1
|
Zhao F, Lu M, Wang H. Ginsenoside Rg1 ameliorates chronic intermittent hypoxia-induced vascular endothelial dysfunction by suppressing the formation of mitochondrial reactive oxygen species through the calpain-1 pathway. J Ginseng Res 2023; 47:144-154. [PMID: 36644390 PMCID: PMC9834019 DOI: 10.1016/j.jgr.2022.07.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/19/2022] [Indexed: 01/18/2023] Open
Abstract
Background As the major pathophysiological feature of obstructive sleep apnea (OSA), chronic intermittent hypoxia (CIH) is vital for the occurrence of cardiovascular complications. The activation of calpain-1 mediates the production of endothelial reactive oxygen species (ROS) and impairs nitric oxide (NO) bioavailability, resulting in vascular endothelial dysfunction (VED). Ginsenoside Rg1 is thought to against endothelial cell dysfunction, but the potential mechanism of CIH-induced VED remains unclear. Methods C57BL/6 mice and human coronary artery endothelial cells (HCAECs) were exposed to CIH following knockout or overexpression of calpain-1. The effect of ginsenoside Rg1 on VED, oxidative stress, mitochondrial dysfunction, and the expression levels of calpain-1, PP2A and p-eNOS were detected both in vivo and in vitro. Results CIH promoted VED, oxidative stress and mitochondrial dysfunction accompanied by enhanced levels of calpain-1 and PP2A and reduced levels of p-eNOS in mice and cellular levels. Ginsenoside Rg1, calpain-1 knockout, OKA, NAC and TEMPOL treatment protected against CIH-induced VED, oxidative stress and mitochondrial dysfunction, which is likely concomitant with the downregulated protein expression of calpain-1 and PP2A and the upregulation of p-eNOS in mice and cellular levels. Calpain-1 overexpression increased the expression of PP2A, reduced the level of p-eNOS, and accelerated the occurrence and development of VED, oxidative stress and mitochondrial dysfunction in HCAECs exposed to CIH. Moreover, scavengers of O2 • -, H2O2, complex Ⅰ or mitoKATP abolished CIH-induced impairment in endothelial-dependent relaxation. Conclusion Ginsenoside Rg1 may alleviate CIH-induced vascular endothelial dysfunction by suppressing the formation of mitochondrial reactive oxygen species through the calpain-1 pathway.
Collapse
Affiliation(s)
| | - Meili Lu
- Corresponding authors. Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, 121001, China.
| | - Hongxin Wang
- Corresponding authors. Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical University, Jinzhou, 121001, China.
| |
Collapse
|
2
|
Jin J, Liu J, Luo Y, He H, Zheng X, Zheng C, Huang Y, Chen Y. High fructose induces dysfunctional vasodilatation via PP2A-mediated eNOS Ser1177 dephosphorylation. Nutr Metab (Lond) 2022; 19:24. [PMID: 35331293 PMCID: PMC8944156 DOI: 10.1186/s12986-022-00659-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 03/08/2022] [Indexed: 11/17/2022] Open
Abstract
Background Processed foods are popular and contain large amounts of industrial fructose, which changes people’s diet and exacerbates the negative health effects of high fructose. Several studies have shown that excessive intake of fructose has a major impact on vascular disease. However, the mechanism of the effect of high fructose on blood vessels is currently unclear. Methods The effect of fructose on the vasodilatation of isolated thoracic aortic rings was observed by using wire myography in wild-type (WT) mice. Cell viability and nitric oxide (NO) production were assessed by the corresponding kits in mouse vascular endothelial cells. The effect of fructose on endothelial nitric oxide synthase (eNOS) and protein phosphatase 2A (PP2A) and their changes in phosphorylation were detected by using Western blots. Moreover, a PP2A inhibitor (okadaic acid, OA) was used to evaluate the relationship between fructose and PP2A. Furthermore, PP2ACα endothelial-specific knockout (PP2A cKO) mice were used to detect the vasodilatation of in vitro fructose-incubated thoracic aortic rings by using wire myography. Results High fructose induced endothelium-dependent dysfunctional vasodilatation. High fructose reduced acetylcholine (Ach)-induced vasodilation but did not affect sodium nitroprusside (SNP)-induced vasodilation. Accordingly, NO production and the phosphorylation level of eNOS at serine (Ser) 1177 (P-eNOS) in vascular endothelial cells were remarkably reduced without changes in cell viability. The expression of protein phosphatase 2A catalytic subunit (PP2AC) was increased and the expression of phosphorylated PP2AC (P-PP2A, tyrosine [Tyr] 307) was significantly decreased. Nevertheless, these effects were reversed by OA. Moreover, knockout of the PP2A gene could recover the response of vessels to Ach under high fructose stimulation. Conclusions Our observations demonstrate an underlying mechanism of fructose-induced dysfunctional vasodilatation. Fructose could activate PP2A, which leads to decrease in the phosphorylation of eNOS at Ser1177 and the reduction of NO release, thus leading to the occurrence of endothelium-dependent dysfunctional vasodilatation.
Collapse
Affiliation(s)
- Jiaqi Jin
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, No. 232 Waihuan Dong Rd., Guangzhou University Town, Panyu District, Guangzhou, 510000, China
| | - Jingya Liu
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, No. 232 Waihuan Dong Rd., Guangzhou University Town, Panyu District, Guangzhou, 510000, China
| | - Yong Luo
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, No. 232 Waihuan Dong Rd., Guangzhou University Town, Panyu District, Guangzhou, 510000, China
| | - Hong He
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, No. 63 Duobao Road, Liwan District, Guangzhou, 510150, China
| | - Xinyue Zheng
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Chaoyang Zheng
- Department of Cardiology, The Second Clinical Medical College and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yi Huang
- Department of Stomatology, The First Affiliated Hospital, The School of Dental Medicine, Jinan University, No. 613W. Huangpu Avenue, Guangzhou, 510630, China.
| | - Yang Chen
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, No. 232 Waihuan Dong Rd., Guangzhou University Town, Panyu District, Guangzhou, 510000, China.
| |
Collapse
|
3
|
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: 1.0] [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
|
4
|
Wu CC, Hung CJ, Wang YY, Lin SY, Chen WY, Kuan YH, Liao SL, Yang CP, Chen CJ. Propofol Improved Glucose Tolerance Associated with Increased FGF-21 and GLP-1 Production in Male Sprague-Dawley Rats. Molecules 2020; 25:3229. [PMID: 32679813 PMCID: PMC7397023 DOI: 10.3390/molecules25143229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/08/2020] [Accepted: 07/13/2020] [Indexed: 11/16/2022] Open
Abstract
Anesthetics, particularly volatile anesthetics, have been shown to impair glucose metabolism and cause hyperglycemia, closely linking them with mortality and morbidity as related to surgery. Beyond being an anesthetic used for general anesthesia and sedation, intravenous hypnotic propofol displays an effect on glucose metabolism. To extend the scope of propofol studies, its effects on glucose metabolism were evaluated in male Sprague-Dawley rats of various ages. Unlike chloral hydrate and isoflurane, propofol had little effect on basal glucose levels in rats at 2 months of age, although it did reduce chloral hydrate- and isoflurane-induced hyperglycemia. Propofol reduced postload glucose levels after either intraperitoneal or oral administration of glucose in both 7- and 12-month-old rats, but not those at 2 months of age. These improved effects regarding propofol on glucose metabolism were accompanied by an increase in insulin, fibroblast growth factor-21 (FGF-21), and glucagon-like peptide-1 (GLP-1) secretion. Additionally, an increase in hepatic FGF-21 expression, GLP-1 signaling, and FGF-21 signaling, along with a decrease in endoplasmic reticulum (ER) stress, were noted in propofol-treated rats at 7 months of age. Current findings imply that propofol may turn into insulin-sensitizing molecules during situations of existing insulin resistance, which involve FGF-21, GLP-1, and ER stress.
Collapse
Affiliation(s)
- Chih-Cheng Wu
- Department of Anesthesiology, Taichung Veterans General Hospital, Taichung City 407, Taiwan; (C.-C.W.); (C.-J.H.)
- Department of Financial Engineering, Providence University, Taichung City 433, Taiwan
- Department of Data Science and Big Data Analytics, Providence University, Taichung City 433, Taiwan
| | - Chih-Jen Hung
- Department of Anesthesiology, Taichung Veterans General Hospital, Taichung City 407, Taiwan; (C.-C.W.); (C.-J.H.)
| | - Ya-Yu Wang
- Department of Family Medicine, Taichung Veterans General Hospital, Taichung City 407, Taiwan;
- Institute of Clinical Medicine, National Yang Ming University, Taipei City 112, Taiwan;
| | - Shih-Yi Lin
- Institute of Clinical Medicine, National Yang Ming University, Taipei City 112, Taiwan;
- Center for Geriatrics and Gerontology, Taichung Veterans General Hospital, Taichung City 407, Taiwan
| | - Wen-Ying Chen
- Department of Veterinary Medicine, National Chung-Hsing University, Taichung City 402, Taiwan;
| | - Yu-Hsiang Kuan
- Department of Pharmacology, Chung Shan Medical University, Taichung City 402, Taiwan;
| | - Su-Lan Liao
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City 407, Taiwan; (S.-L.L.); (C.-P.Y.)
| | - Ching-Ping Yang
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City 407, Taiwan; (S.-L.L.); (C.-P.Y.)
| | - Chun-Jung Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung City 407, Taiwan; (S.-L.L.); (C.-P.Y.)
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung City 404, Taiwan
- Ph.D. Program in Translational Medicine, College of Life Sciences, National Chung Hsing University, Taichung City 402, Taiwan
| |
Collapse
|
5
|
Wang M, Suo L, Yang S, Zhang W. CircRNA 001372 Reduces Inflammation in Propofol-Induced Neuroinflammation and Neural Apoptosis through PIK3CA/Akt/NF-κB by miRNA-148b-3p. J INVEST SURG 2020; 34:1167-1177. [PMID: 32506974 DOI: 10.1080/08941939.2020.1771639] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Objectives: To investigate effects of circular RNA (circRNA) 001372 and its antagonist miRNAs-148b-3p on propofol-induced neurotoxicity and neuroinflammation in rat brain and pheochromocytoma cells.Methods: Sprague Dawley rats in propofol model group (n = 6) were intraperitoneal injected with propofol (50 mg/kg) and in sham control group (n = 6) without any treatment. Twenty-four h later, brain tissues were acquired during pentobarbital anesthesia. PC-12 cells were transfected with or without circRNA001372 mimics, circRNA001372 inhibitor, negative mimics or miRNA-148b-3p for 48 h and then treated with propofol (100 μM) for 48 h. Quantitative reverse transcription PCR and gene chips were used for detecting levels of circRNA001372, Haemotoxylin and Eosin staining for cell morphology, MTT for cell viability, flow cytometry for apoptosis, enzyme-linked immunosorbent assay for lactate dehydrogenase (LDH), interleukin-1β (IL-1β), IL-6, IL17 and IL-18, and Western blots for phosphoinositide 3-kinase (PI3K), Akt, phosphorylated Akt, and nuclear factor (NF) κB, dual-light luminescent reporter gene assay for luciferase reporter.Results: The propofol anesthesia in rats decreases levels of circRNA001372 and increases levels of cytokines including IL-1β, IL-6, IL17 and IL-18, resulting in the neurocyte damage in brain. In propofol-treated PC-12 cells, the inhibition of circRNA001372 increases apoptosis and cell damage makers, including LDH, IL-1β, IL-6, IL17, IL-18, resulting in the reduction of cell viability, which have been revised after over-expression of circRNA001372. MiRNA-148b-3p reduces circRNA001372-incresed PI3K and pAKt levels but enhances the circRNA001372-decreased NFκB level.Conclusions: CircRNA001372 suppresses propofol-induced neurotoxicity and neuroinflammation through PI3K/Akt/NF-κB signaling pathway in rat brain and neurocytes. MiRNA-148b-3p antagonizes the effects of circRNA001372.
Collapse
Affiliation(s)
- Mingyu Wang
- Department of Anesthesia, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | - Liangyuan Suo
- Department of Anesthesia, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | - Shun Yang
- Department of Anesthesia, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | - Weiqing Zhang
- Department of Anesthesia, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| |
Collapse
|
6
|
Wang T, Zhu H, Hou Y, Duan W, Meng F, Liu Y. Ketamine attenuates high-glucose-mediated endothelial inflammation in human umbilical vein endothelial cells. Can J Physiol Pharmacol 2020; 98:156-161. [PMID: 32078386 DOI: 10.1139/cjpp-2019-0185] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hyperglycemia mediates oxidative stress, thus inducing transcription factor nuclear factor kappa B (NF-κB) activation, increasing endothelial adhesion molecule expression and monocyte/endothelial interaction, and resulting in endothelial injury. Ketamine was reported to attenuate oxidative stress in many cases. In this research, we determined whether and how ketamine protects against high-glucose-mediated augmentation of monocyte/endothelial interaction and endothelial adhesion molecule expression in human umbilical vein endothelial cells. High glucose augmented monocyte/endothelial adhesion and endothelial adhesion molecule expression. High glucose induced reactive oxygen species (ROS) production and augmented phospho-protein kinase C (p-PKC) βII expression and PKC activity. Moreover, high glucose inhibited the inhibitory subunit of nuclear factor-κBα (IκBα) expression in the cytoplasm and induced NF-κB nuclear translocation. Importantly, the effects induced by high glucose were counteracted by ketamine treatment. Further, CGP53353, a PKC βII inhibitor, inhibited high-glucose-mediated NF-κB nuclear translocation, attenuated adhesion molecule expression, and reduced monocyte/endothelial interaction. Further, these effects of ketamine against high-glucose-induced endothelial injury were inhibited by phorbol 12-myristate 13-acetate, a PKC βII activator. In conclusion, ketamine, via reducing ROS accumulation, inhibited PKC βII Ser660 phosphorylation and PKC and NF-κB activation and reduced high-glucose-induced expression of endothelial adhesion molecules and monocyte/endothelial interaction.
Collapse
Affiliation(s)
- Tianhai Wang
- Department of Anaesthesiology, Xinjiang Medical University, Affiliated Tumour Hospital, Xinjiang, PR China
| | - Hongge Zhu
- Department of Second Pulmonary Medicine, Xinjiang Medical University, Affiliated Tumour Hospital, Xinjiang, PR China
| | - Yanshen Hou
- Department of Anaesthesiology, Xinjiang Medical University, Affiliated Tumour Hospital, Xinjiang, PR China
| | - Wenming Duan
- Department of Anaesthesiology, Xinjiang Medical University, Affiliated Tumour Hospital, Xinjiang, PR China
| | - Fufen Meng
- Department of Anaesthesiology, Xinjiang Medical University, Affiliated Tumour Hospital, Xinjiang, PR China
| | - Yahua Liu
- Department of Anaesthesiology, Xinjiang Medical University, Affiliated Tumour Hospital, Xinjiang, PR China
| |
Collapse
|
7
|
Xu K, Liu X, Yin D, Ren G, Zhao Y. PP2A alleviates oxidized LDL-induced endothelial dysfunction by regulating LOX-1/ROS/MAPK axis. Life Sci 2020; 243:117270. [PMID: 31923421 DOI: 10.1016/j.lfs.2020.117270] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/03/2020] [Accepted: 01/03/2020] [Indexed: 11/25/2022]
Abstract
AIMS The purpose of this study is to investigate the effect of PP2A on the progression of AS and the special molecular mechanism. MAIN METHODS The expression of PP2A in Human umbilical vein endothelial cells (HUVECs) induced by different concentrations of Ox-LDL was measured by RT-PCR and Western blot. The binding activity of PP2A and LOX-1 was determined by CoIP assay. Western blot was used to measure the protein expression of VCAM-1, ICAM-1 and MCP-1. KEY FINDING The results revealed that the expression of PP2A was decreased with the increase of Ox-LDL concentration in HUVECs. Overexpression of PP2A alleviated Ox-LDL-induced dysfunction and inflammatory response in HUVECs. The results of Co-immunoprecipitation (CoIP) showed that PP2A had direct effect on LOX-1, and PP2A inhibited the expression of LOX-1. In addition, overexpression of LOX-1 reversed the inhibitory effect of PP2A on Ox-LDL-induced dysfunction and inflammatory response in HUVECs. What is more, PP2A inhibited LOX-1/ROS/MAPK axis. SIGNIFICANCE it suggests that PP2A alleviates Ox-LDL-induced dysfunction and inflammatory response of HUVECs potentially by regulating the LOX-1/ROS/MAPK axis,which suggests that PP2A has anti-inflammatory effect during the formation of as, and the molecular therapy of PP2A provides a theoretical basis.
Collapse
Affiliation(s)
- Kaicheng Xu
- Department of Anesthesiology, China-Japan Union hospital JiLin University, Chang chun, JiLin 130033, China
| | - Xiwen Liu
- Department of vascular surgery, China-Japan Union hospital JiLin University, Chang chun, JiLin 130033, China
| | - Dexin Yin
- Department of vascular surgery, China-Japan Union hospital JiLin University, Chang chun, JiLin 130033, China
| | - Guanghao Ren
- Department of vascular surgery, China-Japan Union hospital JiLin University, Chang chun, JiLin 130033, China
| | - Yue Zhao
- Department of vascular surgery, China-Japan Union hospital JiLin University, Chang chun, JiLin 130033, China.
| |
Collapse
|
8
|
Franco-Juárez B, Mejía-Martínez F, Moreno-Arriola E, Hernández-Vázquez A, Gómez-Manzo S, Marcial-Quino J, Arreguín-Espinosa R, Velázquez-Arellano A, Ortega-Cuellar D. A high glucose diet induces autophagy in a HLH-30/TFEB-dependent manner and impairs the normal lifespan of C. elegans. Aging (Albany NY) 2019; 10:2657-2667. [PMID: 30299269 PMCID: PMC6224263 DOI: 10.18632/aging.101577] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 09/24/2018] [Indexed: 12/27/2022]
Abstract
A high-glucose diet (HGD) is associated with the development of metabolic diseases that decrease life expectancy, including obesity and type-2 diabetes (T2D); however, the mechanism through which a HGD does so is still unclear. Autophagy, an evolutionarily conserved mechanism, has been shown to promote both cell and organismal survival. The goal of this study was to determine whether exposure of Caenorhabditis elegans to a HGD affects autophagy and thus contributes to the observed lifespan reduction under a HGD. Unexpectedly, nematodes exposed to a HGD showed increased autophagic flux via an HLH-30/TFEB-dependent mechanism because animals with loss of HLH-30/TFEB, even those with high glucose exposure, had an extended lifespan, suggesting that HLH-30/TFEB might have detrimental effects on longevity through autophagy under this stress condition. Interestingly, pharmacological treatment with okadaic acid, an inhibitor of the PP2A and PP1 protein phosphatases, blocked HLH-30 nuclear translocation, but not TAX-6/calcineurin suppression by RNAi, during glucose exposure. Together, our data support the suggested dual role of HLH-30/TFEB and autophagy, which, depending on the cellular context, may promote either organismal survival or death.
Collapse
Affiliation(s)
- Berenice Franco-Juárez
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas UNAM - Instituto Nacional de Pediatría, Mexico City 04530, Mexico
| | - Fanny Mejía-Martínez
- Laboratorio de Nutrición Experimental, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico
| | - Elizabeth Moreno-Arriola
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas UNAM - Instituto Nacional de Pediatría, Mexico City 04530, Mexico
| | - Alain Hernández-Vázquez
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas UNAM - Instituto Nacional de Pediatría, Mexico City 04530, Mexico
| | - Saul Gómez-Manzo
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico
| | - Jaime Marcial-Quino
- Consejo Nacional de Ciencia y Tecnología (CONACYT), Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico
| | - Roberto Arreguín-Espinosa
- Departamento de Química de Biomacromoléculas, Instituto de Química, Universidad Nacional Autónoma de Mexico, Mexico City 04510, Mexico
| | - Antonio Velázquez-Arellano
- Unidad de Genética de la Nutrición, Instituto de Investigaciones Biomédicas UNAM - Instituto Nacional de Pediatría, Mexico City 04530, Mexico
| | - Daniel Ortega-Cuellar
- Laboratorio de Nutrición Experimental, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico
| |
Collapse
|
9
|
Ding XW, Sun X, Shen XF, Lu Y, Wang JQ, Sun ZR, Miao CH, Chen JW. Propofol attenuates TNF-α-induced MMP-9 expression in human cerebral microvascular endothelial cells by inhibiting Ca 2+/CAMK II/ERK/NF-κB signaling pathway. Acta Pharmacol Sin 2019; 40:1303-1313. [PMID: 31235816 DOI: 10.1038/s41401-019-0258-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/21/2019] [Indexed: 02/07/2023]
Abstract
Metalloproteinase 9 (MMP-9) is able to degrade collagen IV, an important component of blood-brain barrier (BBB). Expression of MMPs, especially MMP-9, correlates with BBB disruption during central nervous system inflammation. Propofol has been reported to have anti-inflammation effects. In this study, we investigated the effects of propofol on TNF-α-induced MMP-9 expression in human cerebral microvascular endothelial cells (hCMEC/D3 cells) and explored the underlying mechanisms. The hCMEC/D3 cells were treated with propofol (25 μM), followed by TNF-α (25 ng/mL). We showed that TNF-α treatment markedly increased MMP-9 expression and decreased collagen IV expression in hCMEC/D3 cells, which was blocked by pretreatment with propofol. TNF-α-induced downregulation of collagen IV was also reversed by MMP-9 knockdown with siRNA. We revealed that TNF-α upregulated MMP-9 expression in hCMEC/D3 cells through activation of Ca2+/CAMK II/ERK/NF-κB signaling pathway; co-treatment with inhibitors of CaMK II (KN93), ERK (LY3214996), NF-κB (PDTC) or Ca2+chelator (BAPTA-AM) abrogated the effect of TNF-α on MMP-9 expression. We further established an in vitro BBB model by co-culturing of hCMEC/D3 cells and human astrocytes for 6 days and measuring trans-endothelial electrical resistance (TEER) to reflect the BBB permeability. TNF-α treatment markedly decreased TEER value, which was attenuated by pretreatment with propofol (25 μM) or MMP-9 knockdown with siRNA. In conclusion, propofol inhibits TNF-α-induced MMP-9 expression in hCMEC/D3 cells via repressing the Ca2+/CAMKII/ERK/NF-κB signaling pathway. TNF-α-impaired BBB integrity could be reversed by propofol, and propofol attenuates the inhibitory effect of TNF-α on collagen IV.
Collapse
|
10
|
Wang J, Qi J, Wu Q, Jiang H, Yin Y, Huan Y, Zhao Y, Zhu M. Propofol attenuates high glucose-induced P66shc expression in human umbilical vein endothelial cells through Sirt1. Acta Biochim Biophys Sin (Shanghai) 2019; 51:197-203. [PMID: 30590376 DOI: 10.1093/abbs/gmy167] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 11/28/2018] [Indexed: 01/02/2023] Open
Abstract
Perioperative hyperglycemia is a common metabolic disorder in clinic settings. Hyperglycemia leads to endothelial inflammation, endothelial cell apoptosis, and dysfunction, thus resulting in endothelial injury. Propofol (2,6-diisopropylphenol) is a widely used intravenous anesthetic in clinic settings. Our previous study indicated that propofol inhibits mitochondrial reactive oxygen species (ROS) production via down-regulation of phosphatase A2 (PP2A) expression, inhibition of Ser36-p66shc dephosphorylation and mitochondrial translocation, thus improving high glucose-induced endothelial injury. The expression of p66shc was inhibited by propofol in hyperglycemic human umbilical vein endothelial cells (HUVECs). However, the mechanism by which propofol inhibits p66shc expression in hyperglycemic HUVECs is still obscure. In the present study, we mainly examined how propofol inhibited high glucose-induced p66shc expression in HUVECs. Compared with 5 mM glucose treatment, high glucose increased p66shc expression and decreased sirt1 expression, which was inhibited by propofol treatment. Moreover, EX527 (a sirt1 inhibitor) reversed the effect of propofol against high glucose-induced p66shc expression. However, EX527 did not reverse the effects of propofol against high glucose-induced ROS accumulation, endothelial inflammation, and apoptosis. Furthermore, when cells were incubated with propofol, EX527, and FTY720 (a PP2A activator) simultaneously, the effects of propofol against high glucose-induced ROS accumulation, inflammation, and apoptosis were reversed. Our results suggested that propofol inhibited high glucose-induced p66shc expression via upregulation of sirt1 expression in hyperglycemic HUVECs. Moreover, propofol protects against high glucose-mediated ROS accumulation and endothelial injury via both inhibition of p66shc expression and dephosphorylation of Ser36-p66shc.
Collapse
Affiliation(s)
- Jing Wang
- Department of Anaesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Anaesthesiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jie Qi
- Department of Anaesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Anaesthesiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qichao Wu
- Department of Anaesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Anaesthesiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hui Jiang
- Department of Anaesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Anaesthesiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuehao Yin
- Department of Anaesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Anaesthesiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yan Huan
- Department of Anaesthesiology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yanjun Zhao
- Department of Anaesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Anaesthesiology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Anaesthesiology, Eye and Ant Hospital of Fudan University, Shanghai, China
| | - Minmin Zhu
- Department of Anaesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Anaesthesiology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Wang Q, Liu J, Guo T, Liu D, Pan J. Epidermal Growth Factor Reverses the Inhibitory Effects of the Bisphosphonate, Zoledronic Acid, on Human Oral Keratinocytes and Human Vascular Endothelial Cells In Vitro via the Epidermal Growth Factor Receptor (EGFR)/Akt/Phosphoinositide 3-Kinase (PI3K) Signaling Pathway. Med Sci Monit 2019; 25:700-710. [PMID: 30675875 PMCID: PMC6357820 DOI: 10.12659/msm.911579] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Background Medication-related osteonecrosis of the jaw (MRONJ) is due to the direct effects of drug toxicity and the effects on angiogenesis. The aims of this study were to evaluate the effects of treatment with the bisphosphonate, zoledronic acid, on human oral keratinocytes (HOKs) and human umbilical vein endothelial cells (HUVECs) in vitro, and whether epidermal growth factor (EGF) could alter these effects. Material/Methods HOKs and HUVECs were incubated with zoledronic acid or EGF. Cell viability was assessed by the cell counting kit-8 (CCK-8), cell apoptosis was studied using Annexin-V conjugated to fluorescein isothiocyanate (FITC). Angiogenesis was studied by observing HUVEC tube formation and cell migrations using a transwell assay. A scratch wound assay investigated cell migration of HOKs. Western blot measured expression levels of phosphorylated epidermal growth factor receptor (EGFR), Akt, phosphoinositide 3-kinase (PI3K), the mechanistic target of rapamycin (mTOR), and endothelial nitric oxide synthase (eNOS). Results Zoledronic acid treatment (5 μmol/L) significantly inhibited cell viability and cell migration of HOKs and HUVECs and angiogenesis of HUVECS (P<0.05); EGF partially reversed these effects (P<0.05). Zoledronic acid treatment of HOKs and HUVECs had no significant effects on apoptosis (P>0.05), but significantly reduced expression levels of p-EGFR, p-Akt, p-PI3K, p-mTOR), and p-eNOS (P<0.05); EGF partially reversed these effects and increased the expression levels (P<0.05). Conclusions EGF partially reversed the effects of the bisphosphonate, zoledronic acid, on HOKs and HUVECs in vitro via the EGFR/Akt/PI3K signaling pathway. Further studies are required to determine the effects of EGF on MRONJ including bisphosphonate-related osteonecrosis of the jaw.
Collapse
Affiliation(s)
- Qizhang Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China (mainland)
| | - Jiyuan Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China (mainland)
| | - Ting Guo
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China (mainland)
| | - Dazhong Liu
- Xindu District Peoples' Hospital of Chengdu, Chengdu, Sichuan, China (mainland)
| | - Jian Pan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China (mainland)
| |
Collapse
|