1
|
Vianello E, Beltrami AP, Aleksova A, Janjusevic M, Fluca AL, Corsi Romanelli MM, La Sala L, Dozio E. The Advanced Glycation End-Products (AGE)-Receptor for AGE System (RAGE): An Inflammatory Pathway Linking Obesity and Cardiovascular Diseases. Int J Mol Sci 2025; 26:3707. [PMID: 40332316 PMCID: PMC12028226 DOI: 10.3390/ijms26083707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/31/2025] [Accepted: 04/11/2025] [Indexed: 05/08/2025] Open
Abstract
The AGE (advanced glycation end-products)-RAGE (receptor for AGE) system is a pro-inflammatory pathway that contributes to the pathogenesis of obesity and obesity-related cardiovascular disorders (CVD). Circulating AGE and the soluble form of RAGE (sRAGE) has been suggested as a potential biomarker of CVD related to obesity. In this study, we aim to (1) summarize the current knowledge about the role of obesity in the onset and progression of CVD, (2) discuss the role of the AGE-RAGE system as a pathway promoting obesity and linking obesity to CVD, and (3) highlight available strategies for reducing AGE-RAGE system activation and the associated beneficial effects.
Collapse
Affiliation(s)
- Elena Vianello
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (E.V.); (M.M.C.R.); (L.L.S.)
- Experimental Laboratory for Research on Organ Damage Biomarkers, IRCCS Istituto Auxologico Italiano, 20149 Milan, Italy
| | - Antonio P. Beltrami
- Department of Medicine, Università degli Studi di Udine, 33100 Udine, Italy;
- Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy
| | - Aneta Aleksova
- Department of Medical Surgical and Health Sciences, Università degli Studi di Trieste, 34129 Trieste, Italy; (A.A.); (M.J.); (A.L.F.)
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina, 34100 Trieste, Italy
| | - Milijana Janjusevic
- Department of Medical Surgical and Health Sciences, Università degli Studi di Trieste, 34129 Trieste, Italy; (A.A.); (M.J.); (A.L.F.)
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina, 34100 Trieste, Italy
| | - Alessandra L. Fluca
- Department of Medical Surgical and Health Sciences, Università degli Studi di Trieste, 34129 Trieste, Italy; (A.A.); (M.J.); (A.L.F.)
- Cardiothoracovascular Department, Azienda Sanitaria Universitaria Giuliano Isontina, 34100 Trieste, Italy
| | - Massimiliano M. Corsi Romanelli
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (E.V.); (M.M.C.R.); (L.L.S.)
- Department of Clinical and Experimental Pathology, IRCCS Istituto Auxologico Italiano, 20149 Milan, Italy
| | - Lucia La Sala
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (E.V.); (M.M.C.R.); (L.L.S.)
- IRCCS Multimedica, 20138 Milan, Italy
| | - Elena Dozio
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (E.V.); (M.M.C.R.); (L.L.S.)
- Experimental Laboratory for Research on Organ Damage Biomarkers, IRCCS Istituto Auxologico Italiano, 20149 Milan, Italy
| |
Collapse
|
2
|
Van den Eynde MDG, Houben AJHM, Scheijen JLJM, Linkens AMA, Niessen PM, Simons N, Hanssen NMJ, Kusters YHAM, Eussen SJMP, Miyata T, Stehouwer CDA, Schalkwijk CG. Pyridoxamine reduces methylglyoxal and markers of glycation and endothelial dysfunction, but does not improve insulin sensitivity or vascular function in abdominally obese individuals: A randomized double-blind placebo-controlled trial. Diabetes Obes Metab 2023; 25:1280-1291. [PMID: 36655410 DOI: 10.1111/dom.14977] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023]
Abstract
AIM To investigate the effects of pyridoxamine (PM), a B6 vitamer and dicarbonyl scavenger, on glycation and a large panel of metabolic and vascular measurements in a randomized double-blind placebo-controlled trial in abdominally obese individuals. MATERIALS AND METHODS Individuals (54% female; mean age 50 years; mean body mass index 32 kg/m2 ) were randomized to an 8-week intervention with either placebo (n = 36), 25 mg PM (n = 36) or 200 mg PM (n = 36). We assessed insulin sensitivity, β-cell function, insulin-mediated microvascular recruitment, skin microvascular function, flow-mediated dilation, and plasma inflammation and endothelial function markers. PM metabolites, dicarbonyls and advanced glycation endproducts (AGEs) were measured using ultra-performance liquid chromatography tandem mass spectrometry. Treatment effects were evaluated by one-way ANCOVA. RESULTS In the high PM dose group, we found a reduction of plasma methylglyoxal (MGO) and protein-bound Nδ-(5-hydro-5-methyl-4-imidazolon-2-yl)-ornithine (MG-H1), as compared to placebo. We found a reduction of the endothelial dysfunction marker soluble vascular cell adhesion molecule-1 (sVCAM-1) in the low and high PM dose group and of soluble intercellular adhesion molecule-1 (sICAM-1) in the high PM dose, as compared to placebo. We found no treatment effects on insulin sensitivity, vascular function or other functional outcome measurements. CONCLUSIONS This study shows that PM is metabolically active and reduces MGO, AGEs, sVCAM-1 and sICAM-1, but does not affect insulin sensitivity and vascular function in abdominally obese individuals. The reduction in adhesion markers is promising because these are important in the pathogenesis of endothelial damage and atherosclerosis.
Collapse
Affiliation(s)
- Mathias D G Van den Eynde
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, The Netherlands
- Top Institute of Food and Nutrition (TIFN), Wageningen, The Netherlands
| | - Alfons J H M Houben
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, The Netherlands
- Top Institute of Food and Nutrition (TIFN), Wageningen, The Netherlands
| | - Jean L J M Scheijen
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, The Netherlands
| | - Armand M A Linkens
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, The Netherlands
| | - Petra M Niessen
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, The Netherlands
| | - Nynke Simons
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, The Netherlands
| | - Nordin M J Hanssen
- Amsterdam Diabetes Center, Department of Internal and Vascular Medicine, Amsterdam UMC Locatie AMC, Amsterdam, The Netherlands
| | - Yvo H A M Kusters
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, The Netherlands
| | - Simone J M P Eussen
- School for Cardiovascular Diseases (CARIM), Maastricht, The Netherlands
- Department of Epidemiology, Maastricht University, Maastricht, The Netherlands
- Care and Public Health Research Institute (CAPHRI), Maastricht, The Netherlands
| | - Toshio Miyata
- Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Coen D A Stehouwer
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, The Netherlands
- Top Institute of Food and Nutrition (TIFN), Wageningen, The Netherlands
| | - Casper G Schalkwijk
- Department of Internal Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Cardiovascular Diseases (CARIM), Maastricht, The Netherlands
- Top Institute of Food and Nutrition (TIFN), Wageningen, The Netherlands
| |
Collapse
|
3
|
Hadzi-Petrushev N, Angelovski M, Mladenov M. Advanced Glycation End Products and Diabetes. CONTEMPORARY ENDOCRINOLOGY 2023:99-127. [DOI: 10.1007/978-3-031-39721-9_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
4
|
Filip A, Taleb S, Bascetin R, Jahangiri M, Bardin M, Lerognon C, Fève B, Lacolley P, Jalkanen S, Mercier N. Increased atherosclerotic plaque in AOC3 knock-out in ApoE−/− mice and characterization of AOC3 in atherosclerotic human coronary arteries. Front Cardiovasc Med 2022; 9:848680. [PMID: 36176983 PMCID: PMC9513161 DOI: 10.3389/fcvm.2022.848680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 08/04/2022] [Indexed: 11/20/2022] Open
Abstract
Introduction Amine oxidase copper containing 3 (AOC3) displays adhesion between leukocytes and endothelial cells and enzymatic functions. Given its controversial role in atherogenesis, we proposed to investigate the involvement of AOC3 in the formation of atherosclerotic plaques in ApoE−/−AOC3−/− mice and human coronary arteries. Methods Lesions, contractile markers, and AOC3 were studied in aortic tissues from 15- and 25-week-old mice and different stages of human coronary atherosclerotic arteries by immunohistochemistry (IHC) and/or western blot. Human VSMCs, treated or not with LJP1586, an AOC3 inhibitor, were used to measure differentiation markers by qPCR. AOC3 co-localization with specific cell markers was studied by using confocal microscopy in mice and human samples. Results At 15 weeks old, the absence of AOC3 was associated with increased lesion size, α-SMA, and CD3 staining in the plaque independently of a cholesterol modification. At 25 weeks old, advanced plaques were larger with equivalent staining for α-SMA while CD3 increased in the media from ApoE−/−AOC3−/− mice. At both ages, the macrophage content of the lesion was not modified. Contractile markers decreased whereas MCP-1 appeared augmented only in the 15-week-old ApoE−/−AOC3. AOC3 is mainly expressed by mice and human VSMC is slightly expressed by endothelium but not by macrophages. Conclusion AOC3 knock-out increased atherosclerotic plaques at an early stage related to a VSMC dedifferentiation associated with a higher T cells recruitment in plaques explained by the MCP-1 augmentation. This suggests that AOC3 may have an important role in atherosclerosis independent of its canonical inflammatory effect. The dual role of AOC3 impacts therapeutic strategies using pharmacological regulators of SSAO activity.
Collapse
Affiliation(s)
- Anna Filip
- Université de Lorraine, Inserm, Défaillance Cardiovasculaire Aigue et Chronique (DCAC), Université de Lorraine, Lorraine, France
| | - Soraya Taleb
- Inserm UMR_S970, Paris Centre de Recherche Cardiovasculaire (PARCC), Paris, France
| | - Rümeyza Bascetin
- Université de Lorraine, Inserm, Défaillance Cardiovasculaire Aigue et Chronique (DCAC), Université de Lorraine, Lorraine, France
| | - Mohammad Jahangiri
- Université de Lorraine, Inserm, Défaillance Cardiovasculaire Aigue et Chronique (DCAC), Université de Lorraine, Lorraine, France
| | - Matthieu Bardin
- Université de Lorraine, Inserm, Défaillance Cardiovasculaire Aigue et Chronique (DCAC), Université de Lorraine, Lorraine, France
| | - Cindy Lerognon
- Université de Lorraine, Inserm, Défaillance Cardiovasculaire Aigue et Chronique (DCAC), Université de Lorraine, Lorraine, France
| | - Bruno Fève
- Sorbonne Université, Inserm UMR_S938, Centre de Recherche Saint Antoine, IHU ICAN, Service d'Endocrinologie, CRMR PRISIS, APHP Hôpital Saint-Antoine, Paris, France
| | - Patrick Lacolley
- Université de Lorraine, Inserm, Défaillance Cardiovasculaire Aigue et Chronique (DCAC), Université de Lorraine, Lorraine, France
| | - Sirpa Jalkanen
- Medicity Laboratory, University of Turku, Turku, Finland
| | - Nathalie Mercier
- Université de Lorraine, Inserm, Défaillance Cardiovasculaire Aigue et Chronique (DCAC), Université de Lorraine, Lorraine, France
- *Correspondence: Nathalie Mercier
| |
Collapse
|
5
|
García-Vega D, González-Juanatey JR, Eiras S. Diabesity in Elderly Cardiovascular Disease Patients: Mechanisms and Regulators. Int J Mol Sci 2022; 23:7886. [PMID: 35887234 PMCID: PMC9318065 DOI: 10.3390/ijms23147886] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 12/04/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death in the world. In 2019, 550 million people were suffering from CVD and 18 million of them died as a result. Most of them had associated risk factors such as high fasting glucose, which caused 134 million deaths, and obesity, which accounted for 5.02 million deaths. Diabesity, a combination of type 2 diabetes and obesity, contributes to cardiac, metabolic, inflammation and neurohumoral changes that determine cardiac dysfunction (diabesity-related cardiomyopathy). Epicardial adipose tissue (EAT) is distributed around the myocardium, promoting myocardial inflammation and fibrosis, and is associated with an increased risk of heart failure, particularly with preserved systolic function, atrial fibrillation and coronary atherosclerosis. In fact, several hypoglycaemic drugs have demonstrated a volume reduction of EAT and effects on its metabolic and inflammation profile. However, it is necessary to improve knowledge of the diabesity pathophysiologic mechanisms involved in the development and progression of cardiovascular diseases for comprehensive patient management including drugs to optimize glucometabolic control. This review presents the mechanisms of diabesity associated with cardiovascular disease and their therapeutic implications.
Collapse
Affiliation(s)
- David García-Vega
- Cardiology and Intensive Cardiac Care Department, University Hospital, 15706 Santiago de Compostela, Spain;
- Cardiology Group, Health Research Institute, 15706 Santiago de Compostela, Spain
| | - José Ramón González-Juanatey
- Cardiology and Intensive Cardiac Care Department, University Hospital, 15706 Santiago de Compostela, Spain;
- Cardiology Group, Health Research Institute, 15706 Santiago de Compostela, Spain
- CIBERCV, 28029 Madrid, Spain
| | - Sonia Eiras
- CIBERCV, 28029 Madrid, Spain
- Translational Cardiology Group (Laboratory 6), Health Research Institute, 15706 Santiago de Compostela, Spain
| |
Collapse
|
6
|
Sen A, Vincent V, Thakkar H, Abraham R, Ramakrishnan L. Beneficial Role of Vitamin D on Endothelial Progenitor Cells (EPCs) in Cardiovascular Diseases. J Lipid Atheroscler 2022; 11:229-249. [PMID: 36212746 PMCID: PMC9515729 DOI: 10.12997/jla.2022.11.3.229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/23/2022] [Accepted: 04/04/2022] [Indexed: 11/23/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death in the world. Endothelial progenitor cells (EPCs) are currently being explored in the context of CVD risk. EPCs are bone marrow derived progenitor cells involved in postnatal endothelial repair and neovascularization. A large body of evidence from clinical, animal, and in vitro studies have shown that EPC numbers in circulation and their functionality reflect endogenous vascular regenerative capacity. Traditionally vitamin D is known to be beneficial for bone health and calcium metabolism and in the last two decades, its role in influencing CVD and cancer risk has generated significant interest. Observational studies have shown that low vitamin D levels are associated with an adverse cardiovascular risk profile. Still, Mendelian randomization studies and randomized control trials (RCTs) have not shown significant effects of vitamin D on cardiovascular events. The criticism regarding the RCTs on vitamin D and CVD is that they were not designed to investigate cardiovascular outcomes in vitamin D-deficient individuals. Overall, the association between vitamin D and CVD remains inconclusive. Recent clinical and experimental studies have demonstrated the beneficial role of vitamin D in increasing the circulatory level of EPC as well as their functionality. In this review we present evidence supporting the beneficial role of vitamin D in CVD through its modulation of EPC homeostasis.
Collapse
Affiliation(s)
- Atanu Sen
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Vinnyfred Vincent
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Himani Thakkar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Ransi Abraham
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Lakshmy Ramakrishnan
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
7
|
Copur S, Rossing P, Afsar B, Sag AA, Siriopol D, Kuwabara M, Ortiz A, Kanbay M. A primer on metabolic memory: why existing diabesity treatments fail. Clin Kidney J 2021; 14:756-767. [PMID: 34512957 PMCID: PMC8422888 DOI: 10.1093/ckj/sfaa143] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Indexed: 11/28/2022] Open
Abstract
Despite massive government and private sector investments into prevention of cardiovascular disease, diabetes mellitus and obesity, efforts have largely failed, and the burden of cost remains in the treatment of downstream morbidity and mortality, with overall stagnating outcomes. A new paradigm shift in the approach to these patients may explain why existing treatment strategies fail, and offer new treatment targets. This review aims to provide a clinician-centred primer on metabolic memory, defined as the sum of irreversible genetic, epigenetic, cellular and tissue-level alterations that occur with long-time exposure to metabolic derangements.
Collapse
Affiliation(s)
- Sidar Copur
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Peter Rossing
- Steno Diabetes Center Copenhagen, University of Copenhagen, Copenhagen, Denmark
| | - Baris Afsar
- Department of Internal Medicine, Division of Nephrology, Suleyman Demirel University School of Medicine, Isparta, Turkey
| | - Alan A Sag
- Department of Radiology, Division of Vascular and Interventional Radiology, Duke University Medical Center, Durham, NC, USA
| | - Dimitrie Siriopol
- Nephrology Clinic, Dialysis and Renal Transplant Center, 'C.I. PARHON' University Hospital, 'Grigore T. Popa' University of Medicine, Iasi, Romania
| | | | - Alberto Ortiz
- School of Medicine, Dialysis Unit, IIS-Fundacion Jimenez Diaz, Universidad Autónoma de Madrid, Madrid, Spain
| | - Mehmet Kanbay
- Department of Medicine, Division of Nephrology, Koc University School of Medicine, Istanbul, Turkey
| |
Collapse
|
8
|
Menini S, Iacobini C, Fantauzzi CB, Pugliese G. L-carnosine and its Derivatives as New Therapeutic Agents for the Prevention and Treatment of Vascular Complications of Diabetes. Curr Med Chem 2020; 27:1744-1763. [PMID: 31296153 DOI: 10.2174/0929867326666190711102718] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/20/2019] [Accepted: 04/25/2019] [Indexed: 02/01/2023]
Abstract
Vascular complications are among the most serious manifestations of diabetes. Atherosclerosis is the main cause of reduced life quality and expectancy in diabetics, whereas diabetic nephropathy and retinopathy are the most common causes of end-stage renal disease and blindness. An effective therapeutic approach to prevent vascular complications should counteract the mechanisms of injury. Among them, the toxic effects of Advanced Glycation (AGEs) and Lipoxidation (ALEs) end-products are well-recognized contributors to these sequelae. L-carnosine (β-alanyl-Lhistidine) acts as a quencher of the AGE/ALE precursors Reactive Carbonyl Species (RCS), which are highly reactive aldehydes derived from oxidative and non-oxidative modifications of sugars and lipids. Consistently, L-carnosine was found to be effective in several disease models in which glyco/lipoxidation plays a central pathogenic role. Unfortunately, in humans, L-carnosine is rapidly inactivated by serum carnosinase. Therefore, the search for carnosinase-resistant derivatives of Lcarnosine represents a suitable strategy against carbonyl stress-dependent disorders, particularly diabetic vascular complications. In this review, we present and discuss available data on the efficacy of L-carnosine and its derivatives in preventing vascular complications in rodent models of diabetes and metabolic syndrome. We also discuss genetic findings providing evidence for the involvement of the carnosinase/L-carnosine system in the risk of developing diabetic nephropathy and for preferring the use of carnosinase-resistant compounds in human disease. The availability of therapeutic strategies capable to prevent both long-term glucose toxicity, resulting from insufficient glucoselowering therapy, and lipotoxicity may help reduce the clinical and economic burden of vascular complications of diabetes and related metabolic disorders.
Collapse
Affiliation(s)
- Stefano Menini
- Department of Clinical and Molecular Medicine, "La Sapienza" University, Rome, Italy
| | - Carla Iacobini
- Department of Clinical and Molecular Medicine, "La Sapienza" University, Rome, Italy
| | | | - Giuseppe Pugliese
- Department of Clinical and Molecular Medicine, "La Sapienza" University, Rome, Italy
| |
Collapse
|
9
|
Gryszczyńska B, Budzyń M, Formanowicz D, Wanic-Kossowska M, Formanowicz P, Majewski W, Iskra M, Kasprzak MP. Selected Atherosclerosis-Related Diseases May Differentially Affect the Relationship between Plasma Advanced Glycation End Products, Receptor sRAGE, and Uric Acid. J Clin Med 2020; 9:E1416. [PMID: 32397681 PMCID: PMC7290396 DOI: 10.3390/jcm9051416] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 05/05/2020] [Accepted: 05/07/2020] [Indexed: 12/11/2022] Open
Abstract
Our study aimed to identify the relationship between advanced glycation end products (AGEs), soluble receptor for advanced glycation end products (sRAGE), the AGEs/sRAGE, and uric acid (UA) levels in selected atherosclerosis diseases, i.e., abdominal aortic aneurysms (AAA), aortoiliac occlusive disease (AIOD), and chronic kidney disease (CKD), resulting from apparent differences in oxidative stress intensity. Furthermore, we suggest that increased AGEs levels may stimulate an antioxidant defense system reflected by the UA level. The studied group size consisted of 70 AAA patients, 20 AIOD patients, 50 patients in the pre-dialyzed group (PRE), and 35 patients in the hemodialyzed group (HD). The enzyme-linked immunosorbent assay was used to measure AGEs and sRAGE levels. We found a significantly higher concentration of AGEs in CKD patients as compared to AAA and AIOD patients. Furthermore, the sRAGE level was higher in the CKD patients in comparison to AIOD and AAA patients. UA level was significantly higher in the PRE group compared to AAA patients. In conclusion, the diseases included in this study differ in the anti- and prooxidant defense system, which is reflected in the relations between the AGEs, the sRAGE, the AGEs/sRAGE ratio, as well as the UA levels.
Collapse
Affiliation(s)
- Bogna Gryszczyńska
- Department of General Chemistry, Chair of Chemistry and Clinical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 8, 60-806 Poznan, Poland; (M.B.); (M.I.); (M.P.K.)
| | - Magdalena Budzyń
- Department of General Chemistry, Chair of Chemistry and Clinical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 8, 60-806 Poznan, Poland; (M.B.); (M.I.); (M.P.K.)
| | - Dorota Formanowicz
- Department of Clinical Biochemistry and Laboratory Medicine, Poznan University of Medical Sciences, Rokietnicka 8, 60-806 Poznan, Poland;
| | - Maria Wanic-Kossowska
- Department of Nephrology, Transplantology and Internal Medicine, Poznan University of Medical Sciences, Przybyszewskiego 49, 60-355 Poznan, Poland;
| | - Piotr Formanowicz
- Institute of Computing Science, Poznan University of Technology, Piotrowo 2, 60-965 Poznan, Poland;
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Wacław Majewski
- Department of General and Vascular Surgery, Poznan University of Medical Sciences, Dluga 1/2, 61-848 Poznan, Poland;
| | - Maria Iskra
- Department of General Chemistry, Chair of Chemistry and Clinical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 8, 60-806 Poznan, Poland; (M.B.); (M.I.); (M.P.K.)
| | - Magdalena P. Kasprzak
- Department of General Chemistry, Chair of Chemistry and Clinical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 8, 60-806 Poznan, Poland; (M.B.); (M.I.); (M.P.K.)
| |
Collapse
|
10
|
Affiliation(s)
- Miriam F. W.eiss
- University Hospitals of Cleveland and Case Western Reserve University; Cleveland, Ohio, U.S.A
| |
Collapse
|
11
|
Tsuboi H, Takahashi M, Minamida Y, Yoshida N. Psychological well-being and green tea consumption are associated with lower pentosidine serum levels among elderly female residents in Japan. J Psychosom Res 2019; 126:109825. [PMID: 31520891 DOI: 10.1016/j.jpsychores.2019.109825] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 09/05/2019] [Accepted: 09/06/2019] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Pentosidine (PEN), a well-defined advanced glycation end product (AGE), may be affected by psychological status, given the recent findings regarding AGE receptor functions. Because AGEs can be a factor in aging and in the development or worsening of many degenerative diseases, it is important to find a way to reduce the PEN levels in our body. This study aims to investigate novel factors associated with PEN levels. METHODS A cross-sectional study involving 106 female participants (aged 59-69) was conducted in a tea-producing district in Japan. The serum concentration of PEN was detected and evaluated in relation to the participants' psychological status, which was assessed using the Japanese version of the 28-item General Health Questionnaire (GHQ) and lifestyle factors. Factors that were significantly associated with PEN were analysed using multiple linear regression analyses. Significance was defined as p < .05. RESULTS The serum PEN concentrations were significantly and positively associated with the total GHQ scores and BMI after controlling for covariates (standardised beta coefficient (B) = 0.26, p < .01; B = 0.27, p < .01, respectively). In addition, the PEN levels were significantly lower in participants who consumed seven cups or more of green tea per day than those who consumed six or fewer cups per day (B = 0.19, p < .05). CONCLUSIONS Low GHQ scores (i.e. better psychological well-being) and green tea consumption may be helpful in decreasing AGEs.
Collapse
Affiliation(s)
- Hirohito Tsuboi
- Institute of Medical, Pharmaceutical & Health Sciences, Kanazawa University, Kanazawa, Japan.
| | - Masaaki Takahashi
- Department of Orthopedic Surgery, Jyuzen Memorial Hospital, Hamamatsu, Japan
| | - Yuuki Minamida
- Institute of Medical, Pharmaceutical & Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Naoko Yoshida
- Institute of Medical, Pharmaceutical & Health Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
12
|
Sánchez E, Betriu À, Yeramian A, Fernández E, Purroy F, Sánchez-de-la-Torre M, Pamplona R, Miquel E, Kerkeni M, Hernández C, Simó R, Lecube A, Hernández M, Rius F, Polanco D, Barbé F, Torres G, Suárez G, Portero-Otin M, Jové M, Colàs-Campàs L, Benabdelhak I, Farràs C, Ortega M, Manuel Valdivielso J, Bermúdez-López M, Martínez-Alonso M. Skin Autofluorescence Measurement in Subclinical Atheromatous Disease: Results from the ILERVAS Project. J Atheroscler Thromb 2019; 26:879-889. [PMID: 30842389 PMCID: PMC6800392 DOI: 10.5551/jat.47498] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Aim: Advanced glycation end-products (AGEs) have been involved in the atherogenic process in the high-risk population. The goal of this study was to demonstrate that AGEs are related to subclinical atheromatous disease in subjects with low to moderate vascular risk. Methods: A cross-sectional study in which 2,568 non-diabetic subjects of both sexes without cardiovascular disease were included. Subcutaneous content of AGEs was assessed by skin autofluorescence (SAF) and subclinical atheromatous disease was measured by assessing the atheromatous plaque burden in carotid and femoral regions using ultrasonography. In addition, serum pentosidine, carboxymethyl-lysine (CML) and AGE receptors (RAGE) were assessed in a nested case-control study with 41 subjects without plaque and 41 individuals subjects with generalized disease. Results: Patients with atheromatous plaque had a higher SAF than those with no plaque (1.9 [1.7 to 2.3] vs. 1.8 [1.6 to 2.1] arbitrary units (AU), p % 0.001). The SAF correlated with the total number of affected regions (r = 0.171, p < 0.001), increasing progressively from 1.8 [1.6 to 2.1] AU in those without atheromatous disease to 2.3 [1.9 to 2.7] AU in patients with ≥ 8 plaques (p < 0.001). A correlation was also observed between SAF and the total plaque area (r = 0.113, p < 0.001). The area under the Receiver Operating Characteristic curve was 0.65 (0.61 to 0.68) for identifying male subjects with atheromatous disease. The multivariable logistic regression model showed a significant and independent association between SAF and the presence of atheromatous disease. However, no significant differences in serum pentosidine, CML, and RAGE were observed. Conclusions: Increased subcutaneous content of AGEs is associated with augmented atheromatous plaque burden. Our results suggest that SAF may provide clinically relevant information to the current strategies for the evaluation of cardiovascular risk, especially among the male population.
Collapse
Affiliation(s)
- Enric Sánchez
- Endocrinology and Nutrition Department. University Hospital Arnau de Vilanova. Obesity, Diabetes and Metabolism (ODIM) research group. IRBLleida. University of Lleida
| | - Àngels Betriu
- Unit for the Detection and Treatment of Atherothrombotic Diseases (UDETMA V&R). University Hospital Arnau de Vilanova. Vascular and Renal Translational Research Group. IRBLleida. University of Lleida
| | - Andree Yeramian
- Endocrinology and Nutrition Department. University Hospital Arnau de Vilanova. Obesity, Diabetes and Metabolism (ODIM) research group. IRBLleida. University of Lleida
| | - Elvira Fernández
- Unit for the Detection and Treatment of Atherothrombotic Diseases (UDETMA V&R). University Hospital Arnau de Vilanova. Vascular and Renal Translational Research Group. IRBLleida. University of Lleida
| | - Francesc Purroy
- Stroke Unit. University Hospital Arnau de Vilanova. Clinical Neurosciences Group. IRBLleida. University of Lleida
| | - Manuel Sánchez-de-la-Torre
- Respiratory Department. University Hospital Arnau de Vilanova-Santa Maria. Translational Research in Respiratory Medicine. IRBLleida. University of Lleida.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII)
| | - Reinald Pamplona
- Department of Experimental Medicine. IRBLleida. University of Lleida
| | - Eva Miquel
- Borges Blanques Primary Health Care Unit
| | - Mohsen Kerkeni
- Laboratory of Biochemistry, LR12ES05, Faculty of Medicine, University of Monastir
| | - Cristina Hernández
- Endocrinology and Nutrition Department, University Hospital Vall d'Hebron. Diabetes and Metabolism Research Unit, Vall d'Hebron Institut de Recerca (VHIR). Universitat Autònoma de Barcelona.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII)
| | - Rafael Simó
- Endocrinology and Nutrition Department, University Hospital Vall d'Hebron. Diabetes and Metabolism Research Unit, Vall d'Hebron Institut de Recerca (VHIR). Universitat Autònoma de Barcelona.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII)
| | - Albert Lecube
- Endocrinology and Nutrition Department. University Hospital Arnau de Vilanova. Obesity, Diabetes and Metabolism (ODIM) research group. IRBLleida. University of Lleida.,Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII)
| | | | | | - Marta Hernández
- Endocrinology and Nutrition Department. University Hospital Arnau de Vilanova. Obesity, Diabetes and Metabolism (ODIM) research group. IRBLleida. University of Lleida
| | - Ferran Rius
- Endocrinology and Nutrition Department. University Hospital Arnau de Vilanova. Obesity, Diabetes and Metabolism (ODIM) research group. IRBLleida. University of Lleida
| | - Dinora Polanco
- Respiratory Department. University Hospital Arnau de Vilanova-Santa Maria. Translational Research in Respiratory Medicine. IRBLleida. University of Lleida
| | - Ferran Barbé
- Respiratory Department. University Hospital Arnau de Vilanova-Santa Maria. Translational Research in Respiratory Medicine. IRBLleida. University of Lleida.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII)
| | - Gerard Torres
- Respiratory Department. University Hospital Arnau de Vilanova-Santa Maria. Translational Research in Respiratory Medicine. IRBLleida. University of Lleida.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII)
| | - Guillermo Suárez
- Respiratory Department. University Hospital Arnau de Vilanova-Santa Maria. Translational Research in Respiratory Medicine. IRBLleida. University of Lleida.,Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII)
| | | | - Mariona Jové
- Department of Experimental Medicine. IRBLleida. University of Lleida
| | - Laura Colàs-Campàs
- Stroke Unit. University Hospital Arnau de Vilanova. Clinical Neurosciences Group. IRBLleida. University of Lleida
| | - Ikram Benabdelhak
- Stroke Unit. University Hospital Arnau de Vilanova. Clinical Neurosciences Group. IRBLleida. University of Lleida
| | | | | | - José Manuel Valdivielso
- Unit for the Detection and Treatment of Atherothrombotic Diseases (UDETMA V&R). University Hospital Arnau de Vilanova. Vascular and Renal Translational Research Group. IRBLleida. University of Lleida
| | - Marcelino Bermúdez-López
- Unit for the Detection and Treatment of Atherothrombotic Diseases (UDETMA V&R). University Hospital Arnau de Vilanova. Vascular and Renal Translational Research Group. IRBLleida. University of Lleida
| | - Montse Martínez-Alonso
- Unit for the Detection and Treatment of Atherothrombotic Diseases (UDETMA V&R). University Hospital Arnau de Vilanova. Vascular and Renal Translational Research Group. IRBLleida. University of Lleida
| |
Collapse
|
13
|
Banarjee R, Sharma A, Bai S, Deshmukh A, Kulkarni M. Proteomic study of endothelial dysfunction induced by AGEs and its possible role in diabetic cardiovascular complications. J Proteomics 2018; 187:69-79. [DOI: 10.1016/j.jprot.2018.06.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 06/12/2018] [Accepted: 06/15/2018] [Indexed: 12/30/2022]
|
14
|
Dhananjayan K, Gunawardena D, Hearn N, Sonntag T, Moran C, Gyengesi E, Srikanth V, Münch G. Activation of Macrophages and Microglia by Interferon-γ and Lipopolysaccharide Increases Methylglyoxal Production: A New Mechanism in the Development of Vascular Complications and Cognitive Decline in Type 2 Diabetes Mellitus? J Alzheimers Dis 2018; 59:467-479. [PMID: 28582854 DOI: 10.3233/jad-161152] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Methylglyoxal (MGO), a dicarbonyl compound derived from glucose, is elevated in diabetes mellitus and contributes to vascular complications by crosslinking collagen and increasing arterial stiffness. It is known that MGO contributes to inflammation as it forms advanced glycation end products (AGEs), which activate macrophages via the receptor RAGE. The aim of study was to investigate whether inflammatory activation can increase MGO levels, thereby completing a vicious cycle. In order to validate this, macrophage (RAW264.7, J774A.1) and microglial (N11) cells were stimulated with IFN-γ and LPS (5 + 5 and 10 + 10 IFN-γ U/ml or μg/ml LPS), and extracellular MGO concentration was determined after derivatization with 5,6-Diamino-2,4-dihydroxypyrimidine sulfate by HPLC. MGO levels in activated macrophage cells (RAW264.7) peaked at 48 h, increasing 2.86-fold (3.14±0.4 μM) at 5 U/ml IFN-γ+5 μg/ml LPS, and 4.74-fold (5.46±0.30 μM) at 10 U/ml IFN-γ+10 μg/ml LPS compared to the non-activated controls (1.15±0.02 μM). The other two cell lines, J774A.1 macrophages and N11 microglia, showed a similar response. We suggest that inflammation increases MGO production, possibly exacerbating arterial stiffness, cardiovascular complications, and diabetes-related cognitive decline.
Collapse
Affiliation(s)
- Karthik Dhananjayan
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Dhanushka Gunawardena
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Nerissa Hearn
- Molecular Medicine Research Group, Western Sydney University, Penrith, NSW, Australia
| | - Tanja Sonntag
- Molecular Medicine Research Group, Western Sydney University, Penrith, NSW, Australia
| | - Chris Moran
- Department of Medicine, Peninsula Health & Peninsula Clinical School, Monash University, VIC, Australia
| | - Erika Gyengesi
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia
| | - Velandai Srikanth
- Department of Medicine, Peninsula Health & Peninsula Clinical School, Monash University, VIC, Australia
| | - Gerald Münch
- Department of Pharmacology, School of Medicine, Western Sydney University, Penrith, NSW, Australia.,Molecular Medicine Research Group, Western Sydney University, Penrith, NSW, Australia.,National Institute of Complementary Medicine, Western Sydney University, Penrith, NSW, Australia
| |
Collapse
|
15
|
Alizadeh M, Kheirouri S. Curcumin against advanced glycation end products (AGEs) and AGEs-induced detrimental agents. Crit Rev Food Sci Nutr 2017; 59:1169-1177. [DOI: 10.1080/10408398.2017.1396200] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Mohammad Alizadeh
- Associate Professor, Department of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sorayya Kheirouri
- Associate Professor, Department of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
16
|
Liu CY, Zhao ZH, Chen ZT, Che CH, Zou ZY, Wu XM, Chen SG, Li YX, Lin HB, Wei XF, You J, Huang HP. DL-3-n-butylphthalide protects endothelial cells against advanced glycation end product-induced injury by attenuating oxidative stress and inflammation responses. Exp Ther Med 2017; 14:2241-2248. [PMID: 28962149 DOI: 10.3892/etm.2017.4784] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 02/14/2017] [Indexed: 12/21/2022] Open
Abstract
Endothelial dysfunction, regarded as a key step in the pathophysiological course of diabetic vascular complications, is initiated and deteriorated by advanced glycation end products (AGEs). DL-3-n-butylphthalide (DL-NBP) has been proven to have protective effects on neurons and vascular endothelial cells against ischemic and anoxic damage. The aim of the present study was to investigate whether NBP is able to attenuate AGE-induced endothelial dysfunction in vitro, and also elucidate the possible underlying mechanism. An injury model of human umbilical vein endothelial cells (HUVECs) induced by AGEs (200 µg/ml) was established. The results demonstrated that pretreatment with NBP (1-100 µM) significantly increased HUVEC viability and inhibited the apoptosis induced by AGEs. In addition, AGEs stimulated the expression levels of the receptor for AGEs protein and the downstream protein nuclear factor-κB in HUVECs, which were inhibited by pretreatment with NBP. Furthermore, it significantly reduced reactive oxygen species generation and the level of the inflammatory cytokines, intercellular cell adhesion molecule-1 and monocyte chemotactic protein-1, in HUVECs mediated by AGEs. The current findings indicated that NBP attenuated AGE-induced endothelial dysfunction by ameliorating inflammation and oxidative stress responses.
Collapse
Affiliation(s)
- Chang-Yun Liu
- Department of Neurology, Union Hospital, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Zhen-Hua Zhao
- Department of Neurology, Fujian Provincial Hospital, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Zhi-Ting Chen
- Department of Neurology, Union Hospital, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Chun-Hui Che
- Department of Neurology, Union Hospital, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Zhang-Yu Zou
- Department of Neurology, Union Hospital, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Xiao-Min Wu
- Department of Neurology, Union Hospital, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Sheng-Gen Chen
- Department of Neurology, Union Hospital, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Yuan-Xiao Li
- Department of Neurology, Union Hospital, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Han-Bin Lin
- Department of Neurology, Union Hospital, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Xiao-Fan Wei
- Department of Neurology, Union Hospital, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Jie You
- Department of Endocrinology, Union Hospital, Fujian Medical University, Fujian Endocrinology Institute, Fuzhou, Fujian 350001, P.R. China
| | - Hua-Pin Huang
- Department of Neurology, Union Hospital, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
17
|
Son WR, Nam MH, Hong CO, Kim Y, Lee KW. Plantamajoside from Plantago asiatica modulates human umbilical vein endothelial cell dysfunction by glyceraldehyde-induced AGEs via MAPK/NF-κB. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:66. [PMID: 28109289 PMCID: PMC5251346 DOI: 10.1186/s12906-017-1570-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 01/07/2017] [Indexed: 01/25/2023]
Abstract
Background Plantago asiatica has been traditionally used for traditional medicine around East Asia. Plantamajoside (PM), which is isolated from this plant, is known for biological properties including anti-inflammation and antioxidant activity. To demonstrate the biological activity of PM against endothelial dysfunction induced by advanced glycation end-products (AGEs), a cellular inflammatory mechanism system was evaluated in human umbilical vein endothelial cells (HUVECs). Methods We obtained PM through previous research in our laboratory. We formed the AGEs from bovine serum albumin with glyceraldehyde in the dark for seven days. To confirm the modulation of the inflammatory mechanism in endothelial dysfunction, we quantified the various pro-inflammatory cytokines and endothelial dysfunction-related proteins in the HUVECs with Western blotting and with real-time and quantitative real-time polymerase chain reactions. Results Co-treatment with PM and AGEs significantly suppressed inflammatory cytokines and adhesion molecule expression. Moreover, the PM treatment for down-regulated inflammatory signals and blocked monocyte adhesion on the HUVECs. Conclusions Theses results demonstrated that PM, as a potential natural compound, protects AGE-induced endothelial cells against inflammatory cellular dysfunction. Electronic supplementary material The online version of this article (doi:10.1186/s12906-017-1570-1) contains supplementary material, which is available to authorized users.
Collapse
|
18
|
Kakde S, Bhopal RS, Bhardwaj S, Misra A. Urbanized South Asians' susceptibility to coronary heart disease: The high-heat food preparation hypothesis. Nutrition 2016; 33:216-224. [PMID: 27776951 DOI: 10.1016/j.nut.2016.07.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 06/17/2016] [Accepted: 07/08/2016] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Known risk factors do not fully explain the comparatively high susceptibility to coronary heart disease (CHD) in South Asians (Indian, Pakistani, Bangladeshi, and Sri Lankan populations in South Asia and overseas). The search for explanatory hypotheses and cofactors that raise susceptibility of South Asians to CHD continues. The aim of this study was to propose "the high-heat food preparation hypothesis," where neo-formed contaminants (NFCs) such as trans-fatty acids (TFAs) and advanced glycation end-products (AGEs) are the cofactors. METHODS We reviewed the actions of AGEs and TFAs, the burden of these products in tissues and blood in South Asians, the relationship between these products and CHD, the effects of preparing food and reheating oils at high temperatures on NFCs, and the foods and mode of preparation in South Asian and Chinese cuisines. RESULTS Animal and human studies show NFCs increase the risk for CHD. Evidence on the consumption and body burden of these products across ethnic groups is not available, and comparable data on the NFC content of the cuisine of South Asians and potential comparison populations (e.g., the Chinese with lower CHD rates) are limited. South Asians' cuisine is dominated by frying and roasting techniques that use high temperatures. South Asian foods have high TFA content primarily through the use of partially hydrogenated fats, reheated oils, and high-heat cooking. Reheating oils greatly increases the TFA content. In comparison, Chinese cuisine involves mostly braising, steaming, and boiling rather than frying. CONCLUSION We hypothesize that South Asians' susceptibility to CHD is partly attributable to high-heat treated foods producing high NFCs. Research to accrue direct evidence is proposed.
Collapse
Affiliation(s)
- Smitha Kakde
- Edinburgh Migration, Ethnicity and Health Research Group, Centre for Population Health Sciences, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Raj S Bhopal
- Edinburgh Migration, Ethnicity and Health Research Group, Centre for Population Health Sciences, Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK.
| | - Swati Bhardwaj
- National Diabetes, Obesity and Cholesterol Diseases Foundation, SDA, New Delhi, India; Diabetes Foundation (India), SDA, New Delhi, India; Center of Nutrition & Metabolic Research (C-NET), Delhi, India
| | - Anoop Misra
- National Diabetes, Obesity and Cholesterol Diseases Foundation, SDA, New Delhi, India; Diabetes Foundation (India), SDA, New Delhi, India; Center of Nutrition & Metabolic Research (C-NET), Delhi, India; Fortis C-DOC Center for Excellence for Diabetes, Metabolic Diseases and Endocrinology, New Delhi, India
| |
Collapse
|
19
|
McGarrity S, Halldórsson H, Palsson S, Johansson PI, Rolfsson Ó. Understanding the Causes and Implications of Endothelial Metabolic Variation in Cardiovascular Disease through Genome-Scale Metabolic Modeling. Front Cardiovasc Med 2016; 3:10. [PMID: 27148541 PMCID: PMC4834436 DOI: 10.3389/fcvm.2016.00010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 04/03/2016] [Indexed: 01/04/2023] Open
Abstract
High-throughput biochemical profiling has led to a requirement for advanced data interpretation techniques capable of integrating the analysis of gene, protein, and metabolic profiles to shed light on genotype-phenotype relationships. Herein, we consider the current state of knowledge of endothelial cell (EC) metabolism and its connections to cardiovascular disease (CVD) and explore the use of genome-scale metabolic models (GEMs) for integrating metabolic and genomic data. GEMs combine gene expression and metabolic data acting as frameworks for their analysis and, ultimately, afford mechanistic understanding of how genetic variation impacts metabolism. We demonstrate how GEMs can be used to investigate CVD-related genetic variation, drug resistance mechanisms, and novel metabolic pathways in ECs. The application of GEMs in personalized medicine is also highlighted. Particularly, we focus on the potential of GEMs to identify metabolic biomarkers of endothelial dysfunction and to discover methods of stratifying treatments for CVDs based on individual genetic markers. Recent advances in systems biology methodology, and how these methodologies can be applied to understand EC metabolism in both health and disease, are thus highlighted.
Collapse
Affiliation(s)
- Sarah McGarrity
- Center for Systems Biology, University of Iceland , Reykjavik , Iceland
| | - Haraldur Halldórsson
- Department of Pharmacology and Toxicology, School of Health Sciences, University of Iceland , Reykjavik , Iceland
| | - Sirus Palsson
- Center for Systems Biology, University of Iceland, Reykjavik, Iceland; Sinopia Biosciences Inc., San Diego, CA, USA
| | - Pär I Johansson
- Section for Transfusion Medicine, Capital Region Blood Bank, Rigshospitalet, University of Copenhagen , Copenhagen , Denmark
| | - Óttar Rolfsson
- Center for Systems Biology, University of Iceland, Reykjavik, Iceland; Department of Biochemistry and Molecular Biology, School of Health Sciences, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
20
|
Paradela-Dobarro B, Rodiño-Janeiro BK, Alonso J, Raposeiras-Roubín S, González-Peteiro M, González-Juanatey JR, Álvarez E. Key structural and functional differences between early and advanced glycation products. J Mol Endocrinol 2016; 56:23-37. [PMID: 26581238 DOI: 10.1530/jme-15-0031] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Most of the studies on advanced glycation end products (AGE) have been carried out with uncharacterized mixtures of AGE, so the observed effects cannot be linked to defined structures. Therefore, we analysed the structural differences between glycated human serum albumin (gHSA), a low glycated protein, and AGE-human serum albumin (AGE-HSA), a high glycated protein, and we compared their effects on endothelial functionality. Specifically, we characterized glycation and composition on both early and advanced stage glycation products of gHSA and AGE-HSA by using the MALDI-TOF-mass spectrometry assay. Furthermore, we studied the effects of both types of glycation products on reactive oxygen species (ROS) production and in the expression of vascular and intercellular cell adhesion molecules (VCAM-1 and ICAM-1) on human umbilical endothelial cells (HUVEC). We also measured the adhesion of peripheral blood mononuclear cells (PBMC) to HUVEC. Low concentrations of gHSA enhanced long-lasting ROS production in HUVEC, whereas lower concentrations of AGE-HSA caused the anticipation of the induced extracellular ROS production. Both gHSA and AGE-HSA up-regulated the expression of VCAM-1 and ICAM-1 at mRNA levels. Nevertheless, only AGE-HSA increased protein levels and enhanced the adhesion of PBMC to HUVEC monolayers. Functional differences were observed between gHSA and AGE-HSA, causing the latter an anticipation of the pro-oxidant effects in comparison to gHSA. Moreover, although both molecules induced genetic up-regulation of adhesion molecules in HUVEC, only the high glycated protein functionally increased mononuclear cell adhesion to endothelial monolayers. These observations could have important clinical consequences in the development of diabetic vascular complications.
Collapse
Affiliation(s)
- Beatriz Paradela-Dobarro
- Laboratorio no 6. Grupo de CardiologíaInstituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, SpainServicio de proteómica. Instituto de Investigación Sanitaria de Santiago (IDIS)Santiago de Compostela, SpainServicio de CardiologíaComplejo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, SpainDepartamento de EnfermeríaUniversidad de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Bruno K Rodiño-Janeiro
- Laboratorio no 6. Grupo de CardiologíaInstituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, SpainServicio de proteómica. Instituto de Investigación Sanitaria de Santiago (IDIS)Santiago de Compostela, SpainServicio de CardiologíaComplejo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, SpainDepartamento de EnfermeríaUniversidad de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Jana Alonso
- Laboratorio no 6. Grupo de CardiologíaInstituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, SpainServicio de proteómica. Instituto de Investigación Sanitaria de Santiago (IDIS)Santiago de Compostela, SpainServicio de CardiologíaComplejo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, SpainDepartamento de EnfermeríaUniversidad de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Sergio Raposeiras-Roubín
- Laboratorio no 6. Grupo de CardiologíaInstituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, SpainServicio de proteómica. Instituto de Investigación Sanitaria de Santiago (IDIS)Santiago de Compostela, SpainServicio de CardiologíaComplejo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, SpainDepartamento de EnfermeríaUniversidad de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Mercedes González-Peteiro
- Laboratorio no 6. Grupo de CardiologíaInstituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, SpainServicio de proteómica. Instituto de Investigación Sanitaria de Santiago (IDIS)Santiago de Compostela, SpainServicio de CardiologíaComplejo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, SpainDepartamento de EnfermeríaUniversidad de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - José R González-Juanatey
- Laboratorio no 6. Grupo de CardiologíaInstituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, SpainServicio de proteómica. Instituto de Investigación Sanitaria de Santiago (IDIS)Santiago de Compostela, SpainServicio de CardiologíaComplejo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, SpainDepartamento de EnfermeríaUniversidad de Santiago de Compostela, Santiago de Compostela 15782, Spain Laboratorio no 6. Grupo de CardiologíaInstituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, SpainServicio de proteómica. Instituto de Investigación Sanitaria de Santiago (IDIS)Santiago de Compostela, SpainServicio de CardiologíaComplejo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, SpainDepartamento de EnfermeríaUniversidad de Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Ezequiel Álvarez
- Laboratorio no 6. Grupo de CardiologíaInstituto de Investigación Sanitaria de Santiago (IDIS), Santiago de Compostela, SpainServicio de proteómica. Instituto de Investigación Sanitaria de Santiago (IDIS)Santiago de Compostela, SpainServicio de CardiologíaComplejo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, SpainDepartamento de EnfermeríaUniversidad de Santiago de Compostela, Santiago de Compostela 15782, Spain
| |
Collapse
|
21
|
Ohkawara H, Ikeda K, Ogawa K, Takeishi Y. MEMBRANE TYPE 1-MATRIX METALLOPROTEINASE (MT1-MMP) IDENTIFIED AS A MULTIFUNCTIONAL REGULATOR OF VASCULAR RESPONSES. Fukushima J Med Sci 2015; 61:91-100. [PMID: 26370683 DOI: 10.5387/fms.2015-15] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Membrane type 1-matrix metalloproteinase (MT1-MMP) functions as a signaling molecules in addition to a transmembrane metalloprotease, which degrades interstitial collagens and extracellular matrix components. This review focuses on the multifunctional roles of MT1-MMP as a signaling molecule in vascular responses to pro-atherosclerotic stimuli in the pathogenesis of cardiovascular diseases. First, the lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1)-MT1-MMP signaling axis contributes to endothelial dysfunction, which is mediated via small GTP-binding protein RhoA and Rac1 activation. Second, MT1-MMP plays a crucial role in reactive oxygen species (ROS) generation through the activation of receptor for advanced glycation end products (AGEs) in smooth muscle cells, indicating that MT1-MMP may be a therapeutic target for diabetic vascular complications. Third, MT1-MMP is involved in RhoA/Rac1 activation and Ca(2+) signaling in the mechanism of thrombin-stimulated endothelial dysfunction and oxidant stress. Fourth, the inhibition of the MT1-MMP/Akt signaling pathway may be an attractive strategy for treating endothelial disordered hemostasis in the development of vascular diseases linked to TNF-α-induced inflammation. Fifth, MT1-MMP through RAGE induced RhoA/Rac1 activation and tissue factor protein upregulation through NF-κB phosphorylation in endothelial cells stimulated by high-mobility group box-1, which plays a key role in the systemic inflammation. These findings suggest that the MT1-MMP-mediated signaling axis may be a promising target for treating atherosclerosis and subsequent cardiovascular diseases.
Collapse
Affiliation(s)
- Hiroshi Ohkawara
- Department of Cardiology and Hematology, Fukushima Medical University
| | | | | | | |
Collapse
|
22
|
Goveia J, Stapor P, Carmeliet P. Principles of targeting endothelial cell metabolism to treat angiogenesis and endothelial cell dysfunction in disease. EMBO Mol Med 2015; 6:1105-20. [PMID: 25063693 PMCID: PMC4197858 DOI: 10.15252/emmm.201404156] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The endothelium is the orchestral conductor of blood vessel function. Pathological blood vessel formation (a process termed pathological angiogenesis) or the inability of endothelial cells (ECs) to perform their physiological function (a condition known as EC dysfunction) are defining features of various diseases. Therapeutic intervention to inhibit aberrant angiogenesis or ameliorate EC dysfunction could be beneficial in diseases such as cancer and cardiovascular disease, respectively, but current strategies have limited efficacy. Based on recent findings that pathological angiogenesis and EC dysfunction are accompanied by EC-specific metabolic alterations, targeting EC metabolism is emerging as a novel therapeutic strategy. Here, we review recent progress in our understanding of how EC metabolism is altered in disease and discuss potential metabolic targets and strategies to reverse EC dysfunction and inhibit pathological angiogenesis.
Collapse
Affiliation(s)
- Jermaine Goveia
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven, Belgium Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center VIB, Leuven, Belgium
| | - Peter Stapor
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven, Belgium Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center VIB, Leuven, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center, Department of Oncology, University of Leuven, Leuven, Belgium Laboratory of Angiogenesis and Neurovascular Link, Vesalius Research Center VIB, Leuven, Belgium
| |
Collapse
|
23
|
Nowotny K, Jung T, Höhn A, Weber D, Grune T. Advanced glycation end products and oxidative stress in type 2 diabetes mellitus. Biomolecules 2015; 5:194-222. [PMID: 25786107 PMCID: PMC4384119 DOI: 10.3390/biom5010194] [Citation(s) in RCA: 737] [Impact Index Per Article: 73.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 02/06/2015] [Accepted: 03/02/2015] [Indexed: 12/25/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a very complex and multifactorial metabolic disease characterized by insulin resistance and β cell failure leading to elevated blood glucose levels. Hyperglycemia is suggested to be the main cause of diabetic complications, which not only decrease life quality and expectancy, but are also becoming a problem regarding the financial burden for health care systems. Therefore, and to counteract the continually increasing prevalence of diabetes, understanding the pathogenesis, the main risk factors, and the underlying molecular mechanisms may establish a basis for prevention and therapy. In this regard, research was performed revealing further evidence that oxidative stress has an important role in hyperglycemia-induced tissue injury as well as in early events relevant for the development of T2DM. The formation of advanced glycation end products (AGEs), a group of modified proteins and/or lipids with damaging potential, is one contributing factor. On the one hand it has been reported that AGEs increase reactive oxygen species formation and impair antioxidant systems, on the other hand the formation of some AGEs is induced per se under oxidative conditions. Thus, AGEs contribute at least partly to chronic stress conditions in diabetes. As AGEs are not only formed endogenously, but also derive from exogenous sources, i.e., food, they have been assumed as risk factors for T2DM. However, the role of AGEs in the pathogenesis of T2DM and diabetic complications—if they are causal or simply an effect—is only partly understood. This review will highlight the involvement of AGEs in the development and progression of T2DM and their role in diabetic complications.
Collapse
Affiliation(s)
- Kerstin Nowotny
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany.
| | - Tobias Jung
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany.
| | - Annika Höhn
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany.
| | - Daniela Weber
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany.
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany.
| |
Collapse
|
24
|
Muthenna P, Raghu G, Kumar PA, Surekha MV, Reddy GB. Effect of cinnamon and its procyanidin-B2 enriched fraction on diabetic nephropathy in rats. Chem Biol Interact 2014; 222:68-76. [PMID: 25199697 DOI: 10.1016/j.cbi.2014.08.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 08/23/2014] [Accepted: 08/27/2014] [Indexed: 01/17/2023]
Abstract
Non-enzymatic protein glycation and resultant accumulation of advanced glycation endproducts (AGE) are implicated in the pathogenesis of diabetic complications including diabetic nephropathy (DN). It is considered that antiglycating agents offer protection against AGE mediated pathologies including DN. Earlier we characterized procyanidin-B2 (PCB2) as the active component from cinnamon (Cinnamomum zeylanicum) that inhibits AGE formation in vitro. In this study, we have investigated the potential of PCB2-enriched fraction of cinnamon to prevent in vivo accumulation of AGE and to ameliorate renal changes in diabetic rats. Streptozotocin-induced diabetic rats were fed with either 3% cinnamon or 0.002% PCB2-fraction in diet for 12weeks. Biochemical analysis of blood and urine was performed at the end of experiment. Evaluation of glomerular markers that serve as indicators of renal function was done by immunohistochemistry, immunoblotting and qRT-PCR. Supplementation of diabetic rats with cinnamon and PCB2-fraction prevented glycation mediated RBC-IgG cross-links and HbA1c accumulation in diabetes rats. Cinnamon and PCB2-fraction also inhibited the accumulation of N-carboxy methyl lysine (CML), a prominent AGE in diabetic kidney. Interestingly, cinnamon and its PCB2-fraction prevented the AGE mediated loss of expression of glomerular podocyte proteins; nephrin and podocin. Inhibition of AGE by cinnamon and PCB2-fraction ameliorated the diabetes mediated renal malfunction in rats as evidenced by reduced urinary albumin and creatinine. In conclusion, PCB2 from cinnamon inhibited AGE accumulation in diabetic rat kidney and ameliorated AGE mediated pathogenesis of DN.
Collapse
Affiliation(s)
- P Muthenna
- National Institute of Nutrition, Hyderabad, India
| | - G Raghu
- National Institute of Nutrition, Hyderabad, India
| | - P Anil Kumar
- National Institute of Nutrition, Hyderabad, India
| | - M V Surekha
- National Institute of Nutrition, Hyderabad, India
| | | |
Collapse
|
25
|
Shikata K, Makino H. Microinflammation in the pathogenesis of diabetic nephropathy. J Diabetes Investig 2014; 4:142-9. [PMID: 24843643 PMCID: PMC4019266 DOI: 10.1111/jdi.12050] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 12/10/2012] [Accepted: 12/10/2012] [Indexed: 12/19/2022] Open
Abstract
Diabetic nephropathy is the leading cause of end‐stage renal failure in developed countries. Furthermore, diabetic nephropathy is related to the risk of cardiovascular diseases and an increase in mortality of diabetic patients. Several factors are involved in the development of nephropathy, including glomerular hyperfiltration, oxidative stress, accumulation of advanced glycation end‐products, activation of protein kinase C, acceleration of the polyol pathway and over‐expression of transforming growth factor‐β. Recently, accumulated data have emphasized the critical roles of chronic low‐grade inflammation, ‘microinflammation’, in the pathogenesis of diabetic nephropathy, suggesting that microinflammation is a common mechanism in the development of diabetic vascular complications. Expression of cell adhesion molecules, chemokines and pro‐inflammatory cytokines are increased in the renal tissues of diabetic patients and animals. Deficiency of pro‐inflammatory molecules results in amelioration of renal injuries after induction of diabetes in mice. Plasma and urinary levels of cytokines, chemokines and cell adhesion molecules, are elevated and correlated with albuminuria. Several kinds of drugs that have anti‐inflammatory actions as their pleiotropic effects showed renoprotective effects on diabetic animals. Modulation of the inflammatory process prevents renal insufficiency in diabetic animal models, suggesting that microinflammation is one of the promising therapeutic targets for diabetic nephropathy, as well as for cardiovascular diseases.
Collapse
Affiliation(s)
- Kenichi Shikata
- Center for Innovative Clinical Medicine Okayama University Hospital Okayama Japan ; Department of Medicine and Clinical Science Okayama University Graduate School of Medicine Dentistry, and Pharmaceutical Science Okayama Japan
| | - Hirofumi Makino
- Center for Innovative Clinical Medicine Okayama University Hospital Okayama Japan ; Department of Medicine and Clinical Science Okayama University Graduate School of Medicine Dentistry, and Pharmaceutical Science Okayama Japan
| |
Collapse
|
26
|
Nishizawa Y, Wada RI, Baba M, Takeuchi M, Hanyu-Itabashi C, Yagihashi S. Neuropathy induced by exogenously administered advanced glycation end-products in rats. J Diabetes Investig 2014; 1:40-9. [PMID: 24843407 PMCID: PMC4020676 DOI: 10.1111/j.2040-1124.2009.00002.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Aims/Introduction: Advanced glycation end‐products (AGE) have been implicated in the development of diabetic neuropathy. It still remains unknown, however, how AGE cause functional and structural changes of the peripheral nerve in diabetes. To explore the role of AGE in diabetic neuropathy, we examined the peripheral nerve by injecting AGE into normal Wistar rats. Materials and Methods: Young, normal male Wistar rats were injected intraperitoneally (i.p.) daily for 12 weeks with purified AGE prepared by incubating D‐glucose with bovine serum albumin (BSA). A control group received BSA alone. A group of rats given AGE were co‐treated with aminoguanidine (50 mg/kg/day, i.p.). Peripheral nerve function and structure, as well as nerve Na+,K+‐ATPase activity, were examined in these rats. Immunohistochemical expressions of 8‐hydroxy‐2′‐deoxyguanosine (8OHdG) and nuclear factor‐κB (NF‐κB)p65 were also examined. Results: Serum AGE levels were increased two to threefold in the AGE‐treated group compared with those in the BSA‐treated control group. AGE‐treated rats showed a marked slowing of motor nerve conduction velocity (MNCV) and decreased nerve Na+,K+‐ATPase activity compared with those in the BSA‐treated group. These changes were accompanied by intensified expressions of 8OHdG and NF‐κBp65 in endothelial cells and Schwann cells. Aminoguanidine treatment corrected MNCV delay, Na+,K+‐ATPase activity, and suppressed the expression of 8OHdG and NF‐κB, despite there being no influence on serum AGE levels. Conclusions: The results suggest that an elevated concentration of blood AGE might be one of the contributing factors to the development of neuropathic changes in diabetes.
Collapse
Affiliation(s)
- Yusuke Nishizawa
- Department of Pathology and Molecular Medicine, Graduate School of Medicine, Hirosaki University, Hirosaki
| | - Ryu-Ichi Wada
- Department of Pathology and Molecular Medicine, Graduate School of Medicine, Hirosaki University, Hirosaki
| | - Masayuki Baba
- Division of Neurology, Aomori Prefectural Hospital, Aomori
| | - Masayoshi Takeuchi
- Department of Pathophysiological Science, Faculty of Pharmaceutical Sciences, Hokuriku University, Kanazawa, Japan
| | - Chieko Hanyu-Itabashi
- Department of Pathology and Molecular Medicine, Graduate School of Medicine, Hirosaki University, Hirosaki
| | - Soroku Yagihashi
- Department of Pathology and Molecular Medicine, Graduate School of Medicine, Hirosaki University, Hirosaki
| |
Collapse
|
27
|
Kiefer AS, Fleming T, Eckert GJ, Poindexter BB, Nawroth PP, Yoder MC. Methylglyoxal concentrations differ in standard and washed neonatal packed red blood cells. Pediatr Res 2014; 75:409-14. [PMID: 24346110 DOI: 10.1038/pr.2013.243] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 09/05/2013] [Indexed: 12/23/2022]
Abstract
BACKGROUND Preterm infants have a greater risk of necrotizing enterocolitis following transfusion. It is hypothesized that high glucose concentrations in red blood cell (RBC) preservatives lead to increased methylglyoxal (MG) metabolism, causing glycation-driven damage to transfused RBCs. Such changes to the RBCs could promote a proinflammatory state in transfusion recipients. METHODS Standard and washed RBCs in Adsol-3, two common neonatal preparations, were studied. Consecutive measurements were performed of glucose, MG, reduced glutathione, glyoxalase I activity (GLO-I), and D-lactate, the stable end product of MG detoxification by glyoxalase enzymes over the 42-d storage period. RESULTS RBC units consume glucose and produceD-lactate and MG during storage. In 28/30 units, the MG concentrations showed only small variations during storage. Two units had elevated MG levels (>10 pmol/mg Hb) during the first half of storage. Washing of the RBCs significantly reduced both MG and D-lactate. CONCLUSION This study shows two patterns of MG metabolism in packed RBCs for neonatal transfusion and raises the possibility that RBC units with higher MG levels may have increased glycation-driven damage in the transfused RBCs. Whether transfused MG could trigger an inflammatory response such as necrotizing enterocolitis in preterm neonates and whether washing could prevent this require further study.
Collapse
Affiliation(s)
- Autumn S Kiefer
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Thomas Fleming
- Department of Internal Medicine and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
| | - George J Eckert
- Department of Biostatistics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Brenda B Poindexter
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| | - Peter P Nawroth
- Department of Internal Medicine and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
| | - Mervin C Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
28
|
Becker RC. Preoteomics, metabolomics and circulating endothelial progenitor cells in acute coronary syndromes. J Thromb Thrombolysis 2014; 21:203-6. [PMID: 16622619 DOI: 10.1007/s11239-006-5969-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
29
|
Brouwers O, Niessen PMG, Miyata T, Østergaard JA, Flyvbjerg A, Peutz-Kootstra CJ, Sieber J, Mundel PH, Brownlee M, Janssen BJA, De Mey JGR, Stehouwer CDA, Schalkwijk CG. Glyoxalase-1 overexpression reduces endothelial dysfunction and attenuates early renal impairment in a rat model of diabetes. Diabetologia 2014; 57:224-35. [PMID: 24162587 DOI: 10.1007/s00125-013-3088-5] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 10/02/2013] [Indexed: 02/02/2023]
Abstract
AIMS/HYPOTHESIS In diabetes, advanced glycation end-products (AGEs) and the AGE precursor methylglyoxal (MGO) are associated with endothelial dysfunction and the development of microvascular complications. In this study we used a rat model of diabetes, in which rats transgenically overexpressed the MGO-detoxifying enzyme glyoxalase-I (GLO-I), to determine the impact of intracellular glycation on vascular function and the development of early renal changes in diabetes. METHODS Wild-type and Glo1-overexpressing rats were rendered diabetic for a period of 24 weeks by intravenous injection of streptozotocin. Mesenteric arteries were isolated to study ex vivo vascular reactivity with a wire myograph and kidneys were processed for histological examination. Glycation was determined by mass spectrometry and immunohistochemistry. Markers for inflammation, endothelium dysfunction and renal dysfunction were measured with ELISA-based techniques. RESULTS Diabetes-induced formation of AGEs in mesenteric arteries and endothelial dysfunction were reduced by Glo1 overexpression. Despite the absence of advanced nephrotic lesions, early markers of renal dysfunction (i.e. increased glomerular volume, decreased podocyte number and diabetes-induced elevation of urinary markers albumin, osteopontin, kidney-inflammation-molecule-1 and nephrin) were attenuated by Glo1 overexpression. In line with this, downregulation of Glo1 in cultured endothelial cells resulted in increased expression of inflammation and endothelium dysfunction markers. In fully differentiated cultured podocytes incubation with MGO resulted in apoptosis. CONCLUSIONS/INTERPRETATION This study shows that effective regulation of the GLO-I enzyme is important in the prevention of vascular intracellular glycation, endothelial dysfunction and early renal impairment in experimental diabetes. Modulating the GLO-I pathway therefore may provide a novel approach to prevent vascular complications in diabetes.
Collapse
Affiliation(s)
- Olaf Brouwers
- Laboratory for Metabolism and Vascular Medicine, Division of General Internal Medicine, Department of Internal Medicine, Maastricht University Medical Center, Universiteitssingel 50, P.O. Box 616 (box 14), 6200 MD, Maastricht, the Netherlands,
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Oxidative Stress and Cardiovascular Disease in Diabetes. OXIDATIVE STRESS IN APPLIED BASIC RESEARCH AND CLINICAL PRACTICE 2014. [DOI: 10.1007/978-1-4899-8035-9_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
31
|
Cabral-Miranda G, Yamashiro-Kanashiro EHG, Gidlund M, Sales MGF. Specific label-free and real-time detection of oxidized low density lipoprotein (oxLDL) using an immunosensor with three monoclonal antibodies. J Mater Chem B 2014; 2:477-484. [DOI: 10.1039/c3tb21048k] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
32
|
Protein glycation during aging and in cardiovascular disease. J Proteomics 2013; 92:248-59. [DOI: 10.1016/j.jprot.2013.05.012] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 05/08/2013] [Accepted: 05/12/2013] [Indexed: 01/11/2023]
|
33
|
Effect of dietary advanced glycation end products on postprandial appetite, inflammation, and endothelial activation in healthy overweight individuals. Eur J Nutr 2013; 53:661-72. [DOI: 10.1007/s00394-013-0574-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 08/01/2013] [Indexed: 01/02/2023]
|
34
|
van Eupen MGA, Schram MT, Colhoun HM, Hanssen NMJ, Niessen HWM, Tarnow L, Parving HH, Rossing P, Stehouwer CDA, Schalkwijk CG. The methylglyoxal-derived AGE tetrahydropyrimidine is increased in plasma of individuals with type 1 diabetes mellitus and in atherosclerotic lesions and is associated with sVCAM-1. Diabetologia 2013; 56:1845-55. [PMID: 23620061 DOI: 10.1007/s00125-013-2919-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Accepted: 04/11/2013] [Indexed: 12/31/2022]
Abstract
AIMS/HYPOTHESIS Methylglyoxal (MGO) is a major precursor for advanced glycation end-products (AGEs), which are thought to play a role in vascular complications in diabetes. Known MGO-arginine-derived AGEs are 5-hydro-5-methylimidazolone (MG-H1), argpyrimidine and tetrahydropyrimidine (THP). We studied THP in relation to type 1 diabetes, endothelial dysfunction, low-grade inflammation, vascular complications and atherosclerosis. METHODS We raised and characterised a monoclonal antibody against MGO-derived THP. We measured plasma THP with a competitive ELISA in two cohort studies: study A (198 individuals with type 1 diabetes and 197 controls); study B (individuals with type 1 diabetes, 175 with normoalbuminuria and 198 with macroalbuminuria [>300 mg/24 h]). We measured plasma markers of endothelial dysfunction and low-grade inflammation, and evaluated the presence of THP and N (ε)-(carboxymethyl)lysine (CML) in atherosclerotic arteries. RESULTS THP was higher in individuals with type 1 diabetes than in those without (median [interquartile range] 115.5 U/μl [102.4-133.2] and 109.8 U/μl [91.8-122.3], respectively; p = 0.03). THP was associated with plasma soluble vascular cell adhesion molecule 1 in both study A (standardised β = 0.48 [95% CI 0.38, 0.58]; p < 0.001) and study B (standardised β = 0.31 [95% CI 0.23, 0.40]; p < 0.001), and with secreted phospholipase A2 (standardised β = 0.26 [95% CI 0.17, 0.36]; p < 0.001) in study B. We found no association of THP with micro- or macro-vascular complications. Both THP and CML were detected in atherosclerotic arteries. CONCLUSIONS/INTERPRETATION Our results suggest that MGO-derived THP may reflect endothelial dysfunction among individuals with and without type 1 diabetes, and therefore may potentially play a role in the development of atherosclerosis and vascular disease.
Collapse
Affiliation(s)
- M G A van Eupen
- Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Advanced glycation end products and diabetic retinopathy. J Ocul Biol Dis Infor 2013; 5:63-9. [PMID: 24596941 DOI: 10.1007/s12177-013-9104-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Accepted: 04/03/2013] [Indexed: 01/08/2023] Open
Abstract
Studies have established hyperglycemia as the most important factor in the progress of vascular complications. Formation of advanced glycation end products (AGEs) correlates with glycemic control. The AGE hypothesis proposes that hyperglycemia contributes to the pathogenesis of diabetic complications including retinopathy. However, their role in diabetic retinopathy remains largely unknown. This review discusses the chemistry of AGEs formation and their patho-biochemistry particularly in relation to diabetic retinopathy. AGEs exert deleterious effects by acting directly to induce cross-linking of long-lived proteins to promote vascular stiffness, altering vascular structure and function and interacting with receptor for AGE, to induce intracellular signaling leading to enhanced oxidative stress and elaboration of key proinflammatory and prosclerotic cytokines. Novel anti-AGE strategies are being developed hoping that in next few years, some of these promising therapies will be successfully evaluated in clinical context aiming to reduce the major economical and medical burden caused by diabetic retinopathy.
Collapse
|
36
|
Wang JC, Bennett M. Aging and atherosclerosis: mechanisms, functional consequences, and potential therapeutics for cellular senescence. Circ Res 2012; 111:245-59. [PMID: 22773427 DOI: 10.1161/circresaha.111.261388] [Citation(s) in RCA: 652] [Impact Index Per Article: 50.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Atherosclerosis is classed as a disease of aging, such that increasing age is an independent risk factor for the development of atherosclerosis. Atherosclerosis is also associated with premature biological aging, as atherosclerotic plaques show evidence of cellular senescence characterized by reduced cell proliferation, irreversible growth arrest and apoptosis, elevated DNA damage, epigenetic modifications, and telomere shortening and dysfunction. Not only is cellular senescence associated with atherosclerosis, there is growing evidence that cellular senescence promotes atherosclerosis. This review examines the pathology of normal vascular aging, the evidence for cellular senescence in atherosclerosis, the mechanisms underlying cellular senescence including reactive oxygen species, replication exhaustion and DNA damage, the functional consequences of vascular cell senescence, and the possibility that preventing accelerated cellular senescence is a therapeutic target in atherosclerosis.
Collapse
Affiliation(s)
- Julie C Wang
- Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | | |
Collapse
|
37
|
Simm A, Navarrete-Santos A, Hofmann B, Bushnaq H, Nass N. [Protein glycation as a pathological mechanism in diabetes]. Z Gerontol Geriatr 2012; 45:95-9. [PMID: 22350390 DOI: 10.1007/s00391-011-0282-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
The incidence of diabetes has increased in the recent years. Diabetes is characterized by increased sugar concentrations in the blood. Due to this dysregulation, more carbohydrate-induced modification of proteins - so-called advanced glycation end products (AGEs) - are formed endogenously by non-enzymatic reactions. These are discussed to be at least in part responsible for diabetes-associated diseases. The accumulation of AGEs in the tissue can be used as a biomarker for patient outcome. In contrast, the effects of the uptake of AGEs from nutrition are still unclear.
Collapse
Affiliation(s)
- A Simm
- Klinik und Poliklinik für Herz und Thoraxchirurgie, Universitätsklinikum Halle (Saale), Ernst-Grube-Str. 40, 06120, Halle, Deutschland.
| | | | | | | | | |
Collapse
|
38
|
Li Y, Liu S, Zhang Z, Xu Q, Xie F, Wang J, Ping S, Li C, Wang Z, Zhang M, Huang J, Chen D, Hu L, Li C. RAGE mediates accelerated diabetic vein graft atherosclerosis induced by combined mechanical stress and AGEs via synergistic ERK activation. PLoS One 2012; 7:e35016. [PMID: 22496883 PMCID: PMC3322163 DOI: 10.1371/journal.pone.0035016] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 03/08/2012] [Indexed: 12/29/2022] Open
Abstract
Aims/Hypothesis Diabetes with hypertension rapidly accelerates vascular disease, but the underlying mechanism remains unclear. We evaluated the hypothesis that the receptor of advanced glycation end products (RAGE) might mediate combined signals initiated by diabetes-related AGEs and hypertension-induced mechanical stress as a common molecular sensor. Methods In vivo surgical vein grafts created by grafting vena cava segments from C57BL/6J mice into the common carotid arteries of streptozotocin (STZ)-treated and untreated isogenic mice for 4 and 8 weeks were analyzed using morphometric and immunohistochemical techniques. In vitro quiescent mouse vascular smooth muscle cells (VSMCs) with either knockdown or overexpression of RAGE were subjected to cyclic stretching with or without AGEs. Extracellular signal-regulated kinase (ERK) phosphorylation and Ki-67 expression were investigated. Results Significant increases in neointimal formation, AGE deposition, Ki-67 expression, and RAGE were observed in the vein grafts of STZ-induced diabetic mice. The highest levels of ERK phosphorylation and Ki-67 expression in VSMCs were induced by simultaneous stretch stress and AGE exposure. The synergistic activation of ERKs and Ki-67 in VSMCs was significantly inhibited by siRNA-RAGE treatment and enhanced by over-expression of RAGE. Conclusion RAGE may mediate synergistically increased ERK activation and VSMC proliferation induced by mechanical stretching with and without AGEs. It may serve as a common molecular bridge between the two, accelerating vascular remodeling. This study provides potential drug targets and novel therapeutic strategies for the treatment of vascular diseases resulting from diabetes with hypertension.
Collapse
MESH Headings
- Animals
- Atherosclerosis/metabolism
- Atherosclerosis/physiopathology
- Cell Proliferation
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/physiopathology
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/physiopathology
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Extracellular Signal-Regulated MAP Kinases/physiology
- Glycation End Products, Advanced/metabolism
- Glycation End Products, Advanced/physiology
- Ki-67 Antigen/analysis
- Male
- Mice
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiopathology
- Receptor for Advanced Glycation End Products
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/physiology
- Stress, Mechanical
- Tunica Intima/growth & development
- Tunica Intima/metabolism
- Veins/metabolism
- Veins/transplantation
Collapse
Affiliation(s)
- Yuhuang Li
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shuying Liu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhengyu Zhang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qingbo Xu
- Cardiovascular Division, King's College London British Heart Foundation Centre, London, United Kingdom
| | - Fukang Xie
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jingjing Wang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Suning Ping
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chen Li
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhaojing Wang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Min Zhang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jintao Huang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Dadi Chen
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Liping Hu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chaohong Li
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- * E-mail: (CHL)
| |
Collapse
|
39
|
Garcia‐Touza M, Sowers JR. Evidence-based hypertension treatment in patients with diabetes. J Clin Hypertens (Greenwich) 2012; 14:97-102. [PMID: 22277142 PMCID: PMC3270694 DOI: 10.1111/j.1751-7176.2011.00570.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 09/19/2011] [Accepted: 09/26/2011] [Indexed: 12/13/2022]
Abstract
Both impaired glucose tolerance and diabetes are associated with substantially increased prevalence of hypertension, cardiovascular and renal disease. The goal for hypertension treatment in diabetic patients is in evolution, because of recent clinical trials. For example, the results of the recent Action to Control Cardiovascular Risk in Diabetes-BP Arm (ACCORD BP) trial failed to show an additional benefit on cardiovascular event reduction at a mean systolic BP of 119 mm Hg. A post hoc analysis of 6,400 patients with type 2 diabetes from the International Verapamil-Trandolapril Study (INVEST) also failed to show additional cardiovascular risk reduction among patients who achieved a BP <130/80 mm Hg. While the evidence fails to support a lower BP goal to reduce coronary events, there was a risk reduction in stroke events both in ACCORD and the Appropriate Blood Pressure Control in NIDDM (ABCD) trial. A number of other clinical trials also demonstrate that when systolic pressures fall to less than 130 mm Hg, a reduction in stroke but not coronary disease events occurs. Thus, the precise BP goal for diabetic patients remains unresolved. We would posit that a BP goal of 135/85 mm Hg may be a reasonable compromise when viewing the impact of BP reduction on composite stroke and coronary artery disease in extant trials.
Collapse
Affiliation(s)
- Mariana Garcia‐Touza
- From the Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, University of Missouri
;
the Department of Medical Physiology and Pharmacology
;
and the
Harry S. Truman VA Medical Center, Columbia, MO
| | - James R. Sowers
- From the Division of Endocrinology, Diabetes & Metabolism, Department of Internal Medicine, University of Missouri
;
the Department of Medical Physiology and Pharmacology
;
and the
Harry S. Truman VA Medical Center, Columbia, MO
| |
Collapse
|
40
|
Nemoto S, Taguchi K, Matsumoto T, Kamata K, Kobayashi T. Aminoguanidine normalizes ET-1-induced aortic contraction in type 2 diabetic Otsuka Long-Evans Tokushima Fatty (OLETF) rats by suppressing Jab1-mediated increase in ET(A)-receptor expression. Peptides 2012; 33:109-19. [PMID: 22154739 DOI: 10.1016/j.peptides.2011.11.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 11/18/2011] [Accepted: 11/18/2011] [Indexed: 01/08/2023]
Abstract
Circulating levels of endothelin (ET)-1 are increased in the diabetic state, as is endogenous ET(A)-receptor-mediated vasoconstriction. However, the responsible mechanisms remain unknown. We hypothesized that ET-1-induced vasoconstriction is augmented in type 2 diabetes with hyperglycemia through an increment in advanced glycation end-products (AGEs). So, we investigated whether treatment with aminoguanidine (AG), an inhibitor of AGEs, would normalize the ET-1-induced contraction induced by ET-1 in strips of thoracic aortas isolated from OLETF rats at the chronic stage of diabetes. In such aortas (vs. those from age-matched genetic control LETO rats): (1) the ET-1-induced contraction was enhanced, (2) the levels of HIF1α/ECE1/plasma ET-1 and plasma CML-AGEs were increased, (3) the ET-1-stimulated ERK phosphorylation mediated by ET(A)-R was increased, (4) the expression level of Jab1-modified ET(A)-R protein was reduced, and (5) the expression level of O-GlcNAcylated ET(A)-R protein was increased. Aortas isolated from such OLETF rats that had been treated with AG (50mg/kg/day for 10 weeks) exhibited reduced ET-1-induced contraction, suppressed ET-1-stimulated ERK phosphorylation accompanied by down-regulation of ET(A)-R, and increased modification of ET(A)-R by Jab1. Such AG-treated rats exhibited normalized plasma ET-1 and CML-AGE levels, and their aortas exhibited decreased HIF1α/ECE1 expression. However, such AG treatment did not alter the elevated levels of plasma glucose or insulin, or systolic blood pressure seen in OLETF rats. These data from the OLETF model suggest that within the timescale studied here, AG normalizes ET-1-induced aortic contraction by suppressing ET(A)-R/ERK activities and/or by normalizing the imbalance between Jab1 and O-GlcNAc in type 2 diabetes.
Collapse
Affiliation(s)
- Shingo Nemoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Shinagawa-ku, Tokyo 142-8501, Japan
| | | | | | | | | |
Collapse
|
41
|
Xu Y, Feng L, Wang S, Zhu Q, Zheng Z, Xiang P, He B, Tang D. Calycosin protects HUVECs from advanced glycation end products-induced macrophage infiltration. JOURNAL OF ETHNOPHARMACOLOGY 2011; 137:359-370. [PMID: 21669275 DOI: 10.1016/j.jep.2011.05.041] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Revised: 05/18/2011] [Accepted: 05/28/2011] [Indexed: 05/30/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Astragali radix is a traditional Chinese medicine that has long been used for treatment of diabetes and diabetes-associated disease, but its active component and mechanism on the disease is not well defined. AIM OF THE STUDY Infiltration of leukocytes within the glomeruli and vasculature is one of the early and characteristic features of diabetic nephropathy. Advanced glycation end products (AGEs) play pivotal role in the progression of diabetic-associated diseases. The present study was designed to explore the therapeutic effect of calycosin, an active component from A. radix, on AGEs-induced macrophages infiltration in HUVECs. MATERIALS AND METHODS Transwell HUVEC-macrophage co-culture system was established to evaluate macrophage migration and adhesion. Immunocytochemistry was applied to examine TGF-beta1, ICAM-1 and RAGE protein expressions; real-time PCR was carried out to determine mRNA expression of TGF-beta1, ICAM-1 and RAGE. Immunofluorescence was carried out to observe estrogen receptor-alpha, ICAM-1, RAGE expression and the phosphorylation status of ERK1/2 and NF-κB. RESULTS Calycosin significantly reduced AGEs-induced macrophage migration and adhesion to HUVEC. Pre-treatment with calycosin strikingly down-regulated HUVEC TGF-beta1, ICAM-1 and RAGE expressions in both protein and mRNA levels. Furthermore, calycosin incubation significantly increased estrogen receptor expression and reversed AGEs-induced ERK1/2 and NF-κB phosphorylation and nuclear translocation in HUVEC, and this effect of calycosin could be inhibited by estrogen receptor inhibitor, ICI182780. CONCLUSIONS These findings suggest that calycosin can reduce AGEs-induced macrophage migration and adhesion to endothelial cells and relieve the local inflammation; furthermore, this effect was via estrogen receptor-ERK1/2-NF-κB pathway.
Collapse
Affiliation(s)
- Youhua Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Faculty of Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Giannini C, Mohn A, Chiarelli F, Kelnar CJH. Macrovascular angiopathy in children and adolescents with type 1 diabetes. Diabetes Metab Res Rev 2011; 27:436-60. [PMID: 21433262 DOI: 10.1002/dmrr.1195] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Diabetes represents one of the most common diseases globally. Worryingly, the worldwide incidence of type 1 diabetes (T1D) is rising by 3% per year. Despite the rapid increase in diabetes incidence, recent advances in diabetes treatment have been successful in decreasing morbidity and mortality from diabetes-related retinopathy, nephropathy, and neuropathy. In contrast, there is clear evidence for the lack of improvement in mortality for cardiovascular diseases (CVDs). This emphasizes the importance of focusing childhood diabetes care strategies for the prevention of CVD in adulthood. Furthermore, although most work on diabetes and macrovascular disease relates to type 2 diabetes, it has been shown that the age-adjusted relative risk of CVD in T1D far exceeds that in type 2 diabetes. As T1D appears predominantly during childhood, those with T1D are at greater risk for coronary events early in life and require lifelong medical attention. Because of the important health effects of CVDs in children and adolescents with T1D, patients, family members, and care providers should understand the interaction of T1D and cardiovascular risk. In addition, optimal cardiac care for the patient with diabetes should focus on aggressive management of traditional cardiovascular risk factors to optimize those well-recognized as well as new specific risk factors which are becoming available. Therefore, a complete characterization of the molecular mechanisms involved in the development and progression of macrovascular angiopathy is needed. Furthermore, as vascular abnormalities begin as early as in childhood, potentially modifiable risk factors should be identified at an early stage of vascular disease development.
Collapse
Affiliation(s)
- Cosimo Giannini
- Department of Pediatrics, University of Chieti, Chieti, Italy.
| | | | | | | |
Collapse
|
43
|
Talmor-Barkan Y, Bernheim J, Green J, Benchetrit S, Rashid G. Calcitriol counteracts endothelial cell pro-inflammatory processes in a chronic kidney disease-like environment. J Steroid Biochem Mol Biol 2011; 124:19-24. [PMID: 21236342 DOI: 10.1016/j.jsbmb.2011.01.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2010] [Revised: 12/15/2010] [Accepted: 01/04/2011] [Indexed: 11/24/2022]
Abstract
In advanced chronic kidney disease (CKD), hypocalcemia, high levels of advanced glycation end products (AGEs), and parathyroid hormone (PTH) coexist and are considered to play a role in the development of chronic vasculopathies. The aim of the present study was to evaluate the impact of a CKD-like environment on cultured endothelial cell (EC) functions and to assess the impact of calcitriol on the expression of parameters such as endothelial nitric oxide synthase (eNOS), receptor of AGEs (RAGE), interleukin 6 (IL-6) and nuclear factor kappa B (NFκB). Human umbilical vein cord endothelial cells (HUVEC) were grown in medium containing low Ca(2+) concentration stimulated with AGE-HSA and PTH and treated with calcitriol for additional incubation. mRNA expression was established by reverse transcriptase-PCR, protein expression by Western blot analysis, IL-6 secretion by ELISA, NOS activity by conversion of [(14)C]arginine to [(14)C]citrulline and DNA-binding activity of NFκB-p65 assayed colorimetrically in nuclear extracts. The CKD-like environment characterized by the association of low Ca(2+) and high levels of AGEs and PTH, depressed eNOS system activity and enhanced RAGE and IL-6 expression/secretion. DNA-binding activity of nuclear NFκB-p65 was increased and the expression of IκBα decreased. Addition of calcitriol normalized the expression, secretion and activity of eNOS, RAGE and IL-6. The enhanced NFκB activity was also counteracted probably due to the increased IκBα expression. The effect of CKD-like environment on EC may partly explain the increased vasculopathies in CKD patients, in contrast to calcitriol, which suggests a vascular protective action.
Collapse
MESH Headings
- Blotting, Western
- Calcitriol/pharmacology
- Endothelial Cells/drug effects
- Endothelial Cells/pathology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/pathology
- Glycation End Products, Advanced/metabolism
- Humans
- Interleukin-6/biosynthesis
- Interleukin-6/genetics
- Kidney Failure, Chronic/drug therapy
- Kidney Failure, Chronic/metabolism
- Kidney Failure, Chronic/pathology
- NF-kappa B/metabolism
- Nitric Oxide Synthase Type III/biosynthesis
- Nitric Oxide Synthase Type III/genetics
- Nitric Oxide Synthase Type III/metabolism
- Parathyroid Hormone/metabolism
- RNA, Messenger/biosynthesis
- RNA, Messenger/chemistry
- RNA, Messenger/genetics
- Receptor for Advanced Glycation End Products
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Serum Albumin/metabolism
- Serum Albumin, Human
- Vasculitis/drug therapy
- Vasculitis/metabolism
- Vasculitis/pathology
Collapse
Affiliation(s)
- Y Talmor-Barkan
- Department of Nephrology and Hypertension, Meir Medical Center, Kfar Saba, Israel
| | | | | | | | | |
Collapse
|
44
|
Smolock AR, Mishra G, Eguchi K, Eguchi S, Scalia R. Protein kinase C upregulates intercellular adhesion molecule-1 and leukocyte-endothelium interactions in hyperglycemia via activation of endothelial expressed calpain. Arterioscler Thromb Vasc Biol 2011; 31:289-96. [PMID: 21071702 PMCID: PMC3086836 DOI: 10.1161/atvbaha.110.217901] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OBJECTIVE We tested the hypothesis of a role for the calcium-dependent protease calpain in the endothelial dysfunction induced by hyperglycemic activation of protein kinase C (PKC). METHODS AND RESULTS Chronic hyperglycemia with insulin deficiency (type 1 diabetes) was induced in rats by streptozotocin. Total PKC and calpain activities, along with activity and expression level of the 2 endothelial-expressed calpains isoforms, μ- and m-calpain, were measured in vascular tissue homogenates by enzymatic assays and Western blot analysis, respectively. Intravital microscopy was used to measure and correlate leukocyte-endothelium interactions with calpain activity in the microcirculation. Expression levels and endothelial localization of the inflammatory adhesion molecule intercellular adhesion molecule-1 were studied by Western blot analysis and immunofluorescence, respectively. The mechanistic role of hyperglycemia alone in the process of PKC-induced calpain activation and actions was also investigated. We found that in the type 1 diabetic vasculature, PKC selectively upregulates the activity of the μ-calpain isoform. Mechanistic studies confirmed a role for hyperglycemia and PKCβ in this process. The functional implications of PKC-induced calpain activation were upregulation of endothelial expressed intercellular adhesion molecule-1 and leukocyte-endothelium interactions. CONCLUSIONS Our results uncover the role of μ-calpain in the endothelial dysfunction of PKC. Calpain may represent a novel molecular target for the treatment of PKC-associated diabetic vascular disease.
Collapse
Affiliation(s)
- Amanda R Smolock
- Department of Physiology and the Cardiovascular Research Center, School of Medicine, Temple University, 3500 N Broad St., Philadelphia, PA 19041, USA
| | | | | | | | | |
Collapse
|
45
|
Soro-Paavonen A, Zhang WZ, Venardos K, Coughlan MT, Harris E, Tong DCK, Brasacchio D, Paavonen K, Chin-Dusting J, Cooper ME, Kaye D, Thomas MC, Forbes JM. Advanced glycation end-products induce vascular dysfunction via resistance to nitric oxide and suppression of endothelial nitric oxide synthase. J Hypertens 2010; 28:780-8. [PMID: 20186099 DOI: 10.1097/hjh.0b013e328335043e] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE A number of factors contribute to diabetes-associated vascular dysfunction. In the present study, we tested whether exposure to advanced glycation end-products (AGEs) impairs vascular reactivity independently of hyperglycemia and examined the potential mechanisms responsible for diabetes and AGE-associated vascular dysfunction. METHODS Vasodilator function was studied using infusion of exogenous AGEs into Sprague-Dawley rats as compared with control and streptozotocin-induced diabetic rats all followed for 16 weeks (n = 10 per group). The level of arginine metabolites and expression of endothelial nitric oxide synthase (eNOS) and downstream mediators of nitric oxide-dependent signaling were examined. To further explore these mechanisms, cultured bovine aortic endothelial cells (BAECs) were exposed to AGEs. RESULTS Both diabetic and animals infused with AGE-modified rat serum albumin (AGE-RSA) had significantly impaired vasodilatory response to acetylcholine. Unlike diabetes-associated endothelial dysfunction, AGE infusion was not associated with changes in plasma arginine metabolites, asymmetric dimethyl-L-arginine levels or eNOS expression. However, expression of the downstream mediator cGMP-dependent protein kinase 1 (PKG-1) was significantly reduced by both AGE exposure and diabetes. AGEs also augmented hyperglycemia-associated depletion in endothelial nitric oxide production and eNOS protein expression in vitro, and the novel AGE inhibitor, alagebrium chloride, partly restored these parameters. CONCLUSION We demonstrate that AGEs represent a potentially important cause of vascular dysfunction, linked to the induction of nitric oxide resistance. These findings also emphasize the deleterious and potentially additive effects of AGEs and hyperglycemia in diabetic vasculature.
Collapse
|
46
|
Rondeau P, Bourdon E. The glycation of albumin: structural and functional impacts. Biochimie 2010; 93:645-58. [PMID: 21167901 DOI: 10.1016/j.biochi.2010.12.003] [Citation(s) in RCA: 309] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Accepted: 12/07/2010] [Indexed: 02/06/2023]
Abstract
Oxidative stress and protein modifications are frequently observed in numerous disease states. Glucose constitutes a vital nutrient necessary to cellular oxygen metabolism. However, hyperglycemia-associated damage is an important factor in diabetes disorders. Albumin, the major circulating protein in blood, can undergo increased glycation in diabetes. From recent studies, it has become evident that protein glycation has important implications for protein activity, unfolding, and degradation, as well as for cell functioning. After giving a brief overview of the key role of albumin in overall antioxidant defense, this review examines its role as a target of glycation reactions. A synthesis of state of the art methods for measuring and characterizing albumin glycation is detailed. In light of recent data, we then report the impact of glycation on the structure of albumin and its various activities, especially its antioxidant and binding capacities. The biological impact of glycated albumin on cell physiology is also discussed, specifically the role of the protein as a biological marker of diabetes.
Collapse
Affiliation(s)
- Philippe Rondeau
- Laboratoire de Biochimie et Génétique Moléculaire (LBGM)-Groupe d'Etude sur l'Inflammation Chronique et l'Obésité (GEICO), Université de La Réunion, Saint Denis de La Réunion, France.
| | | |
Collapse
|
47
|
Abstract
Oxidative stress plays a pivotal role in the development of diabetes complications, both microvascular and cardiovascular. The metabolic abnormalities of diabetes cause mitochondrial superoxide overproduction in endothelial cells of both large and small vessels, as well as in the myocardium. This increased superoxide production causes the activation of 5 major pathways involved in the pathogenesis of complications: polyol pathway flux, increased formation of AGEs (advanced glycation end products), increased expression of the receptor for AGEs and its activating ligands, activation of protein kinase C isoforms, and overactivity of the hexosamine pathway. It also directly inactivates 2 critical antiatherosclerotic enzymes, endothelial nitric oxide synthase and prostacyclin synthase. Through these pathways, increased intracellular reactive oxygen species (ROS) cause defective angiogenesis in response to ischemia, activate a number of proinflammatory pathways, and cause long-lasting epigenetic changes that drive persistent expression of proinflammatory genes after glycemia is normalized ("hyperglycemic memory"). Atherosclerosis and cardiomyopathy in type 2 diabetes are caused in part by pathway-selective insulin resistance, which increases mitochondrial ROS production from free fatty acids and by inactivation of antiatherosclerosis enzymes by ROS. Overexpression of superoxide dismutase in transgenic diabetic mice prevents diabetic retinopathy, nephropathy, and cardiomyopathy. The aim of this review is to highlight advances in understanding the role of metabolite-generated ROS in the development of diabetic complications.
Collapse
Affiliation(s)
- Ferdinando Giacco
- Diabetes Research Center, Departments of Medicine/Endocrinology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, New York 10461-1602, USA
| | | |
Collapse
|
48
|
Abstract
BACKGROUND Advanced glycation end-products (AGEs) are increased in situations with hyperglycemia and oxidative stress such as diabetes mellitus. They are products of nonenzymatic glycation and oxidation of proteins and lipids. The kidney plays an important role in clearance and metabolism of AGEs. METHODS Medline and other relevant databases were searched. In addition, key review articles were scanned for relevant original publication. Finally, original data from our research group were also included. RESULTS Kidney podocytes and endothelial cells express specific receptors for AGEs. Their activation leads to multiple pathophysiological effects including hypertrophy with cell cycle arrest and apoptosis, altered migration, and generation of proinflammatory cytokines. AGEs have been primarily implicated in the pathophysiology of diabetic nephropathy and diabetic microvascular complications. AGEs are also involved in other primary renal diseases as well as in the development and progression of atherosclerosis. However, serum or plasma concentrations of AGEs do not correlate well with cardiovascular events in patients with chronic kidney disease (CKD). This is likely due to the fact that serum concentrations failed to correlate with AGEs deposited in target tissues. Several inhibitors of the AGE-RAGE axis are currently tested for various indications. CONCLUSION AGEs and their receptors are involved in the pathogenesis of vascular and kidney disease. The role of circulating AGEs as biomarkers for cardiovascular risk estimation is questionable. Whether putative inhibitors of AGEs will get the maturity for its therapeutic use in the future remains open.
Collapse
Affiliation(s)
- Martin Busch
- Department of Internal Medicine III, Jena University Hospital - Friedrich Schiller University, Jena, Germany
| | | | | | | |
Collapse
|
49
|
|
50
|
Nobécourt E, Tabet F, Lambert G, Puranik R, Bao S, Yan L, Davies MJ, Brown BE, Jenkins AJ, Dusting GJ, Bonnet DJ, Curtiss LK, Barter PJ, Rye KA. Nonenzymatic glycation impairs the antiinflammatory properties of apolipoprotein A-I. Arterioscler Thromb Vasc Biol 2010; 30:766-72. [PMID: 20110571 DOI: 10.1161/atvbaha.109.201715] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE The goal of this study was to investigate the effects of nonenzymatic glycation on the antiinflammatory properties of apolipoprotein (apo) A-I. METHODS AND RESULTS Rabbits were infused with saline, lipid-free apoA-I from normal subjects (apoA-I(N)), lipid-free apoA-I nonenzymatically glycated by incubation with methylglyoxal (apoA-I(Glyc in vitro)), nonenzymatically glycated lipid-free apoA-I from subjects with diabetes (apoA-I(Glyc in vivo)), discoidal reconstituted high-density lipoproteins (rHDL) containing phosphatidylcholine and apoA-I(N), (A-I(N))rHDL, or apoA-I(Glyc in vitro), (A-I(Glyc in vitro))rHDL. At 24 hours postinfusion, acute vascular inflammation was induced by inserting a nonocclusive, periarterial carotid collar. The animals were euthanized 24 hours after the insertion of the collar. The collars caused intima/media neutrophil infiltration and increased endothelial expression of vascular cell adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1). ApoA-I(N) infusion decreased neutrophil infiltration and VCAM-1 and ICAM-1 expression by 89%, 90%, and 66%, respectively. The apoA-I(Glyc in vitro) infusion decreased neutrophil infiltration by 53% but did not reduce VCAM-1 or ICAM-1 expression. ApoA-I(Glyc in vivo) did not inhibit neutrophil infiltration or adhesion molecule expression. (A-I(Glyc in vitro))rHDL also inhibited vascular inflammation less effectively than (A-I(N))rHDL. The reduced antiinflammatory properties of nonenzymatically glycated apoA-I were attributed to a reduced ability to inhibit nuclear factor-kappaB activation and reactive oxygen species formation. CONCLUSIONS Nonenzymatic glycation impairs the antiinflammatory properties of apoA-I.
Collapse
Affiliation(s)
- Estelle Nobécourt
- Lipid Research Group, The Heart Research Institute, Sydney, New South Wales, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|