased the mRNA and protein level of heterogeneous nuclear ribonucleoprotein E1 (hnRNP E1) in HUVECs. In addition, knockdown of hnRNP E1 by RNA interference inhibited the effects of JL014 on VEC apoptosis and autophagy. Furthermore, we investigated the effect of JL014 on the expression of HMBOX1, a key VEC apoptosis inhibitor and autophagy inducer by inhibiting mTOR signaling and the level of cleaved caspase-3. Our results demonstrated that JL014 enhanced mRNA transcription and increased protein synthesis of HMBOX1. JL014 also inhibited mTOR signaling and the cleaved caspase-3 level. Mechanistic studies revealed that hnRNP E1 could bind to the promoter and 5'UTR of HMBOX1 and active HMBOX1 expression. Therefore, our results firmly establish hnRNP E1 as a new regulator of VEC apoptosis and autophagy through mediating HMBOX1 expression, and opened the door to a novel therapeutic drug for related vascular diseases.
Collapse
Affiliation(s)
- Ning Meng
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Yan Gong
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Xin Mu
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Yan Hong Wang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Le Su
- State Key Laboratory of Biobased Material and Green Papermaking, School of Bioengineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan, 250353, China
| | - Cheng Shi Jiang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Hua Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| |
Collapse
|
32
|
Lim H, Heo MY, Kim HP. Flavonoids: Broad Spectrum Agents on Chronic Inflammation. Biomol Ther (Seoul) 2019; 27:241-253. [PMID: 31006180 PMCID: PMC6513185 DOI: 10.4062/biomolther.2019.034] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/02/2019] [Accepted: 04/04/2019] [Indexed: 12/17/2022] Open
Abstract
Flavonoids are major plant constituents with numerous biological/pharmacological actions both in vitro and in vivo. Of these actions, their anti-inflammatory action is prominent. They can regulate transcription of many proinflammatory genes such as cyclooxygenase-2/inducible nitric oxide synthase and many cytokines/chemokines. Recent studies have demonstrated that certain flavonoid derivatives can affect pathways of inflammasome activation and autophagy. Certain flavonoids can also accelerate the resolution phase of inflammation, leading to avoiding chronic inflammatory stimuli. All these pharmacological actions with newly emerging activities render flavonoids to be potential therapeutics for chronic inflammatory disorders including arthritic inflammation, meta-inflammation, and inflammaging. Recent findings of flavonoids are summarized and future perspectives are presented in this review.
Collapse
Affiliation(s)
- Hyun Lim
- College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Moon Young Heo
- College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Hyun Pyo Kim
- College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
33
|
Fang M, Liu S, Wang Q, Gu X, Ding P, Wang W, Ding Y, Liu J, Wang R. Qualitative and Quantitative Analysis of 24 Components in Jinlianhua Decoction by UPLC-MS/MS. Chromatographia 2019; 82:1801-1825. [PMID: 32214429 PMCID: PMC7088050 DOI: 10.1007/s10337-019-03806-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 09/06/2019] [Accepted: 09/09/2019] [Indexed: 01/30/2023]
Abstract
Jinlianhua Decoction (JD), composed of Flos Trollii, Herba Taraxaci, Folium Isatidis, Radix Puerariae Lobatae, and Folium Perillae in a ratio of 6:15:10:10:6, is a prescription for Fengwen which is a group of febrile diseases due to wind in Chinese medicine. It was originally used for the prevention and treatment of severe acute respiratory syndrome (SARS), and could also be used to treat influenza due to their common pathomechanism. To elucidate the unclear pharmacodynamic basis of JD, the LC-QExactive-MS system was used to qualitatively analyze its main components in this study. As a result, 89 compounds were identified and 24 important ones were selected thereby to further perform the simultaneous quantification in 8 batches of JD samples using LC-QTrap-MS with multiple reaction monitoring (MRM). Based on the qualitative and quantitative results in combination with the bioactivities reported, 16 compounds including orientin, 2″-O-β-l-galactopyranosylorientin, puerarin, trollisin I, rosmarinic acid, 2″-O-(2′″-methylbutanoyl) isoswertisin, daidzin, scutellarin, 3′-methoxy puerarin, vitexin, 3′-hydroxy puerarin, 2″-O-(2′″-methylbutanoyl) vitexin, kaempferol, caffeic acid, 3,4-dimethoxybenzoic acid, and cynaroside were determined as the major components of JD. This study provides a useful combinational method for analyzing the major pharmacodynamic substances of JD and lays a foundation for the quality control research of the decoction.
Collapse
Affiliation(s)
- Mingyue Fang
- 1School of Life Sciences, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan District, Beijing, 102488 China
| | - Shuangyue Liu
- 1School of Life Sciences, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan District, Beijing, 102488 China
| | - Qingqing Wang
- 1School of Life Sciences, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan District, Beijing, 102488 China
| | - Xuan Gu
- 1School of Life Sciences, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan District, Beijing, 102488 China
| | - Pengmin Ding
- 1School of Life Sciences, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan District, Beijing, 102488 China
| | - Weihua Wang
- 2Drug Discovery Facility, Tsinghua University, Zhongguancun North Street, Haidian District, Beijing, 100084 China
| | - Yi Ding
- 2Drug Discovery Facility, Tsinghua University, Zhongguancun North Street, Haidian District, Beijing, 100084 China
| | - Junxiu Liu
- 3Department of Otorhinolaryngology, Peking University Third Hospital, No. 49, Huayuan North Road, Haidian District, Beijing, 100191 China
| | - Rufeng Wang
- 1School of Life Sciences, Beijing University of Chinese Medicine, Yangguang South Street, Fangshan District, Beijing, 102488 China
| |
Collapse
|
34
|
Chen X, Lin J, Hu T, Ren Z, Li L, Hameed I, Zhang X, Men C, Guo Y, Xu D, Zhan Y. Galectin-3 exacerbates ox-LDL-mediated endothelial injury by inducing inflammation via integrin β1-RhoA-JNK signaling activation. J Cell Physiol 2018; 234:10990-11000. [PMID: 30536538 PMCID: PMC6590151 DOI: 10.1002/jcp.27910] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 10/25/2018] [Indexed: 12/30/2022]
Abstract
Oxidized low‐density lipoprotein (Ox‐LDL)‐induced endothelial cell injury plays a crucial role in the pathogenesis of atherosclerosis (AS). Plasma galectin‐3 (Gal‐3) is elevated inside and drives diverse systemic inflammatory disorders, including cardiovascular diseases. However, the exact role of Gal‐3 in ox‐LDL‐mediated endothelial injury remains unclear. This study explores the effects of Gal‐3 on ox‐LDL‐induced endothelial dysfunction and the underlying molecular mechanisms. In this study, Gal‐3, integrin β1, and GTP‐RhoA in the blood and plaques of AS patients were examined by ELISA and western blot respectively. Their levels were found to be obviously upregulated compared with non‐AS control group. CCK8 assay and flow cytometry analysis showed that Gal‐3 significantly decreased cell viability and promoted apoptosis in ox‐LDL‐treated human umbilical vascular endothelial cells (HUVECs). The upregulation of integrinβ1, GTP‐RhoA, p‐JNK, p‐p65, p‐IKKα, and p‐IKKβ induced by ox‐LDL was further enhanced by treatment with Gal‐3. Pretreatment with Gal‐3 increased expression of inflammatory factors (interleukin [IL]‐6, IL‐8, and IL‐1β), chemokines(CXCL‐1 and CCL‐2) and adhesion molecules (VCAM‐1 and ICAM‐1). Furthermore, the promotional effects of Gal‐3 on NF‐κB activation and inflammatory factors in ox‐LDL‐treated HUVECs were reversed by the treatments with integrinβ1‐siRNA or the JNK inhibitor. We also found that integrinβ1‐siRNA decreased the protein expression of GTP‐RhoA and p‐JNK, while RhoA inhibitor partially reduced the upregulated expression of p‐JNK induced by Gal‐3. In conclusion, our finding suggests that Gal‐3 exacerbates ox‐LDL‐mediated endothelial injury by inducing inflammation via integrin β1‐RhoA‐JNK signaling activation.
Collapse
Affiliation(s)
- Xiumei Chen
- Department of Geriatric Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jianzhong Lin
- Department of Urology and Central Laboratory, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Tingting Hu
- Department of Cancer Research, The First Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Zhiyun Ren
- Department of Urology and Central Laboratory, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, China
| | - Linnan Li
- Department of Cancer Research, Academy of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Irbaz Hameed
- Department of Cardiothoracic Surgery, New York Presbyterian Hospital Weill cornell Medicine, New York, New York
| | - Xiaoyu Zhang
- Department of Geriatric Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chen Men
- Department of Geriatric Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yan Guo
- Department of Geriatric Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Di Xu
- Department of Geriatric Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yiyang Zhan
- Department of Geriatric Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
35
|
Che Ahmad Tantowi NA, Lau SF, Mohamed S. Ficus deltoidea Prevented Bone Loss in Preclinical Osteoporosis/Osteoarthritis Model by Suppressing Inflammation. Calcif Tissue Int 2018; 103:388-399. [PMID: 29808374 DOI: 10.1007/s00223-018-0433-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/15/2018] [Indexed: 12/27/2022]
Abstract
Osteoporosis (OP) and osteoarthritis (OA) are debilitating musculoskeletal diseases of the elderly. Ficus deltoidea (FD) or mistletoe fig, a medicinal plant, was pre-clinically evaluated against OP- and OA-related bone alterations, in postmenopausal OA rat model. Thirty twelfth-week-old female rats were divided into groups (n = 6). Four groups were bilateral ovariectomized (OVX) and OA-induced by intra-articular monosodium iodoacetate (MIA) injection into the right knee joints. The Sham control and OVX-OA non-treated groups were given deionized water. The three other OVX-OA groups were orally administered daily with FD extract (200, 400 mg/kg) or diclofenac (5 mg/kg) for 4 weeks. The rats' bones and blood were evaluated for protein and mRNA expressions of osteoporosis and inflammatory indicators, and micro-CT computed tomography for bone microstructure. The non-treated OVX-OA rats developed severe OP bone loss and bone microstructural damage in the subchondral and metaphyseal regions, supported by reduced serum bone formation markers (osteocalcin, osteoprotegerin) and increased bone resorption markers (RANKL and CTX-I). The FD extract significantly (p < 0.05) mitigated these bone microstructural and biomarker changes by dose-dependently down-regulating pro-inflammatory NF-κβ, TNF-α, and IL-6 mRNA expressions. The FD extract demonstrated good anti-osteoporotic properties in this OP/OA preclinical model by stimulating bone formation and suppressing bone resorption via anti-inflammatory pathways. This is among the few reports relating the subchondral bone plate and trabecular thickening with the metaphyseal trabecular osteopenic bone loss under osteoporotic-osteoarthritis conditions, providing some insights on the debated inverse relationship between osteoporosis and osteoarthritis.
Collapse
Affiliation(s)
- Nur Adeelah Che Ahmad Tantowi
- UPM-MAKNA Laboratory of Cancer Research, Institute of Bioscience, Universiti Putra Malaysia, UPM, 43400, Serdang, Selangor, Malaysia
| | - Seng Fong Lau
- Faculty of Veterinary Medicine, Universiti Putra Malaysia, UPM, Serdang, Malaysia
| | - Suhaila Mohamed
- UPM-MAKNA Laboratory of Cancer Research, Institute of Bioscience, Universiti Putra Malaysia, UPM, 43400, Serdang, Selangor, Malaysia.
| |
Collapse
|
36
|
Vitexin attenuates lipopolysaccharide-induced acute lung injury by controlling the Nrf2 pathway. PLoS One 2018; 13:e0196405. [PMID: 29694408 PMCID: PMC5942793 DOI: 10.1371/journal.pone.0196405] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 04/12/2018] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND A major feature of acute lung injury (ALI) is excessive inflammation in the lung. Vitexin is an active component from medicinal plants which has antioxidant and anti-inflammatory activities. Oxidative stress and inflammation play important roles in the pathophysiological processes in ALI. In the current study, we investigate the effect and potential mechanisms of Vitexin on lipopolysaccharide (LPS)-induced ALI. METHODS ALI was induced by LPS intratracheal instillation in C57BL/6 wild-type mice and Nrf2 gene knocked down (Nrf2-/-) mice. One hour before LPS challenge, Vitexin or vehicle intraperitoneal injection was performed. Bronchoalveolar lavage fluid and lung tissues were examined for lung inflammation and injury at 24 h after LPS challenge. RESULTS Our animal study's results showed that LPS-induced recruitment of neutrophils and elevation of proinflammatory cytokine levels were attenuated by Vitexin treatment. Vitexin decreased lung edema and alveolar protein content. Moreover, Vitexin activated nuclear factor erythroid-2-related factor 2 (Nrf2), and increased the activity of its target gene heme oxygenase (HO)-1. The LPS-induced reactive oxygen species were inhibited by Vitexin. In addition, the activation of the nucleotide-binding domain and leucine-rich repeat PYD-containing protein 3 (NLRP3) inflammasome was suppressed by Vitexin. However, these effects of Vitexin were abolished in the Nrf2-/- mice. Our cell studies showed that Vitexin enhanced the expression of Nrf2 and HO-1 activity. Moreover, reactive oxygen species (ROS) and IL-1β productions were reduced in Vitexin-treated cells. However, knockdown of Nrf2 by siRNA in RAW cells reversed the benefit of Vitexin. CONCLUSIONS Vitexin suppresses LPS-induced ALI by controlling Nrf2 pathway.
Collapse
|
37
|
Shi D, Niu P, Heng X, Chen L, Zhu Y, Zhou J. Autophagy induced by cardamonin is associated with mTORC1 inhibition in SKOV3 cells. Pharmacol Rep 2018; 70:908-916. [PMID: 30099297 DOI: 10.1016/j.pharep.2018.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 03/22/2018] [Accepted: 04/12/2018] [Indexed: 01/06/2023]
Abstract
BACKGROUND The mammalian target of rapamycin (mTOR) integrates energy level to modulate cell proliferation and autophagy. Cardamonin exhibits anti-proliferative activity through inhibiting mTOR. In this study, the effect of cardamonin on autophagy and its mechanism on mTOR inhibition were investigated. METHODS Cell viability and proliferation were measured by MTT assay and BrdU incorporation, respectively. Cell apoptosis was assayed by flow cytometry and cell autophagy was detected by electron microscopy and GFP-LC3 fluorescence. The mechanism of cardamonin on mTORC1 inhibition was investigated by Raptor siRNA and Raptor over-expression. RESULTS The cell viability and proliferation were inhibited by cardamonin. The autophagosomes and the protein level of LC3-II were increased by cardamonin. Cell apoptosis and the levels of cleaved PARP and Caspase-3 were increased by cardamonin. Cardamonin inhibited the phosphorylation of mTOR and ribosome S6 protein kinase 1 (S6K1) as well as the protein level of regulatory associated protein of mTOR (Raptor). However, cardamonin had no effect on the component of mTORC2 and its downstream substrate Akt. The inhibitory effect of cardamonin on the phosphorylation of mTOR and S6K1 was eliminated by Raptor knockdown with siRNA, whereas this effect of cardamonin was stronger than that of rapamycin and AZD8055 in Raptor over-expression cells. Cell viability was inhibited by cardamonin in both Raptor knockdown and Raptor over-expression cells, which was consistent with the inhibitory effect of cardamonin on mTOR. CONCLUSION These findings demonstrated that the autophagy induced by cardamonin was associated with mTORC1 inhibition through decreasing the protein level of Raptor in SKOV3 cells.
Collapse
Affiliation(s)
- Daohua Shi
- Department of Pharmacy, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China.
| | - Peiguang Niu
- Department of Pharmacy, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Xiaojie Heng
- Department of Pharmacy, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Lijun Chen
- Department of Pharmacy, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Yanting Zhu
- Department of Pharmacy, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Jintuo Zhou
- Department of Pharmacy, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
38
|
Shah SZA, Zhao D, Hussain T, Yang L. Role of the AMPK pathway in promoting autophagic flux via modulating mitochondrial dynamics in neurodegenerative diseases: Insight into prion diseases. Ageing Res Rev 2017; 40:51-63. [PMID: 28903070 DOI: 10.1016/j.arr.2017.09.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Revised: 09/06/2017] [Accepted: 09/07/2017] [Indexed: 12/15/2022]
Abstract
Neurons are highly energy demanding cells dependent on the mitochondrial oxidative phosphorylation system. Mitochondria generate energy via respiratory complexes that constitute the electron transport chain. Adenosine triphosphate depletion or glucose starvation act as a trigger for the activation of adenosine monophosphate-activated protein kinase (AMPK). AMPK is an evolutionarily conserved protein that plays an important role in cell survival and organismal longevity through modulation of energy homeostasis and autophagy. Several studies suggest that AMPK activation may improve energy metabolism and protein clearance in the brains of patients with vascular injury or neurodegenerative disease. Mild mitochondrial dysfunction leads to activated AMPK signaling, but severe endoplasmic reticulum stress and mitochondrial dysfunction may lead to a shift from autophagy towards apoptosis and perturbed AMPK signaling. Hence, controlling mitochondrial dynamics and autophagic flux via AMPK activation might be a useful therapeutic strategy in neurodegenerative diseases to reinstate energy homeostasis and degrade misfolded proteins. In this review article, we discuss briefly the role of AMPK signaling in energy homeostasis, the structure of AMPK, activation mechanisms of AMPK, regulation of AMPK, the role of AMPK in autophagy, the role of AMPK in neurodegenerative diseases, and finally the role of autophagic flux in prion diseases.
Collapse
Affiliation(s)
- Syed Zahid Ali Shah
- National Animal Transmissible Spongiform Encephalopathy Laboratory and Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing 100193, China
| | - Deming Zhao
- National Animal Transmissible Spongiform Encephalopathy Laboratory and Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing 100193, China
| | - Tariq Hussain
- National Animal Transmissible Spongiform Encephalopathy Laboratory and Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing 100193, China
| | - Lifeng Yang
- National Animal Transmissible Spongiform Encephalopathy Laboratory and Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
39
|
Yan F, Yang Y, Yu L, Zheng X. Effects of C-Glycosides from Apios americana Leaves against Oxidative Stress during Hyperglycemia through Regulating Mitogen-Activated Protein Kinases and Nuclear Factor Erythroid 2-Related Factor 2. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:7457-7466. [PMID: 28758742 DOI: 10.1021/acs.jafc.7b03163] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Main components of Apios americana leaves extract (ALE) were flavonoid C-glycosides, including vitexin (46.7%), schaftoside (18.9%), and orientin (4.32%). In vitro, ALE restored glucose consumption, glucose uptake, and glycogen content in glucose-induced hepatic cells. Exposure of HepG2 cells to high glucose resulted in reactive oxygen species and O2- accumulation, while ALE alleviated these increases by 47 ± 0.68 and 68 ± 0.74%, respectively. Glucose increased c-Jun N-terminal kinase (JNK) and decreased extracellular signal-regulated kinases 1 and 2 (ERK1/2) and p38 phosphorylation, while ALE reduced p-JNK and p-p38 but not p-ERK1/2, accompanied by nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1, and NAD(P)H quinine oxidoreductase 1 downregulation. In vivo, the lifespan of Caenorhabditis elegans was more violently shortened by paraquat under hyperglycemia, while ALE protected this damage in N2 worms (2.6 times extension) but not in daf-16 mutants. Furthermore, p38/PMK-1 and Nrf2/SKN-1 expressions in worms were suppressed by glucose, which were reversed by ALE treatment. These results suggest that ALE prevents glucose-induced damage via regulating specific mitogen-activated protein kinases and Nrf2 pathways.
Collapse
Affiliation(s)
- Fujie Yan
- Department of Food Science and Nutrition, ‡Zhejiang Key Laboratory for Agro-food Processing, and §Fuli Institute of Food Science, Zhejiang University , Hangzhou, Zhejiang 310058, People's Republic of China
| | - Yunyun Yang
- Department of Food Science and Nutrition, ‡Zhejiang Key Laboratory for Agro-food Processing, and §Fuli Institute of Food Science, Zhejiang University , Hangzhou, Zhejiang 310058, People's Republic of China
| | - Lushuang Yu
- Department of Food Science and Nutrition, ‡Zhejiang Key Laboratory for Agro-food Processing, and §Fuli Institute of Food Science, Zhejiang University , Hangzhou, Zhejiang 310058, People's Republic of China
| | - Xiaodong Zheng
- Department of Food Science and Nutrition, ‡Zhejiang Key Laboratory for Agro-food Processing, and §Fuli Institute of Food Science, Zhejiang University , Hangzhou, Zhejiang 310058, People's Republic of China
| |
Collapse
|
40
|
Dai X, Zeng J, Yan X, Lin Q, Wang K, Chen J, Shen F, Gu X, Wang Y, Chen J, Pan K, Cai L, Wintergerst KA, Tan Y. Sitagliptin-mediated preservation of endothelial progenitor cell function via augmenting autophagy enhances ischaemic angiogenesis in diabetes. J Cell Mol Med 2017; 22:89-100. [PMID: 28799229 PMCID: PMC5742710 DOI: 10.1111/jcmm.13296] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 05/27/2017] [Indexed: 01/13/2023] Open
Abstract
Recently, the dipeptidyl peptidase‐4 (DPP‐4) inhibitor sitagliptin, a major anti‐hyperglycaemic agent, has received substantial attention as a therapeutic target for cardiovascular diseases via enhancing the number of circulating endothelial progenitor cells (EPCs). However, the direct effects of sitagliptin on EPC function remain elusive. In this study, we evaluated the proangiogenic effects of sitagliptin on a diabetic hind limb ischaemia (HLI) model in vivo and on EPC culture in vitro. Treatment of db/db mice with sitagliptin (Januvia) after HLI surgery efficiently enhanced ischaemic angiogenesis and blood perfusion, which was accompanied by significant increases in circulating EPC numbers. EPCs derived from the bone marrow of normal mice were treated with high glucose to mimic diabetic hyperglycaemia. We found that high glucose treatment induced EPC apoptosis and tube formation impairment, which were significantly prevented by sitagliptin pretreatment. A mechanistic study found that high glucose treatment of EPCs induced dramatic increases in oxidative stress and apoptosis; pretreatment of EPCs with sitagliptin significantly attenuated high glucose‐induced apoptosis, tube formation impairment and oxidative stress. Furthermore, we found that sitagliptin restored the basal autophagy of EPCs that was impaired by high glucose via activating the AMP‐activated protein kinase/unc‐51‐like autophagy activating kinase 1 signalling pathway, although an autophagy inhibitor abolished the protective effects of sitagliptin on EPCs. Altogether, the results indicate that sitagliptin‐induced preservation of EPC angiogenic function results in an improvement of diabetic ischaemia angiogenesis and blood perfusion, which are most likely mediated by sitagliptin‐induced prevention of EPC apoptosis via augmenting autophagy.
Collapse
Affiliation(s)
- Xiaozhen Dai
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, School of Nursing at the Wenzhou Medical University, Wenzhou, China.,School of Biomedicine, Chengdu Medical College, Chengdu, China.,Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, USA
| | - Jun Zeng
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, USA.,Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Xiaoqing Yan
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, School of Nursing at the Wenzhou Medical University, Wenzhou, China
| | - Qian Lin
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, USA.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, USA
| | - Kai Wang
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, USA.,Departments of Pediatrics, Endocrinology and Metabolism, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jing Chen
- Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, USA
| | - Feixia Shen
- Departments of Pediatrics, Endocrinology and Metabolism, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xuemei Gu
- Departments of Pediatrics, Endocrinology and Metabolism, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuehui Wang
- Departments of Geriatrics, the First Hospital of Jilin University, Changchun, China
| | - Jun Chen
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, School of Nursing at the Wenzhou Medical University, Wenzhou, China.,Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, USA
| | - Kejian Pan
- School of Biomedicine, Chengdu Medical College, Chengdu, China
| | - Lu Cai
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, School of Nursing at the Wenzhou Medical University, Wenzhou, China.,Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, USA.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, USA
| | - Kupper A Wintergerst
- Department of Pediatrics, Division of Endocrinology, Wendy L. Novak Diabetes Care Center, University of Louisville, Louisville, KY, USA
| | - Yi Tan
- Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, School of Nursing at the Wenzhou Medical University, Wenzhou, China.,Pediatric Research Institute, Department of Pediatrics, University of Louisville School of Medicine, Louisville, USA.,Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, USA
| |
Collapse
|
41
|
Liu YM, Ma JH, Zeng QL, Lv J, Xie XH, Pan YJ, Yu ZJ. MiR-19a Affects Hepatocyte Autophagy via Regulating lncRNA NBR2 and AMPK/PPARα in D-GalN/Lipopolysaccharide-Stimulated Hepatocytes. J Cell Biochem 2017; 119:358-365. [PMID: 28586153 DOI: 10.1002/jcb.26188] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 06/05/2017] [Indexed: 12/14/2022]
Abstract
This study aims to evaluate the potential involvement and regulatory mechanism of miR-19a in hepatocytes autophagy of acute liver failure (ALF). The in vitro hepatocytes injury model of primary hepatocyte and hepatocytes line HL-7702 was established by D-galactosamine (D-GalN) and lipopolysaccharide (LPS) co-treatment. Relative expression level of miR-19a and NBR2 was determined by qRT-PCR. Protein expression of AMPK/PPARα and autophagy-related gene was determined by Western blot. In hepatic tissue of 20 ALF patients and D-GalN/LPS-stimulated hepatocytes, miR-19a was upregulated and NBR2 was downregulated. D-GalN/LPS stimulation caused the inactivation of AMPK/PPARα signaling and the decrease of autophagy-related LC3-II/LC3-I ratio and beclin-1 expression in hepatocytes. The expression of both AMPK/PPARα and NBR2 were negatively controlled by miR-19a overexpression or knockdown. Moreover, both NBR2 and PPARα were targeted regulated by miR-19a according to luciferase reporter assay. In D-GalN/LPS-stimulated hepatocytes, AMPK activation promoted PPARα expression. AMPK inactivation inhibited the pro-autophagy effect of miR-19a and caused the decrease of LC3-II/LC3-I ratio and beclin-1 expression. PPARα activation abrogated the anti-autophagy effect of miR-19a mimic and caused the increase of LC3-II/LC3-I ratio and beclin-1 expression. NBR2 knockdown reversed the anti-autophagy impact of miR-19a inhibitor and caused the decrease of LC3-II/LC3-I ratio and beclin-1 expression. In summary, our data suggested that miR-19a negatively controlled the autophagy of hepatocytes attenuated in D-GalN/LPS-stimulated hepatocytes via regulating NBR2 and AMPK/PPARα signaling. J. Cell. Biochem. 119: 358-365, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yan-Min Liu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jin-Hui Ma
- National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing-Lei Zeng
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jun Lv
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xu-Hua Xie
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ya-Jie Pan
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zu-Jiang Yu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|