1
|
Karakasis P, Theofilis P, Patoulias D, Vlachakis PK, Antoniadis AP, Fragakis N. Diabetes-Driven Atherosclerosis: Updated Mechanistic Insights and Novel Therapeutic Strategies. Int J Mol Sci 2025; 26:2196. [PMID: 40076813 PMCID: PMC11900163 DOI: 10.3390/ijms26052196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
The global rise in diabetes prevalence has significantly contributed to the increasing burden of atherosclerotic cardiovascular disease (ASCVD), a leading cause of morbidity and mortality in this population. Diabetes accelerates atherosclerosis through mechanisms such as hyperglycemia, oxidative stress, chronic inflammation, and epigenetic dysregulation, leading to unstable plaques and an elevated risk of cardiovascular events. Despite advancements in controlling traditional risk factors like dyslipidemia and hypertension, a considerable residual cardiovascular risk persists, highlighting the need for innovative therapeutic approaches. Emerging treatments, including sodium-glucose cotransporter 2 (SGLT2) inhibitors, glucagon-like peptide-1 (GLP-1) receptor agonists, epigenetic modulators, and RNA-based therapies, are showing promise in addressing the unique challenges of diabetes-associated ASCVD. Precision medicine strategies, such as nanoparticle-based drug delivery and cell-specific therapies, offer further potential for mitigating cardiovascular complications. Advances in multiomics and systems biology continue to deepen our understanding of the molecular mechanisms driving diabetes-associated atherosclerosis. This review synthesizes recent advances in understanding the pathophysiology and treatment of diabetes-related atherosclerosis, offering a roadmap for future research and precision medicine approaches to mitigate cardiovascular risk in this growing population.
Collapse
Affiliation(s)
- Paschalis Karakasis
- Second Department of Cardiology, Medical School, Hippokration General Hospital, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54124 Thessaloniki, Greece; (A.P.A.); (N.F.)
| | - Panagiotis Theofilis
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece; (P.T.); (P.K.V.)
| | - Dimitrios Patoulias
- Second Propedeutic Department of Internal Medicine, Faculty of Medicine, School of Health Sciences Aristotle, University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Panayotis K. Vlachakis
- First Cardiology Department, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, 15772 Athens, Greece; (P.T.); (P.K.V.)
| | - Antonios P. Antoniadis
- Second Department of Cardiology, Medical School, Hippokration General Hospital, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54124 Thessaloniki, Greece; (A.P.A.); (N.F.)
| | - Nikolaos Fragakis
- Second Department of Cardiology, Medical School, Hippokration General Hospital, Aristotle University of Thessaloniki, Konstantinoupoleos 49, 54124 Thessaloniki, Greece; (A.P.A.); (N.F.)
| |
Collapse
|
2
|
Strunz CMC, Roggerio A, Cruz PL, Benvenuti LA, Irigoyen MC, Mansur ADP. Resveratrol Attenuates Fibrosis and Alters Signaling Pathways in Diabetic Cardiac and Skeletal Muscles and Adipose Tissue Without Reversing Structural Damage. Int J Mol Sci 2025; 26:1672. [PMID: 40004135 PMCID: PMC11855909 DOI: 10.3390/ijms26041672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
Resveratrol (RSV) improves metabolic functions, but its tissue-specific effects on diabetes remain unclear. This study investigated RSV's impact on molecular pathways in an experimental model of diabetes in cardiac and skeletal muscles and adipose tissue. Wistar rats were assigned to control (C), control treated with RSV (RC), diabetic (D), and diabetic treated with RSV (RD). Diabetes was induced using streptozotocin and nicotinamide, and RSV was administered for six weeks. In diabetic rats, RSV treatment significantly reduced collagen accumulation in cardiac and skeletal muscle tissues compared to untreated diabetic controls, although it did not restore muscle mass. Adipose tissue in diabetic rats exhibited a significant reduction of 3.4 times in collagen levels following RSV treatment. However, this reduction was not associated with any measurable improvement in tissue function. In cardiac tissue, RSV downregulated phosphorylated protein kinase B (AKT)/AKT and phosphorylated ribosomal protein S6 (rpS6)/rpS6 while mammalian target of rapamycin (mTOR) activity remained unchanged. In skeletal muscle, RSV suppressed rpS6 phosphorylation without affecting (mTOR) signaling. RSV enhanced mTOR and Beclin-1 expression in adipose tissue, though metabolic dysfunction persisted. RSV reduced receptors for advanced glycation end-product expression in all tissues, indicating the modulation of hyperglycemia-driven pathways. RSV improved fibrosis and signaling pathways but failed to reverse abnormal tissue growth patterns, including cardiac hypertrophy, skeletal muscle atrophy, and adipose tissue atrophy.
Collapse
Affiliation(s)
- Célia Maria Cássaro Strunz
- Laboratório de Análises Clínicas, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil; (A.R.); (M.C.I.)
| | - Alessandra Roggerio
- Laboratório de Análises Clínicas, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil; (A.R.); (M.C.I.)
| | - Paula Lázara Cruz
- Laboratório de Hipertensão Experimental, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil;
| | - Luiz Alberto Benvenuti
- Laboratório de Anatomia Patológica, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil;
| | - Maria Cláudia Irigoyen
- Laboratório de Análises Clínicas, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil; (A.R.); (M.C.I.)
| | - Antonio de Padua Mansur
- Serviço de Prevenção, Cardiopatia na Mulher e Reabilitação Cardiovascular, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo 05403-900, SP, Brazil;
| |
Collapse
|
3
|
Liu Z, Liu J, Wang X, Zhang Y, Ma Y, Guan G, Yuwen Y, He N, Liu H, Yu X, Ma S, Wang J, Zhang J, Zhu L, Zhang Y. N ε-carboxyethyl-lysin influences atherosclerotic plaque stability through ZKSCAN3 acetylation-regulated macrophage autophagy via the RAGE/LKB1/AMPK1/SIRT1 pathway. Cardiovasc Diabetol 2025; 24:36. [PMID: 39844245 PMCID: PMC11755919 DOI: 10.1186/s12933-025-02586-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 01/07/2025] [Indexed: 01/30/2025] Open
Abstract
Atherosclerosis, a chronic inflammatory condition characterized by plaque formation, often leads to instability, particularly under Type 2 diabetes mellitus (T2DM) conditions, which exacerbate cardiovascular risks. However, the molecular mechanisms underlying this process remain incompletely understood. In this study, we investigated the correlation between acute coronary syndrome (ACS) and serum levels of Nε-carboxyethyl-lysin (CEL), a prominent advanced glycation end product (AGE) elevated in T2DM, in a cohort of 225 patients with coronary artery disease. Using a murine model of atherosclerosis complicated by T2DM, we examined the effects of CEL on plaque stability and macrophage autophagy. Our findings revealed that elevated serum CEL levels are independently associated with increased ACS incidence. Metabolomic profiling identified CEL as a key AGE contributing to plaque instability in diabetic conditions. Mechanistically, CEL disrupted macrophage autophagy and plaque stability by perturbing the Receptor for Advanced Glycation End products (RAGE)/Liver Kinase B1 (LKB1)/AMP-activated Protein Kinase 1 (AMPK1)/Sirtuin 1 (SIRT1) signaling cascade. This pathway further regulated autophagic activity through SIRT1-mediated acetylation of Zinc Finger with KRAB and SCAN Domains 3 (ZKSCAN3). These findings highlight CEL's critical role in promoting plaque instability in T2DM by impairing key molecular pathways that regulate autophagy, offering potential therapeutic targets for managing atherosclerosis in diabetic patients.
Collapse
Affiliation(s)
- Zhongwei Liu
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710003, China
- Department of Technology Transfer and Management, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
- Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, 710068, China
- School of Medicine, Xizangminzu University, Xianyang, 712082, China
- School of Medicine, Yan'an University, Yan'an, 716000, China
- Traditional Chinese Medicine Inheritance and Innovation Platform, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Jing Liu
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710003, China
- Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, 710068, China
| | - Xiqiang Wang
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710003, China
- Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, 710068, China
| | - Yong Zhang
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710003, China
- Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, 710068, China
| | - Yanpeng Ma
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710003, China
- Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, 710068, China
| | - Gongchang Guan
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710003, China
- Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, 710068, China
| | - Ya Yuwen
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710003, China
- Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, 710068, China
- School of Medicine, Xizangminzu University, Xianyang, 712082, China
| | - Ni He
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710003, China
- Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, 710068, China
- School of Medicine, Yan'an University, Yan'an, 716000, China
| | - Hanxiu Liu
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710003, China
- Department of Technology Transfer and Management, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
- School of Medicine, Yan'an University, Yan'an, 716000, China
| | - Xingfeng Yu
- Department of Nursing, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Sen Ma
- Department of Technology Transfer and Management, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Junkui Wang
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China.
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710003, China.
- Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, 710068, China.
| | - Jin Zhang
- Department of Otolaryngology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China.
| | - Ling Zhu
- Department of Cardiology, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China.
- Atherosclerosis Integrated Chinese and Western Medicine Key Research Laboratory, Research Office of Shaanxi Administration of Traditional Chinese Medicine, Xi'an, 710003, China.
- Affiliated Shaanxi Provincial People's Hospital, Northwestern Polytechnical University, Xi'an, 710068, China.
| | - Yulian Zhang
- The Director's Office, Shaanxi Provincial People's Hospital, 256 Youyi Xi Rd, Xi'an, 710068, China.
| |
Collapse
|
4
|
Khan AW, Jandeleit-Dahm KAM. Atherosclerosis in diabetes mellitus: novel mechanisms and mechanism-based therapeutic approaches. Nat Rev Cardiol 2025:10.1038/s41569-024-01115-w. [PMID: 39805949 DOI: 10.1038/s41569-024-01115-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/17/2024] [Indexed: 01/16/2025]
Abstract
Atherosclerosis is a disease of large and medium arteries that can lead to life-threatening cardiovascular and cerebrovascular consequences, such as myocardial infarction and stroke. Moreover, atherosclerosis is a major contributor to cardiovascular-related mortality in individuals with diabetes mellitus. Diabetes aggravates the pathobiological mechanisms that underlie the development of atherosclerosis. Currently available anti-atherosclerotic drugs or strategies solely focus on optimal control of systemic risk factors, including hyperglycaemia and dyslipidaemia, but do not adequately target the diabetes-exacerbated mechanisms of atherosclerotic cardiovascular disease, highlighting the need for targeted, mechanism-based therapies. This Review focuses on emerging pathological mechanisms and related novel therapeutic targets in atherosclerotic cardiovascular disease in patients with diabetes.
Collapse
Affiliation(s)
- Abdul Waheed Khan
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| | - Karin A M Jandeleit-Dahm
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- German Diabetes Centre, Leibniz Centre for Diabetes Research at the Heinrich Heine University, Dusseldorf, Germany
| |
Collapse
|
5
|
Lei S, Liu C, Zheng TX, Fu W, Huang MZ. The relationship of redox signaling with the risk for atherosclerosis. Front Pharmacol 2024; 15:1430293. [PMID: 39148537 PMCID: PMC11324460 DOI: 10.3389/fphar.2024.1430293] [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: 05/09/2024] [Accepted: 07/09/2024] [Indexed: 08/17/2024] Open
Abstract
Oxidative balance plays a pivotal role in physiological homeostasis, and many diseases, particularly age-related conditions, are closely associated with oxidative imbalance. While the strategic role of oxidative regulation in various diseases is well-established, the specific involvement of oxidative stress in atherosclerosis remains elusive. Atherosclerosis is a chronic inflammatory disorder characterized by plaque formation within the arteries. Alterations in the oxidative status of vascular tissues are linked to the onset, progression, and outcome of atherosclerosis. This review examines the role of redox signaling in atherosclerosis, including its impact on risk factors such as dyslipidemia, hyperglycemia, inflammation, and unhealthy lifestyle, along with dysregulation, vascular homeostasis, immune system interaction, and therapeutic considerations. Understanding redox signal transduction and the regulation of redox signaling will offer valuable insights into the pathogenesis of atherosclerosis and guide the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Sujuan Lei
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Chen Liu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Tian-Xiang Zheng
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Department of General Surgery (Hepatopancreatobiliary Surgery), Chongqing, Sichuan, China
| | - Wenguang Fu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Department of General Surgery (Hepatopancreatobiliary Surgery), Chongqing, Sichuan, China
| | - Mei-Zhou Huang
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Department of General Surgery (Hepatopancreatobiliary Surgery), Chongqing, Sichuan, China
| |
Collapse
|
6
|
Sun Y, Zhang X, Nie X, Yang R, Zhao X, Cui C, Liu W. Dough-Kneading-Inspired Design of an Adhesive Cardiac Patch to Attenuate Cardiac Fibrosis and Improve Cardiac Function via Regulating Glycometabolism. Adv Healthc Mater 2024; 13:e2303685. [PMID: 38386972 DOI: 10.1002/adhm.202303685] [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: 10/25/2023] [Revised: 02/19/2024] [Indexed: 02/24/2024]
Abstract
Recently, hydrogel adhesive patches have been explored for treating myocardial infarction. However, achieving secure adhesion onto the wet beating heart and local regulation of pathological microenvironment remains challenging. Herein, a dough-kneading-inspired design of hydrogel adhesive cardiac patch is reported, aiming to improve the strength of prevalent powder-formed patch and retain wet adhesion. In mimicking the polysaccharide and protein components of natural flour, methacrylated polyglutamic acid (PGAMA) is electrostatically interacted with hydroxypropyl chitosan (HPCS) to form PGAMA/HPCS coacervate hydrogel. The PGAMA/HPCS hydrogel is freeze-dried and ground into powders, which are further rehydrated with two aqueous solutions of functional drug, 3-acrylamido phenylboronic acid (APBA)/rutin (Rt) complexes for protecting the myocardium from advanced glycation end product (AGEs) injury by reactive oxygen species (ROS) -responsive Rt release, and hypoxanthine-loaded methacrylated hyaluronic acid (HAMA) nanogels for enhancing macrophage targeting ability to regulate glycometabolism for combating inflammation. The rehydrated powders bearing APBA/Rt complexes and HAMA-hypoxanthine nanogels are repeatedly kneaded into a dough-like gel, which is further subjected to thermal-initiated crosslinking to form a stabilized and sticky patch. This biofunctional patch is applied onto the rats' infarcted myocardium, and the outcomes at 28 days post-surgery indicate efficient restoration of cardiac functions and attenuation of cardiac fibrosis.
Collapse
Affiliation(s)
- Yage Sun
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350, China
| | - Xiaoping Zhang
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350, China
| | - Xiongfeng Nie
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350, China
| | - Rong Yang
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350, China
| | - Xinrui Zhao
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350, China
| | - Chunyan Cui
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350, China
| | - Wenguang Liu
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350, China
| |
Collapse
|
7
|
Manrique-Acevedo C, Hirsch IB, Eckel RH. Prevention of Cardiovascular Disease in Type 1 Diabetes. N Engl J Med 2024; 390:1207-1217. [PMID: 38598575 DOI: 10.1056/nejmra2311526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Affiliation(s)
- Camila Manrique-Acevedo
- From the Division of Endocrinology and Metabolism, Department of Medicine, and NextGen Precision Health, University of Missouri, and the Harry S. Truman Memorial Veterans' Hospital - both in Columbia (C.M.-A.); the Department of Medicine, University of Washington School of Medicine, Seattle (I.B.H.); and the Divisions of Endocrinology, Metabolism and Diabetes, and Cardiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora (R.H.E.)
| | - Irl B Hirsch
- From the Division of Endocrinology and Metabolism, Department of Medicine, and NextGen Precision Health, University of Missouri, and the Harry S. Truman Memorial Veterans' Hospital - both in Columbia (C.M.-A.); the Department of Medicine, University of Washington School of Medicine, Seattle (I.B.H.); and the Divisions of Endocrinology, Metabolism and Diabetes, and Cardiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora (R.H.E.)
| | - Robert H Eckel
- From the Division of Endocrinology and Metabolism, Department of Medicine, and NextGen Precision Health, University of Missouri, and the Harry S. Truman Memorial Veterans' Hospital - both in Columbia (C.M.-A.); the Department of Medicine, University of Washington School of Medicine, Seattle (I.B.H.); and the Divisions of Endocrinology, Metabolism and Diabetes, and Cardiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora (R.H.E.)
| |
Collapse
|
8
|
Naser N, Lonj CK, Rikard-Bell M, Sandow SL, Murphy TV. Advanced glycated end-products inhibit dilation through constitutive endothelial RAGE and Nox1/4 in rat isolated skeletal muscle arteries. Microcirculation 2024; 31:e12837. [PMID: 37985248 DOI: 10.1111/micc.12837] [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: 07/17/2023] [Revised: 10/17/2023] [Accepted: 11/07/2023] [Indexed: 11/22/2023]
Abstract
OBJECTIVE This study investigated the actions of advanced glycated end-products (AGE), their receptors (RAGE), and NAD(P)H oxidase (Nox) subtypes 1, 2, and 4 on mechanisms of endothelium-dependent dilation of the rat cremaster muscle artery (CMA). METHODS Immunofluorescence studies were used to examine expression of RAGE in rat arteries. ROS accumulation was measured using luminescence and fluorescence assays. Functional studies were performed using pressure myography. RESULTS High levels of RAGE expression were shown in the endothelial cells of the CMA, compared with low endothelial expression in middle cerebral and mesenteric arteries and the aorta. Exogenous AGE (in vitro glycated bovine serum albumin) stimulated H2O2 accumulation in CMA, which was prevented by the RAGE antagonist FPS-ZM1, the NAD(P)H oxidase (Nox) inhibitor apocynin and inhibited by the Nox1/4 inhibitor setanaxib, but not the Nox2 inhibitor GSK2795039. In functional studies, AGE inhibited vasodilation of CMA stimulated by acetylcholine, sodium nitroprusside, and the BKCa activator NS1619, but not adenosine-induced dilation. FPS-ZM1, apocynin, and setanaxib prevented the inhibitory effects of AGE on responses to acetylcholine and NS-1619. CONCLUSION These observations suggest RAGE are constitutively expressed in the endothelium of the rat CMA and may be activated by AGE to stimulate Nox1/4 and ROS formation with resulting inhibition of NO and BKCa-mediated endothelium-dependent dilation.
Collapse
Affiliation(s)
- Nadim Naser
- Physiology, School of Biomedical Sciences, UNSW Sydney, Sydney, Australia
| | - Chenchel K Lonj
- Physiology, School of Biomedical Sciences, UNSW Sydney, Sydney, Australia
| | - Matthew Rikard-Bell
- Physiology, School of Biomedical Sciences, UNSW Sydney, Sydney, Australia
- Townsville University Hospital, Townsville, Queensland, Australia
| | - Shaun L Sandow
- Biomedical Science, University of the Sunshine Coast, Maroochydore, Australia
- Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Timothy V Murphy
- Physiology, School of Biomedical Sciences, UNSW Sydney, Sydney, Australia
| |
Collapse
|
9
|
Ravi S, Martin LC, Krishnan M, Kumaresan M, Manikandan B, Ramar M. Interactions between macrophage membrane and lipid mediators during cardiovascular diseases with the implications of scavenger receptors. Chem Phys Lipids 2024; 258:105362. [PMID: 38006924 DOI: 10.1016/j.chemphyslip.2023.105362] [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: 10/09/2023] [Revised: 11/06/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
The onset and progression of cardiovascular diseases with the major underlying cause being atherosclerosis, occur during chronic inflammatory persistence in the vascular system, especially within the arterial wall. Such prolonged maladaptive inflammation is driven by macrophages and their key mediators are generally attributed to a disparity in lipid metabolism. Macrophages are the primary cells of innate immunity, endowed with expansive membrane domains involved in immune responses with their signalling systems. During atherosclerosis, the membrane domains and receptors control various active organisations of macrophages. Their scavenger/endocytic receptors regulate the trafficking of intracellular and extracellular cargo. Corresponding influence on lipid metabolism is mediated by their dynamic interaction with scavenger membrane receptors and their integrated mechanisms such as pinocytosis, phagocytosis, cholesterol export/import, etc. This interaction not only results in the functional differentiation of macrophages but also modifies their structural configurations. Here, we reviewed the association of macrophage membrane biomechanics and their scavenger receptor families with lipid metabolites during the event of atherogenesis. In addition, the membrane structure of macrophages and the signalling pathways involved in endocytosis integrated with lipid metabolism are detailed. This article establishes future insights into the scavenger receptors as potential targets for cardiovascular disease prevention and treatment.
Collapse
Affiliation(s)
- Sangeetha Ravi
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | | | - Mahalakshmi Krishnan
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Manikandan Kumaresan
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India
| | - Beulaja Manikandan
- Department of Biochemistry, Annai Veilankanni's College for Women, Chennai 600 015, India
| | - Manikandan Ramar
- Department of Zoology, University of Madras, Guindy Campus, Chennai 600 025, India.
| |
Collapse
|
10
|
Angioni R, Bonfanti M, Caporale N, Sánchez-Rodríguez R, Munari F, Savino A, Pasqualato S, Buratto D, Pagani I, Bertoldi N, Zanon C, Ferrari P, Ricciardelli E, Putaggio C, Ghezzi S, Elli F, Rotta L, Scardua A, Weber J, Cecatiello V, Iorio F, Zonta F, Cattelan AM, Vicenzi E, Vannini A, Molon B, Villa CE, Viola A, Testa G. RAGE engagement by SARS-CoV-2 enables monocyte infection and underlies COVID-19 severity. Cell Rep Med 2023; 4:101266. [PMID: 37944530 PMCID: PMC10694673 DOI: 10.1016/j.xcrm.2023.101266] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 03/16/2023] [Accepted: 10/10/2023] [Indexed: 11/12/2023]
Abstract
The spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has fueled the COVID-19 pandemic with its enduring medical and socioeconomic challenges because of subsequent waves and long-term consequences of great concern. Here, we chart the molecular basis of COVID-19 pathogenesis by analyzing patients' immune responses at single-cell resolution across disease course and severity. This approach confirms cell subpopulation-specific dysregulation in COVID-19 across disease course and severity and identifies a severity-associated activation of the receptor for advanced glycation endproducts (RAGE) pathway in monocytes. In vitro THP1-based experiments indicate that monocytes bind the SARS-CoV-2 S1-receptor binding domain (RBD) via RAGE, pointing to RAGE-Spike interaction enabling monocyte infection. Thus, our results demonstrate that RAGE is a functional receptor of SARS-CoV-2 contributing to COVID-19 severity.
Collapse
Affiliation(s)
- Roberta Angioni
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; Fondazione Istituto di Ricerca Pediatrica - Città Della Speranza, 35127 Padova, Italy
| | - Matteo Bonfanti
- Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy
| | - Nicolò Caporale
- Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Via Santa Sofia 9, 20122 Milan, Italy; Department of Experimental Oncology, European Institute of Oncology IRCCS, Via Adamello 16, 20139 Milan, Italy
| | - Ricardo Sánchez-Rodríguez
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; Fondazione Istituto di Ricerca Pediatrica - Città Della Speranza, 35127 Padova, Italy
| | - Fabio Munari
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; Fondazione Istituto di Ricerca Pediatrica - Città Della Speranza, 35127 Padova, Italy
| | - Aurora Savino
- Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy
| | | | - Damiano Buratto
- Institute of Quantitative Biology, College of Life Sciences, Zhejiang University, Hangzhou 310058, China; Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Isabel Pagani
- Viral Pathogenesis and Biosafety Unit, San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Nicole Bertoldi
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; Fondazione Istituto di Ricerca Pediatrica - Città Della Speranza, 35127 Padova, Italy
| | - Carlo Zanon
- Fondazione Istituto di Ricerca Pediatrica - Città Della Speranza, 35127 Padova, Italy
| | - Paolo Ferrari
- Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy
| | | | - Cristina Putaggio
- Infectious Disease Unit, Padova University Hospital, 35128 Padova, Italy
| | - Silvia Ghezzi
- Viral Pathogenesis and Biosafety Unit, San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | - Francesco Elli
- Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Via Santa Sofia 9, 20122 Milan, Italy
| | - Luca Rotta
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Via Adamello 16, 20139 Milan, Italy
| | | | - Janine Weber
- Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy
| | | | - Francesco Iorio
- Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy
| | - Francesco Zonta
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China; Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou 215123, China
| | | | - Elisa Vicenzi
- Viral Pathogenesis and Biosafety Unit, San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy
| | | | - Barbara Molon
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; Fondazione Istituto di Ricerca Pediatrica - Città Della Speranza, 35127 Padova, Italy
| | - Carlo Emanuele Villa
- Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy; Department of Experimental Oncology, European Institute of Oncology IRCCS, Via Adamello 16, 20139 Milan, Italy
| | - Antonella Viola
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy; Fondazione Istituto di Ricerca Pediatrica - Città Della Speranza, 35127 Padova, Italy.
| | - Giuseppe Testa
- Human Technopole, Viale Rita Levi-Montalcini 1, 20157 Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Via Santa Sofia 9, 20122 Milan, Italy; Department of Experimental Oncology, European Institute of Oncology IRCCS, Via Adamello 16, 20139 Milan, Italy.
| |
Collapse
|
11
|
Oezer K, Kolibabka M, Gassenhuber J, Dietrich N, Fleming T, Schlotterer A, Morcos M, Wohlfart P, Hammes HP. The effect of GLP-1 receptor agonist lixisenatide on experimental diabetic retinopathy. Acta Diabetol 2023; 60:1551-1565. [PMID: 37423944 PMCID: PMC10520173 DOI: 10.1007/s00592-023-02135-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 06/10/2023] [Indexed: 07/11/2023]
Abstract
AIMS Glucagon-like peptide-1 receptor agonists are effective treatments for type 2 diabetes, effectively lowering glucose without weight gain and with low risk for hypoglycemia. However, their influence on the retinal neurovascular unit remains unclear. In this study, we analyzed the effects of the GLP-1 RA lixisenatide on diabetic retinopathy. METHODS Vasculo- and neuroprotective effects were assessed in experimental diabetic retinopathy and high glucose-cultivated C. elegans, respectively. In STZ-diabetic Wistar rats, acellular capillaries and pericytes (quantitative retinal morphometry), neuroretinal function (mfERG), macroglia (GFAP western blot) and microglia (immunohistochemistry) quantification, methylglyoxal (LC-MS/MS) and retinal gene expressions (RNA-sequencing) were determined. The antioxidant properties of lixisenatide were tested in C. elegans. RESULTS Lixisenatide had no effect on glucose metabolism. Lixisenatide preserved the retinal vasculature and neuroretinal function. The macro- and microglial activation was mitigated. Lixisenatide normalized some gene expression changes in diabetic animals to control levels. Ets2 was identified as a regulator of inflammatory genes. In C. elegans, lixisenatide showed the antioxidative property. CONCLUSIONS Our data suggest that lixisenatide has a protective effect on the diabetic retina, most likely due to a combination of neuroprotective, anti-inflammatory and antioxidative effects of lixisenatide on the neurovascular unit.
Collapse
Affiliation(s)
- Kuebra Oezer
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| | - Matthias Kolibabka
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Nadine Dietrich
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Thomas Fleming
- Department of Medicine I and Clinical Chemistry, Heidelberg University, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Andrea Schlotterer
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Michael Morcos
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Stoffwechselzentrum Rhein-Pfalz, Belchenstraße 1-5, 68163, Mannheim, Germany
| | - Paulus Wohlfart
- Sanofi, MSAT M&I Bioassays and Compliance, Frankfurt, Germany
| | - Hans-Peter Hammes
- 5th Medical Department, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
12
|
Khan MI, Ashfaq F, Alsayegh AA, Hamouda A, Khatoon F, Altamimi TN, Alhodieb FS, Beg MMA. Advanced glycation end product signaling and metabolic complications: Dietary approach. World J Diabetes 2023; 14:995-1012. [PMID: 37547584 PMCID: PMC10401445 DOI: 10.4239/wjd.v14.i7.995] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/08/2023] [Accepted: 04/27/2023] [Indexed: 07/12/2023] Open
Abstract
Advanced glycation end products (AGEs) are a heterogeneous collection of compounds formed during industrial processing and home cooking through a sequence of nonenzymatic glycation reactions. The modern western diet is full of heat-treated foods that contribute to AGE intake. Foods high in AGEs in the contemporary diet include processed cereal products. Due to industrialization and marketing strategies, restaurant meals are modified rather than being traditionally or conventionally cooked. Fried, grilled, baked, and boiled foods have the greatest AGE levels. Higher AGE-content foods include dry nuts, roasted walnuts, sunflower seeds, fried chicken, bacon, and beef. Animal proteins and processed plant foods contain furosine, acrylamide, heterocyclic amines, and 5-hydroxymethylfurfural. Furosine (2-furoil-methyl-lysine) is an amino acid found in cooked meat products and other processed foods. High concentrations of carboxymethyl-lysine, carboxyethyl-lysine, and methylglyoxal-O are found in heat-treated nonvegetarian foods, peanut butter, and cereal items. Increased plasma levels of AGEs, which are harmful chemicals that lead to age-related diseases and physiological aging, diabetes, and autoimmune/inflammatory rheumatic diseases such as systemic lupus erythematosus and rheumatoid arthritis. AGEs in the pathophysiology of metabolic diseases have been linked to individuals with diabetes mellitus who have peripheral nerves with high amounts of AGEs and diabetes has been linked to increased myelin glycation. Insulin resistance and hyperglycemia can impact numerous human tissues and organs, leading to long-term difficulties in a number of systems and organs, including the cardiovascular system. Plasma AGE levels are linked to all-cause mortality in individuals with diabetes who have fatal or nonfatal coronary artery disease, such as ventricular dysfunction. High levels of tissue AGEs are independently associated with cardiac systolic dysfunction in diabetic patients with heart failure compared with diabetic patients without heart failure. It is widely recognized that AGEs and oxidative stress play a key role in the cardiovascular complications of diabetes because they both influence and are impacted by oxidative stress. All chronic illnesses involve protein, lipid, or nucleic acid modifications including crosslinked and nondegradable aggregates known as AGEs. Endogenous AGE formation or dietary AGE uptake can result in additional protein modifications and stimulation of several inflammatory signaling pathways. Many of these systems, however, require additional explanation because they are not entirely obvious. This review summarizes the current evidence regarding dietary sources of AGEs and metabolism-related complications associated with AGEs.
Collapse
Affiliation(s)
- Mohammad Idreesh Khan
- Department of Clinical Nutrition, College of Applied Health Sciences in Ar Rass, Qassim University, Ar Rass 51921, Saudi Arabia
| | - Fauzia Ashfaq
- Clinical Nutrition Department, Applied Medical Sciences College, Jazan University, Jazan 82817, Saudi Arabia
| | - Abdulrahman A Alsayegh
- Clinical Nutrition Department, Applied Medical Sciences College, Jazan University, Jazan 82817, Saudi Arabia
| | - Alshaimaa Hamouda
- Clinical Nutrition Department, Applied Medical Sciences College, Jazan University, Jazan 82817, Saudi Arabia
| | - Fahmida Khatoon
- Department of Biochemistry, College of Medicine, University of Hail, Hail 2240, Saudi Arabia
| | - Tahani Nasser Altamimi
- Department of Family and Community Medicine, College of Medicine, University of Hail, Hail 2240, Saudi Arabia
| | - Fahad Saad Alhodieb
- Department of Clinical Nutrition, College of Applied Health Sciences in Ar Rass, Qassim University, Ar Rass 51921, Saudi Arabia
| | | |
Collapse
|
13
|
Hadipour E, Emami SA, Tayarani‐Najaran N, Tayarani‐Najaran Z. Effects of sesame ( Sesamum indicum L.) and bioactive compounds (sesamin and sesamolin) on inflammation and atherosclerosis: A review. Food Sci Nutr 2023; 11:3729-3757. [PMID: 37457142 PMCID: PMC10345702 DOI: 10.1002/fsn3.3407] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 04/13/2023] [Accepted: 04/22/2023] [Indexed: 07/18/2023] Open
Abstract
Inflammation, oxidative stress, obesity, infection, hyperlipidemia, hypertension, and diabetes are the main causes of atherosclerosis, which in the long term lead to hardening of the arteries. In the current study, we reviewed recent findings of the mechanism of sesame and its active compounds of sesamin and sesamolin regulates on atherosclerosis. Sesame can decrease the lipid peroxidation and affect the enzymes, which control the balance of oxidative status in the body. Besides modulating the inflammatory cytokines, sesame regulates the main mediators of the signaling pathways in the process of inflammation, such as prostaglandin E2 (PGE2), nuclear factor kappa light-chain enhancer of activated B cells (NF-kB) and peroxisome proliferator-activated receptor gamma (PPAR-γ). Sesame decreases the growth of different pathogens. It fights against obesity and helps to reduce weight, body mass index (BMI), waist circumference, and lipid count of serum and liver. In addition to lowering fasting blood sugar (FBS), it decreases the hemoglobin A1c (HbA1c) and glucose levels and improves insulin function. With high content of linoleic acid, α-linolenic acid, and total polyunsaturated fatty acid (PUFA), sesame efficiently controls the blood plasma lipids and changes the lipid profile. In the case of hypertension, it maintains the health of endothelium through multiple mechanisms and conserves the response of the arteries to vasodilation. PUFA in sesame suppresses blood clotting and fibrinogen activity. All the mentioned properties combat atherosclerosis and hardening of blood vessels, which are detailed in the present review for sesame.
Collapse
Affiliation(s)
- Elham Hadipour
- Department of Biology, Faculty of ScienceUniversity of GuilanRashtIran
| | - Seyed Ahmad Emami
- Department of Traditional Pharmacy, School of PharmacyMashhad University of Medical SciencesMashhadIran
| | - Niloufar Tayarani‐Najaran
- Department of Dental Prosthesis, School of DentistryMashhad University of Medical SciencesMashhadIran
| | - Zahra Tayarani‐Najaran
- Targeted Drug Delivery Research CenterPharmaceutical Technology Institute, Mashhad University of Medical SciencesMashhadIran
| |
Collapse
|
14
|
Chandrakumar S, Santiago Tierno I, Agarwal M, Matisioudis N, Kern TS, Ghosh K. Subendothelial Matrix Stiffening by Lysyl Oxidase Enhances RAGE-Mediated Retinal Endothelial Activation in Diabetes. Diabetes 2023; 72:973-985. [PMID: 37058096 PMCID: PMC10281239 DOI: 10.2337/db22-0761] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 03/30/2023] [Indexed: 04/15/2023]
Abstract
Endothelial cell (EC) activation is a crucial determinant of retinal vascular inflammation associated with diabetic retinopathy (DR), a major microvascular complication of diabetes. We previously showed that, similar to abnormal biochemical factors, aberrant mechanical cues in the form of lysyl oxidase (LOX)-dependent subendothelial matrix stiffening also contribute significantly to retinal EC activation in diabetes. Yet, how LOX is itself regulated and precisely how it mechanically controls retinal EC activation in diabetes is poorly understood. Here, we show that high-glucose-induced LOX upregulation in human retinal ECs (HRECs) is mediated by proinflammatory receptor for advanced glycation end products (RAGE). HRECs treated with methylglyoxal (MGO), an active precursor to the advanced glycation end product (AGE) MG-H1, exhibited LOX upregulation that was blocked by a RAGE inhibitor, thus confirming the ability of RAGE to promote LOX expression. Crucially, as a downstream effector of RAGE, LOX was found to mediate both the proinflammatory and matrix remodeling effects of AGE/RAGE, primarily through its ability to crosslink or stiffen matrix. Finally, using decellularized HREC-derived matrices and a mouse model of diabetes, we demonstrate that LOX-dependent matrix stiffening feeds back to enhance RAGE, thereby achieving its autoregulation and proinflammatory effects. Collectively, these findings provide fresh mechanistic insights into the regulation and proinflammatory role of LOX-dependent mechanical cues in diabetes while simultaneously implicating LOX as an alternative (downstream) target to block AGE/RAGE signaling in DR. ARTICLE HIGHLIGHTS We investigated the regulation and proinflammatory role of retinal endothelial lysyl oxidase (LOX) in diabetes. Findings reveal that LOX is upregulated by advanced glycation end products (AGE) and receptor for AGE (RAGE) and mediates AGE/RAGE-induced retinal endothelial cell activation and subendothelial matrix remodeling. We also show that LOX-dependent subendothelial matrix stiffening feeds back to enhance retinal endothelial RAGE. These findings implicate LOX as a key proinflammatory factor and an alternative (downstream) target to block AGE/RAGE signaling in diabetic retinopathy.
Collapse
Affiliation(s)
- Sathishkumar Chandrakumar
- Department of Bioengineering, University of California, Riverside, CA
- Department of Ophthalmology, University of California, Los Angeles, CA
- Doheny Eye Institute, Pasadena, CA
| | - Irene Santiago Tierno
- Department of Ophthalmology, University of California, Los Angeles, CA
- Doheny Eye Institute, Pasadena, CA
- Molecular, Cellular, and Integrated Physiology Interdepartmental PhD Program, University of California, Los Angeles, CA
| | - Mahesh Agarwal
- Department of Ophthalmology, University of California, Los Angeles, CA
- Doheny Eye Institute, Pasadena, CA
| | | | - Timothy S. Kern
- Department of Ophthalmology, University of California, Irvine, CA
- Gavin Herbert Eye Institute, University of California, Irvine, CA
| | - Kaustabh Ghosh
- Department of Bioengineering, University of California, Riverside, CA
- Department of Ophthalmology, University of California, Los Angeles, CA
- Doheny Eye Institute, Pasadena, CA
- Molecular, Cellular, and Integrated Physiology Interdepartmental PhD Program, University of California, Los Angeles, CA
| |
Collapse
|
15
|
Ahmad S, Zaib S. An Evaluation of Biomarkers as Determinants of Peripheral Arterial Disease in those with Diabetes Mellitus. ChemistrySelect 2023. [DOI: 10.1002/slct.202300297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Affiliation(s)
- Shabbir Ahmad
- Department of Basic and Applied Chemistry, Faculty of Science and Technology University of Central Punjab Lahore 54590 Pakistan
| | - Sumera Zaib
- Department of Basic and Applied Chemistry, Faculty of Science and Technology University of Central Punjab Lahore 54590 Pakistan
| |
Collapse
|
16
|
Advanced Glycation End-Products and Diabetic Neuropathy of the Retina. Int J Mol Sci 2023; 24:ijms24032927. [PMID: 36769249 PMCID: PMC9917392 DOI: 10.3390/ijms24032927] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/29/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Diabetic retinopathy is a tissue-specific neurovascular impairment of the retina in patients with both type 1 and type 2 diabetes. Several pathological factors are involved in the progressive impairment of the interdependence between cells that consist of the neurovascular units (NVUs). The advanced glycation end-products (AGEs) are one of the major pathological factors that cause the impairments of neurovascular coupling in diabetic retinopathy. Although the exact mechanisms for the toxicities of the AGEs in diabetic retinopathy have not been definitively determined, the AGE-receptor of the AGE (RAGE) axis, production of reactive oxygen species, inflammatory reactions, and the activation of the cell death pathways are associated with the impairment of the NVUs in diabetic retinopathy. More specifically, neuronal cell death is an irreversible change that is directly associated with vision reduction in diabetic patients. Thus, neuroprotective therapies must be established for diabetic retinopathy. The AGEs are one of the therapeutic targets to examine to ameliorate the pathological changes in the NVUs in diabetic retinopathy. This review focuses on the basic and pathological findings of AGE-induced neurovascular abnormalities and the potential therapeutic approaches, including the use of anti-glycated drugs to protect the AGE-induced impairments of the NVUs in diabetic retinopathy.
Collapse
|
17
|
Lind L, Fall T, Ärnlöv J, Elmståhl S, Sundström J. Large-Scale Metabolomics and the Incidence of Cardiovascular Disease. J Am Heart Assoc 2023; 12:e026885. [PMID: 36645074 PMCID: PMC9939066 DOI: 10.1161/jaha.122.026885] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Background The study aimed to show the relationship between a large number of circulating metabolites and subsequent cardiovascular disease (CVD) and subclinical markers of CVD in the general population. Methods and Results In 2278 individuals free from CVD in the EpiHealth study (aged 45-75 years, mean age 61 years, 50% women), 790 annotated nonxenobiotic metabolites were measured by mass spectroscopy (Metabolon). The same metabolites were measured in the PIVUS (Prospective Investigation of Vasculature in Uppsala Seniors) study (n=603, all aged 80 years, 50% women), in which cardiac and carotid artery pathologies were evaluated by ultrasound. During a median follow-up of 8.6 years, 107 individuals experienced a CVD (fatal or nonfatal myocardial infarction, stroke, or heart failure) in EpiHealth. Using a false discovery rate of 0.05 for age- and sex-adjusted analyses and P<0.05 for adjustment for traditional CVD risk factors, 37 metabolites were significantly related to incident CVD. These metabolites belonged to multiple biochemical classes, such as amino acids, lipids, and nucleotides. Top findings were dimethylglycine and N-acetylmethionine. A lasso selection of 5 metabolites improved discrimination when added on top of traditional CVD risk factors (+4.0%, P=0.0054). Thirty-five of the 37 metabolites were related to subclinical markers of CVD evaluated in the PIVUS study. The metabolite 1-carboxyethyltyrosine was associated with left atrial diameter as well as inversely related to both ejection fraction and the echogenicity of the carotid artery. Conclusions Several metabolites were discovered to be associated with future CVD, as well as with subclinical markers of CVD. A selection of metabolites improved discrimination when added on top of CVD risk factors.
Collapse
Affiliation(s)
- Lars Lind
- Department of Medical SciencesUppsala UniversityUppsalaSweden
| | - Tove Fall
- Department of Medical SciencesUppsala UniversityUppsalaSweden
| | - Johan Ärnlöv
- Division of Family Medicine and Primary Care, Department of Neurobiology, Care Science and SocietyKarolinska InstitutetHuddingeSweden,School of Health and Social StudiesDalarna UniversityFalunSweden
| | - Sölve Elmståhl
- Division of Geriatric Medicine, Department of Clinical Sciences in MalmöLund UniversityMalmöSweden
| | - Johan Sundström
- Department of Medical SciencesUppsala UniversityUppsalaSweden
| |
Collapse
|
18
|
Mestdagh J, Koster SBL, Damman J, Thio HB. Diabetes and the Skin. THE DIABETES TEXTBOOK 2023:1003-1020. [DOI: 10.1007/978-3-031-25519-9_61] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
19
|
Pan HT, Xi ZQ, Wei XQ, Wang K. A network pharmacology approach to predict potential targets and mechanisms of " Ramulus Cinnamomi (cassiae) - Paeonia lactiflora" herb pair in the treatment of chronic pain with comorbid anxiety and depression. Ann Med 2022; 54:413-425. [PMID: 35098831 PMCID: PMC8812742 DOI: 10.1080/07853890.2022.2031268] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Traditional Chinese medicine (TCM) prescriptions have multiple bioactive properties. "Gui Zhi-Shao Yao" herb pair is widely used to treat chronic pain (CP), as well as anxiety and depression. However, its related targets and underlying mechanisms have not been deciphered. METHODS In this study, the network pharmacology method was used to explore the bioactive components and targets of "Gui Zhi-Shao Yao" herb pair and further elucidate its potential biological mechanisms of action in the treatment of CP with comorbid anxiety disorder (AD) and mental depression (MD). RESULTS Following a series of analyses, we identified 15 active compounds, hitting 130 potential targets. After the intersections the targets of this herb pair and CP, AD and MD - sorted by the value of degree - nine targets were identified as the vital ones: Akt1, IL6, TNF, PTGS2, JUN, CASP3, MAPK8, PPARγ and NOS3. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis results demonstrated 11 pathways, such as AGE-RAGE signalling pathway, IL-17 signalling pathway, TNF signalling pathway, which primarily participate in the pathological processes. CONCLUSIONS This study preliminarily predicted and verified the pharmacological and molecular mechanisms of "Gui Zhi-Shao Yao" herb pair for treating CP with comorbid AD and MD from a holistic perspective. In vivo and in vitro experiments will be required to further investigate the mechanisms.KEY MESSAGEA network pharmacology approach was applied to identify key targets and molecular mechanisms.Nine targets were regarded as the vital targets for chronic pain with comorbid anxiety and depression.Predicted 11 pathways were the potential therapy targets and pharmacological mechanism of "Gui Zhi-Shao Yao" herb pair.
Collapse
Affiliation(s)
- Hao-Tian Pan
- Acupuncture Anesthesia Clinical Research Institute, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zi-Qi Xi
- Acupuncture Anesthesia Clinical Research Institute, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xu-Qiang Wei
- Acupuncture Anesthesia Clinical Research Institute, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ke Wang
- Acupuncture Anesthesia Clinical Research Institute, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
20
|
Palshikar MG, Palli R, Tyrell A, Maggirwar S, Schifitto G, Singh MV, Thakar J. Executable models of immune signaling pathways in HIV-associated atherosclerosis. NPJ Syst Biol Appl 2022; 8:35. [PMID: 36131068 PMCID: PMC9492768 DOI: 10.1038/s41540-022-00246-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 09/01/2022] [Indexed: 11/09/2022] Open
Abstract
Atherosclerosis (AS)-associated cardiovascular disease is an important cause of mortality in an aging population of people living with HIV (PLWH). This elevated risk has been attributed to viral infection, anti-retroviral therapy, chronic inflammation, and lifestyle factors. However, the rates at which PLWH develop AS vary even after controlling for length of infection, treatment duration, and for lifestyle factors. To investigate the molecular signaling underlying this variation, we sequenced 9368 peripheral blood mononuclear cells (PBMCs) from eight PLWH, four of whom have atherosclerosis (AS+). Additionally, a publicly available dataset of PBMCs from persons before and after HIV infection was used to investigate the effect of acute HIV infection. To characterize dysregulation of pathways rather than just measuring enrichment, we developed the single-cell Boolean Omics Network Invariant Time Analysis (scBONITA) algorithm. scBONITA infers executable dynamic pathway models and performs a perturbation analysis to identify high impact genes. These dynamic models are used for pathway analysis and to map sequenced cells to characteristic signaling states (attractor analysis). scBONITA revealed that lipid signaling regulates cell migration into the vascular endothelium in AS+ PLWH. Pathways implicated included AGE-RAGE and PI3K-AKT signaling in CD8+ T cells, and glucagon and cAMP signaling pathways in monocytes. Attractor analysis with scBONITA facilitated the pathway-based characterization of cellular states in CD8+ T cells and monocytes. In this manner, we identify critical cell-type specific molecular mechanisms underlying HIV-associated atherosclerosis using a novel computational method.
Collapse
Affiliation(s)
- Mukta G Palshikar
- Biophysics, Structural, and Computational Biology Program, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Rohith Palli
- Medical Scientist Training Program, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Alicia Tyrell
- University of Rochester Clinical & Translational Science Institute, Rochester, USA
| | - Sanjay Maggirwar
- Department of Microbiology, Immunology and Tropical Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Giovanni Schifitto
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, USA
- Department of Imaging Sciences, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Meera V Singh
- Department of Neurology, University of Rochester School of Medicine and Dentistry, Rochester, USA
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Juilee Thakar
- Biophysics, Structural, and Computational Biology Program, University of Rochester School of Medicine and Dentistry, Rochester, USA.
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, USA.
- Department of Biostatistics and Computational Biology, University of Rochester School of Medicine and Dentistry, Rochester, USA.
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, USA.
| |
Collapse
|
21
|
Chen X, Shi C, Wang Y, Yu H, Zhang Y, Zhang J, Li P, Gao J. The mechanisms of glycolipid metabolism disorder on vascular injury in type 2 diabetes. Front Physiol 2022; 13:952445. [PMID: 36117707 PMCID: PMC9473659 DOI: 10.3389/fphys.2022.952445] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
Patients with diabetes have severe vascular complications, such as diabetic nephropathy, diabetic retinopathy, cardiovascular disease, and neuropathy. Devastating vascular complications lead to increased mortality, blindness, kidney failure, and decreased overall quality of life in people with type 2 diabetes (T2D). Glycolipid metabolism disorder plays a vital role in the vascular complications of T2D. However, the specific mechanism of action remains to be elucidated. In T2D patients, vascular damage begins to develop before insulin resistance and clinical diagnosis. Endothelial dysregulation is a significant cause of vascular complications and the early event of vascular injury. Hyperglycemia and hyperlipidemia can trigger inflammation and oxidative stress, which impair endothelial function. Furthermore, during the pathogenesis of T2D, epigenetic modifications are aberrant and activate various biological processes, resulting in endothelial dysregulation. In the present review, we provide an overview and discussion of the roles of hyperglycemia- and hyperlipidemia-induced endothelial dysfunction, inflammatory response, oxidative stress, and epigenetic modification in the pathogenesis of T2D. Understanding the connections of glucotoxicity and lipotoxicity with vascular injury may reveal a novel potential therapeutic target for diabetic vascular complications.
Collapse
Affiliation(s)
- Xiatian Chen
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | | | - Yin Wang
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Hua Yu
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao, China
| | - Yu Zhang
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jiaxuan Zhang
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Peifeng Li
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Jinning Gao
- Center for Molecular Genetics, Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
22
|
Advanced Glycation End Products (AGEs) and Chronic Kidney Disease: Does the Modern Diet AGE the Kidney? Nutrients 2022; 14:nu14132675. [PMID: 35807857 PMCID: PMC9268915 DOI: 10.3390/nu14132675] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/22/2022] [Accepted: 06/23/2022] [Indexed: 12/13/2022] Open
Abstract
Since the 1980s, chronic kidney disease (CKD) affecting all ages has increased by almost 25%. This increase may be partially attributable to lifestyle changes and increased global consumption of a “western” diet, which is typically energy dense, low in fruits and vegetables, and high in animal protein and ultra-processed foods. These modern food trends have led to an increase in the consumption of advanced glycation end products (AGEs) in conjunction with increased metabolic dysfunction, obesity and diabetes, which facilitates production of endogenous AGEs within the body. When in excess, AGEs can be pathological via both receptor-mediated and non-receptor-mediated pathways. The kidney, as a major site for AGE clearance, is particularly vulnerable to AGE-mediated damage and increases in circulating AGEs align with risk of CKD and all-cause mortality. Furthermore, individuals with significant loss of renal function show increased AGE burden, particularly with uraemia, and there is some evidence that AGE lowering via diet or pharmacological inhibition may be beneficial for CKD. This review discusses the pathways that drive AGE formation and regulation within the body. This includes AGE receptor interactions and pathways of AGE-mediated pathology with a focus on the contribution of diet on endogenous AGE production and dietary AGE consumption to these processes. We then analyse the contribution of AGEs to kidney disease, the evidence for dietary AGEs and endogenously produced AGEs in driving pathogenesis in diabetic and non-diabetic kidney disease and the potential for AGE targeted therapies in kidney disease.
Collapse
|
23
|
Reddy Addi U, Jakhotia S, Reddy SS, Reddy GB. Advanced glycation end products in brain during aging. Chem Biol Interact 2022; 355:109840. [PMID: 35104490 DOI: 10.1016/j.cbi.2022.109840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 01/18/2022] [Accepted: 01/27/2022] [Indexed: 11/03/2022]
Abstract
Aging is a main risk factor for many diseases including neurodegenerative disorders. Numerous theories and mechanisms including accumulation of advanced glycation end products (AGEs) have been put forward in explaining brain aging. However, a focused study on the status of AGEs in the brain during progressive aging in connection with interrelated cellular processes like ubiquitin-proteasome system (UPS), unfolded protein response, autophagy-lysosome system and apoptosis is lacking. Hence, in this study, we investigated the levels of AGEs in the brain of 5-, 10-, 15- and 20-months old WNIN rats. Endoplasmic reticulum (ER) stress response, UPS components, autophagy flux, neurotrophic and presynaptic markers along with cell death markers were analyzed by immunoblotting. The neuronal architecture was analyzed by H&E and Nissl staining. The results demonstrated progressive accumulation of AGEs in the brain during aging. Adaptive ER stress response was observed by 10-months while maladaptive ER stress response was seen at 15- and 20-months of age along with impaired UPS and autophagy, and perturbations in neuronal growth factors. All these disturbances intensify with age to further exaggerate cell death mechanisms. There was a shrinkage of the cell size with aging and Congo-red staining revealed β-amyloid accumulation in higher ages. Together these results suggest that progressive accumulation of AGEs with aging in the brain may lead to neuronal damage by affecting ER homeostasis, UPS, autophagic flux, and neuronal growth factors.
Collapse
Affiliation(s)
- Utkarsh Reddy Addi
- Biochemistry Division, ICMR-National Institute of Nutrition, Hyderabad, India
| | - Sneha Jakhotia
- Biochemistry Division, ICMR-National Institute of Nutrition, Hyderabad, India
| | - S Sreenivasa Reddy
- Biochemistry Division, ICMR-National Institute of Nutrition, Hyderabad, India.
| | | |
Collapse
|
24
|
Mao L, Yin R, Yang L, Zhao D. Role of advanced glycation end products on vascular smooth muscle cells under diabetic atherosclerosis. Front Endocrinol (Lausanne) 2022; 13:983723. [PMID: 36120471 PMCID: PMC9470882 DOI: 10.3389/fendo.2022.983723] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/11/2022] [Indexed: 11/30/2022] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease and leading cause of cardiovascular diseases. The progression of AS is a multi-step process leading to high morbidity and mortality. Hyperglycemia, dyslipidemia, advanced glycation end products (AGEs), inflammation and insulin resistance which strictly involved in diabetes are closely related to the pathogenesis of AS. A growing number of studies have linked AGEs to AS. As one of the risk factors of cardiac metabolic diseases, dysfunction of VSMCs plays an important role in AS pathogenesis. AGEs are increased in diabetes, participate in the occurrence and progression of AS through multiple molecular mechanisms of vascular cell injury. As the main functional cells of vascular, vascular smooth muscle cells (VSMCs) play different roles in each stage of atherosclerotic lesions. The interaction between AGEs and receptor for AGEs (RAGE) accelerates AS by affecting the proliferation and migration of VSMCs. In addition, increasing researches have reported that AGEs promote osteogenic transformation and macrophage-like transformation of VSMCs, and affect the progression of AS through other aspects such as autophagy and cell cycle. In this review, we summarize the effect of AGEs on VSMCs in atherosclerotic plaque development and progression. We also discuss the AGEs that link AS and diabetes mellitus, including oxidative stress, inflammation, RAGE ligands, small noncoding RNAs.
Collapse
Affiliation(s)
| | | | | | - Dong Zhao
- *Correspondence: Longyan Yang, ; Dong Zhao,
| |
Collapse
|
25
|
Khan SN, Shaheen F, Aleem U, Sheikh S, Tamfu AN, Ashraf S, Ul-Haq Z, Ullah S, Wahab AT, Choudhary MI, Jahan H. Peptide conjugates of 18β-glycyrrhetinic acid as potent inhibitors of α-glucosidase and AGEs-induced oxidation. Eur J Pharm Sci 2021; 168:106045. [PMID: 34666184 DOI: 10.1016/j.ejps.2021.106045] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/20/2021] [Accepted: 10/14/2021] [Indexed: 01/06/2023]
Abstract
18β-Glycyrrhetinic acid (18β-GA) is known for several biological activities, and has been the focus of extensive research for the development of therapeutic agents. In the current study, 18β-GA-peptide conjugates 2-11 were evaluated for their in vitro α-glucosidase inhibitory and antiglycation activities. Structure-activity relationship (SAR) established and molecular interactions of active bioconjugates with the enzyme's binding sites were predicted through molecular modeling approach. In tripeptide moiety of conjugates 2-11, peptide residue at position 1 was found to have a significant role on α-glucosidase inhibition. The most active 18β-GA-peptide conjugates 5 (18β-GA-Cys1-Tyr2-Gly3), and 8 (18β-GA-Pro1-Tyr2-Gly3) exhibited several-fold potent α-glucosidase inhibition (IC50 values 20-28 μM), as compared to standard drug acarbose (IC50 = 875.8 ± 2.10 µM). Kinetic studies of potent compounds, 4-8 revealed that conjugate 5 exhibits competitive-type of inhibition, while conjugates 6-8 showed a non-competitive type of inhibition. The simulation studies also supported the kinetic results that conjugate 5 (18β-GA-Cys1-Tyr2-Gly3) inhibits the α-glucosidase enzyme by blocking its substrate binding site. AGEs-induced NO• inhibitors play an important role in controlling the inflammation associated with diabetes mellitus. The peptide conjugates 2-11 were also evaluated in vitro for AGEs-induced NO• inhibition using RAW 264.7 macrophage cell line. Our data revealed that conjugates 7-10 were the more potent AGEs-induced NO• inhibitors, comparable to standards rutin, and PDTC. The peptide conjugate 5 (a competitive inhibitor of α-glucosidase) also exhibited a strong inhibitory activity against AGEs-induced NO• production. Furthermore, peptide conjugates 2-11 were found non-cytotoxic to mouse fibroblast NIH-3T3, and murine macrophages RAW 264.7 cell lines. In conclusion, our data demonstrates that besides possessing strong α-glucosidase inhibition, the newly synthesized peptide conjugates also alleviated the AGEs-induced NO• production in RAW macrophages. Dual inhibition of α-glucosidase enzyme, and AGEs-induced NO• production by 18β-GA-peptide conjugates qualify them for further research in anti-diabetic drug discovery.
Collapse
Affiliation(s)
- Sadiq Noor Khan
- Third World Center for Science and Technology, H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Farzana Shaheen
- Third World Center for Science and Technology, H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| | - Umair Aleem
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Sumbla Sheikh
- Third World Center for Science and Technology, H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; Institute for Medical Virology and Epidemiology of Viral Diseases, University of Tübingen, Elfriede-Aulhorn-str. 6, 72076 Tübingen, Germany
| | - Alfred Ngenge Tamfu
- Department of Chemical Engineering, School of Chemical Engineering and Mineral Industries, University of Ngaoundere, 454 Ngaoundere, Cameroon
| | - Sajda Ashraf
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Saeed Ullah
- Third World Center for Science and Technology, H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Atia-Tul- Wahab
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - M Iqbal Choudhary
- Third World Center for Science and Technology, H. E. J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan; Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21412, Saudi Arabia
| | - Humera Jahan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| |
Collapse
|
26
|
Huang HL, Kuo CS, Chang TY, Chou RH, Chen IC, Yang FC, Chen NJ, Lin SJ, Wu CC, Huang PH. An oral absorbent, AST-120, restores vascular growth and blood flow in ischemic muscles in diabetic mice via modulation of macrophage transition. J Mol Cell Cardiol 2021; 155:99-110. [PMID: 33713645 DOI: 10.1016/j.yjmcc.2021.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 02/06/2021] [Accepted: 03/04/2021] [Indexed: 10/21/2022]
Abstract
Background Diabetes has a pronounced effect on the peripheral vasculature. The accumulation of advanced glycation end products (AGEs) is regarded as the crucial mechanism responsible for vascular damage in diabetes, but it is not easy to be avoided from food. In this study, we aimed to investigate the effects of an oral absorbent, AST-120, on the accumulation of AGEs and changes in blood flow recovery in diabetic mice. Methods The mice were divided into four groups, wild-type (WT) mice without treatment, WT mice treated with 5% AST-120 mixed into pulverized chow, streptozotocin-induced diabetes mellitus (DM) mice, and DM mice treated with 5% AST-120. Six weeks after hind-limb ischemia surgery, blood flow reperfusion, histology, plasma AGE, and cytokine were examined. Bone marrow cells were cultured and derived into macrophages to evaluate the effects of AGEs on macrophage polarization. Results Plasma AGEs were significantly increased in diabetic mice. AST-120 could bind to AGEs and reduced their plasma concentrations. Histological analysis revealed fewer collateral vessels with corresponding impairment of blood flow recovery in diabetic mice. In these mice, AGE-positive and AGE receptor-positive macrophages were numerous in ischemic limbs compared with non- diabetic mice. In diabetic mice, macrophages in ischemic tissues demonstrated greater M1 polarization than M2 polarization; this pattern was reversed in the AST-120 treatment group. The change in macrophage polarization was associated with the corresponding expression of pro-inflammatory cytokines in the ischemic tissues. In cell cultures, AGEs triggered the transformation of bone marrow-derived macrophages into the M1 phenotype. The alterations in the polarization of macrophages were reversed after treatment with AST-120. Conclusions Oral administration of AST-120 decreased the serum levels of AGEs in diabetic mice and improved neovascularization of ischemic limbs. This benefit may be due to, at least partially, the alterations in macrophage polarization and the associated changes in inflammatory cytokines.
Collapse
Affiliation(s)
- Hsin-Lei Huang
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; National Taipei University of Nursing and Health Sciences, Taiwan
| | - Chin-Sung Kuo
- Division of Endocrinology and Metabolism, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ting-Yung Chang
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Cardiovascular Research Center, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ruey-Hsing Chou
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Cardiovascular Research Center, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - I-Chun Chen
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Fu-Chen Yang
- Institute of Microbiology and Immunology, School of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Nien-Jung Chen
- Institute of Microbiology and Immunology, School of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan; Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shing-Jong Lin
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; Cardiovascular Research Center, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Taipei Medical University, Taipei, Taiwan; Division of Cardiology, Heart Center, Cheng-Hsin General Hospital, Taipei, Taiwan
| | - Chih-Cheng Wu
- National Tsing-Hua University, Institute of Biomedical Engineering, Hsinchu, Taiwan; Cardiovascular Center, National Taiwan University Hospital, Hsinchu Branch, Taipei, Taiwan; National Taiwan University Hospital, College of Medicine, Taipei, Taiwan; Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan.
| | - Po-Hsun Huang
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Department of Critical Care Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Cardiovascular Research Center, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
27
|
Shen L, Zhang T, Yang Y, Lu D, Xu A, Li K. FPS-ZM1 Alleviates Neuroinflammation in Focal Cerebral Ischemia Rats via Blocking Ligand/RAGE/DIAPH1 Pathway. ACS Chem Neurosci 2021; 12:63-78. [PMID: 33300334 DOI: 10.1021/acschemneuro.0c00530] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Receptor for advanced glycation end products (RAGEs), a multiligand receptor belonging to the cell-surface immunoglobulin superfamily, has been reported to play a crucial role in neuroinflammation and neurodegenerative diseases. Here, we tested our hypothesis that the RAGE-specific antagonist FPS-ZM1 is neuroprotective against ischemic brain injury. Distal middle cerebral artery occlusion (MCAO) or sham operation was performed on anesthetized Sprague-Dawley male rats (n = 60), which were then treated with FPS-ZM1 or vehicle (four groups in total = Vehicle + MCAO, FPS-ZM1 + MCAO, Vehicle + sham, and FPS-ZM1 + sham). After 1 week, neurological function was evaluated, and then, brain tissues were collected for 2,3,5-triphenyltetrazolium chloride staining, Nissl staining, TUNEL staining, Western blotting, and immunohistochemical analyses. FPS-ZM1 treatment after MCAO markedly attenuated neurological deficits and reduced the infarct area. More interestingly, FPS-ZM1 inhibited ischemia-induced astrocytic activation and microgliosis and decreased the elevated levels of proinflammatory cytokines. Furthermore, FPS-ZM1 blocked the increase in the level of RAGE and, notably, of DIAPH1, the key cytoplasmic hub for RAGE-ligand-mediated activation of cellular signaling. Accordingly, FPS-ZM1 also reversed the MCAO-induced increase in phosphorylation of NF-κB targets that are potentially downstream from RAGE/DIAPH1. Our findings reveal that FPS-ZM1 treatment reduces neuroinflammation in rats with focal cerebral ischemia and further suggest that the ligand/RAGE/DIAPH1 pathway contributes to this FPS-ZM1-mediated alleviation of neuroinflammation.
Collapse
Affiliation(s)
- Lingling Shen
- Department of Neurology and Stroke Centre, the Fist Affiliated Hospital of Jinan University, Guangzhou 510632, China
- Clinical Neuroscience Institute of Jinan University, Guangzhou 510632, China
| | - Tianyuan Zhang
- Department of Neurology and Stroke Centre, the Fist Affiliated Hospital of Jinan University, Guangzhou 510632, China
- Clinical Neuroscience Institute of Jinan University, Guangzhou 510632, China
| | - Yu Yang
- Department of Neurology and Stroke Centre, the Fist Affiliated Hospital of Jinan University, Guangzhou 510632, China
- Clinical Neuroscience Institute of Jinan University, Guangzhou 510632, China
| | - Dan Lu
- Department of Neurology and Stroke Centre, the Fist Affiliated Hospital of Jinan University, Guangzhou 510632, China
- Clinical Neuroscience Institute of Jinan University, Guangzhou 510632, China
| | - Anding Xu
- Department of Neurology and Stroke Centre, the Fist Affiliated Hospital of Jinan University, Guangzhou 510632, China
- Clinical Neuroscience Institute of Jinan University, Guangzhou 510632, China
| | - Keshen Li
- Clinical Neuroscience Institute of Jinan University, Guangzhou 510632, China
| |
Collapse
|
28
|
Anwar S, Khan S, Almatroudi A, Khan AA, Alsahli MA, Almatroodi SA, Rahmani AH. A review on mechanism of inhibition of advanced glycation end products formation by plant derived polyphenolic compounds. Mol Biol Rep 2021; 48:787-805. [PMID: 33389535 DOI: 10.1007/s11033-020-06084-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/11/2020] [Indexed: 12/15/2022]
Abstract
Advanced glycation end products (AGEs) are naturally occurring biomolecules formed by interaction of reducing sugars with biomolecules such as protein and lipids etc., Long term high blood sugar level and glycation accelerate the formation of AGEs. Unchecked continuous formation and accumulation of AGEs are potential risks for pathogenesis of various chronic diseases. Current mode of antidiabetic therapy is based on synthetic drugs that are often linked with severe adverse effects. Polyphenolic compounds derived from plants are supposed to inhibit glycation and formation of AGEs at multiple levels. Some polyphenolic compounds regulate the blood glucose metabolism by amplification of cell insulin resistance and activation of insulin like growth factor binding protein signaling pathway. Their antioxidant nature and metal chelating activity, ability to trap intermediate dicarbonyl compounds could be possible mechanisms against glycation and AGEs formation and hence, against AGEs induced health complications. Although, few species of polyphenolic compounds are being used in in vitro trials and their in vivo study is still in progress, increasing the area of research in this field may produce a fruitful approach in management of overall diabetic complications.
Collapse
Affiliation(s)
- Shehwaz Anwar
- Department of Medical Laboratories, College of Applied Medical Science, Qassim University, Buraydah, 52571, Saudi Arabia
| | - Shifa Khan
- Department of Biochemistry, Faculty of Medicine, JNMC, Aligarh Muslim University, Aligarh, Uttar Pradesh, 202002, India
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Science, Qassim University, Buraydah, 52571, Saudi Arabia
| | - Amjad Ali Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, P.O. Box 6699, Buraydah, 51452, Saudi Arabia
| | - Mohammed A Alsahli
- Department of Medical Laboratories, College of Applied Medical Science, Qassim University, Buraydah, 52571, Saudi Arabia
| | - Saleh A Almatroodi
- Department of Medical Laboratories, College of Applied Medical Science, Qassim University, Buraydah, 52571, Saudi Arabia
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Science, Qassim University, Buraydah, 52571, Saudi Arabia.
| |
Collapse
|
29
|
Abstract
The advanced glycosylation end product receptor (RAGE) acts as a recognition receptor and interacts with different types of ligands that form and accumulate in the tissues and circulation, such as diabetes, inflammation, insulin resistance, and obesity. In these environments, RAGE is expressed on the surface of various cells associated with tissue disturbance. This review mainly summarizes the characteristics of RAGE-related signalling, with a particular emphasis on the role of RAGE in the development of obesity. We also briefly describe the phenotypes and characteristics of macrophages and focus on the role of adipose tissue macrophages (ATMs) and the regulatory mechanisms in obesity, diabetes, and other related metabolic diseases. Besides, we will also elaborate on the prospect of new strategies for treating diabetes and obesity-related metabolic diseases by inhibiting RAGE signalling and regulating ATMs recruitment and polarization.
Collapse
Affiliation(s)
- Ziqian Feng
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Luochen Zhu
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Jianbo Wu
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Drug Discovery Research Center, Southwest Medical University, Luzhou, China
- Laboratory for Cardiovascular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
30
|
Joseph S, Walejko JM, Zhang S, Edison AS, Keller-Wood M. Maternal hypercortisolemia alters placental metabolism: a multiomics view. Am J Physiol Endocrinol Metab 2020; 319:E950-E960. [PMID: 32954824 PMCID: PMC7790119 DOI: 10.1152/ajpendo.00190.2020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Previous studies have suggested that increases in maternal cortisol or maternal stress in late pregnancy increase the risk of stillbirth at term. In an ovine model with increased maternal cortisol over the last 0.20 of gestation, we have previously found evidence of disruption of fetal serum and cardiac metabolomics and altered expression of genes related to mitochondrial function and metabolism in biceps femoris, diaphragm, and cardiac muscle. The present studies were designed to test for effects of chronically increased maternal cortisol on gene expression and metabolomics in placentomes near term. We hypothesized that changes in placenta might underlie or contribute to the alterations in fetal serum metabolomics and thereby contribute to changes in striated muscle metabolism. Placentomes were collected from pregnancies in early labor (143 ± 1 days gestation) of control ewes (n = 7) or ewes treated with cortisol (1 mg·kg-1·day-1 iv; n = 5) starting at day 115 of gestation. Transcriptomics and metabolomics were performed using an ovine gene expression microarray (Agilent 019921) and HR-MAS NMR, respectively. Multiomic analysis indicates that amino acid metabolism, particularly of branched-chain amino acids and glutamate, occur in placenta; changes in amino acid metabolism, degradation, or biosynthesis in placenta were consistent with changes in valine, isoleucine, leucine, and glycine in fetal serum. The analysis also indicates changes in glycerophospholipid metabolism and suggests changes in endoplasmic reticulum stress and antioxidant status in the placenta. These findings suggest that changes in placental function occurring with excess maternal cortisol in late gestation may contribute to metabolic dysfunction at birth.
Collapse
Affiliation(s)
- Serene Joseph
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida
| | - Jacquelyn M Walejko
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville Florida
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia
| | - Sicong Zhang
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia
| | - Arthur S Edison
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia
- Department of Genetics, Institute of Bioinformatics, University of Georgia, Athens, Georgia
| | - Maureen Keller-Wood
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida
- D.H. Barron Reproductive and Perinatal Biology Research Program, University of Florida, Gainesville, Florida
| |
Collapse
|
31
|
Haddad M, Perrotte M, Landri S, Lepage A, Fülöp T, Ramassamy C. Circulating and Extracellular Vesicles Levels of N-(1-Carboxymethyl)-L-Lysine (CML) Differentiate Early to Moderate Alzheimer's Disease. J Alzheimers Dis 2020; 69:751-762. [PMID: 31127773 DOI: 10.3233/jad-181272] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Both advanced glycation end products (AGEs) N-(1-carboxymethyl)-L-lysine (CML) and pentosidine were found in the brain from Alzheimer's disease (AD) patients and were associated with the neuropathological hallmarks of AD. In AD patients, the circulating level of both AGEs remains unknown. Moreover, their levels in peripheral extracellular vesicles (EVs) and their association with AD remain to be determined. Finally, it is not known if neuronal cells can release AGEs via EVs and propagate AGEs. OBJECTIVE To determine the levels of circulating CML and pentosidine during the progression of AD. Moreover, their levels in circulating EVs were determined and their association with the clinical cognitive scores were analyzed. Finally, we have studied the possibility that neuronal cells eliminate and transfer these AGEs through EVs. METHODS CML and pentosidine levels were measured in serum and in circulating EVs. Released-EVs from SK-N-SH neuronal cells were isolated and CML levels were also determined. RESULTS The levels of CML in albumin-free serum proteins were higher in the early stage of AD while the levels of pentosidine remained unchanged. In contrast, the levels of CML in the EVs were lower in the moderate stage of AD. Interestingly, the levels of CML in serum were negatively correlated with the clinical cognitive scores MMSE and MoCA. For the first time, we were able to demonstrate that CML was present in EVs released from neuronal cells in culture. CONCLUSION Peripheral and circulating EVs levels of CML can differentiate early to moderate AD. In the brain, neuronal CML can propagate from cells-to-cells via EVs.
Collapse
Affiliation(s)
- Mohamed Haddad
- INRS-Institut Armand-Frappier, Laval, QC, Canada.,Institute of Nutrition and Functional Foods, Université Laval, Québec, QC, Canada
| | - Morgane Perrotte
- INRS-Institut Armand-Frappier, Laval, QC, Canada.,Institute of Nutrition and Functional Foods, Université Laval, Québec, QC, Canada
| | - Sarra Landri
- INRS-Institut Armand-Frappier, Laval, QC, Canada.,Institute of Nutrition and Functional Foods, Université Laval, Québec, QC, Canada
| | - Aurelie Lepage
- Department of Medicine, Geriatric Division, Research Center on Aging, Sherbrooke University, Sherbrooke, QC, Canada
| | - Tamàs Fülöp
- Department of Medicine, Geriatric Division, Research Center on Aging, Sherbrooke University, Sherbrooke, QC, Canada
| | - Charles Ramassamy
- INRS-Institut Armand-Frappier, Laval, QC, Canada.,Institute of Nutrition and Functional Foods, Université Laval, Québec, QC, Canada
| |
Collapse
|
32
|
Liu Z, Ma Y, Cui Q, Xu J, Tang Z, Wang Y, He C, Wang X. Toll-like receptor 4 plays a key role in advanced glycation end products-induced M1 macrophage polarization. Biochem Biophys Res Commun 2020; 531:602-608. [PMID: 32814631 DOI: 10.1016/j.bbrc.2020.08.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 08/06/2020] [Indexed: 12/30/2022]
Abstract
OBJECTIVE This study was aimed to investigate the role of Toll-like receptor 4 (TLR4) in advanced glycation end products (AGEs)- induced macrophage polarization toward M1. METHODS Isolated primary macrophages were exposed to prepared AGEs at concentrations of 0, 2.5, 5 and 10 μmol/L. Macrophages were also exposed to hydrogen peroxide (H2O2) which provided exogenous reactive oxygen species (ROS). Receptor for AGEs (RAGE) was over-expressed by a vector. Specific siRNA silencing TLR4 and inhibitor TAK-242 were used to pre-treat the macrophages. Intracellular ROS was determined by DCFH-DA. Immunofluorescence staining was used to evaluate the expression of inducible nitric oxide synthase (iNOS) which is the marker of M1 macrophage phenotype. Real-time PCR was used to assess the mRNA expression level of TLR4 and RAGE. Protein expression levels of cytoplasmic RAGE, TLR4, nuclear signal transducers and activators of transcription 1 (STAT1) and phosphorylation levels of cytoplasmic STAT1 were evaluated by Western blotting. ELISA was used to measure concentrations of interleukin 6 (IL6), IL12 and tumor necrosis factor (TNF)α in supernatant of cell culture medium of macrophages. RESULTS AGEs significantly elevated intracellular ROS generation, expression levels of iNOS, cytoplasmic RAGE, TLR4, nuclear STAT1, phosphorylation levels of cytoplasmic STAT1, as well as IL6, IL12 and TNFα contents in a concentration-dependent manner. TLR4 silencing and inhibitor pre-treatment reduced expression levels of cytoplasmic RAGE, TLR4, phosphorylation of STAT1 and nuclear STAT1 in AGEs-exposed macrophages without affecting RAGE expression and intracellular ROS production levels. RAGE over-expression elevated both ROS and TLR4 expression levels in macrophages. TLR4 expression elevation was also found in H2O2-treat macrophages. CONCLUSION AGEs induced macrophage polarization toward M1 via activating RAGE/ROS/TLR4/STAT1 signaling pathway.
Collapse
Affiliation(s)
- Zhongwei Liu
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Yanpeng Ma
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Qianwei Cui
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Jing Xu
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Zhiguo Tang
- Department of Cardiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China
| | - Yuan Wang
- Department of Medical Prevention, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi, China.
| | - Chunhui He
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.
| | - Xi Wang
- Department of Obstetrics and Gynecology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
33
|
Le Bagge S, Fotheringham AK, Leung SS, Forbes JM. Targeting the receptor for advanced glycation end products (RAGE) in type 1 diabetes. Med Res Rev 2020; 40:1200-1219. [PMID: 32112452 DOI: 10.1002/med.21654] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/09/2019] [Accepted: 11/12/2019] [Indexed: 12/18/2022]
Abstract
Type 1 diabetes (T1D) is one of the most common chronic diseases manifesting in early life, with the prevalence increasing worldwide at a rate of approximately 3% per annum. The prolonged hyperglycaemia characteristic of T1D upregulates the receptor for advanced glycation end products (RAGE) and accelerates the formation of RAGE ligands, including advanced glycation end products, high-mobility group protein B1, S100 calcium-binding proteins, and amyloid-beta. Interestingly, changes in the expression of RAGE and these ligands are evident in patients before the onset of T1D. RAGE signals via various proinflammatory cascades, resulting in the production of reactive oxygen species and cytokines. A large number of proinflammatory ligands that can signal via RAGE have been implicated in several chronic diseases, including T1D. Therefore, it is unsurprising that RAGE has become a potential therapeutic target for the treatment and prevention of disease. In this review, we will explore how RAGE might be targeted to prevent the development of T1D.
Collapse
Affiliation(s)
- Selena Le Bagge
- Glycation and Diabetes, Translational Research Institute (TRI), Mater Research Institute-The University of Queensland (MRI-UQ), Brisbane, Queensland, Australia.,School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Amelia K Fotheringham
- Glycation and Diabetes, Translational Research Institute (TRI), Mater Research Institute-The University of Queensland (MRI-UQ), Brisbane, Queensland, Australia.,School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Sherman S Leung
- Glycation and Diabetes, Translational Research Institute (TRI), Mater Research Institute-The University of Queensland (MRI-UQ), Brisbane, Queensland, Australia.,School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Josephine M Forbes
- Glycation and Diabetes, Translational Research Institute (TRI), Mater Research Institute-The University of Queensland (MRI-UQ), Brisbane, Queensland, Australia.,Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia.,Mater Clinical School, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
34
|
Lee TW, Kao YH, Chen YJ, Chao TF, Lee TI. Therapeutic potential of vitamin D in AGE/RAGE-related cardiovascular diseases. Cell Mol Life Sci 2019; 76:4103-4115. [PMID: 31250032 PMCID: PMC11105755 DOI: 10.1007/s00018-019-03204-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 06/15/2019] [Accepted: 06/19/2019] [Indexed: 12/18/2022]
Abstract
Cardiovascular diseases (CVDs) are among the leading threats to human health. The advanced glycation end product (AGE) and receptor for AGE (RAGE) signaling pathway regulates the pathogenesis of CVDs, through its effects on arterial stiffness, atherosclerosis, mitochondrial dysfunction, oxidative stress, calcium homeostasis, and cytoskeletal function. Targeting the AGE/RAGE pathway is a potential therapeutic strategy for ameliorating CVDs. Vitamin D has several beneficial effects on the cardiovascular system. Experimental findings have shown that vitamin D regulates AGE/RAGE signaling and its downstream effects. This article provides a comprehensive review of the mechanistic insights into AGE/RAGE involvement in CVDs and the modulation of the AGE/RAGE signaling pathways by vitamin D.
Collapse
Affiliation(s)
- Ting-Wei Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, 111 Xinglong Road, Section 3 Wenshan District, Taipei, 11696, Taiwan
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tze-Fan Chao
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine and Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Ting-I Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, 111 Xinglong Road, Section 3 Wenshan District, Taipei, 11696, Taiwan.
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
35
|
Inflammation‐regulatory microRNAs: Valuable targets for intracranial atherosclerosis. J Neurosci Res 2019; 97:1242-1252. [DOI: 10.1002/jnr.24487] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/24/2019] [Accepted: 06/10/2019] [Indexed: 12/16/2022]
|
36
|
Koregol AC, Kalburgi NB, Kanniappa Sadasivan S, Warad S, Kamat Wagh A, Thomas T, Sinha P. 8-Isoprostane in chronic periodontitis and type II diabetes: Exploring the link. J Dent Res Dent Clin Dent Prospects 2019; 12:252-257. [PMID: 30774790 PMCID: PMC6368953 DOI: 10.15171/joddd.2018.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 12/09/2018] [Indexed: 12/31/2022] Open
Abstract
Background. Reactive oxygen species (ROS) are associated with the pathogenesis of inflammatory diseases and have a direct or indirect role in tissue damage constituting oxidative stress. ROS are also involved in impairment of β-cell function during development of diabetes, which leads to genetic ablation of KATP channels, triggering up-regulation of antioxidant enzymes. Several markers of lipid peroxidation, protein oxidation and DNA damage induced by ROS can be measured. Over the last decade, isoprostanes have been considered as the best markers of lipid peroxidation. The aim of this study was to determine the presence of 8-isoprostane in healthy, chronic periodontitis and chronic periodontitis subjects with type II diabetes and to find the correlation between 8-isoprostane levels among groups and with clinical parameters like gingival index, probing depth and clinical attachment levels.
Methods. Ninety subjects were selected and divided into 3 groups: healthy, chronic periodontitis and chronic periodontitis subjects with type II diabetes (n=30 each). Saliva was collected from these subjects after obtaining consent and analyzed for 8-isoprostane levels using ELISA kit. Statistical analysis was performed using Kruskal-Wallis test, Mann-Whitney U test and Spearman’s correlation coefficient (P<0.001).
Results. Statistically significant difference was found in the levels of 8-isoprostane between healthy, chronic periodontitis and chronic periodontitis subjects with type II diabetes and with all clinical parameters.
Conclusion. 8-isoprostane can be considered as a pathophysiological marker to measure oxidative stress in periodontal diseases.
Collapse
Affiliation(s)
- Arati C Koregol
- Department of Periodontics, P.M. Nadagouda Memorial Dental College and Hospital, Bagalkot, India
| | - Nagaraj B Kalburgi
- Department of Periodontics, P.M. Nadagouda Memorial Dental College and Hospital, Bagalkot, India
| | | | - Shivaraj Warad
- Department of Periodontics, P.M. Nadagouda Memorial Dental College and Hospital, Bagalkot, India
| | - Apoorva Kamat Wagh
- Department of Periodontics, P.M. Nadagouda Memorial Dental College and Hospital, Bagalkot, India
| | - Tivin Thomas
- Department of Periodontics, P.M. Nadagouda Memorial Dental College and Hospital, Bagalkot, India
| | - Priya Sinha
- Department of Periodontics, P.M. Nadagouda Memorial Dental College and Hospital, Bagalkot, India
| |
Collapse
|
37
|
Jud P, Sourij H. Therapeutic options to reduce advanced glycation end products in patients with diabetes mellitus: A review. Diabetes Res Clin Pract 2019; 148:54-63. [PMID: 30500546 DOI: 10.1016/j.diabres.2018.11.016] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 11/14/2018] [Accepted: 11/23/2018] [Indexed: 12/18/2022]
Abstract
Diabetes mellitus (DM) defines metabolic disorders, characterised by elevated levels of blood glucose. Chronic hyperglycaemic state promotes consequently the formation of advanced glycation end products (AGEs) and the expression of their receptor (RAGE) which aggravate many diabetic complications. Due to the relevant role of AGEs and RAGE, several therapeutic approaches with an anti-AGE or RAGE-antagonizing effect are investigated. These therapeutic options include AGE cross-link breakers, AGE inhibitors, RAGE antagonists, drugs clinically approved for various indications like antidiabetic, antihypertensive drugs or statins, as well as dietary and phytotherapeutic approaches. The aim of this review is to give an overview of these therapeutic approaches, their outcomes in clinical studies and their role in the management of diabetes and its complications.
Collapse
Affiliation(s)
- Philipp Jud
- Division of Angiology, Department of Internal Medicine, Medical University of Graz, Graz, Austria.
| | - Harald Sourij
- Division of Endocrinology and Diabetology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
38
|
Srivastava RAK. Life-style-induced metabolic derangement and epigenetic changes promote diabetes and oxidative stress leading to NASH and atherosclerosis severity. J Diabetes Metab Disord 2018; 17:381-391. [PMID: 30918873 DOI: 10.1007/s40200-018-0378-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 11/20/2018] [Indexed: 12/15/2022]
Abstract
Energy imbalance resulting from high calorie food intake and insufficient metabolic activity leads to increased body mass index (BMI) and sets the stage for metabolic derangement influencing lipid and carbohydrate metabolism and ultimately leading to insulin resistance, dyslipidemia, and type 2 diabetes. 70% of cardiovascular disease (CVD) deaths occur in patients with diabetes. Environment-induced physiological perturbations trigger epigenetic changes through chromatin modification and leads to type 2 diabetes and progression of nonalcoholic fatty liver disease (NAFLD) and CVD. Thus, in terms of disease progression and pathogenesis, energy homeostasis, metabolic dysregulation, diabetes, fatty liver, and CVD are interlinked. Since advanced glycation end products (AGEs) and low-grade inflammation in type 2 diabetes play definitive roles in the pathogenesis of liver and vascular diseases, a natural checkpoint to prevent diabetes and associated complications appears to be the identification and management of prediabetes together with weight management, since 70% of prediabetic individuals develop diabetes during their life time, and every kg of weight increase is associated with up to 9% increase in diabetes risk. A good proportion of diabetes and obesity population have fatty liver that progresses to non-alcoholic steatohepatitis (NASH) and cirrhosis, and increased risk of hepatocellular carcinoma. Diabetes and NASH both have elevated oxidative stress, impaired cholesterol elimination, and increased inflammation that leads to CVD risk. This review addresses life-style-induced metabolic pathway derangement and how it contributes to epigenetic changes, type 2 diabetes and NASH progression, which collectively lead to increased risk of CVD.
Collapse
Affiliation(s)
- Rai Ajit K Srivastava
- Integrated Pharma Solutions, Philadelphia, PA USA.,2Department of Nutrition, Wayne State University, Detroit, MI USA
| |
Collapse
|
39
|
Schlotterer A, Kolibabka M, Lin J, Acunman K, Dietrich N, Sticht C, Fleming T, Nawroth P, Hammes HP. Methylglyoxal induces retinopathy-type lesions in the absence of hyperglycemia: studies in a rat model. FASEB J 2018; 33:4141-4153. [PMID: 30485119 DOI: 10.1096/fj.201801146rr] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The aim of this study was to evaluate whether damage to the neurovascular unit in diabetes depends on reactive metabolites such as methylglyoxal (MG), and to assess its impact on retinal gene expression. Male Wistar rats were supplied with MG (50 mM) by drinking water and compared with age-matched streptozotocin-diabetic animals and untreated controls. Retinal damage was evaluated for the accumulation of MG-derived advanced glycation end products, changes in hexosamine and PKC pathway activation, microglial activation, vascular alterations (pericyte loss and vasoregression), neuroretinal function assessed by electroretinogram, and neurodegeneration. Retinal gene regulation was studied by microarray analysis, and transcription factor involvement was identified by upstream regulator analysis. Systemic application of MG by drinking water increased retinal MG to levels comparable with diabetic animals. Elevated retinal MG resulted in MG-derived hydroimidazolone modifications in the ganglion cell layer, inner nuclear layer, and outer nuclear layer, a moderate activation of the hexosamine pathway, a pan-retinal activation of microglia, loss of pericytes, increased formation of acellular capillaries, decreased function of bipolar cells, and increased expression of the crystallin gene family. MG mimics important aspects of diabetic retinopathy and plays a pathogenic role in microglial activation, vascular damage, and neuroretinal dysfunction. In response to MG, the retina induces expression of neuroprotective crystallins.-Schlotterer, A., Kolibabka, M., Lin, J., Acunman, K., Dietrich, N., Sticht, C., Fleming, T., Nawroth, P., Hammes, H.-P. Methylglyoxal induces retinopathy-type lesions in the absence of hyperglycemia: studies in a rat model.
Collapse
Affiliation(s)
- Andrea Schlotterer
- Fifth Medical Department, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Matthias Kolibabka
- Fifth Medical Department, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Jihong Lin
- Fifth Medical Department, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Kübra Acunman
- Fifth Medical Department, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Nadine Dietrich
- Fifth Medical Department, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Carsten Sticht
- Medical Research Center, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany; and
| | - Thomas Fleming
- Department of Medicine I and Clinical Chemistry, Heidelberg University, Heidelberg, Germany
| | - Peter Nawroth
- Department of Medicine I and Clinical Chemistry, Heidelberg University, Heidelberg, Germany
| | - Hans-Peter Hammes
- Fifth Medical Department, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
40
|
RAGE deficiency does not affect non-alcoholic steatohepatitis and atherosclerosis in Western type diet-fed Ldlr -/- mice. Sci Rep 2018; 8:15256. [PMID: 30323247 PMCID: PMC6189204 DOI: 10.1038/s41598-018-33661-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 09/28/2018] [Indexed: 01/11/2023] Open
Abstract
Non-alcoholic fatty liver disease is a spectrum of liver diseases ranging from steatosis only to non-alcoholic steatohepatitis (NASH). The latter is characterized by hepatic inflammation, which increases the risk of cardiovascular disease. It is poorly understood which factors contribute to the onset of hepatic inflammation characterizing the progression from steatosis to NASH. Previously, we demonstrated increased advanced glycation endproducts (AGEs) in the livers of NASH patients. We hypothesise that AGEs play a key role in NASH development by activating their proinflammatory receptor, RAGE. RAGE-deficient mice and wildtype littermates, both on Ldlr−/− background, were fed a Western type diet (WTD) for 3 or 12 weeks. Flow cytometry, histology, gene expression and AGE measurements were performed to evaluate the effects of RAGE deficiency. RAGE-deficient mice displayed reduced weight gain and visceral fat expansion compared to control mice. No difference in adipose tissue inflammation was observed between groups. RAGE deficiency did not affect WTD-induced monocytosis, circulating lipids or hepatic steatosis. WTD-induced hepatic neutrophil and macrophage accumulation and atherosclerotic plaque development was comparable between control and RAGE-deficient mice. No difference in AGE levels was observed. RAGE does not seem to play a major role in the development of NASH or atherosclerosis in a hyperlipidemic mouse model.
Collapse
|
41
|
Zanotto C, Hansen F, Galland F, Batassini C, Federhen BC, da Silva VF, Leite MC, Nardin P, Gonçalves CA. Glutamatergic Alterations in STZ-Induced Diabetic Rats Are Reversed by Exendin-4. Mol Neurobiol 2018; 56:3538-3551. [PMID: 30145785 DOI: 10.1007/s12035-018-1320-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 08/14/2018] [Indexed: 01/25/2023]
Abstract
Diabetes mellitus is a metabolic disorder that results in glucotoxicity and the formation of advanced glycated end products (AGEs), which mediate several systemic adverse effects, particularly in the brain tissue. Alterations in glutamatergic neurotransmission and cognitive impairment have been reported in DM. Exendin-4 (EX-4), an analogue of glucagon-like peptide-1 (GLP-1), appears to have beneficial effects on cognition in rats with chronic hyperglycemia. Herein, we investigated the ability of EX-4 to reverse changes in AGE content and glutamatergic transmission in an animal model of DM looking principally at glutamate uptake and GluN1 subunit content of the N-methyl-D-aspartate (NMDA) receptor. Additionally, we evaluated the effects of EX-4 on in vitro models and the signaling pathway involved in these effects. We found a decrease in glutamate uptake and GluN1 content in the hippocampus of diabetic rats; EX-4 was able to revert these parameters, but had no effect on the other parameters evaluated (glycemia, C-peptide, AGE levels, RAGE, and glyoxalase 1). EX-4 abrogated the decrease in glutamate uptake and GluN1 content caused by methylglyoxal (MG) in hippocampal slices, in addition to leading to an increase in glutamate uptake in astrocyte culture cells and hippocampal slices under basal conditions. The effect of EX-4 on glutamate uptake was mediated by the phosphatidylinositide 3-kinases (PI3K) signaling pathway, which could explain the protective effect of EX-4 in the brain tissue, since PI3K is involved in cell metabolism, inhibition of apoptosis, and reduces inflammatory responses. These results suggest that EX-4 could be used as an adjuvant treatment for brain impairment associated with excitotoxicity.
Collapse
Affiliation(s)
- Caroline Zanotto
- Department of Biochemistry, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.
| | - Fernanda Hansen
- Department of Nutrition, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Fabiana Galland
- Department of Biochemistry, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Cristiane Batassini
- Department of Biological Sciences, Integrated Regional University of Alto Uruguai and Missões, Frederico Westphalen, Brazil
| | | | | | - Marina Concli Leite
- Department of Biochemistry, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Patrícia Nardin
- Department of Biochemistry, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Carlos-Alberto Gonçalves
- Department of Biochemistry, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| |
Collapse
|
42
|
Şakul A, Arı N, Sotnikova R, Ozansoy G, Karasu Ç. A pyridoindole antioxidant SMe1EC2 regulates contractility, relaxation ability, cation channel activity, and protein-carbonyl modifications in the aorta of young and old rats with or without diabetes mellitus. GeroScience 2018; 40:377-392. [PMID: 30054861 DOI: 10.1007/s11357-018-0034-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 07/17/2018] [Indexed: 12/18/2022] Open
Abstract
We studied the effects of treatment with SMe1EC, a hexahydropyridoindole antioxidant, on vascular reactivity, endothelial function, and oxidonitrosative stress level of thoracic aorta in young and old rats with or without diabetes mellitus. The rats were grouped as young control (YC 3 months old), old control (OC 15 months old), young diabetic (YD), old diabetic (OD), young control treated (YCT), old control treated (OCT), young diabetic treated (YDT), and old diabetic treated (ODT). Diabetes was induced by streptozotocin injection and subsequently SMe1EC2 (10 mg/kg/day, p.o.) was administered to YCT, OCT, YDT, and ODT rats for 5 months. In young and old rats, diabetes resulted in hypertension, weight loss, hyperglycemia, and hypertriglyceridemia, which were partially prevented by SMe1EC2. SMe1EC2 also inhibited the diabetes-induced increase in aorta levels of AGEs (advanced glycosylation end-protein adducts), 4-HNE (4-hydroxy-nonenal-histidine), 3-NT (3-nitrotyrosine), and RAGEs (receptors for AGEs). The contractions of the aorta rings to phenylephrine (Phe) and KCL did not significantly change, but acetylcholine (ACh) and salbutamol relaxations were reduced in OC compared to YC rats. Diabetes induction increased Phe contractions in YC and OC rats, KCL contractions in YC rats, and did not cause further inhibition in already inhibited ACh and salbutamol relaxations in OC rats. We have achieved the lowest levels of ACh relaxation in YD rats compared to other groups. SMe1EC2 did not change the response of aorta to ACh, salbutamol and Phe in YC rats, and ameliorated ACh relaxations in OC and YD but not in OD rats. In YDT and ODT rats, increased Phe and KCL contractions, high blood pressure, and impaired salbutamol relaxations were amended by SMe1EC2. Phe contractions observed in YD and OD rats as well as KCl contractions observed in OC rats were the lowest levels when the rats were treated with SMe1EC2. When the bath solution was shifted to cyclopiazonic acid (CYP) or CYP plus Ca2+-free medium, the contraction induced by a single dose of Phe (3 × 10-6 M) was more inhibited in YD and OD than in YC but not in OC rats. In SMe1EC2-treated rats, neither the presence of CFM nor CFM plus CYP exhibited a significant change in response of aorta to a single dose of Phe. These findings suggest that α1-adrenergic receptor signaling is activated in both age groups of diabetic rats, diabetes activates K+-depolarization and calcium mobilization via CaV especially in the aorta of young rats, and sensitizes the aorta of old rats to the regulating effect of SMe1EC2. ACh relaxations were inhibited in YC rats, increased in OC rats and unchanged in YD and OD rats when aortic rings pretreated with TEA, an inhibitor of calcium-activated K+ channels (KCa), or 4-aminopyridine (4-AP), an inhibitor of voltage-sensitive K+ channels (KV). ACh relaxations were inhibited in YCT, OCT, and YDT rats in the presence of 4-AP or TEA. In ODT rats, 4-AP did not change ACh relaxation but TEA inhibited. These findings suggest that the contribution of Kv and KCa to ACh relaxation is likely upregulated by SMe1EC2 when the relaxations were inhibited by aging or diabetes. We conclude that SMe1EC2 might be a promising agent for aging and diabetes related vascular disorders.
Collapse
Affiliation(s)
- Arzu Şakul
- Department of Pharmacology, Istanbul Medipol University, Istanbul, Turkey
| | - Nuray Arı
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Ruzenna Sotnikova
- Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Gülgün Ozansoy
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Çimen Karasu
- Laboratory for Cellular Stress Response and Signal Transduction Research, Department of Medical Pharmacology, Faculty of Medicine, Gazi University, Ankara, Turkey.
| |
Collapse
|
43
|
Tang SG, Liu XY, Ye JM, Hu TT, Yang YY, Han T, Tan W. Isosteviol ameliorates diabetic cardiomyopathy in rats by inhibiting ERK and NF-κB signaling pathways. J Endocrinol 2018; 238:47-60. [PMID: 29720537 DOI: 10.1530/joe-17-0681] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 04/16/2018] [Indexed: 12/23/2022]
Abstract
Diabetes-induced injury of myocardium, defined as diabetic cardiomyopathy (DCM), accounts for significant mortality and morbidity in diabetic population. Alleviation of DCM by a potent drug remains considerable interests in experimental and clinical researches because hypoglycemic drugs cannot effectively control this condition. Here, we explored the beneficial effects of isosteviol sodium (STVNa) on type 1 diabetes-induced DCM and the potential mechanisms involved. Male Wistar rats were induced to diabetes by injection of streptozotocin (STZ). One week later, diabetic rats were randomly grouped to receive STVNa (STZ/STVNa) or its vehicle (STZ). After 11 weeks of treatment or 11 weeks treatment following 4 weeks of removal of the treatment, the cardiac function and structure were evaluated and related mechanisms were investigated. In diabetic rats, oxidative stress, inflammation, blood glucose and plasma advanced glycation end products (AGEs) were significantly increased, whereas superoxide dismutase 2 (SOD-2) expression and activity were decreased. STVNa treatment inhibited cardiac hypertrophy, fibrosis and inflammation, showed similar ratio of heart to body weight and antioxidant capacities almost similar to the normal controls, which can be sustained at least 4 weeks. Moreover, STVNa inhibited diabetes-inducted stimulation of both extracellular signal-regulated kinase (ERK) and nuclear factor κB (NF-κB) signal pathways. However, blood glucose, plasma AGE and insulin levels were not altered by STVNa treatment. These results indicate that STVNa may be developed into a potent therapy for DCM. The mechanism underlying this therapeutic effect involves the suppression of oxidative stress and inflammation by inhibiting ERK and NF-κB without changing blood glucose or AGEs.
Collapse
Affiliation(s)
- Sheng-Gao Tang
- School of Bioscience and BioengineeringSouth China University of Technology, Guangzhou, China
| | - Xiao-Yu Liu
- School of Bioscience and BioengineeringSouth China University of Technology, Guangzhou, China
| | - Ji-Ming Ye
- Molecular Pharmacology for DiabetesSchool of Health and Biomedical Sciences, RMIT University, Melbourne, Victoria, Australia
| | - Ting-Ting Hu
- School of Bioscience and BioengineeringSouth China University of Technology, Guangzhou, China
| | - Ying-Ying Yang
- School of Bioscience and BioengineeringSouth China University of Technology, Guangzhou, China
| | - Ting Han
- School of Bioscience and BioengineeringSouth China University of Technology, Guangzhou, China
| | - Wen Tan
- Institute of Biomedical & Pharmaceutical ScienceGuangdong University of Technology, Guangzhou, China
| |
Collapse
|
44
|
The Ability of Different Ketohexoses to Alter Apo-A-I Structure and Function In Vitro and to Induce Hepatosteatosis, Oxidative Stress, and Impaired Plasma Lipid Profile in Hyperlipidemic Zebrafish. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3124364. [PMID: 29951163 PMCID: PMC5987316 DOI: 10.1155/2018/3124364] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 10/25/2017] [Indexed: 01/22/2023]
Abstract
In the current study, we have tested the nonenzymatic glycation activities of ketohexoses, such as tagatose and psicose. Although tagatose-treated apoA-I (t-A-I) and psicose-treated apoA-I (p-A-I) exerted more inhibitory activity you cupric ion-mediated low-density lipoprotein (LDL) oxidation and oxidized LDL (oxLDL) phagocytosis into macrophage than fructose-treated apoA-I (f-A-I). In the lipid-free state, t-A-I and f-A-I showed more multimerized band without crosslinking. Since t-A-I lost its phospholipid binding ability, the rHDL formation was not as successful as f-A-I. However, injecting t-A-I showed more antioxidant activities in zebrafish embryo under the presence of oxLDL. Three weeks of consumption of fructose (50% of wt in Tetrabit/4% cholesterol) showed a 14% elevation of serum triacylglycerol (TG), while tagatose-administered group showed 30% reduction in serum TG compared to high cholesterol control. Fructose-fed group showed the biggest area of Oil Red O staining with the intensity as strong as the HCD control. However, tagatose-consumed group showed much lesser Oil Red O-stained area with the reduction of lipid accumulation. In conclusion, although tagatose treatment caused modification of apoA-I, the functional loss was not as much severe as the fructose treatment in macrophage cell model, zebrafish embryo, and hypercholesterolemic zebrafish model.
Collapse
|
45
|
Dhulekar J, Simionescu A. Challenges in vascular tissue engineering for diabetic patients. Acta Biomater 2018; 70:25-34. [PMID: 29396167 PMCID: PMC5871600 DOI: 10.1016/j.actbio.2018.01.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 01/06/2018] [Accepted: 01/09/2018] [Indexed: 12/16/2022]
Abstract
Hyperglycemia and dyslipidemia coexist in diabetes and result in inflammation, degeneration, and impaired tissue remodeling, processes which are not conducive to the desired integration of tissue engineered products into the surrounding tissues. There are several challenges for vascular tissue engineering such as non-thrombogenicity, adequate burst pressure and compliance, suturability, appropriate remodeling responses, and vasoactivity, but, under diabetic conditions, an additional challenge needs to be considered: the aggressive oxidative environment generated by the high glucose and lipid concentrations that lead to the formation of advanced glycation end products (AGEs) in the vascular wall. Extracellular matrix-based scaffolds have adequate physical properties and are biocompatible, however, these scaffolds are altered in diabetes by the formation AGEs and impaired collagen degradation, consequently increasing vascular wall stiffness. In addition, vascular cells detect and respond to altered stimuli from the matrix by pathological remodeling of the vascular wall. Due to the immunomodulatory effects of mesenchymal stem cells (MSCs), they are frequently used in tissue engineering in order to protect the scaffolds from inflammation. MSCs together with antioxidant treatments of the scaffolds are expected to protect the vascular grafts from diabetes-induced alterations. In conclusion, as one of the most daunting environments that could damage the ECM and its interaction with cells is progressively built in diabetes, we recommend that cells and scaffolds used in vascular tissue engineering for diabetic patients are tested in diabetic animal models, in order to obtain valuable results regarding their resistance to diabetic adversities. STATEMENT OF SIGNIFICANCE Almost 25 million Americans have diabetes, characterized by high levels of blood sugar that binds to tissues and disturbs the function of cardiovascular structures. Therefore, patients with diabetes have a high risk of cardiovascular diseases. Surgery is required to replace diseased arteries with implants, but these fail after 5-10 years because they are made of non-living materials, not resistant to diabetes. New tissue engineering materials are developed, based on the patients' own stem cells, isolated from fat, and added to extracellular matrix-based scaffolds. Our main concern is that diabetes could damage the tissue-like implants. Thus we review studies related to the effect of diabetes on tissue components and recommend antioxidant treatments to increase the resistance of implants to diabetes.
Collapse
|
46
|
Balbino KP, Hermsdorff HHM, Bressan J. Polymorphism related to cardiovascular risk in hemodialysis subjects: a systematic review. J Bras Nefrol 2018; 40:179-192. [PMID: 29944163 PMCID: PMC6533983 DOI: 10.1590/2175-8239-jbn-3857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 08/23/2017] [Indexed: 02/01/2023] Open
Abstract
Cardiovascular disease (CVD) is one of the leading causes of mortality in hemodialysis (HD) subjects. In addition to the traditional risk factors that are common in these individuals, genetic factors are also involved, with emphasis on single nucleotide polymorphs (SNPs). In this context, the present study aims to systematically review the studies that investigated the polymorphisms associated with cardiovascular risk in this population. In general, the SNPs present in HD individuals are those of genes related to inflammation, oxidative stress and vascular calcification, also able of interfering in the cardiovascular risk of this population. In addition, polymorphisms in genes related to recognized risk factors for CVD, such as dyslipidemia, arterial hypertension and left ventricular hypertrophy, also influence cardiovascular morbidity and mortality.
Collapse
Affiliation(s)
- Karla Pereira Balbino
- Universidade Federal de Viçosa, Departamento de Nutrição e Saúde,
Viçosa, MG, Brasil
| | | | - Josefina Bressan
- Universidade Federal de Viçosa, Departamento de Nutrição e Saúde,
Viçosa, MG, Brasil
| |
Collapse
|
47
|
Madhurantakam S, Selvaraj S, Rayappan JBB, Krishnan UM. Exploring hesperidin-copper complex as an enzyme mimic for monitoring macrophage activity. J Solid State Electrochem 2018. [DOI: 10.1007/s10008-018-3883-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
48
|
Lee TI, Bai KJ, Chen YC, Lee TW, Chung CC, Tsai WC, Tsao SY, Kao YH. Histone deacetylase inhibition of cardiac autophagy in rats on a high‑fat diet with low‑dose streptozotocin-induced type 2 diabetes mellitus. Mol Med Rep 2017; 17:594-601. [PMID: 29115461 DOI: 10.3892/mmr.2017.7905] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 08/03/2017] [Indexed: 11/06/2022] Open
Abstract
Autophagy serves a role in preserving cellular homeostasis. Diabetes mellitus (DM) impairs cardiac autophagy and is associated with an accumulation of cytotoxic proteins that may provoke apoptosis and damage cardiomyocytes. Histone deacetylase (HDAC) inhibitors attenuate cardiac fibrosis and inflammation, and improve cardiomyopathy resulting from DM. However, the effect of HDAC inhibition on autophagy in DM cardiomyopathy has not been investigated. The purpose of the present study was to evaluate whether HDAC inhibition modulates cardiac autophagy and to investigate the potential mechanisms in type 2 DM (T2DM) hearts. Electrocardiography was used to evaluate cardiac function and western blotting was used to evaluate protein expression in autophagy, the serine/threonine protein kinase mTOR (mTOR) signaling pathway, poly adenosine diphosphate ribose polymerase 1 (PARP1), insulin signaling, advanced glycosylation end product‑specific receptor (RAGE), and proinflammatory cytokines in control rats and in rats treated with a high‑fat diet (60% fat) and low‑dose streptozotocin (35 mg/kg) in order to induce T2DM, with or without an HDAC inhibitor (MPT0E014; 50 mg/kg/rat daily for 7 days). Compared with the control rats, T2DM and T2DM rats treated with MPT0E014 exhibited elevated blood glucose levels and similar body weights. However, T2DM rats treated with MPT0E014 and control rats had a smaller left ventricular end‑diastolic diameter compared with the T2DM rats. The control and T2DM rats treated with MPT0E014 had greater protein expression of cardiac phosphorylated (p)‑5' adenosine monophosphate‑activated protein kinase α 2, light chain 3‑II, Beclin‑1, glucose transporter 4, p‑protein kinase B, and insulin receptor substrate‑1 (Ser 307) compared with T2DM rats. In addition, control and T2DM rats treated with MPT0E014 had decreased cardiac protein expression of cleaved PARP1, p‑mTOR‑S2448, p‑P70S6K‑Thr‑389, RAGE, tumor necrosis factor‑α, and interleukin‑6 compared with T2DM rats. The present study demonstrated that MPT0E014 may improve cardiac function in T2DM rats by modulating myocardial autophagy, inflammation and insulin signaling.
Collapse
Affiliation(s)
- Ting-I Lee
- Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
| | - Kuan-Jen Bai
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei 11490, Taiwan, R.O.C
| | - Ting-Wei Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei 11696, Taiwan, R.O.C
| | - Cheng-Chih Chung
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
| | - Wen-Chih Tsai
- Division of Cardiology, Tzu‑Chi General Hospital, Institute of Medical Sciences, Tzu‑Chi University, Hualien 97004, Taiwan, R.O.C
| | - Shin-Yi Tsao
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Sijhih Cathay General Hospital, New Taipei City 22174, Taiwan, R.O.C
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan, R.O.C
| |
Collapse
|
49
|
Tsujinaka H, Itaya-Hironaka A, Yamauchi A, Sakuramoto-Tsuchida S, Shobatake R, Makino M, Masuda N, Hirai H, Takasawa S, Ogata N. Statins decrease vascular epithelial growth factor expression via down-regulation of receptor for advanced glycation end-products. Heliyon 2017; 3:e00401. [PMID: 28971147 PMCID: PMC5612812 DOI: 10.1016/j.heliyon.2017.e00401] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 07/06/2017] [Accepted: 08/31/2017] [Indexed: 12/13/2022] Open
Abstract
Aims Statins, inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A reductase, possess pleiotropic effects that have been extended to modulation of various cellular behaviors. This study aimed to examine whether statins modulate vascular endothelial growth factor A (VEGF-A) expression in human retinal pigment epithelium (RPE) cells. Main methods Human RPE cells (h1RPE7), damaged by hydroquinone (HQ) + advanced glycation endproducts (AGE) in an in vitro AMD model, were treated with atorvastatin or lovastatin for 24 h. The expression of VEGF-A and receptor for AGE (RAGE) was evaluated by real-time RT-PCR. VEGF-A secretion was measured by ELISA. To investigate the impact of RAGE on VEGF-A expression, small interfering RNA (siRNA) for RAGE (siRAGE) was introduced into h1RPE7 cells and VEGF-A expression was measured by real-time RT-PCR. Deletions of VEGF-A and RAGE promoters were performed and transcriptional activities were measured after the addition of statins to HQ + AGE-damaged RPE cells. Key findings The mRNA levels of VEGF-A and RAGE and the levels of VEGF-A in the culture medium were increased by HQ + AGE. Both atorvastatin and lovastatin attenuated HQ + AGE-induced VEGF-A and RAGE expression. These statins also decreased VEGF-A levels in the culture medium. RNA interference of RAGE attenuated the up-regulation of VEGF-A in the HQ + AGE treated cells. The deletion analysis demonstrated that these statins attenuated RAGE promoter activation in HQ + AGE-damaged RPE cells. Significance Statins attenuated HQ + AGE-induced VEGF expression by decreasing RAGE expression. As VEGF is an important factor in developing wet AMD, statins could decrease the risk of wet-type AMD and be used as preventive medicines.
Collapse
Affiliation(s)
- Hiroki Tsujinaka
- Department of Ophthalmology, Nara Medical University, Kashihara 634-8522, Japan.,Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | | | - Akiyo Yamauchi
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | | | - Ryogo Shobatake
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Mai Makino
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Naonori Masuda
- Department of Ophthalmology, Nara Medical University, Kashihara 634-8522, Japan
| | - Hiromasa Hirai
- Department of Ophthalmology, Nara Medical University, Kashihara 634-8522, Japan
| | - Shin Takasawa
- Department of Biochemistry, Nara Medical University, Kashihara 634-8521, Japan
| | - Nahoko Ogata
- Department of Ophthalmology, Nara Medical University, Kashihara 634-8522, Japan
| |
Collapse
|
50
|
Lee TW, Kao YH, Lee TI, Chen YJ. ADAM10 modulates calcitriol-regulated RAGE in cardiomyocytes. Eur J Clin Invest 2017; 47:675-683. [PMID: 28722189 DOI: 10.1111/eci.12789] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 07/15/2017] [Indexed: 12/28/2022]
Abstract
BACKGROUND Receptor for advanced glycation end products (RAGE) signalling plays a critical role in the pathogenesis of cardiovascular disease. Calcitriol modulates cardiac RAGE expression. This study explored the mechanisms underlying the effect of calcitriol on RAGE and soluble RAGE (sRAGE) expression in cardiomyocytes. MATERIALS AND METHODS Western blot, ELISA, fluorometric assay and PCR analyses were used to evaluate the RAGE, sRAGE, endogenous secretory RAGE (esRAGE), Jun N-terminal kinase (JNK), and a disintegrin and metalloprotease 10 (ADAM10) expression and enzyme activity in HL-1 atrial myocytes without and with calcitriol (10 and 100 nM), nuclear factor-κB (NF-κB) inhibitor (50 μg/mL), or ADAM10 inhibitor (5 μM) incubation for 48 h. RESULTS Calcitriol (10 nM) significantly reduced RAGE protein expression and increased sRAGE concentrations in HL-1 cardiomyocytes compared with control cells. These changes were associated with increased protein expression and enzyme activity of ADAM10 and higher mRNA expression of esRAGE. In the presence of ADAM10 inhibitor, however, the suppressive effect of calcitriol on RAGE was diminished. Methylglyoxal (500 μM for 10 min)-mediated JNK phosphorylation was attenuated in the presence of calcitriol (10 nM). Moreover, control and NF-κB inhibitor-treated HL-1 cells had similar RAGE and sRAGE expression, suggesting that calcitriol-mediated RAGE modulation was independent of NF-κB signalling. CONCLUSIONS We showed that RAGE downregulation and increased sRAGE production by calcitriol were mediated through ADAM10 activation in cardiomyocytes. The results suggest that calcitriol has therapeutic potential in treating RAGE-mediated cardiovascular complications.
Collapse
Affiliation(s)
- Ting-Wei Lee
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Ting-I Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|