1
|
Zhu LR, He XH, Yuan YH, Yuan H, Xia XH. [Changes and significance of oxidized phospholipids and endothelial nitric oxide synthase in the acute stage of Kawasaki disease]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:829-834. [PMID: 39148387 PMCID: PMC11334547 DOI: 10.7499/j.issn.1008-8830.2403056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 07/04/2024] [Indexed: 08/17/2024]
Abstract
OBJECTIVES To investigate the changes in the serum levels of oxidized phospholipids (OxPLs) and endothelial nitric oxide synthase (eNOS) and their association with coronary artery disease (CAL) in children in the acute stage of Kawasaki disease (KD), as well as the clinical significance of OxPLs and eNOS. METHODS A prospective study was conducted on 95 children in the acute stage of KD (KD group). According to the presence of absence of CAL, the KD group was further divided into a CAL subgroup and a non-CAL (NCAL) subgroup. Thirty children with fever due to lower respiratory tract infection were enrolled as the fever group. Thirty healthy children who underwent physical examination were enrolled as the healthy control group. The above groups were compared in terms of general information and serum levels of OxPLs, eNOS and other laboratory indexes, and the correlation between OxPLs level and eNOS level was analyzed. RESULTS The KD group had a significantly higher level of OxPLs and a significantly lower level of eNOS compared with the fever group and the healthy control group (P<0.05). After treatment, the children with KD had a significantly decreased OxPLs level and a significantly increased eNOS level (P<0.05). Compared with the NCAL subgroup, the CAL subgroup had a significantly higher level of OxPLs and a significantly lower level of eNOS (P<0.05). Among the children of KD, the level of OxPLs was negatively correlated with that of eNOS (rs=-0.353, P<0.05). CONCLUSIONS Serum OxPLs and eNOS in the acute stage of KD may be involved in the development of CAL in children with KD, and therefore, they may be used as the biomarkers to predict CAL in these children.
Collapse
Affiliation(s)
- Liu-Rong Zhu
- Department of Pediatrics, First Affiliated Hospital of Hunan Normal University, Changsha 410005, China
| | - Xue-Hua He
- Department of Pediatrics, First Affiliated Hospital of Hunan Normal University, Changsha 410005, China
| | - Yong-Hua Yuan
- Department of Pediatrics, First Affiliated Hospital of Hunan Normal University, Changsha 410005, China
| | | | | |
Collapse
|
2
|
Wang Y, Shou X, Wu Y, Chen J, Zeng R, Liu Q. Identification of key biomarkers of endothelial dysfunction in hypertension with carotid atherosclerosis based on WGCNA and the LASSO algorithm. Heliyon 2024; 10:e32966. [PMID: 38984304 PMCID: PMC11231533 DOI: 10.1016/j.heliyon.2024.e32966] [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: 11/16/2023] [Revised: 06/03/2024] [Accepted: 06/12/2024] [Indexed: 07/11/2024] Open
Abstract
Background Endothelial dysfunction is the early stage of carotid atherosclerosis (CAS) in patients with hypertension. It is worth identifying the potential hub genes of endothelial dysfunction to elucidate pathological mechanism in the progression of the disease. Method We obtained gene expression profiles of GSE43292 from the Gene Expression Omnibus (GEO) database. Hub genes associated with CAS were identified through weighted gene correlation network analysis (WGCNA) and least absolute shrinkage and selection operator (LASSO) regression. Subsequently, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed to explore potential biological mechanisms and signaling pathways. Finally, in vitro experiments on human umbilical vein endothelial cells (HUVECs) were conducted to validate these hub genes. Results The microarray dataset GSE43292 included 32 CAS plaques samples and corresponding macroscopically intact tissues from patients with hypertension. A total of 161 differentially expressed genes were discovered. Through WGCNA analysis, the gray60 module emerged as the most significant module associated with clinical features. The GO and KEGG enrichment analyses of genes in the gray60 module highlighted the substantial involvement of immune response-related signaling pathways. Two key hub genes (CCR1 and NCKAP1L) were pinpointed via LASSO regression. We found a significant increase in the mRNA expression level of the hub genes in oxidized low density lipoprotein (ox-LDL) treated HUVECs. Conclusions Our study indicated that the hub genes related to immune responses are involved in the development of CAS. Two hub genes (CCR1 and NCKAP1L) of endothelial dysfunction were identified. These genes may provide a valuable therapeutic target of CAS in patients with hypertension.
Collapse
Affiliation(s)
- Yimin Wang
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310005, China
| | - Xinyang Shou
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Yuteng Wu
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Jun Chen
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Rui Zeng
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310005, China
| | - Qiang Liu
- The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310005, China
| |
Collapse
|
3
|
Zuo X, Ding X, Zhang Y, Kang YJ. Reversal of atherosclerosis by restoration of vascular copper homeostasis. Exp Biol Med (Maywood) 2024; 249:10185. [PMID: 38978540 PMCID: PMC11228934 DOI: 10.3389/ebm.2024.10185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 06/04/2024] [Indexed: 07/10/2024] Open
Abstract
Atherosclerosis has traditionally been considered as a disorder characterized by the accumulation of cholesterol and thrombotic materials within the arterial wall. However, it is now understood to be a complex inflammatory disease involving multiple factors. Central to the pathogenesis of atherosclerosis are the interactions among monocytes, macrophages, and neutrophils, which play pivotal roles in the initiation, progression, and destabilization of atherosclerotic lesions. Recent advances in our understanding of atherosclerosis pathogenesis, coupled with results obtained from experimental interventions, lead us to propose the hypothesis that atherosclerosis may be reversible. This paper outlines the evolution of this hypothesis and presents corroborating evidence that supports the potential for atherosclerosis regression through the restoration of vascular copper homeostasis. We posit that these insights may pave the way for innovative therapeutic approaches aimed at the reversal of atherosclerosis.
Collapse
Affiliation(s)
- Xiao Zuo
- Tasly Stem Cell Biology Laboratory, Tasly Biopharmaceutical Co., Tianjin, China
| | - Xueqin Ding
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yaya Zhang
- Tasly Stem Cell Biology Laboratory, Tasly Biopharmaceutical Co., Tianjin, China
| | - Y James Kang
- Tasly Stem Cell Biology Laboratory, Tasly Biopharmaceutical Co., Tianjin, China
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Huang X, Jiang F, Ma Y, Zhu K, Wang Z, Hua Z, Yu J, Zhang L. A bibliometric analysis of endoplasmic reticulum stress and atherosclerosis. Front Physiol 2024; 15:1392454. [PMID: 38938744 PMCID: PMC11210825 DOI: 10.3389/fphys.2024.1392454] [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: 03/25/2024] [Accepted: 05/24/2024] [Indexed: 06/29/2024] Open
Abstract
The mechanisms underlying the occurrence and development of atherosclerosis (AS) are diverse, among which endoplasmic reticulum stress (ERS) is an important mechanism that should not be overlooked. However, up to now, there has been no bibliometric study on the relationship between ERS and AS. To understand the research progress in ERS and AS, this paper conducted a statistical analysis of publications in this field using bibliometrics. A total of 1,035 records were retrieved from the Web of Science Core Collection. CiteSpace, VOSviewer, and the R package "bibliometric" were used to analyze the spatiotemporal distribution, countries, authors, institutions, journals, references, and keywords of the literature, and to present the basic information of this field through visualized maps, as well as determine the collaboration relationships among researchers in this field. This field has gradually developed and stabilized over the past 20 years. The current research hotspots in this field mainly include the relationship between ERS and AS-related cells, the mechanisms by which ERS promotes AS, related diseases, and associated cytokines, etc. Vascular calcification, endothelial dysfunction, NLRP3 inflammasome, and heart failure represent the frontier research in this field and are becoming new research hotspots. It is hoped that this study will provide new insights for research and clinical work in the field of ERS and AS.
Collapse
Affiliation(s)
- Xinyu Huang
- Shandong University of Traditional Chinese Medicine, Shandon, China
| | - Feng Jiang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandon, China
| | - Yongbo Ma
- Shandong University of Traditional Chinese Medicine, Shandon, China
| | - Kunpeng Zhu
- Shandong University of Traditional Chinese Medicine, Shandon, China
| | - Zhenyuan Wang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandon, China
| | - Zhen Hua
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandon, China
| | - Jie Yu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandon, China
| | - Lei Zhang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandon, China
| |
Collapse
|
5
|
Sánchez-León ME, Loaeza-Reyes KJ, Matias-Cervantes CA, Mayoral-Andrade G, Pérez-Campos EL, Pérez-Campos-Mayoral L, Hernández-Huerta MT, Zenteno E, Pérez-Cervera Y, Pina-Canseco S. LOX-1 in Cardiovascular Disease: A Comprehensive Molecular and Clinical Review. Int J Mol Sci 2024; 25:5276. [PMID: 38791315 PMCID: PMC11121106 DOI: 10.3390/ijms25105276] [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: 04/03/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
LOX-1, ORL-1, or lectin-like oxidized low-density lipoprotein receptor 1 is a transmembrane glycoprotein that binds and internalizes ox-LDL in foam cells. LOX-1 is the main receptor for oxidized low-density lipoproteins (ox-LDL). The LDL comes from food intake and circulates through the bloodstream. LOX-1 belongs to scavenger receptors (SR), which are associated with various cardiovascular diseases. The most important and severe of these is the formation of atherosclerotic plaques in the intimal layer of the endothelium. These plaques can evolve into complicated thrombi with the participation of fibroblasts, activated platelets, apoptotic muscle cells, and macrophages transformed into foam cells. This process causes changes in vascular endothelial homeostasis, leading to partial or total obstruction in the lumen of blood vessels. This obstruction can result in oxygen deprivation to the heart. Recently, LOX-1 has been involved in other pathologies, such as obesity and diabetes mellitus. However, the development of atherosclerosis has been the most relevant due to its relationship with cerebrovascular accidents and heart attacks. In this review, we will summarize findings related to the physiologic and pathophysiological processes of LOX-1 to support the detection, diagnosis, and prevention of those diseases.
Collapse
Affiliation(s)
- Maria Eugenia Sánchez-León
- Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.E.S.-L.); (K.J.L.-R.); (C.A.M.-C.); (G.M.-A.); (L.P.-C.-M.)
| | - Karen Julissa Loaeza-Reyes
- Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.E.S.-L.); (K.J.L.-R.); (C.A.M.-C.); (G.M.-A.); (L.P.-C.-M.)
- Centro de Estudios en Ciencias de la Salud y la Enfermedad, Facultad de Odontología, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico
| | - Carlos Alberto Matias-Cervantes
- Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.E.S.-L.); (K.J.L.-R.); (C.A.M.-C.); (G.M.-A.); (L.P.-C.-M.)
| | - Gabriel Mayoral-Andrade
- Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.E.S.-L.); (K.J.L.-R.); (C.A.M.-C.); (G.M.-A.); (L.P.-C.-M.)
| | | | - Laura Pérez-Campos-Mayoral
- Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.E.S.-L.); (K.J.L.-R.); (C.A.M.-C.); (G.M.-A.); (L.P.-C.-M.)
| | - María Teresa Hernández-Huerta
- Consejo Nacional de Humanidades, Ciencias y Tecnologías, Facultad de Medicina y Cirugía, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68120, Mexico;
| | - Edgar Zenteno
- Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Yobana Pérez-Cervera
- Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.E.S.-L.); (K.J.L.-R.); (C.A.M.-C.); (G.M.-A.); (L.P.-C.-M.)
- Centro de Estudios en Ciencias de la Salud y la Enfermedad, Facultad de Odontología, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico
| | - Socorro Pina-Canseco
- Centro de Investigación Facultad de Medicina-UNAM-UABJO, Universidad Autónoma “Benito Juárez” de Oaxaca, Oaxaca 68020, Mexico; (M.E.S.-L.); (K.J.L.-R.); (C.A.M.-C.); (G.M.-A.); (L.P.-C.-M.)
| |
Collapse
|
6
|
Munno M, Mallia A, Greco A, Modafferi G, Banfi C, Eligini S. Radical Oxygen Species, Oxidized Low-Density Lipoproteins, and Lectin-like Oxidized Low-Density Lipoprotein Receptor 1: A Vicious Circle in Atherosclerotic Process. Antioxidants (Basel) 2024; 13:583. [PMID: 38790688 PMCID: PMC11118168 DOI: 10.3390/antiox13050583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Atherosclerosis is a complex condition that involves the accumulation of lipids and subsequent plaque formation in the arterial intima. There are various stimuli, cellular receptors, and pathways involved in this process, but oxidative modifications of low-density lipoprotein (ox-LDL) are particularly important in the onset and progression of atherosclerosis. Ox-LDLs promote foam-cell formation, activate proinflammatory pathways, and induce smooth-muscle-cell migration, apoptosis, and cell death. One of the major receptors for ox-LDL is LOX-1, which is upregulated in several cardiovascular diseases, including atherosclerosis. LOX-1 activation in endothelial cells promotes endothelial dysfunction and induces pro-atherogenic signaling, leading to plaque formation. The binding of ox-LDLs to LOX-1 increases the generation of reactive oxygen species (ROS), which can induce LOX-1 expression and oxidize LDLs, contributing to ox-LDL generation and further upregulating LOX-1 expression. This creates a vicious circle that is amplified in pathological conditions characterized by high plasma levels of LDLs. Although LOX-1 has harmful effects, the clinical significance of inhibiting this protein remains unclear. Further studies both in vitro and in vivo are needed to determine whether LOX-1 inhibition could be a potential therapeutic target to counteract the atherosclerotic process.
Collapse
Affiliation(s)
- Marco Munno
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino, 20138 Milan, Italy; (M.M.); (A.M.); (A.G.); (G.M.); (S.E.)
| | - Alice Mallia
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino, 20138 Milan, Italy; (M.M.); (A.M.); (A.G.); (G.M.); (S.E.)
- Dipartimento di Biologia e Biotecnologie “Lazzaro Spallanzani”, Università di Pavia, 27100 Pavia, Italy
| | - Arianna Greco
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino, 20138 Milan, Italy; (M.M.); (A.M.); (A.G.); (G.M.); (S.E.)
| | - Gloria Modafferi
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino, 20138 Milan, Italy; (M.M.); (A.M.); (A.G.); (G.M.); (S.E.)
| | - Cristina Banfi
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino, 20138 Milan, Italy; (M.M.); (A.M.); (A.G.); (G.M.); (S.E.)
| | - Sonia Eligini
- Unit of Functional Proteomics, Metabolomics and Network Analysis, Centro Cardiologico Monzino, 20138 Milan, Italy; (M.M.); (A.M.); (A.G.); (G.M.); (S.E.)
| |
Collapse
|
7
|
Zhang H, Cai J, Zhang R, Shuai S, Tang M, Ju R, Hu Y, Zuo T, Yang Y. The role of serum lipid in predicting coronary artery lesions and intravenous immunoglobulin resistance in Kawasaki disease: a cohort study. J Int Med Res 2024; 52:3000605241252115. [PMID: 38713460 PMCID: PMC11080733 DOI: 10.1177/03000605241252115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 04/15/2024] [Indexed: 05/08/2024] Open
Abstract
OBJECTIVE To assess the predictive value of the serum lipid profile for initial intravenous immunoglobulin (IVIG) resistance and coronary artery lesions (CALs) in patients with Kawasaki disease (KD). METHODS This retrospective cohort study enrolled patients with KD and divided them into IVIG-responsive and IVIG-resistant groups. They were also stratified based on the presence of CALs (CALs and non-CALs groups). Clinical, echocardiographic and biochemical values were evaluated. A subgroup analysis was performed on complete and incomplete KD. Predictors of initial IVIG resistance and CALs were determined by multivariate logistic regression analysis. RESULTS A total of 649 KD patients were enrolled: 151 had CALs and 76 had initial IVIG resistance. Low-density lipoprotein cholesterol (LDL-C) was significantly lower in the IVIG-resistant group than in the IVIG-responsive group. LDL-C and apolipoprotein (Apo) B were significantly lower in the CALs group compared with the non-CALs group. Multivariate logistic regression failed to identify the serum lipid profile (LDL-C, Apo A or Apo B) as an independent risk factor for initial IVIG resistance or CALs in KD patients. CONCLUSION KD patients might have dyslipidaemia in the acute phase, but the serum lipid profile might not be suitable as a single predictor for initial IVIG resistance or CALs.
Collapse
Affiliation(s)
- Hongxi Zhang
- Department of Pharmacy, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Jianghui Cai
- Department of Pharmacy, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
- Department of Pharmacy, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Rui Zhang
- Department of Pharmacy, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Shuping Shuai
- Department of Pharmacy, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Mi Tang
- Department of Pharmacy, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
- Office of Good Clinical Practice, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Rong Ju
- Department of Paediatrics, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Ying Hu
- Department of Pharmacy, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Tianrui Zuo
- Department of Pharmacy, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Yanfeng Yang
- Department of Paediatrics, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| |
Collapse
|
8
|
Liu G, Li B, Qin S, Nice EC, Yang J, Yang L, Huang C. Redox signaling-mediated tumor extracellular matrix remodeling: pleiotropic regulatory mechanisms. Cell Oncol (Dordr) 2024; 47:429-445. [PMID: 37792154 DOI: 10.1007/s13402-023-00884-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2023] [Indexed: 10/05/2023] Open
Abstract
BACKGROUND The extracellular matrix (ECM), a fundamental constituent of all tissues and organs, is crucial for shaping the tumor microenvironment. Dysregulation of ECM remodeling has been closely linked to tumor initiation and progression, where specific signaling pathways, including redox signaling, play essential roles. Reactive oxygen species (ROS) are risk factors for carcinogenesis whose excess can facilitate the oxidative damage of biomacromolecules, such as DNA and proteins. Emerging evidence suggests that redox effects can aid the modification, stimulation, and degradation of ECM, thus affecting ECM remodeling. These alterations in both the density and components of the ECM subsequently act as critical drivers for tumorigenesis. In this review, we provide an overview of the functions and primary traits of the ECM, and it delves into our current understanding of how redox reactions participate in ECM remodeling during cancer progression. We also discuss the opportunities and challenges presented by clinical strategies targeting redox-controlled ECM remodeling to overcome cancer. CONCLUSIONS The redox-mediated ECM remodeling contributes importantly to tumor survival, progression, metastasis, and poor prognosis. A comprehensive investigation of the concrete mechanism of redox-mediated tumor ECM remodeling and the combination usage of redox-targeted drugs with existing treatment means may reveal new therapeutic strategy for future antitumor therapies.
Collapse
Affiliation(s)
- Guowen Liu
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, and , Chengdu, 610041, China
| | - Bowen Li
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, and , Chengdu, 610041, China
| | - Siyuan Qin
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, and , Chengdu, 610041, China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, 3800, Australia
| | - Jinlin Yang
- Department of Gastroenterology & Hepatology, West China Hospital of Sichuan University, Sichuan Province, No.37 Guoxue Alley, Chengdu, 610041, China.
- Department of Gastroenterology & Hepatology, Sichuan University-Oxford University Huaxi Gastrointestinal Cancer Centre, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
| | - Li Yang
- Department of Gastroenterology & Hepatology, West China Hospital of Sichuan University, Sichuan Province, No.37 Guoxue Alley, Chengdu, 610041, China.
- Department of Gastroenterology & Hepatology, Sichuan University-Oxford University Huaxi Gastrointestinal Cancer Centre, West China Hospital, Sichuan University, No.37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, and , Chengdu, 610041, China.
| |
Collapse
|
9
|
Nègre-Salvayre A, Salvayre R. Reactive Carbonyl Species and Protein Lipoxidation in Atherogenesis. Antioxidants (Basel) 2024; 13:232. [PMID: 38397830 PMCID: PMC10886358 DOI: 10.3390/antiox13020232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Atherosclerosis is a multifactorial disease of medium and large arteries, characterized by the presence of lipid-rich plaques lining the intima over time. It is the main cause of cardiovascular diseases and death worldwide. Redox imbalance and lipid peroxidation could play key roles in atherosclerosis by promoting a bundle of responses, including endothelial activation, inflammation, and foam cell formation. The oxidation of polyunsaturated fatty acids generates various lipid oxidation products such as reactive carbonyl species (RCS), including 4-hydroxy alkenals, malondialdehyde, and acrolein. RCS covalently bind to nucleophilic groups of nucleic acids, phospholipids, and proteins, modifying their structure and activity and leading to their progressive dysfunction. Protein lipoxidation is the non-enzymatic post-translational modification of proteins by RCS. Low-density lipoprotein (LDL) oxidation and apolipoprotein B (apoB) modification by RCS play a major role in foam cell formation. Moreover, oxidized LDLs are a source of RCS, which form adducts on a huge number of proteins, depending on oxidative stress intensity, the nature of targets, and the availability of detoxifying systems. Many systems are affected by lipoxidation, including extracellular matrix components, membranes, cytoplasmic and cytoskeletal proteins, transcription factors, and other components. The mechanisms involved in lipoxidation-induced vascular dysfunction are not fully elucidated. In this review, we focus on protein lipoxidation during atherogenesis.
Collapse
Affiliation(s)
- Anne Nègre-Salvayre
- Inserm Unité Mixte de Recherche (UMR), 1297 Toulouse, Centre Hospitalier Universitaire (CHU) Rangueil—BP 84225, 31432 Toulouse CEDEX 4, France;
- Faculty of Medicine, University of Toulouse, 31432 Toulouse, France
| | - Robert Salvayre
- Inserm Unité Mixte de Recherche (UMR), 1297 Toulouse, Centre Hospitalier Universitaire (CHU) Rangueil—BP 84225, 31432 Toulouse CEDEX 4, France;
- Faculty of Medicine, University of Toulouse, 31432 Toulouse, France
| |
Collapse
|
10
|
Lankin VZ, Sharapov MG, Tikhaze AK, Goncharov RG, Antonova OA, Konovalova GG, Novoselov VI. Dicarbonyl-Modified Low-Density Lipoproteins Are Key Inducers of LOX-1 and NOX1 Gene Expression in the Cultured Human Umbilical Vein Endotheliocytes. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:2125-2136. [PMID: 38462455 DOI: 10.1134/s0006297923120143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/24/2023] [Accepted: 09/26/2023] [Indexed: 03/12/2024]
Abstract
Expression of LOX-1 and NOX1 genes in the human umbilical vein endotheliocytes (HUVECs) cultured in the presence of low-density lipoproteins (LDL) modified with various natural dicarbonyls was investigated for the first time. It was found that among the investigated dicarbonyl-modified LDLs (malondialdehyde (MDA)-modified LDLs, glyoxal-modified LDLs, and methylglyoxal-modified LDLs), the MDA-modified LDLs caused the greatest induction of the LOX-1 and NOX1 genes, as well as of the genes of antioxidant enzymes and genes of proapoptotic factors in HUVECs. Key role of the dicarbonyl-modified LDLs in the molecular mechanisms of vascular wall damage and endothelial dysfunction is discussed.
Collapse
Affiliation(s)
- Vadim Z Lankin
- Chazov National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | - Mars G Sharapov
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| | - Alla K Tikhaze
- Chazov National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | - Ruslan G Goncharov
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Olga A Antonova
- Chazov National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | - Galina G Konovalova
- Chazov National Medical Research Center for Cardiology, Moscow, 121552, Russia
| | - Vladimir I Novoselov
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| |
Collapse
|
11
|
Yang J, Cai R, Xun J, Zhang R, Liu L, Shen Y, Qi T, Wang Z, Song W, Tang Y, Sun J, Xu S, Zhao B, Lu H, Chen J. Elevated indoleamine 2,3-dioxygenase activity is associated with endothelial dysfunction in people living with HIV and ROS production in human aortic endothelial cells in vitro. Drug Discov Ther 2023; 17:312-319. [PMID: 37880104 DOI: 10.5582/ddt.2023.01069] [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] [Indexed: 10/27/2023]
Abstract
The precise role of indoleamine 2,3-dioxygenase (IDO) in cardiovascular diseases (CVD) among people living with HIV (PLWH) is still under debate, despite recognized links. This study aimed to investigate the impact of elevated IDO activity on endothelial dysfunction in PLWH. A total of 38 PLWH, who had not previously received anti-retroviral therapy (ART), were enrolled in the study. These participants were monitored for 36 months following the initiation of ART. Measurements including plasma levels of IDO activity, markers of endothelial dysfunction, inflammatory factors, and lipids. In vitro, human aortic endothelial cells (HAEC) were exposed to interferon-γ, an IDO inhibitor, a kynurenine 3-hydroxylase (KMO) inhibitor, as well as different concentrations of kynurenine. Pre-ART, PLWH demonstrated notably elevated plasma concentrations of soluble intercellular adhesion molecule 1 (sICAM-1), soluble vascular cell adhesion molecule 1(sVCAM-1), and IDO activity in comparison to healthy controls. Post-ART, both IDO activity and sICAM-1 levels experienced a significant decrease, with IDO activity reaching levels comparable to those observed in healthy controls. Furthermore, a positive correlation was observed between IDO activity and sICAM-1 (p = 0.0002), as well as sVCAM-1 (p < 0.0001) before ART. In vitro, the augmentation of kynurenine concentration in the medium and the induction of IDO expression in HAEC resulted in increased production of reactive oxygen species (ROS), with minimal impact on endothelial dysfunction. From these findings, it can be concluded that long-term ART has the potential to restore the heightened IDO activity observed in PLWH. The overexpression of IDO primarily influences the expression of ROS in HAEC.
Collapse
Affiliation(s)
- Junyang Yang
- Department of Infectious and Immune Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Rentian Cai
- Department of Infectious Disease, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Jingna Xun
- Department of Infectious and Immune Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Renfang Zhang
- Department of Infectious and Immune Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Li Liu
- Department of Infectious and Immune Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yinzhong Shen
- Department of Infectious and Immune Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Tangkai Qi
- Department of Infectious and Immune Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Zhenyan Wang
- Department of Infectious and Immune Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Wei Song
- Department of Infectious and Immune Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yang Tang
- Department of Infectious and Immune Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jianjun Sun
- Department of Infectious and Immune Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Shuibao Xu
- Department of Infectious and Immune Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Bihe Zhao
- Department of Infectious and Immune Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Hongzhou Lu
- Department of Infectious Diseases and Nursing Research Institution, National Clinical Research Center for Infectious Diseases, The Third People's Hospital of Shenzhen, Shenzhen, China
| | - Jun Chen
- Department of Infectious and Immune Diseases, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Lankin VZ, Tikhaze AK, Konovalova GG. Differences in Structural Changes and Pathophysiological Effects of Low-Density Lipoprotein Particles upon Accumulation of Acylhydroperoxy Derivatives in Their Outer Phospholipid Monolayer or upon Modification of Apoprotein B-100 by Natural Dicarbonyls. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:1910-1919. [PMID: 38105208 DOI: 10.1134/s0006297923110196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/20/2023] [Accepted: 09/20/2023] [Indexed: 12/19/2023]
Abstract
Nanoparticles of the lipid-transporting system of the organism, low-density lipoproteins (LDL) of blood plasma, are prone to free radical peroxidation with formation of their main modified forms - oxidized LDL itself (containing hydroperoxy-acyls in phospholipids of the outer layer of particles) and dicarbonyl-modified LDL (apoprotein B-100 in which chemically modified via the Maillard reaction). Based on the study of free radical oxidation kinetics of LDLs, it was found that the existing in the literature designation of "oxidized lipoproteins" is incorrect because it does not reveal the nature of oxidative modification of LDLs. It was shown in this study that the "atherogenic" LDLs (particles of which are actively captured by the cultured macrophages) are not the oxidized LDL (in which LOOH-derivatives of phospholipids are formed by enzymatic oxidation by C-15 lipoxygenase of rabbit reticulocytes), but dicarbonyl-modified LDLs. Important role of the dicarbonyl-modified LDLs in the molecular mechanisms of atherogenesis and endothelial dysfunction is discussed.
Collapse
Affiliation(s)
- Vadim Z Lankin
- Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia.
| | - Alla K Tikhaze
- Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
| | - Galina G Konovalova
- Chazov National Medical Research Center of Cardiology, Ministry of Health of the Russian Federation, Moscow, 121552, Russia
| |
Collapse
|
13
|
Villalobos-Labra R, Liu R, Spaans F, Sáez T, Semeria Maitret T, Quon A, Sawamura T, Cooke CLM, Davidge ST. Placenta-Derived Extracellular Vesicles From Preeclamptic Pregnancies Impair Vascular Endothelial Function via Lectin-Like Oxidized LDL Receptor-1. Hypertension 2023; 80:2226-2238. [PMID: 37615097 DOI: 10.1161/hypertensionaha.123.21205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 08/08/2023] [Indexed: 08/25/2023]
Abstract
BACKGROUND Preeclampsia is a complex syndrome that includes maternal vascular dysfunction. Syncytiotrophoblast-derived extracellular vesicles from preeclampsia placentas (preeclampsia-STBEVs) were shown to induce endothelial dysfunction, but an endothelial transmembrane mediator is still unexplored. The LOX-1 (lectin-like oxidized low-density lipoprotein receptor-1) is a transmembrane scavenger receptor that can cause endothelial dysfunction, and its expression is increased in the endothelium of preeclampsia women. In this study, we hypothesized that LOX-1 mediates the effects of preeclampsia-STBEVs on endothelial function. METHODS Preeclampsia-STBEVs were collected by perfusion of placentas from women with preeclampsia and in vitro and ex vivo endothelial cell function were assessed. RESULTS In human umbilical vein endothelial cells, inhibition of LOX-1 with LOX-1 blocking antibody (TS20) reduced the uptake of preeclampsia-STBEVs (61.3±8.8%). TS20 prevented the activation of ERK (extracellular signal-regulated kinase, a kinase downstream of LOX-1) and reduced the activation of NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells; 21.1±8.0%) and nitrative stress (23.2±10.3%) that was induced by preeclampsia-STBEVs. Vascular function was assessed by wire myography in isolated mesenteric arteries from pregnant rats that were incubated overnight with preeclampsia-STBEVs±TS20. TS20 prevented endothelium-dependent vasodilation impairment induced by preeclampsia-STBEVs. Nitric oxide contribution to the relaxation was reduced by preeclampsia-STBEVs, which was prevented by TS20. Superoxide dismutase or apocynin, an inhibitor of NOX (nicotinamide adenine dinucleotide phosphate oxidase), restored the impaired endothelium-dependent vasodilation in arteries exposed to preeclampsia-STBEVs. CONCLUSIONS Taken together, our findings demonstrate that LOX-1 mediates the endothelial dysfunction induced by preeclampsia-STBEVs. Our study further expands on the mechanisms that may lead to adverse outcomes in preeclampsia and proposes LOX-1 as a potential target for future interventions.
Collapse
Affiliation(s)
- Roberto Villalobos-Labra
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Escuela de Medicina sede San Felipe (R.V.-L.), Universidad de Valparaíso, Chile
| | - Ricky Liu
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Department of Physiology (R.L., S.T.D.), University of Alberta, Edmonton, Canada
| | - Floor Spaans
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| | - Tamara Sáez
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Departamento de Medicina Interna (T. Sáez), Universidad de Valparaíso, Chile
- Centro de Investigaciones Biomédicas, Escuela de Medicina, Facultad de Medicina (T. Sáez), Universidad de Valparaíso, Chile
| | - Tamara Semeria Maitret
- Department of Laboratory Medicine and Pathology (T.S.M.), University of Alberta, Edmonton, Canada
| | - Anita Quon
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| | - Tatsuya Sawamura
- Departments of Molecular Pathophysiology and Life Innovation, Shinshu University, Matsumoto, Japan (T. Sawamura)
| | - Christy-Lynn M Cooke
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
| | - Sandra T Davidge
- Department of Obstetrics and Gynecology (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute (R.V.-L., R.L., F.S., T. Sáez, A.Q., C.-L.M.C., S.T.D.), University of Alberta, Edmonton, Canada
- Department of Physiology (R.L., S.T.D.), University of Alberta, Edmonton, Canada
| |
Collapse
|
14
|
Grande T, Vornoli A, Lubrano V, Vizzarri F, Raffaelli A, Gabriele M, Novoa J, Sandoval C, Longo V, Echeverria MC, Pozzo L. Chlamydomonas agloeformis from the Ecuadorian Highlands: Nutrients and Bioactive Compounds Profiling and In Vitro Antioxidant Activity. Foods 2023; 12:3147. [PMID: 37685081 PMCID: PMC10487033 DOI: 10.3390/foods12173147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/27/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
Green microalgae are single-celled eukaryotic organisms that, in recent years, are becoming increasingly important in the nutraceutical, cosmetic, and pharmaceutical fields because of their high content of bioactive compounds. In this study, a particular green microalga was isolated from freshwater highland lakes of Ecuador and morphologically and molecularly identified as Chlamydomonas agloeformis (ChA), and it was studied for nutritional and nutraceutical properties. The phenolic composition and the fatty acids profile of lyophilized cells were determined. The methanolic extract was analyzed for the phenolic compounds profile and the antioxidant capacity by means of in vitro tests. Finally, Human Microvascular Endothelial Cells (HMEC-1) were exploited to explore the capacity of ChA to reduce the endothelial damage induced by oxidized LDL-mediated oxidative stress. The extract showed a good antioxidant ability thanks to the high content in polyphenolic compounds. The observed decrease in HMEC-1 cells endothelial damage also was probably due to the antioxidant compounds present in the extract. Based on the outcomes of our in vitro assays, ChA demonstrated to be a promising source of bioactive compounds possessing exceptional antioxidant capacities which make it a prospective functional food.
Collapse
Affiliation(s)
- Teresa Grande
- Institute of Agricultural Biology and Biotechnology-National Research Council (IBBA-CNR), Via Moruzzi 1, 56124 Pisa, Italy; (T.G.); (A.V.); (A.R.); (M.G.); (V.L.)
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Andrea Vornoli
- Institute of Agricultural Biology and Biotechnology-National Research Council (IBBA-CNR), Via Moruzzi 1, 56124 Pisa, Italy; (T.G.); (A.V.); (A.R.); (M.G.); (V.L.)
| | - Valter Lubrano
- Fondazione G. Monasterio, CNR/Regione Toscana, 56124 Pisa, Italy;
| | - Francesco Vizzarri
- National Agricultural and Food Centre Nitra, Hlohovecká 2, 95141 Lužianky, Slovakia;
| | - Andrea Raffaelli
- Institute of Agricultural Biology and Biotechnology-National Research Council (IBBA-CNR), Via Moruzzi 1, 56124 Pisa, Italy; (T.G.); (A.V.); (A.R.); (M.G.); (V.L.)
- Crop Science Research Center, Scuola Superiore Sant’Anna, Piazza Martiri della Libertà 33, 56127 Pisa, Italy
| | - Morena Gabriele
- Institute of Agricultural Biology and Biotechnology-National Research Council (IBBA-CNR), Via Moruzzi 1, 56124 Pisa, Italy; (T.G.); (A.V.); (A.R.); (M.G.); (V.L.)
| | - Jeniffer Novoa
- eCIER Research Group, Department of Biotechnology, Universidad Técnica del Norte, Av. 17 de Julio 5–21 y Gral. José María Córdova, Ibarra 100150, Ecuador; (J.N.); (C.S.); (M.C.E.)
| | - Carla Sandoval
- eCIER Research Group, Department of Biotechnology, Universidad Técnica del Norte, Av. 17 de Julio 5–21 y Gral. José María Córdova, Ibarra 100150, Ecuador; (J.N.); (C.S.); (M.C.E.)
| | - Vincenzo Longo
- Institute of Agricultural Biology and Biotechnology-National Research Council (IBBA-CNR), Via Moruzzi 1, 56124 Pisa, Italy; (T.G.); (A.V.); (A.R.); (M.G.); (V.L.)
| | - Maria Cristina Echeverria
- eCIER Research Group, Department of Biotechnology, Universidad Técnica del Norte, Av. 17 de Julio 5–21 y Gral. José María Córdova, Ibarra 100150, Ecuador; (J.N.); (C.S.); (M.C.E.)
| | - Luisa Pozzo
- Institute of Agricultural Biology and Biotechnology-National Research Council (IBBA-CNR), Via Moruzzi 1, 56124 Pisa, Italy; (T.G.); (A.V.); (A.R.); (M.G.); (V.L.)
| |
Collapse
|
15
|
Lankin VZ, Konovalova GG, Domogatsky SP, Tikhaze AK, Klots IN, Ezhov MV. Clearance and Utilization of Dicarbonyl-Modified LDL in Monkeys and Humans. Int J Mol Sci 2023; 24:10471. [PMID: 37445648 DOI: 10.3390/ijms241310471] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
The kinetics of elimination of various dicarbonyl-modified low-density lipoproteins from the bloodstream of Macaca mulatta monkeys were investigated. The low-density lipoproteins (LDL) in the monkey blood plasma were isolated by density gradient ultracentrifugation and labeled in vitro with the fluorescent dye FITC; thereupon, they were modified with different natural low molecular-weight dicarbonyls: malondialdehyde (MDA), glyoxal, or methylglyoxal. The control native FITC-labeled LDL and dicarbonyl-modified FITC-labeled LDL were injected into the monkey's ulnar vein; thereafter, blood samples were taken at fixed time intervals during 24 h. The plasma level of FITC-labeled LDL was determined with spectrofluorimetry. The study established that glyoxal- and monkeysglyoxal-labeled LDL circulated in monkey virtually at the same time as native (non-modified) LDL. In contrast, MDA-modified LDL disappeared from the blood extremely rapidly. Administration of the PCSK9 inhibitor involocumab (which increases LDL utilization) to patients with coronary heart disease (CHD) was found to significantly reduce levels of MDA-modified LDL.
Collapse
Affiliation(s)
- Vadim Z Lankin
- Department for Free Radical Biochemistry, E.I. Chazov' National Medical Research Center of Cardiology, Russian Ministry of Health, Moscow 121552, Russia
| | - Galina G Konovalova
- Department for Free Radical Biochemistry, E.I. Chazov' National Medical Research Center of Cardiology, Russian Ministry of Health, Moscow 121552, Russia
| | - Sergey P Domogatsky
- Department for Free Radical Biochemistry, E.I. Chazov' National Medical Research Center of Cardiology, Russian Ministry of Health, Moscow 121552, Russia
| | - Alla K Tikhaze
- Department for Free Radical Biochemistry, E.I. Chazov' National Medical Research Center of Cardiology, Russian Ministry of Health, Moscow 121552, Russia
| | - Igor N Klots
- Research Institute of Medical Primatology, National Research Center "Kurchatov' Institute", Sochi 354376, Russia
| | - Marat V Ezhov
- Laboratory of Lipid Disorders, E.I. Chazov' National Medical Research Center of Cardiology, Russian Ministry of Health, Moscow 121552, Russia
| |
Collapse
|
16
|
Vornoli A, Grande T, Lubrano V, Vizzarri F, Gorelli C, Raffaelli A, Della Croce CM, Baca SZ, Sandoval C, Longo V, Pozzo L, Echeverria C. In Vitro Characterization of Antioxidant, Antibacterial and Antimutagenic Activities of the Green Microalga Ettlia pseudoalveolaris. Antioxidants (Basel) 2023; 12:1308. [PMID: 37372038 DOI: 10.3390/antiox12061308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/07/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023] Open
Abstract
Recently, green microalgae have gained importance due to their nutritional and bioactive compounds, which makes them some of the most promising and innovative functional foods. The aim of this study was to evaluate the chemical profile and the in vitro antioxidant, antimicrobial and antimutagenic activity of an aqueous extract of the green microalga Ettlia pseudoalveolaris, obtained from the freshwater lakes of the Ecuadorian Highlands. Human microvascular endothelial cells (HMEC-1) were used to determine the ability of the microalga to reduce the endothelial damage caused by hydrogen peroxide-induced oxidative stress. Furthermore, the eukaryotic system Saccharomyces cerevisiae was used to evaluate the possible cytotoxic, mutagenic and antimutagenic effect of E. pseudoalveolaris. The extract showed a notable antioxidant capacity and a moderate antibacterial activity mostly due to the high content in polyphenolic compounds. It is likely that the antioxidant compounds present in the extract were also responsible for the observed reduction in endothelial damage of HMEC-1 cells. An antimutagenic effect through a direct antioxidant mechanism was also found. Based on the results of in vitro assays, E. pseudoalveolaris proved to be a good source of bioactive compounds and antioxidant, antibacterial and antimutagenic capacities making it a potential functional food.
Collapse
Affiliation(s)
- Andrea Vornoli
- Institute of Agricultural Biology and Biotechnology (IBBA), National Research Council (CNR), Via Moruzzi 1, 56124 Pisa, Italy
| | - Teresa Grande
- Institute of Agricultural Biology and Biotechnology (IBBA), National Research Council (CNR), Via Moruzzi 1, 56124 Pisa, Italy
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Viale Morgagni 50, 50134 Florence, Italy
| | - Valter Lubrano
- Fondazione G. Monasterio, CNR/Regione Toscana, 56124 Pisa, Italy
| | - Francesco Vizzarri
- National Agricultural and Food Centre Nitra, Hlohoveck'a 2, 95141 Lužianky, Slovakia
| | - Chiara Gorelli
- Institute of Agricultural Biology and Biotechnology (IBBA), National Research Council (CNR), Via Moruzzi 1, 56124 Pisa, Italy
| | - Andrea Raffaelli
- Institute of Agricultural Biology and Biotechnology (IBBA), National Research Council (CNR), Via Moruzzi 1, 56124 Pisa, Italy
- Crop Science Research Center, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, 56127 Pisa, Italy
| | - Clara Maria Della Croce
- Institute of Agricultural Biology and Biotechnology (IBBA), National Research Council (CNR), Via Moruzzi 1, 56124 Pisa, Italy
| | - Santiago Zarate Baca
- eCIER Research Group, Department of Biotechnology, Universidad Técnica del Norte, Av. 17 de Julio 5-21 y Gral. José María Córdova, Ibarra 100150, Ecuador
| | - Carla Sandoval
- eCIER Research Group, Department of Biotechnology, Universidad Técnica del Norte, Av. 17 de Julio 5-21 y Gral. José María Córdova, Ibarra 100150, Ecuador
| | - Vincenzo Longo
- Institute of Agricultural Biology and Biotechnology (IBBA), National Research Council (CNR), Via Moruzzi 1, 56124 Pisa, Italy
| | - Luisa Pozzo
- Institute of Agricultural Biology and Biotechnology (IBBA), National Research Council (CNR), Via Moruzzi 1, 56124 Pisa, Italy
| | - Cristina Echeverria
- eCIER Research Group, Department of Biotechnology, Universidad Técnica del Norte, Av. 17 de Julio 5-21 y Gral. José María Córdova, Ibarra 100150, Ecuador
| |
Collapse
|
17
|
LOX-1 Activation by oxLDL Induces AR and AR-V7 Expression via NF-κB and STAT3 Signaling Pathways Reducing Enzalutamide Cytotoxic Effects. Int J Mol Sci 2023; 24:ijms24065082. [PMID: 36982155 PMCID: PMC10049196 DOI: 10.3390/ijms24065082] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/18/2023] [Accepted: 02/22/2023] [Indexed: 03/09/2023] Open
Abstract
The oxidized low-density lipoprotein receptor 1 (LOX-1) is one of the most important receptors for modified LDLs, such as oxidated (oxLDL) and acetylated (acLDL) low-density lipoprotein. LOX-1 and oxLDL are fundamental in atherosclerosis, where oxLDL/LOX1 promotes ROS generation and NF-κB activation inducing the expression of IL-6, a STAT3 activator. Furthermore, LOX-1/oxLDL function has been associated with other diseases, such as obesity, hypertension, and cancer. In prostate cancer (CaP), LOX-1 overexpression is associated with advanced stages, and its activation by oxLDL induces an epithelial-mesenchymal transition, increasing angiogenesis and proliferation. Interestingly, enzalutamide-resistant CaP cells increase the uptake of acLDL. Enzalutamide is an androgen receptor (AR) antagonist for castration-resistant prostate cancer (CRPC) treatment, and a high percentage of patients develop a resistance to this drug. The decreased cytotoxicity is promoted in part by STAT3 and NF-κB activation that induces the secretion of the pro-inflammatory program and the expression of AR and its splicing variant AR-V7. Here, we demonstrate for the first time that oxLDL/LOX-1 increases ROS levels and activates NF-κB, inducing IL-6 secretion and the activation of STAT3 in CRPC cells. Furthermore, oxLDL/LOX1 increases AR and AR-V7 expression and decreases enzalutamide cytotoxicity in CRPC. Thus, our investigation suggests that new factors associated with cardiovascular pathologies, such as LOX-1/oxLDL, may also promote important signaling axes for the progression of CRPC and its resistance to drugs used for its treatment.
Collapse
|
18
|
Lankin VZ, Tikhaze AK, Melkumyants AM. Malondialdehyde as an Important Key Factor of Molecular Mechanisms of Vascular Wall Damage under Heart Diseases Development. Int J Mol Sci 2022; 24:ijms24010128. [PMID: 36613568 PMCID: PMC9820205 DOI: 10.3390/ijms24010128] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/12/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
This mini review is devoted to a specific issue: the role of malondialdehyde (MDA)-a secondary product of free radical lipid peroxidation-in the molecular mechanisms of the formation of primary atherosclerotic vascular wall lesions. The principal difference between this review and the available literature is that it discusses in detail the important role in atherogenesis not of "oxidized" LDL (i.e., LDL particles containing lipohydroperoxides), but of LDL particles chemically modified by the natural low-molecular weight dicarbonyl MDA. To confirm this, we consider the data obtained by us earlier, indicating that "atherogenic" are not LDL oxidized as a result of free radical lipoperoxidation and containing lipohydroperoxy derivatives of phospholipids in the outer layer of particles, but LDL whose apoprotein B-100 has been modified due to the chemical reaction of terminal lysine residue amino groups of the apoB-100 with the aldehyde groups of the MDA (Maillard reaction). In addition, we present our original data proving that MDA injures endothelial glycocalyx that suppress the ability of the endothelium to control arterial tone according to changes in wall shear stress. In summary, this mini review for the first time exhaustively discloses the key role of MDA in atherogenesis.
Collapse
|
19
|
Huang Y, Song C, He J, Li M. Research progress in endothelial cell injury and repair. Front Pharmacol 2022; 13:997272. [PMID: 36176426 PMCID: PMC9513221 DOI: 10.3389/fphar.2022.997272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/24/2022] [Indexed: 11/30/2022] Open
Abstract
Endothelial cells, which are important metabolic and endocrine cells, play an important role in regulating vascular function. The occurrence and development of various cardiovascular and cerebrovascular diseases are associated with endothelial dysfunction. However, the underlying mechanism of vascular endothelial injury is not fully understood. It has been reported that the mechanism of endothelial injury mainly involves inflammation and oxidative stress. Moreover, endothelial progenitor cells are regarded as important contributors in repairing damaged endothelium. Multiple interventions (including chemical drugs and traditional Chinese medicines) exert endothelial protection by decreasing the release of inducing factors, suppressing inflammation and oxidative stress, and preventing endothelial cell senescence.
Collapse
Affiliation(s)
- Yongpan Huang
- Medicine School, Changsha Social Work College, Changsha, Hunan, China
| | - Chong Song
- Medicine School, Changsha Social Work College, Changsha, Hunan, China
| | - Jianbin He
- Department of Respiratory and Critical Care Medicine, The First People’s Hospital of Huaihua, Affiliated to University of South China, Huaihua, Hunan, China
- *Correspondence: Jianbin He, ; Min Li,
| | - Min Li
- Medicine School, Changsha Social Work College, Changsha, Hunan, China
- *Correspondence: Jianbin He, ; Min Li,
| |
Collapse
|
20
|
Dicarbonyl-Dependent Modification of LDL as a Key Factor of Endothelial Dysfunction and Atherosclerotic Vascular Wall Damage. Antioxidants (Basel) 2022; 11:antiox11081565. [PMID: 36009284 PMCID: PMC9405452 DOI: 10.3390/antiox11081565] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 08/02/2022] [Accepted: 08/10/2022] [Indexed: 12/05/2022] Open
Abstract
The review presents evidence that the main damage to the vascular wall occurs not from the action of “oxidized” LDL, which contain hydroperoxy acyls in the phospholipids located in their outer layer, but from the action of LDL particles whose apoprotein B-100 is chemically modified with low molecular weight dicarbonyls, such as malondialdehyde, glyoxal, and methylglyoxal. It has been argued that dicarbonyl-modified LDL, which have the highest cholesterol content, are particularly “atherogenic”. High levels of dicarbonyl-modified LDL have been found to be characteristic of some mutations of apoprotein B-100. Based on the reviewed data, we hypothesized a common molecular mechanism underlying vascular wall damage in atherosclerosis and diabetes mellitus. The important role of oxidatively modified LDL in endothelial dysfunction is discussed in detail. In particular, the role of the interaction of the endothelial receptor LOX-1 with oxidatively modified LDL, which leads to the expression of NADPH oxidase, which in turn generates superoxide anion radical, is discussed. Such hyperproduction of ROS can cause destruction of the glycocalyx, a protective layer of endotheliocytes, and stimulation of apoptosis in these cells. On the whole, the accumulated evidence suggests that carbonyl modification of apoprotein B-100 of LDL is a key factor responsible for vascular wall damage leading to atherogenesis and endothelial dysfunction. Possible ways of pharmacological correction of free radical processes in atherogenesis and diabetogenesis are also discussed.
Collapse
|
21
|
Pigazzani F, Gorni D, Dyar KA, Pedrelli M, Kennedy G, Costantino G, Bruno A, Mackenzie I, MacDonald TM, Tietge UJF, George J. The Prognostic Value of Derivatives-Reactive Oxygen Metabolites (d-ROMs) for Cardiovascular Disease Events and Mortality: A Review. Antioxidants (Basel) 2022; 11:antiox11081541. [PMID: 36009260 PMCID: PMC9405117 DOI: 10.3390/antiox11081541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022] Open
Abstract
Oxidative stress participates in the development and exacerbation of cardiovascular diseases (CVD). The ability to promptly quantify an imbalance in an individual reductive-oxidative (RedOx) state could improve cardiovascular risk assessment and management. Derivatives-reactive oxygen metabolites (d-ROMs) are an emerging biomarker of oxidative stress quantifiable in minutes through standard biochemical analysers or by a bedside point-of-care test. The current review evaluates available data on the prognostic value of d-ROMs for CVD events and mortality in individuals with known and unknown CVD. Outcome studies involving small and large cohorts were analysed and hazard ratio, risk ratio, odds ratio, and mean differences were used as measures of effect. High d-ROM plasma levels were found to be an independent predictor of CVD events and mortality. Risk begins increasing at d-ROM levels higher than 340 UCARR and rises considerably above 400 UCARR. Conversely, low d-ROM plasma levels are a good negative predictor for CVD events in patients with coronary artery disease and heart failure. Moreover, combining d-ROMs with other relevant biomarkers routinely used in clinical practice might support a more precise cardiovascular risk assessment. We conclude that d-ROMs represent an emerging oxidative-stress-related biomarker with the potential for better risk stratification both in primary and secondary cardiovascular prevention.
Collapse
Affiliation(s)
- Filippo Pigazzani
- MEMO Research, Division of Molecular and Clinical Medicine, University of Dundee, Dundee DD2 1GZ, UK
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD2 1GZ, UK
- Correspondence: (F.P.); (A.B.)
| | - Davide Gorni
- Research and Development Department, H&D S.r.l., 43124 Parma, Italy
| | - Kenneth A. Dyar
- German Center for Diabetes Research (DZD), 40225 Neuherberg, Germany
- Metabolic Physiology, Institute for Diabetes and Cancer (IDC), Helmholtz Diabetes Center, Helmholtz Zentrum München, 85764 Neuherberg, Germany
| | - Matteo Pedrelli
- CardioMetabol Unit, Department of Laboratory Medicine and Department of Medicine, Karolinska Institutet, 17177 Huddinge, Sweden
- Medicine Unit Endocrinology, Theme Inflammation and Ageing, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - Gwen Kennedy
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee DD2 1GZ, UK
| | | | - Agostino Bruno
- Research and Development Department, Cor.Con. International S.r.l., 43124 Parma, Italy
- Correspondence: (F.P.); (A.B.)
| | - Isla Mackenzie
- MEMO Research, Division of Molecular and Clinical Medicine, University of Dundee, Dundee DD2 1GZ, UK
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD2 1GZ, UK
| | - Thomas M. MacDonald
- MEMO Research, Division of Molecular and Clinical Medicine, University of Dundee, Dundee DD2 1GZ, UK
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD2 1GZ, UK
| | - Uwe J. F. Tietge
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
- Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - Jacob George
- Division of Molecular and Clinical Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD2 1GZ, UK
| |
Collapse
|
22
|
Relationship between Brain Metabolic Disorders and Cognitive Impairment: LDL Receptor Defect. Int J Mol Sci 2022; 23:ijms23158384. [PMID: 35955522 PMCID: PMC9369234 DOI: 10.3390/ijms23158384] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 02/07/2023] Open
Abstract
The low-density-lipoprotein receptor (LDLr) removes low-density lipoprotein (LDL), an endovascular transporter that carries cholesterol from the bloodstream to peripheral tissues. The maintenance of cholesterol content in the brain, which is important to protect brain function, is affected by LDLr. LDLr co-localizes with the insulin receptor and complements the internalization of LDL. In LDLr deficiency, LDL blood levels and insulin resistance increase, leading to abnormal cholesterol control and cognitive deficits in atherosclerosis. Defects in brain cholesterol metabolism lead to neuroinflammation and blood–brain-barrier (BBB) degradation. Moreover, interactions between endoplasmic reticulum stress (ER stress) and mitochondria are induced by ox-LDL accumulation, apolipoprotein E (ApoE) regulates the levels of amyloid beta (Aβ) in the brain, and hypoxia is induced by apoptosis induced by the LDLr defect. This review summarizes the association between neurodegenerative brain disease and typical cognitive deficits.
Collapse
|
23
|
Santamaria-Juarez C, Atonal-Flores F, Diaz A, Sarmiento-Ortega VE, Garcia-Gonzalez M, Aguilar-Alonso P, Lopez-Lopez G, Brambila E, Treviño S. Aortic dysfunction by chronic cadmium exposure is linked to multiple metabolic risk factors that converge in anion superoxide production. Arch Physiol Biochem 2022; 128:748-756. [PMID: 32067514 DOI: 10.1080/13813455.2020.1726403] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
CONTEXT The chronic exposure to Cadmium (Cd) constitute an risk to develop hypertension and cardiovascular diseases associated with the increase of oxidative stress. OBJECTIVE In this study, we investigate the role of metabolic changes produced by exposure to Cd on the endothelial dysfunction via oxidative stress. METHODS Male Wistar rats were exposed to Cd (32.5-ppm) for 2-months. The zoometry and blood pressure were evaluated, also glucose and lipids profiles in serum and vascular reactivity evaluated in isolated aorta rings. RESULTS Rats exposed to Cd showed an increase of blood pressure and biochemical parameters similar to metabolic syndrome. Additionally, rats exposed to Cd showed a reduced relaxation in aortic rings, which was reversed after the addition of SOD and apocynin an inhibitor of NADPH. CONCLUSION The Cd-exposition induced hypertension and endothelial injury by that modifying the vascular relaxation and develop oxidative stress via NADPH oxidase, superoxide and loss nitric oxide bioavailability.
Collapse
Affiliation(s)
- Celeste Santamaria-Juarez
- Department of Pharmacy, Faculty of Chemistry Science, University Autonomous of Puebla, Puebla, Mexico
| | - Fausto Atonal-Flores
- Department of Physiology, Faculty of Medicine, University Autonomous of Puebla, The Volcano, Mexico
| | - Alfonso Diaz
- Department of Pharmacy, Faculty of Chemistry Science, University Autonomous of Puebla, Puebla, Mexico
| | - Victor E Sarmiento-Ortega
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Faculty of Chemistry Science, University Autonomous of Puebla, Puebla, Mexico
| | - Miguel Garcia-Gonzalez
- Department of Pharmacy, Faculty of Chemistry Science, University Autonomous of Puebla, Puebla, Mexico
| | - Patricia Aguilar-Alonso
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Faculty of Chemistry Science, University Autonomous of Puebla, Puebla, Mexico
| | - Gustavo Lopez-Lopez
- Department of Pharmacy, Faculty of Chemistry Science, University Autonomous of Puebla, Puebla, Mexico
| | - Eduardo Brambila
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Faculty of Chemistry Science, University Autonomous of Puebla, Puebla, Mexico
| | - Samuel Treviño
- Laboratory of Chemical-Clinical Investigations, Department of Clinical Chemistry, Faculty of Chemistry Science, University Autonomous of Puebla, Puebla, Mexico
| |
Collapse
|
24
|
Jamuna S, Ashokkumar R, Devaraj SN. Amelioration of C-Reactive Protein and Lectin Like Oxidized Low Density Lipoprotein Receptor Complex Induced Endothelial Dysfunction by Oligomeric Proanthocyanidins. Appl Biochem Biotechnol 2022; 195:2664-2686. [PMID: 35357665 DOI: 10.1007/s12010-021-03792-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2021] [Indexed: 01/08/2023]
Abstract
C-reactive protein (CRP) is a well-established biochemical marker for atherosclerosis. Modification of LDL inside the artery wall favors the elevation of this acute phase protein. Hence, this mechanism is considered an important factor to trigger the monocyte to macrophages differentiation which results in the formation of foam cells. Therefore, this key event should be targeted and focused on how this complex (OxLDL + CRP) proceeds to endothelial dysfunction. Oligomeric proanthocyanidins (OPC) is a well-known cardioprotective flavon-3-ols. The present study is challenged between the cardioprotective roles of OPC against the deleterious effect of OxLDL + CRP complex upon endothelial cells. Protein-protein docking was carried out between CRP and LOX-1. This docked protein complex was again docked with OPC to show the inhibitory mechanism of CRP binding with LOX-1. OPC showed a promising inhibitory mechanism against OxLDL + CRP complex. Docking studies showed that in the absence of ligands (OPC), binding of CRP and LOX-1 was greater and vice versa in the presence of ligands. Based on these molecular docking results, in vitro studies have been carried out. The monolayer of endothelial cells was incubated with THP-1 monocytes for 48 h, induced with OxLDL (10 μg/ml) + CRP (15 μg/ml) and cotreated with OPC (100 μg/ml). Morphological changes, cell migration assay, and capillary tube forming assay were carried out. Myeloperoxidase levels were estimated to determine the adhesion of monocytes onto EC monolayer. RT-PCR analysis of L-Selectin was also done. The quantification of NO levels and analysis of mRNA expressions of eNOS was to determine the nitric oxide demand caused due to OxLDL + CRP complex. LOX-1, scavenger receptor levels were analyzed by mRNA expression. Proinflammatory markers such as IL-6, MCP-1, and IL-1β were studied. Accumulation of ROS levels was measured fluorimetrically using DCF-DA staining. Mitochondrial membrane potential was determined by JC-1 dye and cell cycle analysis was done by FACS analysis. To emphasis the results, the OPC-treated group showed decreased levels of proinflammatory markers, LOX-1 and L-selectin levels. Endothelial nitric oxide levels were increased upon OPC treatment and reduction in the ROS levels was also observed. Endothelial cells apoptosis was prevented by OPC. To conclude, OxLDL + CRP complex inhibitory effects of OPC could maintain the normal homeostasis.
Collapse
Affiliation(s)
- Sankar Jamuna
- Department of Biochemistry, University of Madras, Guindy campus, Chennai, 600025, India
| | - Rathinavel Ashokkumar
- Department of Biochemistry, University of Madras, Guindy campus, Chennai, 600025, India
| | | |
Collapse
|
25
|
Zhu Z, Li J, Tong R, Zhang X, Yu B. Astaxanthin suppresses End MT by LOX-1 pathway in ox-LDL-induced HUVECs. EUR J INFLAMM 2022. [DOI: 10.1177/1721727x221105131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Introduction Astaxanthin (ASX) is carotenoid with the highest antioxidant activity in various cell types and reverse atherosclerosis. However, the roles and detailed mechanisms of ASX in atherosclerosis associated endothelial injury remains unclear. Methods In vitro atherosclerosis model was established in HUVECs by incubation with oxidized low-density lipoprotein (ox-LDL). Cell viability and oxidative stress were measured. The mRNA and protein expressions of lectin-like ox-LDL receptor (LOX-1) and other related genes were determined. Results ox-LDL reduced cell viability of HUVECs, and induced oxidative stress, as evidenced by elevated cellular malondialdehyde (MDA) and decreased superoxide dismutase (SOD). Pretreatment with ASX (50, 100, 200, and 400 μM) markedly reversed the reduction in cell viability and an increase in migration of HUVECs induced by ox-LDL (50 μg/mL). ASX attenuated the increase in the endothelial-to-mesenchymal transition (EndMT) process, as evidenced by increased CD31 and decreased α-SMA and vimentin proteins by ASX treatment in HUVECs. Furthermore, ASX attenuated the increase in MDA and decrease in SOD induced by ox-LDL, increased supernatant NO production, attenuated the increase in iNOS and decrease in eNOS in HUVECs with ox-LDL. ASX enhanced mRNA and protein expressions of the lectin-like ox-LDL receptor (LOX-1), which was dependent on ASX’s antioxidant activity. The inhibitory effect of ASX on EndMT could be abolished by overexpression of LOX-1 in HUVECs induced by ox-LDL. Conclusions Our data speculate that ASX prevents ox-LDL-induced endothelial cell injury and EndMT by inducing antioxidant property (SOD and NO) and decreasing LOX-1 expression.
Collapse
Affiliation(s)
- Zhongsheng Zhu
- Department of Cardiology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Jinyu Li
- Department of Cardiology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Rui Tong
- Department of Cardiology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Xiaorong Zhang
- Department of Cardiology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Bo Yu
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| |
Collapse
|
26
|
Wang Z, Li Y, Yao J, Yu S, Yu H, Men L, Du J. Selenoprotein S attenuates high glucose and/or ox-LDL-induced endothelium injury by regulating Akt/mTOR signaling and autophagy. Int J Biochem Cell Biol 2021; 141:106111. [PMID: 34715363 DOI: 10.1016/j.biocel.2021.106111] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 09/30/2021] [Accepted: 10/22/2021] [Indexed: 12/17/2022]
Abstract
Glucolipid metabolism disorder in diabetes mellitus (DM) causes human endothelial injury and autophagy dysfunction is an important cause of endothelial dysfunction (ED). Selenoprotein S (SelS) could protect endothelium from oxidative stress, inflammatory responses, and apoptosis. This study assessed the effect of SelS on autophagy in glucolipid metabolic disorders and protection of the resulted vascular endothelial injury. The results showed that high glucose (HG), high oxidized low-density lipoprotein (HL), and HG combined with HL (HGL) could reduce viability of human aortic endothelial cells (HAECs), induce HAECs injury and increase SelS expression in a time-dependent manner. HG, HL, and HGL also initially induced autophagy but later reduced it in HAECs, while activity of the Akt/mTOR signaling was inhibited, especially in HGL culture of HAECs. SelS overexpression reduced the endothelial injury and autophagy and activated the Akt/mTOR signaling in HG, HL and HGL-cultured HAECs, compared to the control. Conversely, knockdown of SelS expression had the opposite effects on HAECs. In conclusion, SelS demonstrated a protective effect on endothelial injury induced by high glucose and/or ox-LDL and the underlying molecular events might be related to its regulation of HAECs autophagy by activating the Akt/mTOR signaling. SelS could be a potential intervention target in prevention and treatment of diabetic vascular complications.
Collapse
Affiliation(s)
- Zinan Wang
- Department of Endocrinology, The First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Xigang District, Dalian 116011, China; Department of Nutrition, The First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Xigang District, Dalian 116011, China. sunny27---@163.com
| | - Yu Li
- Department of Endocrinology, The First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Xigang District, Dalian 116011, China; Dalian Key Laboratory of Prevention and Treatment of Metabolic Diseases and the Vascular Complications, 222 Zhongshan Road, Xigang District, Dalian 116011, China.
| | - Junjie Yao
- Department of Endocrinology, The First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Xigang District, Dalian 116011, China; Dalian Key Laboratory of Prevention and Treatment of Metabolic Diseases and the Vascular Complications, 222 Zhongshan Road, Xigang District, Dalian 116011, China.
| | - Shanshan Yu
- Department of Endocrinology, The First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Xigang District, Dalian 116011, China; Dalian Key Laboratory of Prevention and Treatment of Metabolic Diseases and the Vascular Complications, 222 Zhongshan Road, Xigang District, Dalian 116011, China.
| | - Hao Yu
- Department of Endocrinology, The First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Xigang District, Dalian 116011, China; Dalian Key Laboratory of Prevention and Treatment of Metabolic Diseases and the Vascular Complications, 222 Zhongshan Road, Xigang District, Dalian 116011, China.
| | - Lili Men
- Department of Endocrinology, The First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Xigang District, Dalian 116011, China; Dalian Key Laboratory of Prevention and Treatment of Metabolic Diseases and the Vascular Complications, 222 Zhongshan Road, Xigang District, Dalian 116011, China.
| | - Jianling Du
- Department of Endocrinology, The First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Xigang District, Dalian 116011, China; Dalian Key Laboratory of Prevention and Treatment of Metabolic Diseases and the Vascular Complications, 222 Zhongshan Road, Xigang District, Dalian 116011, China.
| |
Collapse
|
27
|
Knockdown of circular RNA hsa_circ_0003204 inhibits oxidative stress and apoptosis through the miR-330-5p/Nod2 axis to ameliorate endothelial cell injury induced by low-density lipoprotein. Cent Eur J Immunol 2021; 46:140-151. [PMID: 34764783 PMCID: PMC8568026 DOI: 10.5114/ceji.2021.108174] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 04/30/2020] [Indexed: 11/17/2022] Open
Abstract
Introduction Atherosclerosis (AS) is the leading cause of cardiovascular disease. Circular RNA hsa_circ_0003204 (hsa_circ_0003204) was elevated in oxidized low-density lipoprotein (ox-LDL)-induced human umbilical vein endothelial cells. However, the role and molecular mechanism of hsa_circ_0003204 in the AS process have not been studied. Material and methods Human primary aortic endothelial cells (HAECs) were treated with low-density lipoprotein (ox-LDL) to establish the AS model. The viability of ox-LDL-induced HAECs was assessed by counting kit-8 (CCK8) assay. Reactive oxygen species (ROS), malondialdehyde (MDA) and superoxide dismutase (SOD) levels in ox-LDL-induced HAECs supernatant were evaluated with the relevant kits. The apoptosis of ox-LDL-induced HAECs was determined via flow cytometry assay. The expression of hsa_circ_0003204, miR-330-5p, and nucleotide-binding oligomerization domain 2 (Nod2) was analyzed through quantitative real-time polymerase chain reaction (qRT-PCR). The relationship between hsa_circ_0003204 or Nod2 and miR-330-5p was verified by dual-luciferase reporter assay. Protein level of Nod2 was detected using western blot analysis. Results Hsa_circ_0003204 and Nod2 were upregulated while miR-330-5p was decreased in ox-LDL-induced HAECs. Hsa_circ_0003204 depletion restrained the oxidative stress and apoptosis of ox-LDL-induced HAECs. Notably, hsa_circ_0003204 regulated Nod2 expression via sponging miR-330-5p in HAECs. Moreover, miR-330-5p inhibition restored the constraint of the oxidative stress and apoptosis of ox-LDL-induced HAECs caused by hsa_circ_0003204 silencing. Additionally, miR-330-5p targeted Nod2 and Nod2 enhancement abolished the repressive effects of miR-330-5p overexpression on the oxidative stress and apoptosis of ox-LDL-induced HAECs. Conclusions Hsa_circ_0003204 exhaustion mitigated endothelial cell injury through suppressing the oxidative stress and apoptosis in ox-LDL-induced HAECs via the miR-330-5p/Nod2 axis.
Collapse
|
28
|
Marchenko VA, Barashkova SV, Zelinskaya IA, Toropova YG, Ramsay ES, Zhilinskaya IN. [Modulation of endothelial factors activity in human endothelial cells in influenza A(H1N1)pdm09 virus infection]. Vopr Virusol 2021; 66:198-210. [PMID: 34251157 DOI: 10.36233/0507-4088-48] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 07/09/2021] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Influenza A virus infection can lead to endothelial dysfunction (ED), including apoptosis of endothelial cells and modulation of endothelial factor activities. Affected biochemical factors may include those playing important roles in vascular homeostasis. However, the effect of this pathogen on the expression pattern of key endothelial factors is still unknown.The aim of this work was to study the expression of endothelial nitric oxide synthase (eNOS) and plasminogen activator inhibitor-1 (PAI-1, serpin E1) in the EA.hy926 endothelial cells. RESEARCH OBJECTIVES to assess expression of eNOS and PAI-1 in endothelial cells infected with influenza virus A(H1N1)pdm09, and to identify homologous fragments in structure of viral proteins and endothelial factors. MATERIAL AND METHODS Cells were infected with influenza virus A/St. Petersburg/48/16 (H1N1)pdm09 and analyzed in dynamics in 6, 12, 18, 24, 48, and 72 hrs post infection (hpi). Detection of endothelial factors expression levels was performed by immunocytochemical method (ICC) using antibodies for eNOS and PAI-1 while quantitative assessment of expression levels was carried out by program Nis-Elements F3.2 («Nikon», Japan). The search for homologous sequences between viral proteins and eNOS and PAI-1 was performed by computer comparison. Sequences were analyzed as fragments 12 amino acid residues (aar) in length. RESULTS AND DISCUSSION eNOS expression in infected cells had decreased to 7.9% by 6 hpi (control was taken as 100%) to 3.3% at 72 hpi. PAI-1 expression varied significantly over the course of the experiment: by 6 hpi it had decreased to 49.6%, and to 43.2% by 12 hpi. Later PAI-1 levels were: 116.3% (18 hpi); 18.9% (24 hpi); 23.5% (48 hpi), and 35% (72 hpi). CONCLUSION These results indicate that influenza A infection of endothelial cells causes a significant decrease in eNOS expression, while modulating PAI-1 one. The described phenomenon can be used in the further development of directions of pathogenetic therapy of vascular complications of infection caused by this pathogen.
Collapse
Affiliation(s)
- V A Marchenko
- FSBI «A.A. Smorodintsev Research Institute of Influenza» of the Ministry of Health of Russia
| | - S V Barashkova
- SPB SBIH «K.A. Rauhfus Children's Municipal Multi-Specialty Clinical Center of High Medical Technologies»
| | - I A Zelinskaya
- FSBI «Almazov National Medical Research Centre» of the Ministry of Health of Russia
| | - Ya G Toropova
- FSBI «Almazov National Medical Research Centre» of the Ministry of Health of Russia
| | - E S Ramsay
- FSBI «A.A. Smorodintsev Research Institute of Influenza» of the Ministry of Health of Russia
| | - I N Zhilinskaya
- FSBI «A.A. Smorodintsev Research Institute of Influenza» of the Ministry of Health of Russia
| |
Collapse
|
29
|
Zhu HY, Hong FF, Yang SL. The Roles of Nitric Oxide Synthase/Nitric Oxide Pathway in the Pathology of Vascular Dementia and Related Therapeutic Approaches. Int J Mol Sci 2021; 22:ijms22094540. [PMID: 33926146 PMCID: PMC8123648 DOI: 10.3390/ijms22094540] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/18/2021] [Accepted: 04/21/2021] [Indexed: 12/16/2022] Open
Abstract
Vascular dementia (VaD) is the second most common form of dementia worldwide. It is caused by cerebrovascular disease, and patients often show severe impairments of advanced cognitive abilities. Nitric oxide synthase (NOS) and nitric oxide (NO) play vital roles in the pathogenesis of VaD. The functions of NO are determined by its concentration and bioavailability, which are regulated by NOS activity. The activities of different NOS subtypes in the brain are partitioned. Pathologically, endothelial NOS is inactivated, which causes insufficient NO production and aggravates oxidative stress before inducing cerebrovascular endothelial dysfunction, while neuronal NOS is overactive and can produce excessive NO to cause neurotoxicity. Meanwhile, inflammation stimulates the massive expression of inducible NOS, which also produces excessive NO and then induces neuroinflammation. The vicious circle of these kinds of damage having impacts on each other finally leads to VaD. This review summarizes the roles of the NOS/NO pathway in the pathology of VaD and also proposes some potential therapeutic methods that target this pathway in the hope of inspiring novel ideas for VaD therapeutic approaches.
Collapse
Affiliation(s)
- Han-Yan Zhu
- Department of Physiology, College of Medicine, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China;
- Queen Marry College, College of Medicine, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China
| | - Fen-Fang Hong
- Teaching Center, Department of Experimental, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China
- Correspondence: (F.-F.H.); (S.-L.Y.)
| | - Shu-Long Yang
- Department of Physiology, College of Medicine, Nanchang University, 461 Bayi Avenue, Nanchang 330006, China;
- Correspondence: (F.-F.H.); (S.-L.Y.)
| |
Collapse
|
30
|
Dou F, Wu B, Chen J, Liu T, Yu Z, Chen C. PPAR α Targeting GDF11 Inhibits Vascular Endothelial Cell Senescence in an Atherosclerosis Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:2045259. [PMID: 33728018 PMCID: PMC7935606 DOI: 10.1155/2021/2045259] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 02/08/2021] [Accepted: 02/15/2021] [Indexed: 01/01/2023]
Abstract
Atherosclerosis (AS) is a complex vascular disease that seriously harms the health of the elderly. It is closely related to endothelial cell aging, but the role of senescent cells in atherogenesis remains unclear. Studies have shown that peroxisome proliferator-activated receptor alpha (PPARα) inhibits the development of AS by regulating lipid metabolism. Our previous research showed that PPARα was involved in regulating the repair of damaged vascular endothelial cells. Using molecular biology and cell biology approaches to detect senescent cells in atherosclerosis-prone apolipoprotein E-deficient (Apoe -/-) mice, we found that PPARα delayed atherosclerotic plaque formation by inhibiting vascular endothelial cell senescence, which was achieved by regulating the expression of growth differentiation factor 11 (GDF11). GDF11 levels declined with age in several organs including the myocardium, bone, central nervous system, liver, and spleen in mice and participated in the regulation of aging. Our results showed that PPARα inhibited vascular endothelial cell senescence and apoptosis and promoted vascular endothelial cell proliferation and angiogenesis by increasing GDF11 production. Taken together, these results demonstrated that PPARα inhibited vascular endothelial cell aging by promoting the expression of the aging-related protein GDF11, thereby delaying the occurrence of AS.
Collapse
Affiliation(s)
- Fangfang Dou
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, China
| | - Beiling Wu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, China
| | - Jiulin Chen
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, China
| | - Te Liu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, China
| | - Zhihua Yu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, China
| | - Chuan Chen
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, China
| |
Collapse
|
31
|
Luchetti F, Crinelli R, Nasoni MG, Benedetti S, Palma F, Fraternale A, Iuliano L. LDL receptors, caveolae and cholesterol in endothelial dysfunction: oxLDLs accomplices or victims? Br J Pharmacol 2020; 178:3104-3114. [PMID: 32986849 DOI: 10.1111/bph.15272] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 07/29/2020] [Accepted: 09/11/2020] [Indexed: 12/14/2022] Open
Abstract
Oxidized LDLs (oxLDLs) and oxysterols play a key role in endothelial dysfunction and the development of atherosclerosis. The loss of vascular endothelium function negatively impacts vasomotion, cell growth, adhesiveness and barrier functions. While for some of these disturbances, a reasonable explanation can be provided from a mechanistic standpoint, for many others, the molecular mediators that are involved are unknown. Caveolae, specific plasma membrane domains, have recently emerged as targets and mediators of oxLDL-induced endothelial dysfunction. Caveolae and their associated protein caveolin-1 (Cav-1) are involved in oxLDLs/LDLs transcytosis, mainly through the scavenger receptor class B type 1 (SR-B1 or SCARB1). In contrast, oxLDLs endocytosis is mediated by the lectin-like oxidized LDL receptor 1 (LOX-1), whose activity depends on an intact caveolae system. In addition, LOX-1 regulates the expression of Cav-1 and vice versa. On the other hand, oxLDLs may affect cholesterol plasma membrane content/distribution thus influencing caveolae architecture, Cav-1 localization and the associated signalling. Overall, the evidence indicate that caveolae have both active and passive roles in oxLDL-induced endothelial cell dysfunction. First, as mediators of lipid uptake and transfer in the subendothelial space and, later, as targets of changes in composition/dynamics of plasma membrane lipids resulting from increased levels of circulating oxLDLs. Gaining a better understanding of how oxLDLs interact with endothelial cells and modulate caveolae-mediated signalling pathways, leading to endothelial dysfunction, is crucial to find new targets for intervention to tackle atherosclerosis and the related clinical entities. LINKED ARTICLES: This article is part of a themed issue on Oxysterols, Lifelong Health and Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.16/issuetoc.
Collapse
Affiliation(s)
- Francesca Luchetti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Rita Crinelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Maria Gemma Nasoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Serena Benedetti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Francesco Palma
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | | | - Luigi Iuliano
- Department of Medico-Surgical Sciences and Biotechnologies Vascular Biology, Atherothrombosis & Mass Spectrometry, Sapienza University of Rome, Latina, Italy
| |
Collapse
|
32
|
Le Master E, Ahn SJ, Levitan I. Mechanisms of endothelial stiffening in dyslipidemia and aging: Oxidized lipids and shear stress. CURRENT TOPICS IN MEMBRANES 2020; 86:185-215. [PMID: 33837693 PMCID: PMC8168803 DOI: 10.1016/bs.ctm.2020.08.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Vascular stiffening of the arterial walls is well-known as a key factor in aging and the development of cardiovascular disease; however, the role of endothelial stiffness in vascular dysfunction is still an emerging topic. In this review, the authors discuss the impact of dyslipidemia, oxidized lipids, substrate stiffness, age and pro-atherogenic disturbed flow have on endothelial stiffness. Furthermore, we investigate several mechanistic pathways that are key contributors in endothelial stiffness and discuss their physiological effects in the onset of atherogenesis in the disturbed flow regions of the aortic vasculature. The findings in this chapter describe a novel paradigm of synergistic interaction of plasma dyslipidemia/oxidized lipids and pro-atherogenic disturbed shear stress, as well as aging has on endothelial stiffness and vascular dysfunction.
Collapse
Affiliation(s)
- Elizabeth Le Master
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Sang Joon Ahn
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Irena Levitan
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Illinois at Chicago, Chicago, IL, United States.
| |
Collapse
|
33
|
Dou F, Wu B, Sun L, Chen J, Liu T, Yu Z, Chen C. Identification of a novel regulatory pathway for PPARα by RNA-seq characterization of the endothelial cell lipid peroxidative injury transcriptome. Open Biol 2019; 9:190141. [PMID: 31847785 PMCID: PMC6936254 DOI: 10.1098/rsob.190141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Endothelial dysfunction caused by endothelial cell injuries is the initiating factor for atherosclerosis (AS), and lipid peroxidative injury is one of a dominant factor for AS pathogenesis. Using RNA-seq, we compared changes in transcriptome expression before and after endothelial cell injury, and found 311 differentially expressed genes (DEGs), of which 258 genes were upregulated and 53 genes were downregulated. The protein–protein interactions (PPIs) between the genes were analysed using the STRING database, and a PPI network of DEGs was constructed. The relationship distributions among these PPIs were analysed by performing network node statistics. We found that in the top 20 DEGs with high connected protein nodes in the PPI network, 16 were upregulated and 4 were downregulated. Gene ontology (GO) functional enrichment analysis and KEGG pathway enrichment analysis on the DEGs were also performed. By comparing the upregulated expressed genes with high connected protein nodes in the PPI network to those related to endothelial cell lipid damage and repair in the GO analysis, we identified seven genes (NOX4, PPARA, CCL2, PDGFB, IL8, VWF, CD36) and verified their expression levels by real-time polymerase chain reaction. The protein interactions between the seven genes were then analysed using the STRING database. The results predicted that CCL2 interacts with NOX4, PPARα, PDGFβ and VWF individually. Thus, we examined the protein expression levels of CCL2, NOX4, PPARα, PDGFβ and VWF, and found that the expression levels of all proteins were significantly upregulated after the lipid peroxidative injury, with CCL2 and PPARα exhibiting the highest expression levels. Therefore, we investigated the interregulatory relationship between CCL2 and PPARα and their roles in the repair of endothelial cell injury. With the help of gene overexpression and knockdown techniques, we discovered that PPARα promotes the repair of endothelial cell injury by upregulating CCL2 expression in human umbilical vein endothelial cells but that CCL2 cannot regulate PPARα expression. Therefore, we believe that PPARα participates in the repair of endothelial cell lipid peroxidative injury through regulating the expression of CCL2.
Collapse
Affiliation(s)
- Fangfang Dou
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, People's Republic of China
| | - Beiling Wu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, People's Republic of China
| | - Lin Sun
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, People's Republic of China
| | - Jiulin Chen
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, People's Republic of China
| | - Te Liu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, People's Republic of China
| | - Zhihua Yu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, People's Republic of China
| | - Chuan Chen
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200031, People's Republic of China
| |
Collapse
|
34
|
Akhmedov A, Bonetti NR, Reiner MF, Spescha RD, Amstalden H, Merlini M, Gaul DS, Diaz-Cañestro C, Briand-Schumacher S, Spescha RS, Semerano A, Giacalone G, Savarese G, Montecucco F, Kulic L, Nitsch RM, Matter CM, Kullak-Ublick GA, Sessa M, Lüscher TF, Beer JH, Liberale L, Camici GG. Deleterious role of endothelial lectin-like oxidized low-density lipoprotein receptor-1 in ischaemia/reperfusion cerebral injury. J Cereb Blood Flow Metab 2019; 39:2233-2245. [PMID: 30073881 PMCID: PMC6827115 DOI: 10.1177/0271678x18793266] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 06/14/2018] [Accepted: 07/01/2018] [Indexed: 11/17/2022]
Abstract
Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is implicated in cardiovascular disease by modulating apoptosis and oxidative stress. We hypothesized that LOX-1 may be involved in pathophysiology of stroke by mediating ischaemia/reperfusion (I/R)-dependent cell death. Transient middle cerebral artery occlusion (tMCAO) was performed in wild-type (WT) mice, endothelial-specific LOX-1 transgenic mice (eLOX-1TG) and WT animals treated with LOX-1 silencing RNA (siRNA). In WT mice exposed to tMCAO, LOX-1 expression and function were increased in the MCA. Compared to WT animals, eLOX-1TG mice displayed increased stroke volumes and worsened outcome after I/R. Conversely, LOX-1-silencing decreased both stroke volume and neurological impairment. Similarly, in HBMVECs, hypoxia/reoxygenation increased LOX-1 expression, while LOX-1 overexpressing cells showed increased death following hypoxia reoxygenation. Increased caspase-3 activation was observed following LOX-1 overexpression both in vivo and in vitro, thus representing a likely mediator. Finally, monocytes from ischaemic stroke patients exhibited increased LOX-1 expression which also correlated with disease severity. Our data unequivocally demonstrate a key role for LOX-1 in determining outcome following I/R brain damage. Our findings could be corroborated in human brain endothelial cells and monocytes from patients, underscoring their translational relevance and suggesting siRNA-mediated LOX-1 knockdown as a novel therapeutic strategy for stroke patients.
Collapse
Affiliation(s)
- Alexander Akhmedov
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Schlieren, Switzerland
| | - Nicole R Bonetti
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Schlieren, Switzerland
- Department of Internal Medicine, Cantonal Hospital of Baden, Baden, Switzerland
| | - Martin F Reiner
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Schlieren, Switzerland
- Department of Internal Medicine, Cantonal Hospital of Baden, Baden, Switzerland
| | - Remo D Spescha
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Schlieren, Switzerland
| | - Heidi Amstalden
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Schlieren, Switzerland
| | - Mario Merlini
- Gladstone Institute of Neurological Disease, University of California, San Francisco, CA, USA
| | - Daniel S Gaul
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Schlieren, Switzerland
| | - Candela Diaz-Cañestro
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Schlieren, Switzerland
| | | | - Rebecca S Spescha
- Division of Psychiatry Research, University of Zurich, Schlieren, Switzerland
- Zurich Neuroscience Center, University of Zurich, Zurich, Switzerland
| | - Aurora Semerano
- Department of Neurology, San Raffaele Scientific Institute, Milano, Italy
| | - Giacomo Giacalone
- Department of Neurology, San Raffaele Scientific Institute, Milano, Italy
| | - Gianluigi Savarese
- Division of Cardiology, Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- Ospedale Policlinico San Martino, Genoa, Italy
- Centre of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, Italy
| | - Luka Kulic
- Division of Psychiatry Research, University of Zurich, Schlieren, Switzerland
- Zurich Neuroscience Center, University of Zurich, Zurich, Switzerland
| | - Roger M Nitsch
- Division of Psychiatry Research, University of Zurich, Schlieren, Switzerland
- Zurich Neuroscience Center, University of Zurich, Zurich, Switzerland
| | - Christian M Matter
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Schlieren, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Gerd A Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, Switzerland
| | - Maria Sessa
- Department of Neurology, San Raffaele Scientific Institute, Milano, Italy
| | - Thomas F Lüscher
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Schlieren, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, Zurich, Switzerland
| | - Jürg H Beer
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Schlieren, Switzerland
- Department of Internal Medicine, Cantonal Hospital of Baden, Baden, Switzerland
| | - Luca Liberale
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Schlieren, Switzerland
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Giovanni G Camici
- Center for Molecular Cardiology, Schlieren Campus, University of Zurich, Schlieren, Switzerland
- Zurich Neuroscience Center, University of Zurich, Zurich, Switzerland
| |
Collapse
|
35
|
ASK1 Mediates Apoptosis and Autophagy during oxLDL-CD36 Signaling in Senescent Endothelial Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2840437. [PMID: 31772703 PMCID: PMC6854215 DOI: 10.1155/2019/2840437] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/04/2019] [Accepted: 09/11/2019] [Indexed: 01/29/2023]
Abstract
Vessel damage by oxidized low-density lipoprotein (oxLDL) increases reactive oxygen species (ROS) and the membrane receptor cluster of differentiation 36 (CD36), involving various vascular pathological processes. In this study, the role of apoptosis signal-regulating kinase 1 (ASK1) as a cellular effector via the oxLDL-CD36 signaling axis, and its related mechanism as a downstream responder of CD36, was investigated in senescent human aortic endothelial cells (HAECs). To inhibit oxLDL-triggered vascular damage, HAECs and monocytes were treated with the CD36-neutralizing antibody or the ASK1 inhibitor NQDI-1. The oxLDL-triggered increases in ROS and CD36 elevated active ASK1 in the senescent HAECs. The ROS increase induced apoptosis, whereas CD36 neutralization or ASK1 inhibition protected against cell death. The blocking of CD36 increased senescent HAEC autophagy. In monocytes, oxLDL also induced CD36 expression and autophagy, the latter of which still occurred following ASK1 inhibition but not after CD36 neutralization. These findings suggest that oxLDL exposure activates ASK1, as a CD36 downstream responder, to accelerate apoptosis, particularly in senescent HAECs. ASK1's involvement in monocytic autophagy was due to endoplasmic reticulum stress resulting from the oxLDL load, suggesting that oxLDL loading on aged vessels causes atherosclerotic endothelial dysfunction mediated by active ASK1.
Collapse
|
36
|
Bayatpoor ME, Mirzaee S, Karami Abd M, Mohammadi MT, Shahyad S, Bahari Z, Raouf Sarshoori J. Crocin treatment decreased pancreatic atrophy, LOX-1 and RAGE mRNA expression of pancreas tissue in cholesterol-fed and streptozotocin-induced diabetic rats. ACTA ACUST UNITED AC 2019; 17:/j/jcim.ahead-of-print/jcim-2019-0117/jcim-2019-0117.xml. [DOI: 10.1515/jcim-2019-0117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/14/2019] [Indexed: 12/21/2022]
Abstract
AbstractObjectiveOxidative stress in diabetic mellitus is a consequence of oxidative stress, which plays a critical role in the pathogenesis of diabetic tissue damage. Receptors for advanced glycation end products and for oxidized low-density lipoproteins (LDL) have critical contribution in oxidative tissue damage. The present study investigated whether anti-diabetic effects of Crocin via modulation of mRNA expression of RAGE and LOX-1 receptors in diabetic rats.MethodsIn the current study, high-fat cholesterol (HFC) and streptozotocin (40 mg/kg) used to induce type II diabetes. Experimental groups as follows: (Group 1: control); (Group 2: control treatment [Crocin]); (Group 3: DM [STZ]); (Group 4: DM treatment [STZ + Crocin]); (Group 5; DM + HFC [STZ + HFC]); (Group 6; DM + HFC treatment [STZ + HFC + Crocin]). Crocin (20 mg/kg/day, i.p.) administered in treatment groups for 60 days. Serum glucose and cholesterol levels evaluated on days 5, 30 and 60 after induction of DM. Pancreatic tissue from all group removed on day 60 for histological and RT-PCR analysis.ResultsApplication of Crocin significantly decreased serum cholesterol levels on day 60 after induction of DM in diabetic + HFC rats. Moreover, Crocin significantly decreased serum glucose levels on days 30 and 60 both in diabetic and diabetic + HFC rats. Crocin partially prevented the atrophic effects of STZ on both exocrine and endocrine parts of pancreas. Additionally, Crocin significantly decreased LOX-1 and RAGE mRNA expression OF pancreas in diabetic rats.ConclusionThe current study suggested that Crocin suppressed atrophic change of the pancreas by decrease of LOX-1 and RAGE mRNA expression in diabetic rats.
Collapse
Affiliation(s)
- Mohammad Ehsan Bayatpoor
- Student Research Committee, Baqiyatallah University of Medical Sciences, Tehran, Iran (Islamic Republic of)
| | - Saeed Mirzaee
- Student Research Committee, Baqiyatallah University of Medical Sciences, Tehran, Iran (Islamic Republic of)
| | - Mohammad Karami Abd
- Student Research Committee, Baqiyatallah University of Medical Sciences, Tehran, Iran (Islamic Republic of)
| | - Mohammad Taghi Mohammadi
- Department of Physiology and Medical Physics, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Shima Shahyad
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Zahra Bahari
- Department of Physiology and Medical Physics, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Javad Raouf Sarshoori
- Department of Anatomy, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
37
|
Oxidised Low-Density Lipoprotein and Its Receptor-Mediated Endothelial Dysfunction Are Associated with Coronary Artery Lesions in Kawasaki Disease. J Cardiovasc Transl Res 2019; 13:204-214. [PMID: 31428922 DOI: 10.1007/s12265-019-09908-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 08/07/2019] [Indexed: 12/14/2022]
Abstract
The study aimed to investigate the role of oxidised low-density lipoprotein (oxLDL)/lectin-like-oxLDL receptor-1 (LOX-1) in coronary artery lesions (CALs) in Kawasaki disease (KD) and of plasma oxLDL concentration in the early prediction of CALs in KD. This prospective study included 80 KD patients, 20 febrile and 20 healthy children. oxLDL, LOX-1 and other parameters were analysed in the acute phase. Plasma oxLDL concentration and LOX-1 mRNA expression in peripheral blood mononuclear cells (PBMCs) were significantly increased in KD patients compared with febrile and healthy children (P < 0.001 and P = 0.022, respectively), particularly in the group with CALs (P < 0.001 and P = 0.027, respectively). Coronary Z-score was significantly correlated with plasma oxLDL concentration and LOX-1 mRNA expression (r = 0.739 and 0.637, respectively; P < 0.01). The sensitivity and specificity of predicting CALs were 71.4% and 77.2%, respectively, at plasma oxLDL concentration ≥ 12.38 mU/L. oxLDL/LOX-1 may be involved in CAL development. The plasma oxLDL concentration in the acute phase is a potentially useful biological indicator for predicting CAL in KD patients.
Collapse
|
38
|
Gresele P, Momi S, Guglielmini G. Nitric oxide-enhancing or -releasing agents as antithrombotic drugs. Biochem Pharmacol 2019; 166:300-312. [DOI: 10.1016/j.bcp.2019.05.030] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 05/31/2019] [Indexed: 12/16/2022]
|
39
|
Modulation of Nitric Oxide Synthases by Oxidized LDLs: Role in Vascular Inflammation and Atherosclerosis Development. Int J Mol Sci 2019; 20:ijms20133294. [PMID: 31277498 PMCID: PMC6651385 DOI: 10.3390/ijms20133294] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022] Open
Abstract
The maintenance of physiological levels of nitric oxide (NO) produced by eNOS represents a key element for vascular endothelial homeostasis. On the other hand, NO overproduction, due to the activation of iNOS under different stress conditions, leads to endothelial dysfunction and, in the late stages, to the development of atherosclerosis. Oxidized LDLs (oxLDLs) represent the major candidates to trigger biomolecular processes accompanying endothelial dysfunction and vascular inflammation leading to atherosclerosis, though the pathophysiological mechanism still remains to be elucidated. Here, we summarize recent evidence suggesting that oxLDLs produce significant impairment in the modulation of the eNOS/iNOS machinery, downregulating eNOS via the HMGB1-TLR4-Caveolin-1 pathway. On the other hand, increased oxLDLs lead to sustained activation of the scavenger receptor LOX-1 and, subsequently, to NFkB activation, which, in turn, increases iNOS, leading to EC oxidative stress. Finally, these events are associated with reduced protective autophagic response and accelerated apoptotic EC death, which activates atherosclerotic development. Taken together, this information sheds new light on the pathophysiological mechanisms of oxLDL-related impairment of EC functionality and opens new perspectives in atherothrombosis prevention.
Collapse
|
40
|
Zhu Z, Li J, Zhang X. Salidroside protects against ox-LDL-induced endothelial injury by enhancing autophagy mediated by SIRT1-FoxO1 pathway. Altern Ther Health Med 2019; 19:111. [PMID: 31146723 PMCID: PMC6543685 DOI: 10.1186/s12906-019-2526-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 05/21/2019] [Indexed: 02/05/2023]
Abstract
Background Atherosclerosis is a condition with the vascular accumulation of lipid plaques, and its main major contributing factor is endothelial injury induced by oxidized low-density lipoprotein (ox-LDL). Salidroside (SAL) is the primary active ingredient of Rhodiola rosea, and exhibits antioxidant properties on endothelial cells and alleviates atherosclerosis. However, the effect of SAL on autophagy in ox-LDL-induced vascular endothelial injury remains unclear. Here, we investigated the effect and underlying mechanisms of SAL on autophagy in human umbilical vein endothelial cells (HUVECs). Methods HUVECs were incubated with ox-LDL to induce in vitro atherosclerosis model. The cell viability and injury were evaluated by cell counting kit-8 (CCK-8) assay and lactate dehydrogenase (LDH) release assay. The oxidative stress was evaluated by NADPH oxidase, malondialdehyde (MDA) and superoxide dismutase (SOD) activities. Immunofluorescence was performed to detect autophagy using LC3β antibody. Quantitative real-time PCR (qRT-PCR) and western blot were performed to measure the mRNA expressions of SIRT1 and Forkhead box O1 (FOXO1). Nicotinamide (NAM) and AS1842856 were used to inhibit activities of SIRT1 and FOXO1, respectively. Results Exposure of HUVECs to ox-LDL (100 μg/mL) reduced cell viability, increased cellular MDA, and reduced SOD in a concentration-dependent manner. The pretreatment with SAL (20, 50 and 100 μM) significantly enhanced the cell viability and decreased LDH release in HUVECs exposed to ox-LDL (100 μg/mL). ox-LDL induced autophagy in HUVECs, which was further enhanced by pretreatment with SAL. However, SAL attenuated increase in oxidative stress in HUVECs induced by ox-LDL. ox-LDL reduced mRNA and protein expressions of SIRT1 and FOXO1, which could be reversed by SAL. The protective, anti-oxidative and pro-autophagic effects of SAL could be obviously abolished by cotreatment with SIRT1 inhibitor or FOXO1 inhibitor. Conclusion Salidroside shows protective effect on endothelial cell induced by ox-LDL, and the mechanisms might be related to autophagy induction via increasing SIRT1 and FoxO1 expressions.
Collapse
|
41
|
Zhu Z, Li J, Zhang X. Astragaloside IV Protects Against Oxidized Low-Density Lipoprotein (ox-LDL)-Induced Endothelial Cell Injury by Reducing Oxidative Stress and Inflammation. Med Sci Monit 2019; 25:2132-2140. [PMID: 30901320 PMCID: PMC6441302 DOI: 10.12659/msm.912894] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Background Endothelial injury is the main mechanism of atherosclerosis, and is caused by oxidized low-density lipoprotein (ox-LDL). Astragaloside IV (AS-IV) is the primary active ingredient of the Chinese herb Huangqi, and exhibits antioxidant and anti-inflammatory properties in cardiovascular diseases. This study investigated the protective effect of AS-IV in human umbilical vein endothelial cells (HUVECs). Material/Methods HUVEC cells were induced with ox-LDL to establish an in vitro atherosclerosis model. Then HUVECs were pretreated for 1 h with AS-IV at different concentrations (10, 20, and 50 μM) and then exposed to ox-LDL (100 μg/mL) for 48 h. The cell viability, lactate dehydrogenase (LDH) release, apoptosis, migration, intracellular reactive oxygen species (ROS), and NADPH oxidase activity of HUVECs were measured. qRT-PCR was performed to measure the mRNA expressions of Nrf2, HO-1, TNFα, and IL-6. Enzyme-linked immunosorbent assay (ELISA) was performed to measure the supernatant contents of TNFα and IL-6. Results Exposure of HUVECs to ox-LDL reduced cell viability and migration, induced apoptosis, and increased intracellular ROS production and NADPH oxidase. Pretreatment with AS-IV (10, 20, and 50 μM) significantly enhanced the cell viability and migration, suppressed LDH release, apoptosis, ROS production, and NADPH oxidase in HUVECs, in a concentration-dependent manner. The AS-IV (50 μM) alone did not show significant differences from control. AS-IV increased mRNA expressions of Nrf2 and HO-1 and decreased mRNA expressions of TNFα and IL-6 in the ox-LDL-HUEVC cells. Furthermore, AS-IV reduced supernatant contents of TNFα and IL-6. Conclusions Astragaloside IV prevents ox-LDL-induced endothelial cell injury by reducing apoptosis, oxidative stress, and inflammatory response.
Collapse
Affiliation(s)
- Zhongsheng Zhu
- Department of Cardiology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China (mainland)
| | - Jinyu Li
- Department of Cardiology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China (mainland)
| | - Xiaorong Zhang
- Department of Cardiology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China (mainland)
| |
Collapse
|
42
|
OxHDL controls LOX-1 expression and plasma membrane localization through a mechanism dependent on NOX/ROS/NF-κB pathway on endothelial cells. J Transl Med 2019; 99:421-437. [PMID: 30664710 DOI: 10.1038/s41374-018-0151-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 10/17/2018] [Accepted: 10/19/2018] [Indexed: 12/20/2022] Open
Abstract
Systemic inflammatory diseases enhance circulating oxidative stress levels, which results in the oxidation of circulating high-density lipoprotein (oxHDL). Endothelial cell function can be negatively impacted by oxHDL, but the underlying mechanisms for this remain unclear. Some reports indicate that the lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is also a receptor for oxHDL. However, it is unknown if oxHDL induces increased LOX-1 expression at the plasma membrane, as an event that supports endothelial dysfunction. Therefore, the aims of this study were to determine if oxHDL induces plasma-membrane level changes in LOX-1 and, if so, to describe the underlying mechanisms in endothelial cells. Our results demonstrate that the incubation of arterial or vein endothelial cells with oxHDL (and not HDL) induces the increase of LOX-1 expression at the plasma membrane; effect prevented by LOX-1 inhibition. Importantly, same results were observed in endothelial cells from oxHDL-treated rats. Furthermore, the observed oxHDL-induced LOX-1 expression is abolished by the down-regulation of NOX-2 expression with siRNA (and no others NOX isoforms), by the pharmacological inhibition of NAD(P)H oxidase (with DPI or apocynin) or by the inhibition of NF-κB transcription factor. Coherently, LOX-1 expression is augmented by the incubation of endothelial cells with H2O2 or GSSG even in absence of oxHDL, indicating that the NOX-2/ROS/ NF-κB axis is involved. Interestingly, oxHDL incubation also increases TNF-α expression, cytokine that induces LOX-1 expression. Thus, our results suggest a positive feedback mechanism for LOX-1 receptor during inflammatory condition where an oxidative burst will generate oxHDL from native HDL, activating LOX-1 receptor which in turn will increase the expression of NOX-2, TNF-α and LOX-1 receptor at the plasma membrane. In conclusion, oxHDL-induced translocation of LOX-1 to the plasma membrane could constitute an induction mechanism of endothelial dysfunction in systemic inflammatory diseases.
Collapse
|
43
|
Zhou Z, Chen Y, Ni W, Liu T. Upregulation of Nuclear Factor IA Suppresses Oxidized Low-Density Lipoprotein-Induced Endoplasmic Reticulum Stress and Apoptosis in Human Umbilical Vein Endothelial Cells. Med Sci Monit 2019; 25:1009-1016. [PMID: 30721172 PMCID: PMC6373224 DOI: 10.12659/msm.912132] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background Endoplasmic reticulum stress (ERS) is part of the cardiovascular pathological processes, including atherosclerosis. Nuclear factor IA (NFIA) influences atherosclerosis development; however, its effects on ERS remain unknown. This study investigated the effect of NFIA on oxidized low-density lipoprotein (ox-LDL)-induced ERS and apoptosis in endothelial cells. Material/Methods Ox-LDL was used to induce lipotoxicity in human umbilical vein endothelial cells (HUVECs) to establish an in vitro oxidative injury model transfected with pcDNA3.0-NFIA. The cytotoxic response was detected using an assay to determine the release of lactate dehydrogenase (LDH). Morphological changes in cell apoptosis were detected using Hoechst 33258 staining. The proportion of apoptotic cells, releases of reactive oxygen species (ROS), and mitochondrial membrane potential (ΔΨm) were determined using flow cytometry. The expression levels of apoptosis- and ERS-related molecules were detected through Western blotting. Results NFIA expression was downregulated in the in vitro oxidative cell-injury model. Exposure of HUVECs to ox-LDL resulted in a significant increase in apoptosis, decrease in ROS levels, and loss of ΔΨm. Overexpression of NFIA remarkably inhibited ERS and mitochondrial-mediated apoptosis induced by ox-LDL in HUVECs by reversing the effect of ox-LDL on the expression of JNK1, p-JNK1, CHOP, Cyt C, and Bax. Conclusions These results demonstrated that NFIA might have beneficial effects in the prevention of ox-LDL-induced ERS and apoptosis in vascular endothelial cells. This study provided new insights into the mechanism of atherosclerosis.
Collapse
Affiliation(s)
- Zhenyu Zhou
- Department of Cardiology, Central Hospital of Nanchong, The Second Clinical School of North Sichuan Medical College, Nanchong, Sichuan, China (mainland)
| | - Yu Chen
- Comprehensive Ward, Central Hospital of Nanchong, The Second Clinical School of North Sichuan Medical College, Nanchong, Sichuan, China (mainland)
| | - Wei Ni
- Department of Cardiology, Central Hospital of Nanchong, The Second Clinical School of North Sichuan Medical College, Nanchong, Sichuan, China (mainland)
| | - Tao Liu
- Department of Cardiology, Central Hospital of Nanchong, The Second Clinical School of North Sichuan Medical College, Nanchong, Sichuan, China (mainland)
| |
Collapse
|
44
|
Tang ZH, Li TH, Peng J, Zheng J, Li TT, Liu LS, Jiang ZS, Zheng XL. PCSK9: A novel inflammation modulator in atherosclerosis? J Cell Physiol 2018; 234:2345-2355. [PMID: 30246446 DOI: 10.1002/jcp.27254] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/24/2018] [Indexed: 12/11/2022]
Abstract
Proprotein convertase subtilisin/kexin 9 (PCSK9) is the ninth member of the secretory serine protease family. It binds to low-density lipoprotein receptor (LDLR) for endocytosis and lysosome degradation in the liver, resulting in an increasing in circulating LDL-cholesterol (LDL-c) level. Since a PCSK9 induced increase in plasma LDL-c contributes to atherosclerosis, PCSK9 inhibition has become a new strategy in preventing and treating atherosclerosis. However, in addition to the effect of PCSK9 on elevating blood LDL-c levels, accumulating evidence shows that PCSK9 plays an important role in inflammation, likely representing another major mechanism for PCSK9 to promote atherosclerosis. In this review, we discuss the association of PCSK9 and inflammation, and highlight the specific effects of PCSK9 on different vascular cellular components involved in the atherosclerotic inflammation. We also discuss the clinical evidence for the association between PCSK9 and inflammation in atherosclerotic cardiovascular disease. A better understanding of the direct association of PCSK9 with atherosclerotic inflammation might help establish a new role for PCSK9 in vascular biology and identify a novel molecular mechanism for PCSK9 therapy.
Collapse
Affiliation(s)
- Zhi-Han Tang
- Department of Pathophysiology, Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China.,Department of Biochemistry and Molecular Biology, The Libin Cardiovascular Institute of Alberta, The University of Calgary, Health Sciences Center, Calgary, Alberta, Canada
| | - Tao-Hua Li
- Department of Pathophysiology, Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Juan Peng
- Department of Pathophysiology, Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China.,Department of Biochemistry and Molecular Biology, The Libin Cardiovascular Institute of Alberta, The University of Calgary, Health Sciences Center, Calgary, Alberta, Canada
| | - Jie Zheng
- Department of Pathophysiology, Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Ting-Ting Li
- Department of Pathophysiology, Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Lu-Shan Liu
- Department of Pathophysiology, Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Zhi-Sheng Jiang
- Department of Pathophysiology, Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, The Libin Cardiovascular Institute of Alberta, The University of Calgary, Health Sciences Center, Calgary, Alberta, Canada
| |
Collapse
|
45
|
Schmidt A, Bekeschus S, Wende K, Vollmar B, von Woedtke T. A cold plasma jet accelerates wound healing in a murine model of full-thickness skin wounds. Exp Dermatol 2018; 26:156-162. [PMID: 27492871 DOI: 10.1111/exd.13156] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2016] [Indexed: 12/24/2022]
Abstract
Cold plasma has been successfully applied in several fields of medicine that require, for example, pathogen inactivation, implant functionalization or alteration of cellular activity. Previous studies have provided evidence that plasma supports the healing of wounds owing to its beneficial mixtures of reactive species and modulation of inflammation in cells and tissues. To investigate the wound healing activity of an atmospheric pressure plasma jet in vivo, we examined the cold plasma's efficacy on dermal regeneration in a murine model of dermal full-thickness ear wound. Over 14 days, female mice received daily plasma treatment. Quantitative analysis by transmitted light microscopy demonstrated a significantly accelerated wound re-epithelialization at days 3-9 in comparison with untreated controls. In vitro, cold plasma altered keratinocyte and fibroblast migration, while both cell types showed significant stimulation resulting in accelerated closure of gaps in scratch assays. This plasma effect correlated with the downregulation of the gap junctional protein connexin 43 which is thought to be important in the regulation of wound healing. In addition, plasma induced profound changes in adherence junctions and cytoskeletal dynamics as shown by downregulation of E-cadherin and several integrins as well as actin reorganization. Our results theorize cold plasma to be a beneficial treatment option supplementing existing wound therapies.
Collapse
Affiliation(s)
- Anke Schmidt
- Plasma Life Science, Leibniz-Institute for Plasma Science and Technology (INP Greifswald), Greifswald, Germany
| | - Sander Bekeschus
- ZIK Plasmatis, Leibniz-Institute for Plasma Science and Technology (INP Greifswald), Greifswald, Germany
| | - Kristian Wende
- ZIK Plasmatis, Leibniz-Institute for Plasma Science and Technology (INP Greifswald), Greifswald, Germany
| | - Brigitte Vollmar
- Institute for Experimental Surgery, University of Rostock, Rostock, Germany
| | - Thomas von Woedtke
- Plasma Life Science, Leibniz-Institute for Plasma Science and Technology (INP Greifswald), Greifswald, Germany.,ZIK Plasmatis, Leibniz-Institute for Plasma Science and Technology (INP Greifswald), Greifswald, Germany.,Department of Hygiene and Environmental Medicine, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
46
|
Balzan S, Lubrano V. LOX-1 receptor: A potential link in atherosclerosis and cancer. Life Sci 2018; 198:79-86. [PMID: 29462603 DOI: 10.1016/j.lfs.2018.02.024] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 02/07/2018] [Accepted: 02/16/2018] [Indexed: 12/19/2022]
Abstract
Altered production of reactive oxygen species (ROS), causing lipid peroxidation and DNA damage, contributes to the progression of atherosclerosis and cancer. Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is a lectin-like receptor for oxidized low-density lipoproteins (ox-LDL) primarily expressed in endothelial cells and vasculature-rich organs. LOX-1 receptors is a marker for atherosclerosis, and once activated by ox-LDL or other ligands, stimulates the expression of adhesion molecules, pro-inflammatory signaling pathways and proangiogenic proteins, including NF-kB and VEGF, in vascular endothelial cells and macrophages. Several different types of cancer reported LOX-1 gene upregulation, and numerous interplays exist concerning LOX-1 in atherosclerosis, metabolic diseases and cancer. One of them involves NF-kB, an oncogenic protein that regulates the transcription of several inflammatory genes response. In a model of cellular transformation, the MCF10A ER-Src, inhibition of LOX-1 gene reduces NF-kB activation and the inflammatory and hypoxia pathways, suggesting a mechanistic connection between cellular transformation and atherosclerosis. The remodeling proteins MMP-2 and MMP-9 have been found increased in angiogenesis in atherosclerotic plaque and also in human prostate cancer cells. In this review, we outlined the role of LOX-1 in atherogenesis and tumorigenesis as a potential link in these diseases, suggesting that LOX-1 inhibition could represent a promising strategy in the treatment of atherosclerosis and tumors.
Collapse
Affiliation(s)
- Silvana Balzan
- Institute of Clinical Physiology, CNR, Via Moruzzi 1, Pisa 56124, Italy.
| | - Valter Lubrano
- Fondazione CNR/Regione Toscana G. Monasterio, Via Moruzzi 1, Pisa 56124, Italy
| |
Collapse
|
47
|
PRR Function of Innate Immune Receptors in Recognition of Bacteria or Bacterial Ligands. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1112:255-280. [DOI: 10.1007/978-981-13-3065-0_18] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
48
|
Le Master E, Huang RT, Zhang C, Bogachkov Y, Coles C, Shentu TP, Sheng Y, Fancher IS, Ng C, Christoforidis T, Subbaiah PV, Berdyshev E, Qain Z, Eddington DT, Lee J, Cho M, Fang Y, Minshall RD, Levitan I. Proatherogenic Flow Increases Endothelial Stiffness via Enhanced CD36-Mediated Uptake of Oxidized Low-Density Lipoproteins. Arterioscler Thromb Vasc Biol 2018; 38:64-75. [PMID: 29025707 PMCID: PMC5746473 DOI: 10.1161/atvbaha.117.309907] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 09/26/2017] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Disturbed flow (DF) is well-known to induce endothelial dysfunction and synergistically with plasma dyslipidemia facilitate plaque formation. Little is known, however, about the synergistic impact of DF and dyslipidemia on endothelial biomechanics. Our goal was to determine the impact of DF on endothelial stiffness and evaluate the role of dyslipidemia/oxLDL (oxidized low-density lipoprotein) in this process. APPROACH AND RESULTS Endothelial elastic modulus of intact mouse aortas ex vivo and of human aortic endothelial cells exposed to laminar flow or DF was measured using atomic force microscopy. Endothelial monolayer of the aortic arch is found to be significantly stiffer than the descending aorta (4.2+1.1 versus 2.5+0.2 kPa for aortic arch versus descending aorta) in mice maintained on low-fat diet. This effect is significantly exacerbated by short-term high-fat diet (8.7+2.5 versus 4.5+1.2 kPa for aortic arch versus descending aorta). Exposure of human aortic endothelial cells to DF in vitro resulted in 50% increase in oxLDL uptake and significant endothelial stiffening in the presence but not in the absence of oxLDL. DF also increased the expression of oxLDL receptor CD36 (cluster of differentiation 36), whereas downregulation of CD36 abrogated DF-induced endothelial oxLDL uptake and stiffening. Furthermore, genetic deficiency of CD36 abrogated endothelial stiffening in the aortic arch in vivo in mice fed either low-fat diet or high-fat diet. We also show that the loss of endothelial stiffening in CD36 knockout aortas is not mediated by the loss of CD36 in circulating cells. CONCLUSIONS DF facilitates endothelial CD36-dependent uptake of oxidized lipids resulting in local increase of endothelial stiffness in proatherogenic areas of the aorta.
Collapse
Affiliation(s)
- Elizabeth Le Master
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Ru-Ting Huang
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Chongxu Zhang
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Yedida Bogachkov
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Cassandre Coles
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Tzu-Pin Shentu
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Yue Sheng
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Ibra S Fancher
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Carlos Ng
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Theodore Christoforidis
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Pappasani V Subbaiah
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Evgeny Berdyshev
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Zhijian Qain
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - David T Eddington
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - James Lee
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Michael Cho
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Yun Fang
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Richard D Minshall
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.)
| | - Irena Levitan
- From the Division of Pulmonary and Critical Care (E.L.M., C.Z., T.-P.S., I.S.F., I.L.), Division of Endocrinology (P.V.S.), Division of Hematology and Oncology, Department of Medicine (Y.S., Z.Q.), and Departments of Bioengineering (E.L.M., T.-P.S., C.N., T.C., D.T.E., J.L., M.C., I.L.), Pharmacology (Y.B., C.C., R.D.M., I.L.), and Anesthesiology (R.D.M.), University of Illinois at Chicago; Department of Medicine, University of Chicago, IL (R.-T.H., Y.F.); and Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, CO (E.B.).
| |
Collapse
|
49
|
Salusin- β Is Involved in Diabetes Mellitus-Induced Endothelial Dysfunction via Degradation of Peroxisome Proliferator-Activated Receptor Gamma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:6905217. [PMID: 29359008 PMCID: PMC5735326 DOI: 10.1155/2017/6905217] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 10/08/2017] [Indexed: 12/18/2022]
Abstract
The pathophysiological mechanisms for vascular lesions in diabetes mellitus (DM) are complex, among which endothelial dysfunction plays a vital role. Therapeutic target against endothelial injury may provide critical venues for treatment of diabetic vascular diseases. We recently identified that salusin-β contributed to high glucose-induced endothelial cell apoptosis. However, the roles of salusin-β in DM-induced endothelial dysfunction remain largely elusive. Male C57BL/6J mice were used to induce type 2 diabetes mellitus (T2DM) model. Human umbilical vein endothelial cells (HUVECs) were cultured in high glucose/high fat (HG/HF) medium. We demonstrated increased expression of salusin-β in diabetic aortic tissues and high-glucose/high-fat- (HG/HF-) incubated HUVECs. Disruption of salusin-β by shRNA abrogated the reactive oxygen species (ROS) production, inflammation, and nitrotyrosine content of HUVECs cultured in HG/HF medium. The HG/HF-mediated decrease in peroxisome proliferator-activated receptor γ (PPARγ) expression was restored by salusin-β shRNA, and PPARγ inhibitor T0070907 abolished the protective actions of salusin-β shRNA on endothelial injury in HG/HF-treated HUVECs. Salusin-β silencing obviously improved endothelium-dependent vasorelaxation, oxidative stress, inflammatory response, and nitrative stress in diabetic aorta. Taken together, our results highlighted the essential role of salusin-β in pathological endothelial dysfunction, and salusin-β may be a promising target in treatment of vascular complications of DM.
Collapse
|
50
|
Chen JY, Ye ZX, Wang XF, Chang J, Yang MW, Zhong HH, Hong FF, Yang SL. Nitric oxide bioavailability dysfunction involves in atherosclerosis. Biomed Pharmacother 2017; 97:423-428. [PMID: 29091892 DOI: 10.1016/j.biopha.2017.10.122] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 10/22/2017] [Accepted: 10/22/2017] [Indexed: 12/25/2022] Open
Abstract
The pathological characteristics of atherosclerosis (AS) include lipid accumulation, fibrosis formation and atherosclerotic plaque produced in artery intima, which leads to vascular sclerosis, lumen stenosis and irritates the ischemic changes of corresponding organs. Endothelial dysfunction was closely associated with AS. Nitric oxide (NO) is a multifunctional signaling molecule involved in the maintenance of metabolic and cardiovascular homeostasis. NO is also a potent endogenous vasodilator and enters for the key processes that suppresses the formation vascular lesion even AS. NO bioavailability indicates the production and utilization of endothelial NO in organisms, its decrease is related to oxidative stress, lipid infiltration, the expressions of some inflammatory factors and the alteration of vascular tone, which plays an important role in endothelial dysfunction. The enhancement of arginase activity and the increase in asymmetric dimethylarginine and hyperhomocysteinemia levels all contribute to AS by intervening NO bioavailability in human beings. Diabetes mellitus, obesity, chronic kidney disease and smoking, etc., also participate in AS by influencing NO bioavailability and NO level. Here, we reviewed the relationship between NO bioavailability and AS according the newest literatures.
Collapse
Affiliation(s)
- Jing-Yi Chen
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Zi-Xin Ye
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Xiu-Fen Wang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Jian Chang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Mei-Wen Yang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Hua-Hua Zhong
- Department of Experimental Teaching Center, Nanchang University, Nanchang 330031, China
| | - Fen-Fang Hong
- Department of Experimental Teaching Center, Nanchang University, Nanchang 330031, China.
| | - Shu-Long Yang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China.
| |
Collapse
|