151
|
Lipidomic approach provides new clues toward solving the mystery of accelerated atherosclerosis in diabetes. Atherosclerosis 2016; 251:507-509. [DOI: 10.1016/j.atherosclerosis.2016.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 06/03/2016] [Indexed: 11/23/2022]
|
152
|
Specific enrichment of 2-arachidonoyl-lysophosphatidylcholine in carotid atheroma plaque from type 2 diabetic patients. Atherosclerosis 2016; 251:339-347. [DOI: 10.1016/j.atherosclerosis.2016.05.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 04/22/2016] [Accepted: 05/03/2016] [Indexed: 11/23/2022]
|
153
|
Mosch J, Gleissner CA, Body S, Aikawa E. Histopathological assessment of calcification and inflammation of calcific aortic valves from patients with and without diabetes mellitus. Histol Histopathol 2016; 32:293-306. [PMID: 27353274 DOI: 10.14670/hh-11-797] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Calcific aortic valve disease (CAVD) is the most common valvular heart disease and likely evolves from inflammatory pre-conditions in the valve. Type II diabetes mellitus (DMII) has been associated with pathogenesis of CAVD, however, the mechanism initiating CAVD in DMII is not well understood and the human valve pathology in DMII has not been described. We therefore performed quantitative histological analyses of aortic valves of CAVD patients with and without DMII. METHODS CAVD human aortic valves (n=45) obtained after surgical valve replacement were examined macroscopically with gross measurements of calcified areas. Inflammation and calcification were assessed by immunohistochemistry and immunofluorescence staining. RESULTS Calcification was increased in diabetic patients according to gross measurements (p<0.01) and alizarin red staining (p=0.05). Early calcification markers, including Runx2 (p=0.02) and alkaline phosphatase (ALP, p=0.03) were significantly elevated in diabetic patients. Furthermore, in diabetic patients we found significantly increased expression of annexin II (p=0.04) and annexin V (p=0.04), both of which are thought to play a role in microcalcification formation via apoptosis or extracellular vesicle release. Macrophage numbers were comparable in both groups (p=0.41), while the expression of the pro-inflammatory protein S100A9 (p<0.01) was significantly decreased in diabetic individuals. Evaluation of lymphocytes revealed similar CD8 (p=0.45) and CD4 (p=0.92) T cell counts in diabetic and non-diabetic aortic valves. CONCLUSION Aortic valves from diabetic patients show more calcification, while inflammation is similar in both patient populations. Considering the generally accepted theory of an inflammation-dependent mechanism of calcification, these data suggest that in patients with CAVD requiring valve replacement, diabetic patients could be molecularly in a more advanced disease stage with a higher grade of mineralization than non-diabetic patients.
Collapse
Affiliation(s)
- Josephin Mosch
- Center of Excellence in Vascular Biology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA.,Department of Cardiology, University Hospital, Heidelberg, Germany
| | | | - Simon Body
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Elena Aikawa
- Center of Excellence in Vascular Biology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, USA.,Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, USA.
| |
Collapse
|
154
|
Fassio F, Losappio L, Antolin-Amerigo D, Peveri S, Pala G, Preziosi D, Massaro I, Giuliani G, Gasperini C, Caminati M, Heffler E. Kounis syndrome: A concise review with focus on management. Eur J Intern Med 2016; 30:7-10. [PMID: 26795552 DOI: 10.1016/j.ejim.2015.12.004] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Revised: 12/07/2015] [Accepted: 12/13/2015] [Indexed: 01/02/2023]
Abstract
Kounis syndrome is defined as the co-incidental occurrence of an acute coronary syndrome with hypersensitivity reactions following an allergenic event and was first described by Kounis and Zavras in 1991 as an allergic angina syndrome. Multiple causes have been described and most of the data in the literature are derived from the description of clinical cases - mostly in adult patients - and the pathophysiology remains only partly explained. Three different variants of Kounis syndrome have been defined: type I (without coronary disease) is defined as chest pain during an acute allergic reaction in patients without risk factors or coronary lesions in which the allergic event induces coronary spasm that electrocardiographic changes secondary to ischemia; type II (with coronary disease) includes patients with pre-existing atheromatous disease, either previously quiescent or symptomatic, in whom acute hypersensitive reactions cause plaque erosion or rupture, culminating in acute myocardial infarction; more recently a type-III variant of Kounis syndrome has been defined in patients with preexisting coronary disease and drug eluting coronary stent thrombosis. The pathogenesis of the syndrome is discussed, and a therapeutic algorithm is proposed.
Collapse
Affiliation(s)
- Filippo Fassio
- UO Medicina, Ospedale San Jacopo, ASL3 Pistoia, Via Ciliegiole, 97, 51100 Pistoia, Italy.
| | - Laura Losappio
- University of Foggia, Viale Pinto, 1, 71121 Foggia, Italy.
| | - Dario Antolin-Amerigo
- Servicio de Enfermedades del Sistema Inmune-Alergia, Hospital Principe de Asturias, Departamento de Medicina y Especialidades Médicas, Universidad de Alcalá, Carretera Alcalá, Meco, s/n, 28805 Alcalá de Henares, Madrid, Spain.
| | - Silvia Peveri
- U.O.s.D. Allergologia, Ospedale Guglielmo Da Saliceto, Via Campagna, 68, Piacenza, Italy.
| | - Gianni Pala
- Servizio del Medico Competente, Azienda Sanitaria Locale di Sassari, via Catalocchino 11, 07100 Sassari, Italy.
| | - Donatella Preziosi
- IRCCS Policlinico San Donato, Piazza Edmondo Malan, 1, 20097 San Donato Milanese (MI), Italy.
| | - Ilaria Massaro
- Centro di Ricerca, Trasferimento ed Alta Formazione Denothe, Università di Firenze, Viale Pieraccini, 6, Firenze, Italy.
| | - Gabriele Giuliani
- UO Cardiologia, Ospedale San Giovanni di Dio, via di Torregalli 3, 50143 Firenze, Italy.
| | - Chiara Gasperini
- UO Anestesia e Rianimazione, Ospedale San Jacopo, ASL3 Pistoia, Via Ciliegiole, 97, 51100 Pistoia, Italy.
| | - Marco Caminati
- UO Allergologia, Azienda Ospedaliero Universitaria Integrata, Piazzale Aristide Stefani, 1, Verona, Italy.
| | - Enrico Heffler
- Dipartimento di Medicina Clinica e Sperimentale, Pneumologia Riabilitativa e Allergologia, Università degli Studi di Catania, Via Santa Sofia 78, 95126 Catania, Italy.
| |
Collapse
|
155
|
Ofstad AP. Myocardial dysfunction and cardiovascular disease in type 2 diabetes. Scandinavian Journal of Clinical and Laboratory Investigation 2016; 76:271-81. [PMID: 27071642 DOI: 10.3109/00365513.2016.1155230] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is strongly associated with increased risk of myocardial dysfunction and cardiovascular disease (CVD), two separate conditions which often co-exist and influence each other's course. The prevalence of myocardial dysfunction may be as high as 75% in T2DM populations but is often overlooked due to the initial asymptomatic nature of the disease, complicating co-morbidities such as coronary artery disease (CAD) and obesity, and the lack of consensus on diagnostic criteria. More sensitive echocardiographic applications are furthermore needed to improve detection of early subclinical changes in myocardial function which do not affect conventional echocardiographic parameters. The pathophysiology of the diabetic myocardial dysfunction is not fully elucidated, but involves hyperglycemia and high levels of free fatty acids. It evolves over several years and increases the risk of developing overt HF, and is suggested to at least in part account for the worse outcome seen in T2DM individuals after cardiac events. CAD and stroke are the most frequent CV manifestations among T2DM patients and relate to a large degree to the accelerated atherosclerosis driven by inflammation. Diagnosing CAD is challenging due to the lower sensitivity inherent in the diagnostic tests and there is thus a need for new biomarkers to improve prediction and detection of CAD. It seems that a multi-factorial approach (i.e. targeting several CV risk factors simultaneously) is superior to a strict glucose lowering strategy in reducing risk for macrovascular events, and recent research may even support an effect also on HF outcomes.
Collapse
Affiliation(s)
- Anne Pernille Ofstad
- a Department of Medical Research , Bærum Hospital, Vestre Viken Hospital Trust , Drammen , Norway
| |
Collapse
|
156
|
Maltais JS, Simard E, Froehlich U, Denault JB, Gendron L, Grandbois M. iRAGE as a novel carboxymethylated peptide that prevents advanced glycation end product-induced apoptosis and endoplasmic reticulum stress in vascular smooth muscle cells. Pharmacol Res 2015; 104:176-85. [PMID: 26707030 DOI: 10.1016/j.phrs.2015.12.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 11/06/2015] [Accepted: 12/15/2015] [Indexed: 01/11/2023]
Abstract
Advanced glycation end-products (AGE) and the receptor for AGE (RAGE) have been linked to numerous diabetic vascular complications. RAGE activation promotes a self-sustaining state of chronic inflammation and has been shown to induce apoptosis in various cell types. Although previous studies in vascular smooth muscle cells (VSMC) showed that RAGE activation increases vascular calcification and interferes with their contractile phenotype, little is known on the potential of RAGE to induce apoptosis in VSMC. Using a combination of apoptotic assays, we showed that RAGE stimulation with its ligand CML-HSA promotes apoptosis of VSMC. The formation of stress granules and the increase in the level of the associated protein HuR point toward RAGE-dependent endoplasmic reticulum (ER) stress, which is proposed as a key contributor of RAGE-induced apoptosis in VSMC as it has been shown to promote cell death via numerous mechanisms, including up-regulation of caspase-9. Chronic NF-κB activation and modulation of Bcl-2 homologs are also suspected to contribute to RAGE-dependent apoptosis in VSMC. With the goal of reducing RAGE signaling and its detrimental impact on VSMC, we designed a RAGE antagonist (iRAGE) derived from the primary amino acid sequence of HSA. The resulting CML peptide was selected for the high glycation frequency of the primary sequence in the native protein in vivo. Pretreatment with iRAGE blocked 69.6% of the increase in NF-κB signaling caused by RAGE activation with CML-HSA after 48h. Preincubation with iRAGE was successful in reducing RAGE-induced apoptosis, as seen through enhanced cell survival by SPR and reduced PARP cleavage. Activation of executioner caspases was 63.5% lower in cells treated with iRAGE before stimulation with CML-HSA. To our knowledge, iRAGE is the first antagonist shown to block AGE-RAGE interaction and we propose the molecule as an initial candidate for drug discovery.
Collapse
Affiliation(s)
- Jean-Sébastien Maltais
- Département de pharmacologie et physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Elie Simard
- Département de pharmacologie et physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Ulrike Froehlich
- Département de pharmacologie et physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Jean-Bernard Denault
- Département de pharmacologie et physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Louis Gendron
- Département de pharmacologie et physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Michel Grandbois
- Département de pharmacologie et physiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada.
| |
Collapse
|
157
|
Daffu G, Shen X, Senatus L, Thiagarajan D, Abedini A, Hurtado Del Pozo C, Rosario R, Song F, Friedman RA, Ramasamy R, Schmidt AM. RAGE Suppresses ABCG1-Mediated Macrophage Cholesterol Efflux in Diabetes. Diabetes 2015; 64:4046-60. [PMID: 26253613 PMCID: PMC4657581 DOI: 10.2337/db15-0575] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 07/23/2015] [Indexed: 01/13/2023]
Abstract
Diabetes exacerbates cardiovascular disease, at least in part through suppression of macrophage cholesterol efflux and levels of the cholesterol transporters ATP binding cassette transporter A1 (ABCA1) and ABCG1. The receptor for advanced glycation end products (RAGE) is highly expressed in human and murine diabetic atherosclerotic plaques, particularly in macrophages. We tested the hypothesis that RAGE suppresses macrophage cholesterol efflux and probed the mechanisms by which RAGE downregulates ABCA1 and ABCG1. Macrophage cholesterol efflux to apolipoprotein A1 and HDL and reverse cholesterol transport to plasma, liver, and feces were reduced in diabetic macrophages through RAGE. In vitro, RAGE ligands suppressed ABCG1 and ABCA1 promoter luciferase activity and transcription of ABCG1 and ABCA1 through peroxisome proliferator-activated receptor-γ (PPARG)-responsive promoter elements but not through liver X receptor elements. Plasma levels of HDL were reduced in diabetic mice in a RAGE-dependent manner. Laser capture microdissected CD68(+) macrophages from atherosclerotic plaques of Ldlr(-/-) mice devoid of Ager (RAGE) displayed higher levels of Abca1, Abcg1, and Pparg mRNA transcripts versus Ager-expressing Ldlr(-/-) mice independently of glycemia or plasma levels of total cholesterol and triglycerides. Antagonism of RAGE may fill an important therapeutic gap in the treatment of diabetic macrovascular complications.
Collapse
MESH Headings
- ATP Binding Cassette Transporter 1/genetics
- ATP Binding Cassette Transporter 1/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 1
- ATP-Binding Cassette Transporters/antagonists & inhibitors
- ATP-Binding Cassette Transporters/genetics
- ATP-Binding Cassette Transporters/metabolism
- Animals
- Aorta/immunology
- Aorta/metabolism
- Aorta/pathology
- Biological Transport
- Cell Line
- Cells, Cultured
- Cholesterol/metabolism
- Diabetic Angiopathies/blood
- Diabetic Angiopathies/immunology
- Diabetic Angiopathies/metabolism
- Diabetic Angiopathies/pathology
- Glycation End Products, Advanced/blood
- Glycation End Products, Advanced/metabolism
- Humans
- Ligands
- Lipoproteins/antagonists & inhibitors
- Lipoproteins/genetics
- Lipoproteins/metabolism
- Macrophages/cytology
- Macrophages/immunology
- Macrophages/metabolism
- Macrophages/pathology
- Male
- Mice, Knockout
- PPAR gamma/genetics
- PPAR gamma/metabolism
- Plaque, Atherosclerotic/blood
- Plaque, Atherosclerotic/immunology
- Plaque, Atherosclerotic/metabolism
- Plaque, Atherosclerotic/pathology
- Promoter Regions, Genetic
- Receptor for Advanced Glycation End Products/agonists
- Receptor for Advanced Glycation End Products/blood
- Receptor for Advanced Glycation End Products/genetics
- Receptor for Advanced Glycation End Products/metabolism
- Recombinant Proteins/chemistry
- Recombinant Proteins/metabolism
Collapse
Affiliation(s)
- Gurdip Daffu
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Xiaoping Shen
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Laura Senatus
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Devi Thiagarajan
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Andisheh Abedini
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Carmen Hurtado Del Pozo
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Rosa Rosario
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Fei Song
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Richard A Friedman
- Biomedical Informatics Shared Resource, Herbert Irving Comprehensive Cancer Center, and Department of Biomedical Informatics, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Ravichandran Ramasamy
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Department of Medicine, New York University School of Medicine, New York, NY
| |
Collapse
|
158
|
Ramasamy R, Shekhtman A, Schmidt AM. The multiple faces of RAGE--opportunities for therapeutic intervention in aging and chronic disease. Expert Opin Ther Targets 2015; 20:431-46. [PMID: 26558318 DOI: 10.1517/14728222.2016.1111873] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION This review focuses on the multi-ligand receptor of the immunoglobulin superfamily--receptor for advanced glycation endproducts (RAGE). The accumulation of the multiple ligands of RAGE in cellular stress milieux links RAGE to the pathobiology of chronic disease and natural aging. AREAS COVERED In this review, we present a discussion on the ligands of RAGE and the implications of these ligand families in disease. We review the recent literature on the role of ligand-RAGE interaction in the consequences of natural aging; the macro- and microvascular complications of diabetes; obesity and insulin resistance; autoimmune disorders and chronic inflammation; and tumors and Alzheimer's disease. We discuss the mechanisms of RAGE signaling through its intracellular binding effector molecule--the formin DIAPH1. Physicochemical evidence of how the RAGE cytoplasmic domain binds to the FH1 (formin homology 1) domain of DIAPH1, and the consequences thereof, are also reviewed. EXPERT OPINION We discuss the modalities of RAGE antagonism currently in preclinical and clinical studies. Finally, we present the rationale behind potentially targeting the RAGE cytoplasmic domain-DIAPH1 interaction as a logical strategy for therapeutic intervention in the pathological settings of chronic diseases and aging wherein RAGE ligands accumulate and signal.
Collapse
Affiliation(s)
- Ravichandran Ramasamy
- a Diabetes Research Program, Division of Endocrinology, Department of Medicine , New York University Langone Medical Center , New York , NY 10016 , USA
| | - Alexander Shekhtman
- b Department of Chemistry , University at Albany, State University of New York , Albany , NY 12222 , USA
| | - Ann Marie Schmidt
- a Diabetes Research Program, Division of Endocrinology, Department of Medicine , New York University Langone Medical Center , New York , NY 10016 , USA
| |
Collapse
|
159
|
Avogaro A, Fadini GP. Mechanisms of ectopic calcification: implications for diabetic vasculopathy. Cardiovasc Diagn Ther 2015; 5:343-52. [PMID: 26543821 DOI: 10.3978/j.issn.2223-3652.2015.06.05] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Vascular calcification (VC) is the deposition of calcium/phosphate in the vasculature, which portends a worse clinical outcome and predicts major adverse cardiovascular events. VC is an active process initiated and regulated via a variety of molecular signalling pathways. There are mainly two types of calcifications: the media VC and the intima VC. All major risk factors for cardiovascular disease (CVD) have been linked to the presence/development of VC. Besides the risk factors, a genetic component is also operative to determine arterial calcification. Several events take place before VC is established, including inflammation, trans-differentiation of vascular cells and homing of circulating pro-calcific cells. Diabetes is an important predisposing factor for VC. Compared with non-diabetic subjects, patients with diabetes show increased VC and higher expression of bone-related proteins in the medial layer of the vessels. In this review we will highlight the mechanisms underlying vascular calcification in diabetic patients.
Collapse
Affiliation(s)
- Angelo Avogaro
- 1 Division of Metabolic Diseases, Department of Medicine, University of Padova, Padova, Italy ; 2 Laboratory of Experimental Diabetology, Venetian Institute of Molecular Medicine, Padova, Italy
| | - Gian Paolo Fadini
- 1 Division of Metabolic Diseases, Department of Medicine, University of Padova, Padova, Italy ; 2 Laboratory of Experimental Diabetology, Venetian Institute of Molecular Medicine, Padova, Italy
| |
Collapse
|
160
|
Oesterle A, Bowman MAH. S100A12 and the S100/Calgranulins: Emerging Biomarkers for Atherosclerosis and Possibly Therapeutic Targets. Arterioscler Thromb Vasc Biol 2015; 35:2496-507. [PMID: 26515415 DOI: 10.1161/atvbaha.115.302072] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 10/14/2015] [Indexed: 11/16/2022]
Abstract
Atherosclerosis is mediated by local and systematic inflammation. The multiligand receptor for advanced glycation end products (RAGE) has been studied in animals and humans and is an important mediator of inflammation and atherosclerosis. This review focuses on S100/calgranulin proteins (S100A8, S100A9, and S100A12) and their receptor RAGE in mediating vascular inflammation. Mice lack the gene for S100A12, which in humans is located on chromosome 3 between S100A8 and S100A9. Transgenic mice with smooth muscle cell-targeted expression of S100A12 demonstrate increased coronary and aortic calcification, as well as increased plaque vulnerability. Serum S100A12 has recently been shown to predict future cardiovascular events in a longitudinal population study, underscoring a role for S100A12 as a potential biomarker for coronary artery disease. Genetic ablation of S100A9 or RAGE in atherosclerosis-susceptible apolipoprotein E null mice results in reduced atherosclerosis. Importantly, S100A12 and the RAGE axis can be modified pharmacologically. For example, soluble RAGE reduces murine atherosclerosis and vascular inflammation. Additionally, a class of compounds currently in phase III clinical trials for multiple sclerosis and rheumatologic conditions, the quinoline-3-carboxamides, reduce atherosclerotic plaque burden and complexity in transgenic S100A12 apolipoprotein E null mice, but have not been tested with regards to human atherosclerosis. The RAGE axis is an important mediator for inflammation-induced atherosclerosis, and S100A12 has emerged as biomarker for human atherosclerosis. Decreasing inflammation by inhibiting S100/calgranulin-mediated activation of RAGE attenuates murine atherosclerosis, and future studies in patients with coronary artery disease are warranted to confirm S100/RAGE as therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Adam Oesterle
- From the Department of Medicine, The University of Chicago, IL
| | | |
Collapse
|
161
|
Abstract
Plaque rupture, usually of a precursor lesion known as a 'vulnerable plaque' or 'thin-cap fibroatheroma', is the leading cause of thrombosis. Less-frequent aetiologies of coronary thrombosis are erosion, observed with greatest incidence in women aged <50 years, and eruptive calcified nodules, which are occasionally identified in older individuals. Various treatments for patients with coronary artery disease, such as CABG surgery and interventional therapies, have led to accelerated atherosclerosis. These processes occur within months to years, compared with the decades that it generally takes for native disease to develop. Morphological identifiers of accelerated atherosclerosis include macrophage-derived foam cells, intraplaque haemorrhage, and thin fibrous cap. Foam-cell infiltration can be observed within 1 year of a saphenous vein graft implantation, with subsequent necrotic core formation and rupture ensuing after 7 years in over one-third of patients. Neoatherosclerosis occurs early and with greater prevalence in drug-eluting stents than in bare-metal stents and, although rare, complications of late stent thrombosis from rupture are associated with high mortality. Comparison of lesion progression in native atherosclerotic disease, atherosclerosis in saphenous vein grafts, and in-stent neoatherosclerosis provides insight into the pathogenesis of atheroma formation in natural and iatrogenic settings.
Collapse
|
162
|
Li M, Lin F, Lin Y, Peng W. Extracellular polysaccharide from Bordetella species reduces high glucose-induced macrophage apoptosis via regulating interaction between caveolin-1 and TLR4. Biochem Biophys Res Commun 2015; 466:748-54. [DOI: 10.1016/j.bbrc.2015.09.125] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 09/23/2015] [Indexed: 11/26/2022]
|
163
|
Soluble Receptor for Advanced Glycation End Product: A Biomarker for Acute Coronary Syndrome. BIOMED RESEARCH INTERNATIONAL 2015; 2015:815942. [PMID: 26491690 PMCID: PMC4605229 DOI: 10.1155/2015/815942] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 09/14/2015] [Accepted: 09/15/2015] [Indexed: 02/08/2023]
Abstract
The receptor of advanced glycation end products (RAGE) and its ligands are linked to the pathogenesis of coronary artery disease (CAD), and circulating soluble receptor of advanced glycation end products (sRAGE), reflecting the RAGE activity, is suggested as a potential biomarker. Elevated sRAGE levels are reported in relation to acute ischemia and this review focuses on the role of sRAGE as a biomarker for the acute coronary syndrome (ACS). The current studies demonstrated that sRAGE levels are elevated in relation to ACS, however during a very narrow time period, indicating that the time of sampling needs attention. Interestingly, activation of RAGE may influence the pathogenesis and reflection in sRAGE levels in acute and stable CAD differently.
Collapse
|
164
|
Cai X, Bao L, Dai X, Ding Y, Zhang Z, Li Y. Quercetin protects RAW264.7 macrophages from glucosamine-induced apoptosis and lipid accumulation via the endoplasmic reticulum stress pathway. Mol Med Rep 2015; 12:7545-53. [PMID: 26398703 DOI: 10.3892/mmr.2015.4340] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 08/24/2015] [Indexed: 11/05/2022] Open
Abstract
It is increasingly recognized that macrophages are a key cell in the development of atherosclerosis. Glucosamine, the product of the hexosamine biosynthetic pathway in diabetes mellitus, can disturb lipid metabolism, induce apoptosis and accelerate atherosclerosis via endoplasmic reticulum (ER) stress in various types of cells. Previous studies have indicated that quercetin possesses antidiabetic, anti‑oxidative, anti‑inflammatory and anti‑apoptotic activities as a flavonoid. Studies have also demonstrated its novel pharmacological properties for inhibiting ER stress. The present study focussed on the effects of quercetin on cell injury and ER stress in glucosamine‑induced macrophages. RAW264.7 macrophages were cultured with 15 mM glucosamine, following which the levels of apoptosis, intracellular total and free cholesterol, and apoptosis‑ and ER stress‑associated proteins were measured in the macrophages treated with or without quercetin. Additionally, the ratio of cholestryl ester/total cholesterol was calculated to observe the formation of foam cells. The results demonstrated that apoptosis and abnormal lipid accumulation in the RAW264.7 cells, which was induced by glucosamine, were significantly reversed by quercetin. In addition, quercetin treatment suppressed the increase of C/EBP homologous protein, and inhibited the activation of JNK and caspase‑12, which was induced by glucosamine. Quercetin also increased the expression level of full length activating transcriptional factor 6 and decreased the expression of glucose regulated protein 78. Of note, the beneficial effects of quercetin on the glucosamine‑induced RAW264.7 cells were reversed by treatment with tunicamycin. These findings suggest that quercetin may have properties to prevent glucosamine‑induced apoptosis and lipid accumulation via the ER stress pathway in RAW264.7 macrophages.
Collapse
Affiliation(s)
- Xiaxia Cai
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, P.R. China
| | - Lei Bao
- Department of Clinical Nutrition, Peking University International Hospital, Beijing 102206, P.R. China
| | - Xiaoqian Dai
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, P.R. China
| | - Ye Ding
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, P.R. China
| | - Zhaofeng Zhang
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, P.R. China
| | - Yong Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, P.R. China
| |
Collapse
|
165
|
Cohort comparison study of cardiac disease and atherosclerotic burden in type 2 diabetic adults using whole body cardiovascular magnetic resonance imaging. Cardiovasc Diabetol 2015; 14:122. [PMID: 26382729 PMCID: PMC4574534 DOI: 10.1186/s12933-015-0284-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 09/08/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Whole body cardiovascular MR (WB CVMR) combines whole body angiography and cardiac MR assessment. It is accepted that there is a high disease burden in patients with diabetes, however the quantification of the whole body atheroma burden in both arterial and cardiac disease has not been previously reported. In this study we compare the quantified atheroma burden in those individuals with and without diabetes by clinical cardiovascular disease (CVD) status. METHODS 158 participants underwent WB CVMR, and were categorised into one of four groups: (1) type 2 diabetes mellitus (T2DM) with CVD; (2) T2DM without CVD; (3) CVD without T2DM; (4) healthy controls. The arterial tree was subdivided into 31 segments and each scored according to the degree of stenosis. From this a standardised atheroma score (SAS) was calculated. Cardiac MR and late gadolinium enhancement images of the left ventricle were obtained for assessment of mass, volume and myocardial scar assessment. RESULTS 148 participants completed the study protocol--61% male, with mean age of 64 ± 8.2 years. SAS was highest in those with cardiovascular disease without diabetes [10.1 (0-39.5)], followed by those with T2DM and CVD [4 (0-41.1)], then those with T2DM only [3.23 (0-19.4)] with healthy controls having the lowest atheroma score [2.4 (0-19.4)]. Both groups with a prior history of CVD had a higher SAS and left ventricular mass than those without (p < 0.001 for both). However after accounting for known cardiovascular risk factors, only the SAS in the group with CVD without T2DM remained significantly elevated. 6% of the T2DM group had evidence of silent myocardial infarct, with this subcohort having a higher SAS than the remainder of the T2DM group [7.7 (4-19) vs. 2.8 (0-17), p = 0.024]. CONCLUSIONS Global atheroma burden was significantly higher in those with known cardiovascular disease and without diabetes but not in those with diabetes and cardiovascular disease suggesting that cardiovascular events may occur at a lower atheroma burden in diabetes.
Collapse
|
166
|
Koulis C, Watson A, Gray S, Jandeleit-Dahm K. Linking RAGE and Nox in diabetic micro- and macrovascular complications. DIABETES & METABOLISM 2015; 41:272-281. [DOI: 10.1016/j.diabet.2015.01.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 01/24/2015] [Accepted: 01/30/2015] [Indexed: 12/31/2022]
|
167
|
Malmstedt J, Kärvestedt L, Swedenborg J, Brismar K. The receptor for advanced glycation end products and risk of peripheral arterial disease, amputation or death in type 2 diabetes: a population-based cohort study. Cardiovasc Diabetol 2015. [PMID: 26216409 PMCID: PMC4517412 DOI: 10.1186/s12933-015-0257-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background Patients with type 2 diabetes have a high risk for early and extensive development of peripheral arterial disease (PAD) and this excess risk is not explained by increased burden of traditional atherosclerotic risk factors. Activation of the receptor for advanced glycation end products (RAGE) could be one additional mechanism for accelerated PAD and increased risk for amputation and death. We investigated the association between RAGE plasma components and the risk for PAD, amputation and death in patients with type 2 diabetes. We also estimated the rate of amputation-free survival and survival without PAD. Methods We investigated if plasma levels of carboxymethyl-lysine, S100A12 and endosecretory RAGE (esRAGE) were associated with two endpoints: survival without development of PAD and survival without amputation in a 12 years prospective population-based cohort of 146 patients with type 2 diabetes, free from PAD at inclusion. Influence of baseline plasma levels of RAGE ligands (individually and combined by a RAGE-score) were evaluated for both endpoints in the Cox-regression analysis. Results 106 patients survived without amputation and 93 survived without signs of PAD during follow up. Higher levels of S100A12 and RAGE-score were associated with increased risk for amputation or death, hazard ratios (HR) 1.29; 95% confidence interval (CI) [1.04, 1.59] and 1.79; 95% CI [1.07, 2.99] and with increased risk for PAD or death, HR 1.22; 95% CI [1.00, 1.49] and 1.56; [1.00, 2.44] after adjustment for age and sex. The effect was decreased after adjustment for Framingham cardiovascular disease score: risk for amputation or death, HR 1.17; 95% CI [0.94, 1.46] and 1.54; [0.95, 2.49], and risk for PAD or death, HR 1.12; 95% CI [0.91, 1.38] and 1.38; [0.91, 2.11] for S100A12 and RAGE-score respectively. The incidence for amputation or death was 2.8 per 100 person-years; 95% CI [2.0, 3.7] and the incidence rate for PAD or death was 3.6 per 100 person-years; 95% CI [2.7, 4.8]. Conclusion Higher plasma levels of S100A12 and the combined effect (RAGE-score) of esRAGE, carboxymethyl-lysine and S100A12 seem to be associated with shorter PAD- and amputation-free survival in patients with type 2 diabetes. This may indicate a role for S100A12 in PAD by activation of the RAGE system. Electronic supplementary material The online version of this article (doi:10.1186/s12933-015-0257-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jonas Malmstedt
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden. .,Division of Vascular Surgery, Department of Surgery, South Hospital, 118 83, Stockholm, Sweden.
| | - Lars Kärvestedt
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
| | - Jesper Swedenborg
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
| | - Kerstin Brismar
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden. .,Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
168
|
Nichols TC, Merricks EP, Bellinger DA, Raymer RA, Yu J, Lam D, Koch GG, Busby WH, Clemmons DR. Oxidized LDL and Fructosamine Associated with Severity of Coronary Artery Atherosclerosis in Insulin Resistant Pigs Fed a High Fat/High NaCl Diet. PLoS One 2015; 10:e0132302. [PMID: 26147990 PMCID: PMC4492503 DOI: 10.1371/journal.pone.0132302] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 06/11/2015] [Indexed: 12/22/2022] Open
Abstract
Background Insulin-resistant subjects develop more severe and diffuse coronary artery atherosclerosis than insulin-sensitive controls but the mechanisms that mediate this atherosclerosis phenotype are unknown. Research Objective To determine the metabolic parameters that associate with the severity of coronary atherosclerosis in insulin resistant pigs fed a high fat/high NaCl diet. Key Methods The primary endpoint was severity of coronary atherosclerosis in adult pigs (Sus scrofa, n = 37) fed a high fat diet that also contained high NaCl (56% above recommended levels) for 1 year. Principal Findings Twenty pigs developed severe and diffuse distal coronary artery atherosclerosis (i.e., severe = intimal area as a percent medial area > 200% in at least 2 coronary artery cross sections and diffuse distal = intimal area as a percent medial area ≥ 150% over 3 sections separated by 2 cm in the distal half of the coronary artery). The other 17 pigs had substantially less coronary artery atherosclerosis. All 37 pigs had blood pressure in a range that would be considered hypertensive in humans and developed elevations in total and LDL and HDL cholesterol, weight gain, increased backfat, and increased insulin resistance (Bergman Si) without overt diabetes. Insulin resistance was not associated with atherosclerosis severity. Five additional pigs fed regular pig chow also developed increased insulin resistance but essentially no change in the other variables and little to no detectible coronary atherosclerosis. Most importantly, the 20 high fat/high NaCl diet -fed pigs with severe and diffuse distal coronary artery atherosclerosis had substantially greater increases (p< 0.05) in oxidized LDL (oxLDL) and fructosamine consistent with increased protein glycation. Conclusion In pigs fed a high fat/high NaCl diet, glycated proteins are induced in the absence of overt diabetes and this degree of increase is associated with the development of severe and diffuse distal coronary artery atherosclerosis.
Collapse
Affiliation(s)
- Timothy C. Nichols
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- * E-mail:
| | - Elizabeth P. Merricks
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Dwight A. Bellinger
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Robin A. Raymer
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Jing Yu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Diana Lam
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Gary G. Koch
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Walker H. Busby
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - David R. Clemmons
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| |
Collapse
|
169
|
Comparison of Neoatherosclerosis and Neovascularization Between Patients With and Without Diabetes. JACC Cardiovasc Interv 2015; 8:1044-1052. [DOI: 10.1016/j.jcin.2015.02.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 01/21/2015] [Accepted: 02/12/2015] [Indexed: 11/30/2022]
|
170
|
Christensen MHE, Fenne IS, Nordbø Y, Varhaug JE, Nygård KO, Lien EA, Mellgren G. Novel inflammatory biomarkers in primary hyperparathyroidism. Eur J Endocrinol 2015; 173:9-17. [PMID: 25850829 DOI: 10.1530/eje-14-1038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 04/07/2015] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Primary hyperparathyroidism (PHPT) has been associated with low-grade inflammation and increased risk of cardiovascular disease (CVD). The aim of the study was to investigate systemic levels of pro-inflammatory proteins that previously have not been examined in patients with PHPT. The selection of the pro-inflammatory biomarkers included in this study, MMP9, S100A4, S100A8/A9 and the receptors sCD14 and RAGE, was based on a previous microarray screen of mRNAs in adipose tissue from PHPT patients. DESIGN A prospective study was conducted on a total of 57 patients with PHPT and a control group of 20 healthy blood donors. METHODS PHPT patients with normalisation of serum calcium levels after parathyroidectomy were followed for 6 months. Forty-two patients participated in the longitudinal study, in which blood samples were taken at inclusion, and 1, 3 and 6 months after surgery. RESULTS We observed increased serum levels of MMP9 (P=0.029), S100A4 (P<0.001) and sCD14 (P=0.002) in the 57 patients with PHPT compared to the control-group. During 6 months of follow up, S100A4 (P=0.022) and sCD14 (0.002) decreased significantly, while serum levels of MMP9 increased (P=0.025). CONCLUSIONS The results demonstrate an increased inflammatory response in PHPT patients shown by elevated MMP9, S100A4 and sCD14 at inclusion. During the 6 months of follow-up, MMP9 increased further, possibly due to the tissue repair process after parathyroidectomy. S100A4 and sCD14 decreased after surgery demonstrating a partial reversal of the systemic inflammation.
Collapse
Affiliation(s)
- M H E Christensen
- Department of Clinical ScienceUniversity of Bergen, Bergen, NorwayHormone LaboratoryDepartment of SurgeryHaukeland University Hospital, 5021 Bergen, NorwayDepartment of Surgical ScienceUniversity of Bergen, Bergen, NorwayKG Jebsen Center for Diabetes ResearchBergen, NorwayDepartment of Heart DiseaseHaukeland University Hospital, Bergen, Norway Department of Clinical ScienceUniversity of Bergen, Bergen, NorwayHormone LaboratoryDepartment of SurgeryHaukeland University Hospital, 5021 Bergen, NorwayDepartment of Surgical ScienceUniversity of Bergen, Bergen, NorwayKG Jebsen Center for Diabetes ResearchBergen, NorwayDepartment of Heart DiseaseHaukeland University Hospital, Bergen, Norway
| | - I S Fenne
- Department of Clinical ScienceUniversity of Bergen, Bergen, NorwayHormone LaboratoryDepartment of SurgeryHaukeland University Hospital, 5021 Bergen, NorwayDepartment of Surgical ScienceUniversity of Bergen, Bergen, NorwayKG Jebsen Center for Diabetes ResearchBergen, NorwayDepartment of Heart DiseaseHaukeland University Hospital, Bergen, Norway Department of Clinical ScienceUniversity of Bergen, Bergen, NorwayHormone LaboratoryDepartment of SurgeryHaukeland University Hospital, 5021 Bergen, NorwayDepartment of Surgical ScienceUniversity of Bergen, Bergen, NorwayKG Jebsen Center for Diabetes ResearchBergen, NorwayDepartment of Heart DiseaseHaukeland University Hospital, Bergen, Norway
| | - Y Nordbø
- Department of Clinical ScienceUniversity of Bergen, Bergen, NorwayHormone LaboratoryDepartment of SurgeryHaukeland University Hospital, 5021 Bergen, NorwayDepartment of Surgical ScienceUniversity of Bergen, Bergen, NorwayKG Jebsen Center for Diabetes ResearchBergen, NorwayDepartment of Heart DiseaseHaukeland University Hospital, Bergen, Norway
| | - J E Varhaug
- Department of Clinical ScienceUniversity of Bergen, Bergen, NorwayHormone LaboratoryDepartment of SurgeryHaukeland University Hospital, 5021 Bergen, NorwayDepartment of Surgical ScienceUniversity of Bergen, Bergen, NorwayKG Jebsen Center for Diabetes ResearchBergen, NorwayDepartment of Heart DiseaseHaukeland University Hospital, Bergen, Norway Department of Clinical ScienceUniversity of Bergen, Bergen, NorwayHormone LaboratoryDepartment of SurgeryHaukeland University Hospital, 5021 Bergen, NorwayDepartment of Surgical ScienceUniversity of Bergen, Bergen, NorwayKG Jebsen Center for Diabetes ResearchBergen, NorwayDepartment of Heart DiseaseHaukeland University Hospital, Bergen, Norway
| | - K O Nygård
- Department of Clinical ScienceUniversity of Bergen, Bergen, NorwayHormone LaboratoryDepartment of SurgeryHaukeland University Hospital, 5021 Bergen, NorwayDepartment of Surgical ScienceUniversity of Bergen, Bergen, NorwayKG Jebsen Center for Diabetes ResearchBergen, NorwayDepartment of Heart DiseaseHaukeland University Hospital, Bergen, Norway Department of Clinical ScienceUniversity of Bergen, Bergen, NorwayHormone LaboratoryDepartment of SurgeryHaukeland University Hospital, 5021 Bergen, NorwayDepartment of Surgical ScienceUniversity of Bergen, Bergen, NorwayKG Jebsen Center for Diabetes ResearchBergen, NorwayDepartment of Heart DiseaseHaukeland University Hospital, Bergen, Norway Department of Clinical ScienceUniversity of Bergen, Bergen, NorwayHormone LaboratoryDepartment of SurgeryHaukeland University Hospital, 5021 Bergen, NorwayDepartment of Surgical ScienceUniversity of Bergen, Bergen, NorwayKG Jebsen Center for Diabetes ResearchBergen, NorwayDepartment of Heart DiseaseHaukeland University Hospital, Bergen, Norway
| | - E A Lien
- Department of Clinical ScienceUniversity of Bergen, Bergen, NorwayHormone LaboratoryDepartment of SurgeryHaukeland University Hospital, 5021 Bergen, NorwayDepartment of Surgical ScienceUniversity of Bergen, Bergen, NorwayKG Jebsen Center for Diabetes ResearchBergen, NorwayDepartment of Heart DiseaseHaukeland University Hospital, Bergen, Norway Department of Clinical ScienceUniversity of Bergen, Bergen, NorwayHormone LaboratoryDepartment of SurgeryHaukeland University Hospital, 5021 Bergen, NorwayDepartment of Surgical ScienceUniversity of Bergen, Bergen, NorwayKG Jebsen Center for Diabetes ResearchBergen, NorwayDepartment of Heart DiseaseHaukeland University Hospital, Bergen, Norway
| | - G Mellgren
- Department of Clinical ScienceUniversity of Bergen, Bergen, NorwayHormone LaboratoryDepartment of SurgeryHaukeland University Hospital, 5021 Bergen, NorwayDepartment of Surgical ScienceUniversity of Bergen, Bergen, NorwayKG Jebsen Center for Diabetes ResearchBergen, NorwayDepartment of Heart DiseaseHaukeland University Hospital, Bergen, Norway Department of Clinical ScienceUniversity of Bergen, Bergen, NorwayHormone LaboratoryDepartment of SurgeryHaukeland University Hospital, 5021 Bergen, NorwayDepartment of Surgical ScienceUniversity of Bergen, Bergen, NorwayKG Jebsen Center for Diabetes ResearchBergen, NorwayDepartment of Heart DiseaseHaukeland University Hospital, Bergen, Norway Department of Clinical ScienceUniversity of Bergen, Bergen, NorwayHormone LaboratoryDepartment of SurgeryHaukeland University Hospital, 5021 Bergen, NorwayDepartment of Surgical ScienceUniversity of Bergen, Bergen, NorwayKG Jebsen Center for Diabetes ResearchBergen, NorwayDepartment of Heart DiseaseHaukeland University Hospital, Bergen, Norway
| |
Collapse
|
171
|
Gary T, Belaj K, Hafner F, Eller P, Rief P, Hackl G, Brodmann M. Graz Critical Limb Ischemia Score: A Risk Score for Critical Limb Ischemia in Peripheral Arterial Occlusive Disease. Medicine (Baltimore) 2015; 94:e1054. [PMID: 26166076 PMCID: PMC4504640 DOI: 10.1097/md.0000000000001054] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Critical limb ischemia (CLI), a frequently encountered disorder, is associated with a high rate of limb amputation and mortality. To identify patients at high risk for CLI, we developed a simple risk score for peripheral arterial occlusive disease (PAOD).In our cross-sectional study, we first evaluated 1000 consecutive PAOD patients treated at our institution from 2005 to 2007, documenting clinical symptoms, comorbidities, and concomitant medication. We calculated odds ratios (OR) in a binary logistic regression model to find possible risk factors for CLI. We then verified the score in a second step that included the 1124 PAOD patients we treated between 2007 and 2011.In the first patient group, the greatest risk factors for CLI were age ≥75 years (OR 2.0), type 2 diabetes (OR 3.1), prior myocardial infarction (OR 2.5), and therapy with low molecular weight heparins (2.8). We scored 1 point for each of those conditions. One point was given for age between 65 and 75 years (OR 1.6) as well as for therapy with cardiac glycosides (OR 1.9) or loop diuretic therapy (OR 1.5). As statin therapy was protective for CLI with an OR of 0.5, we subtracted 1 point for those patients.In the second group, we could prove that frequency of CLI was significantly higher in patients with a high CLI score. The score correlated well with inflammatory parameters (c-reactive protein and fibrinogen). We were also able to define 3 different risk groups for low (score -1 to 1), intermediate (score 2-4), and high CLI risk (score >4).We developed a simple risk stratification scheme that is based on conditions that can be easily assessed from the medical history, without any laboratory parameters. This score should help to identify PAOD patients at high risk for CLI.
Collapse
Affiliation(s)
- Thomas Gary
- From the Division of Angiology, Department of Internal Medicine, Medical University Graz, Graz, Austria
| | | | | | | | | | | | | |
Collapse
|
172
|
Eun SY, Ko YS, Park SW, Chang KC, Kim HJ. IL-1β enhances vascular smooth muscle cell proliferation and migration via P2Y2 receptor-mediated RAGE expression and HMGB1 release. Vascul Pharmacol 2015; 72:108-17. [PMID: 25956731 DOI: 10.1016/j.vph.2015.04.013] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 03/31/2015] [Accepted: 04/20/2015] [Indexed: 01/11/2023]
Abstract
Vascular smooth muscle cells (VSMCs) are the major cell type in blood vessel walls, and their proliferation and migration play important roles in the development of atherosclerosis. Recently, it has been reported that IL-1β mediates the inflammatory response through the upregulation of the P2Y2 receptor (P2Y2R). Thus, we examined the role of P2Y2R in IL-1β-mediated proliferation and migration of VSMCs and the underlying molecular mechanisms. VSMCs were pretreated with IL-1β for 24h to upregulate P2Y2R expression. The cells were then stimulated with UTP or ATP for the indicated times, and cell proliferation and migration and the related signaling pathways were examined. The equipotent P2Y2R agonists ATP and UTP enhanced proliferation, RAGE expression and HMGB1 secretion in IL-1β-pretreated VSMCs. Additionally, pretreatment with IL-1β enhanced UTP-mediated VSMC migration and MMP-2 release, but these effects were not observed in the P2Y2R-siRNA- or RAGE-siRNA-transfected VSMCs. Next, the signaling molecules involved in P2Y2R-mediated cell proliferation and migration were determined. The ERK, AKT, PKC, Rac-1 and ROCK2 pathways were involved in UTP-induced cell proliferation and migration, MMP-2 and HMGB1 secretion and RAGE expression in the IL-1β-pretreated VSMCs. UTP induced the phosphorylation of ERK, AKT and PKC and the translocation of Rac-1 and ROCK2 from cytosol to membrane as well as stress fiber formation, which were markedly increased in the IL-1β-pretreated VSMCs but not in the P2Y2R-siRNA-transfected VSMCs. These results demonstrate that pro-inflammatory cytokines associated with atherosclerosis, such as IL-1β, can accelerate the process of atherosclerosis through the upregulation of P2Y2R.
Collapse
Affiliation(s)
- So Young Eun
- Department of Pharmacology, School Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju 660-290, Republic of Korea
| | - Young Shin Ko
- Department of Pharmacology, School Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju 660-290, Republic of Korea
| | - Sang Won Park
- Department of Pharmacology, School Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju 660-290, Republic of Korea
| | - Ki Churl Chang
- Department of Pharmacology, School Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju 660-290, Republic of Korea
| | - Hye Jung Kim
- Department of Pharmacology, School Medicine, Institute of Health Sciences, Gyeongsang National University, Jinju 660-290, Republic of Korea.
| |
Collapse
|
173
|
Frasnelli SCT, de Medeiros MC, Bastos ADS, Costa DL, Orrico SRP, Rossa Junior C. Modulation of immune response by RAGE and TLR4 signalling in PBMCs of diabetic and non-diabetic patients. Scand J Immunol 2015; 81:66-71. [PMID: 25223881 DOI: 10.1111/sji.12241] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 08/09/2014] [Indexed: 11/29/2022]
Abstract
Diabetes is associated with increased glucose levels and accumulation of glycated products. It is also associated with impairment in the immune response, such as increased susceptibility to infections. In this study, we assessed the possible interactions between TLR4 and RAGE signalling on apoptosis and on the expression of inflammatory cytokines in PBMC from individuals with and without diabetes. PBMCs were isolated from seven diabetic patients and six individuals without diabetes and stimulated in vitro with bacterial LPS (1 μg/ml) associated or not with BSA-AGE (200 μg/ml). This stimulation was performed for 6 h, both in the presence and in the absence of inhibitors of TLR4 (R. sphaeroides LPS, 20 μg/ml) and RAGE (blocking monoclonal antibody). Apoptosis at early and late stages was assessed by the annexin-V/PI staining using flow cytometry. Regulation of TNF-α and IL-10 gene expression was determined by RT-qPCR. PBMCs from diabetes patients tended to be more resistant apoptosis. There were no synergistic or antagonistic effects with the simultaneous activation of TLR4 and RAGE in PBMCs from either diabetes or non-diabetes group. Activation of TLR4 is more potent for the induction of TNF-α and IL-10; RAGE signalling had a negative regulatory effect on TNF-α expression induced by LPS. TLR and RAGE do not have relevant roles in apoptosis of PBMCs. The activation of TLR has greater role than RAGE in regulating the gene expression of IL-10 and TNF-α.
Collapse
Affiliation(s)
- S C T Frasnelli
- Department of Diagnosis and Surgery, School of Dentistry at Araraquara, UNESP - Univ Estadual Paulista, Araraquara, SP, Brazil
| | | | | | | | | | | |
Collapse
|
174
|
Ramasamy R, Yan SF, Schmidt AM. Glycation, Inflammation and RAGE. Atherosclerosis 2015. [DOI: 10.1002/9781118828533.ch3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
175
|
Affiliation(s)
- Erik A L Biessen
- Experimental Vascular Pathology Group, Department of Pathology, CARIM, Maastricht University Medical Center, Maastricht, The Netherlands (E.L.B., J.C.S.)
| | - Judith C Sluimer
- Experimental Vascular Pathology Group, Department of Pathology, CARIM, Maastricht University Medical Center, Maastricht, The Netherlands (E.L.B., J.C.S.)
| |
Collapse
|
176
|
Vascular effects of advanced glycation end-products: content of immunohistochemically detected AGEs in radial artery samples as a predictor for arterial calcification and cardiovascular risk in asymptomatic patients with chronic kidney disease. DISEASE MARKERS 2015; 2015:153978. [PMID: 25852219 PMCID: PMC4380091 DOI: 10.1155/2015/153978] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 02/24/2015] [Indexed: 11/17/2022]
Abstract
OBJECTIVES Our aim was to determine whether vascular deposition of advanced glycation end-products (AGEs) is associated with arterial calcification and cardiovascular mortality in chronic kidney disease (CKD) patients and to assess the relationships between vascular content of AGEs and selected clinical and biochemical parameters. MATERIALS AND METHODS The study comprised 54 CKD patients (33 hemodialyzed, 21 predialyzed). Examined parameters included BMI, incidence of diabetes, plasma fasting glucose, AGEs, soluble receptor for AGEs and 2,2-diphenyl-1-picrylhydrazyl (DPPH) scavenging, serum C-reactive protein (hsCRP), plasminogen activator inhibitor-1 (PAI-1), and fetuin-A. Fragments of radial artery obtained during creation of hemodialysis access were stained for calcifications using alizarin red. AGEs deposits were identified immunohistochemically and their relative content was quantified. RESULTS Vascular content of AGEs was positively correlated with BMI, hsCRP, fetuin-A, PAI-1, and DPPH scavenging in simple regression; only fetuin-A was an independent predictor in multiple regression. There was a significant positive trend in the intensity of AGEs immunostaining among patients with grades 1, 2, and 3 calcifications. AGEs immunostaining intensity predicted 3-year cardiovascular mortality irrespective of patient's age. CONCLUSIONS The present study demonstrates an involvement of AGEs in the development of medial arterial calcification and the impact of arterial AGE deposition on cardiovascular mortality in CKD patients.
Collapse
|
177
|
Davidson MH. Potential Impact of Dipeptidyl Peptidase-4 Inhibitors on Cardiovascular Pathophysiology in Type 2 Diabetes Mellitus. Postgrad Med 2015; 126:56-65. [DOI: 10.3810/pgm.2014.05.2756] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
178
|
Strang AC, van Wijk DF, Mutsaerts HJMM, Stroes ESG, Nederveen AJ, Rotmans JI, Rabelink TJ, Box FMA. Guideline treatment results in regression of atherosclerosis in type 2 diabetes mellitus. Diab Vasc Dis Res 2015; 12:126-32. [PMID: 25589481 DOI: 10.1177/1479164114559511] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Efficacy of guideline cardiovascular disease prevention regimens may differ between patients with or without type II diabetes mellitus. We therefore compared change in carotid artery wall dimensions in type II diabetes mellitus and non-type II diabetes mellitus patients with a history of a major cardiovascular disease event, using magnetic resonance imaging. METHODS Thirty type II diabetes mellitus patients and 29 age- and sex-matched non-diabetes mellitus patients with a history of stroke or myocardial infarction and a carotid artery stenosis (15%-70%) were included. In all patients, treatment was according to cardiovascular risk management guidelines. At baseline and follow-up, carotid artery vessel wall dimensions were measured using 1.5 T magnetic resonance imaging. RESULTS After 2 years of follow-up, total wall volume of the carotid artery in type II diabetes mellitus patients decreased by 9.6% (p = 0.016). In contrast, stabilization rather than regression of carotid artery wall dimensions was observed in non-diabetes mellitus patients over a 2-year period. Body mass index was identified as a predictor of total wall volume decrease. CONCLUSIONS Guideline treatment arrests atherogenesis in non-diabetes mellitus patients and even decreases vessel wall dimensions in type II diabetes mellitus patients. Baseline body mass index predicts cardiovascular disease prevention efficacy expressed as decrease in total wall volume. These data emphasize the importance of optimal cardiovascular-prevention, particularly in diabetes patients with a high body mass index.
Collapse
Affiliation(s)
- Aart C Strang
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Diederik F van Wijk
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | | | - Erik S G Stroes
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Aart J Nederveen
- Department of Radiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Joris I Rotmans
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ton J Rabelink
- Department of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Frieke M A Box
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
179
|
Ligthart S, Sedaghat S, Ikram MA, Hofman A, Franco OH, Dehghan A. EN-RAGE. Arterioscler Thromb Vasc Biol 2014; 34:2695-9. [DOI: 10.1161/atvbaha.114.304306] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Symen Ligthart
- From the Department of Epidemiology (S.L., S.S., M.A.I., A.H., O.H.F., A.D.), Department of Neurology (M.A.I.), and Department of Radiology (M.A.I.), Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sanaz Sedaghat
- From the Department of Epidemiology (S.L., S.S., M.A.I., A.H., O.H.F., A.D.), Department of Neurology (M.A.I.), and Department of Radiology (M.A.I.), Erasmus University Medical Center, Rotterdam, The Netherlands
| | - M. Arfan Ikram
- From the Department of Epidemiology (S.L., S.S., M.A.I., A.H., O.H.F., A.D.), Department of Neurology (M.A.I.), and Department of Radiology (M.A.I.), Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Albert Hofman
- From the Department of Epidemiology (S.L., S.S., M.A.I., A.H., O.H.F., A.D.), Department of Neurology (M.A.I.), and Department of Radiology (M.A.I.), Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Oscar H. Franco
- From the Department of Epidemiology (S.L., S.S., M.A.I., A.H., O.H.F., A.D.), Department of Neurology (M.A.I.), and Department of Radiology (M.A.I.), Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Abbas Dehghan
- From the Department of Epidemiology (S.L., S.S., M.A.I., A.H., O.H.F., A.D.), Department of Neurology (M.A.I.), and Department of Radiology (M.A.I.), Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
180
|
Stegman B, Puri R, Cho L, Shao M, Ballantyne CM, Barter PJ, Chapman MJ, Erbel R, Libby P, Raichlen JS, Uno K, Kataoka Y, Nissen SE, Nicholls SJ. High-intensity statin therapy alters the natural history of diabetic coronary atherosclerosis: insights from SATURN. Diabetes Care 2014; 37:3114-20. [PMID: 25190674 DOI: 10.2337/dc14-1121] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Although statins can induce coronary atheroma regression, this benefit has yet to be demonstrated in diabetic individuals. We tested the hypothesis that high-intensity statin therapy may promote coronary atheroma regression in patients with diabetes. RESEARCH DESIGN AND METHODS The Study of Coronary Atheroma by Intravascular Ultrasound: Effect of Rosuvastatin Versus Atorvastatin (SATURN) used serial intravascular ultrasound measures of coronary atheroma volume in patients treated with rosuvastatin 40 mg or atorvastatin 80 mg for 24 months. This analysis compared changes in biochemistry and coronary percent atheroma volume (PAV) in patients with (n = 159) and without (n = 880) diabetes. RESULTS At baseline, patients with diabetes had lower LDL cholesterol (LDL-C) and HDL cholesterol (HDL-C) levels but higher triglyceride and CRP levels compared with patients without diabetes. At follow-up, diabetic patients had lower levels of LDL-C (61.0 ± 20.5 vs. 66.4 ± 22.9 mg/dL, P = 0.01) and HDL-C (46.3 ± 10.6 vs. 49.9 ± 12.0 mg/dL, P < 0.001) but higher levels of triglycerides (127.6 [98.8, 163.0] vs. 113.0 mg/dL [87.6, 151.9], P = 0.001) and CRP (1.4 [0.7, 3.3] vs. 1.0 [0.5, 2.1] mg/L, P = 0.001). Both patients with and without diabetes demonstrated regression of coronary atheroma as measured by change in PAV (-0.83 ± 0.13 vs. -1.15 ± 0.13%, P = 0.08). PAV regression was less in diabetic compared with nondiabetic patients when on-treatment LDL-C levels were >70 mg/dL (-0.31 ± 0.23 vs. -1.01 ± 0.21%, P = 0.03) but similar when LDL-C levels were ≤70 mg/dL (-1.09 ± 0.16 vs. -1.24 ± 0.16%, P = 0.50). CONCLUSIONS High-intensity statin therapy alters the progressive nature of diabetic coronary atherosclerosis, yielding regression of disease in diabetic and nondiabetic patients.
Collapse
Affiliation(s)
- Brian Stegman
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH
| | - Rishi Puri
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH C5Research, Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH
| | - Leslie Cho
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH
| | - Mingyuan Shao
- C5Research, Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH
| | - Christie M Ballantyne
- Section of Cardiovascular Research, Baylor College of Medicine, and the Methodist DeBakey Heart and Vascular Center, Houston, TX
| | - Phillip J Barter
- Centre for Vascular Research, University of New South Wales, Sydney, Australia
| | - M John Chapman
- INSERM Dyslipidaemia and Atherosclerosis Research Unit, Pitié-Salpètrière University Hospital, Paris, France
| | | | - Peter Libby
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA
| | | | - Kiyoko Uno
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH
| | - Yu Kataoka
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| | - Steven E Nissen
- Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH C5Research, Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, OH
| | - Stephen J Nicholls
- South Australian Health and Medical Research Institute, University of Adelaide, Adelaide, Australia
| |
Collapse
|
181
|
de Ferranti SD, de Boer IH, Fonseca V, Fox CS, Golden SH, Lavie CJ, Magge SN, Marx N, McGuire DK, Orchard TJ, Zinman B, Eckel RH. Type 1 diabetes mellitus and cardiovascular disease: a scientific statement from the American Heart Association and American Diabetes Association. Diabetes Care 2014; 37:2843-63. [PMID: 25114297 PMCID: PMC4170130 DOI: 10.2337/dc14-1720] [Citation(s) in RCA: 268] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
182
|
Yan L, Bowman MAH. Chronic sustained inflammation links to left ventricular hypertrophy and aortic valve sclerosis: a new link between S100/RAGE and FGF23. INFLAMMATION AND CELL SIGNALING 2014; 1. [PMID: 26082935 DOI: 10.14800/ics.279] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cardiovascular disease including left ventricular hypertrophy, diastolic dysfunction and ectopic valvular calcification are common in patients with chronic kidney disease (CKD). Both S100A12 and fibroblast growth factor 23 (FGF23) have been identified as biomarkers of cardiovascular morbidity and mortality in patients with CKD. We tested the hypothesis that human S100/calgranulin would accelerate cardiovascular disease in mice subjected to CKD. METHODS This review paper focuses on S100 proteins and their receptor for advanced glycation end products (RAGE) and summarizes recent findings obtained in novel developed transgenic hBAC-S100 mice that express S100A12 and S100A8/9 proteins. A bacterial artificial chromosome of the human S100/calgranulin gene cluster containing the genes and regulatory elements for S100A8, S100A9 and S100A12 was expressed in C57BL/6J mice (hBAC-S100). CKD was induced by ureteral ligation, and hBAC-S100 mice and WT mice were studied after 10 weeks of chronic uremia. RESULTS hBAC-S100 mice with CKD showed increased FGF23 in the heart, left ventricular hypertrophy (LVH), diastolic dysfunction, focal cartilaginous metaplasia and calcification of the mitral and aortic valve annulus together with aortic valve sclerosis. This phenotype was not observed in WT mice with CKD or in hBAC-S100 mice lacking RAGE with CKD, suggesting that the inflammatory milieu mediated by S100/RAGE promotes pathological cardiac hypertrophy in CKD. In vitro, inflammatory stimuli including IL-6, TNFα, LPS, or serum from hBAC-S100 mice up regulated FGF23 mRNA and protein in primary murine neonatal and adult cardiac fibroblasts. CONCLUSIONS Taken together, our study shows that myeloid-derived human S100/calgranulin is associated with the development of cardiac hypertrophy and ectopic cardiac calcification in a RAGE dependent manner in a mouse model of CKD. We speculate that FGF23 produced by cardiac fibroblasts in response to cytokines may act in a paracrine manner to accelerate LVH and diastolic dysfunction in hBAC-S100 mice with CKD. We suggest that S100/RAGE-mediated chronic sustained systemic inflammation is linked to pathological cardiac remodeling via direct up regulation of FGF23 in cardiac fibroblasts, thereby providing a new mechanistic understanding for the common association between CKD, diabetes, metabolic syndrome, or hypertension with left ventricular hypertrophy with diastolic dysfunction.
Collapse
Affiliation(s)
- Ling Yan
- Department of Medicine, Section of Cardiology, The University of Chicago, Chicago, IL 60637 USA
| | - Marion A Hofmann Bowman
- Department of Medicine, Section of Cardiology, The University of Chicago, Chicago, IL 60637 USA
| |
Collapse
|
183
|
Edsfeldt A, Gonçalves I, Grufman H, Nitulescu M, Dunér P, Bengtsson E, Mollet IG, Persson A, Nilsson M, Orho-Melander M, Melander O, Björkbacka H, Nilsson J. Impaired Fibrous Repair. Arterioscler Thromb Vasc Biol 2014; 34:2143-50. [DOI: 10.1161/atvbaha.114.303414] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Diabetes mellitus (DM) type II is increasing rapidly worldwide. Patients with DM II have a greater atherosclerotic burden and higher risk of developing cardiovascular complications. Inflammation has been proposed as the main cause for the high risk of atherosclerotic disease in DM II. In this study, we compared markers of inflammation and fibrous repair in plaques from subjects with and without DM II.
Approach and Results—
Carotid endarterectomy specimens were obtained from 63 patients with and 131 without DM. Plaque structure, connective tissue proteins, inflammatory cells, and markers were analyzed by immunohistochemistry, ELISA, Mesoscale, and Luminex technology. Carotid plaques from diabetics had lower levels of extracellular matrix proteins, elastin, and collagen, which are critical for plaque stability. Plaques from diabetics had reduced levels of platelet-derived growth factor and matrix metalloproteinase-2, both important for tissue repair. No differences were observed in inflammatory markers in plaques from diabetic and nondiabetic patients.
Conclusion—
This study suggests that atherosclerotic plaques in subjects with DM II are more prone to rupture because of impaired repair responses rather than to increased vascular inflammation. Although this study did not have a mechanistic design, our findings suggest that targeting impaired repair responses in carotid plaques may help to increase our understanding of atherosclerotic plaque development and vulnerability in patients with DM II.
Collapse
Affiliation(s)
- Andreas Edsfeldt
- From the Experimental Cardiovascular Research Unit (A.E., I.G., H.G., M.N., P.D., E.B., A.P., M.N., H.B., J.N.) and Department of Clinical Sciences (I.G.M., M.O.-M., O.M.), Clinical Research Center, Clinical Sciences, Lund University, Malmö, Sweden; and Department of Cardiology—Coronary Diseases, Skåne University Hospital, Malmö, Sweden (A.E., I.G., H.G., A.P., M.N.)
| | - Isabel Gonçalves
- From the Experimental Cardiovascular Research Unit (A.E., I.G., H.G., M.N., P.D., E.B., A.P., M.N., H.B., J.N.) and Department of Clinical Sciences (I.G.M., M.O.-M., O.M.), Clinical Research Center, Clinical Sciences, Lund University, Malmö, Sweden; and Department of Cardiology—Coronary Diseases, Skåne University Hospital, Malmö, Sweden (A.E., I.G., H.G., A.P., M.N.)
| | - Helena Grufman
- From the Experimental Cardiovascular Research Unit (A.E., I.G., H.G., M.N., P.D., E.B., A.P., M.N., H.B., J.N.) and Department of Clinical Sciences (I.G.M., M.O.-M., O.M.), Clinical Research Center, Clinical Sciences, Lund University, Malmö, Sweden; and Department of Cardiology—Coronary Diseases, Skåne University Hospital, Malmö, Sweden (A.E., I.G., H.G., A.P., M.N.)
| | - Mihaela Nitulescu
- From the Experimental Cardiovascular Research Unit (A.E., I.G., H.G., M.N., P.D., E.B., A.P., M.N., H.B., J.N.) and Department of Clinical Sciences (I.G.M., M.O.-M., O.M.), Clinical Research Center, Clinical Sciences, Lund University, Malmö, Sweden; and Department of Cardiology—Coronary Diseases, Skåne University Hospital, Malmö, Sweden (A.E., I.G., H.G., A.P., M.N.)
| | - Pontus Dunér
- From the Experimental Cardiovascular Research Unit (A.E., I.G., H.G., M.N., P.D., E.B., A.P., M.N., H.B., J.N.) and Department of Clinical Sciences (I.G.M., M.O.-M., O.M.), Clinical Research Center, Clinical Sciences, Lund University, Malmö, Sweden; and Department of Cardiology—Coronary Diseases, Skåne University Hospital, Malmö, Sweden (A.E., I.G., H.G., A.P., M.N.)
| | - Eva Bengtsson
- From the Experimental Cardiovascular Research Unit (A.E., I.G., H.G., M.N., P.D., E.B., A.P., M.N., H.B., J.N.) and Department of Clinical Sciences (I.G.M., M.O.-M., O.M.), Clinical Research Center, Clinical Sciences, Lund University, Malmö, Sweden; and Department of Cardiology—Coronary Diseases, Skåne University Hospital, Malmö, Sweden (A.E., I.G., H.G., A.P., M.N.)
| | - Inês G. Mollet
- From the Experimental Cardiovascular Research Unit (A.E., I.G., H.G., M.N., P.D., E.B., A.P., M.N., H.B., J.N.) and Department of Clinical Sciences (I.G.M., M.O.-M., O.M.), Clinical Research Center, Clinical Sciences, Lund University, Malmö, Sweden; and Department of Cardiology—Coronary Diseases, Skåne University Hospital, Malmö, Sweden (A.E., I.G., H.G., A.P., M.N.)
| | - Ana Persson
- From the Experimental Cardiovascular Research Unit (A.E., I.G., H.G., M.N., P.D., E.B., A.P., M.N., H.B., J.N.) and Department of Clinical Sciences (I.G.M., M.O.-M., O.M.), Clinical Research Center, Clinical Sciences, Lund University, Malmö, Sweden; and Department of Cardiology—Coronary Diseases, Skåne University Hospital, Malmö, Sweden (A.E., I.G., H.G., A.P., M.N.)
| | - Marie Nilsson
- From the Experimental Cardiovascular Research Unit (A.E., I.G., H.G., M.N., P.D., E.B., A.P., M.N., H.B., J.N.) and Department of Clinical Sciences (I.G.M., M.O.-M., O.M.), Clinical Research Center, Clinical Sciences, Lund University, Malmö, Sweden; and Department of Cardiology—Coronary Diseases, Skåne University Hospital, Malmö, Sweden (A.E., I.G., H.G., A.P., M.N.)
| | - Marju Orho-Melander
- From the Experimental Cardiovascular Research Unit (A.E., I.G., H.G., M.N., P.D., E.B., A.P., M.N., H.B., J.N.) and Department of Clinical Sciences (I.G.M., M.O.-M., O.M.), Clinical Research Center, Clinical Sciences, Lund University, Malmö, Sweden; and Department of Cardiology—Coronary Diseases, Skåne University Hospital, Malmö, Sweden (A.E., I.G., H.G., A.P., M.N.)
| | - Olle Melander
- From the Experimental Cardiovascular Research Unit (A.E., I.G., H.G., M.N., P.D., E.B., A.P., M.N., H.B., J.N.) and Department of Clinical Sciences (I.G.M., M.O.-M., O.M.), Clinical Research Center, Clinical Sciences, Lund University, Malmö, Sweden; and Department of Cardiology—Coronary Diseases, Skåne University Hospital, Malmö, Sweden (A.E., I.G., H.G., A.P., M.N.)
| | - Harry Björkbacka
- From the Experimental Cardiovascular Research Unit (A.E., I.G., H.G., M.N., P.D., E.B., A.P., M.N., H.B., J.N.) and Department of Clinical Sciences (I.G.M., M.O.-M., O.M.), Clinical Research Center, Clinical Sciences, Lund University, Malmö, Sweden; and Department of Cardiology—Coronary Diseases, Skåne University Hospital, Malmö, Sweden (A.E., I.G., H.G., A.P., M.N.)
| | - Jan Nilsson
- From the Experimental Cardiovascular Research Unit (A.E., I.G., H.G., M.N., P.D., E.B., A.P., M.N., H.B., J.N.) and Department of Clinical Sciences (I.G.M., M.O.-M., O.M.), Clinical Research Center, Clinical Sciences, Lund University, Malmö, Sweden; and Department of Cardiology—Coronary Diseases, Skåne University Hospital, Malmö, Sweden (A.E., I.G., H.G., A.P., M.N.)
| |
Collapse
|
184
|
Reith S, Battermann S, Hellmich M, Marx N, Burgmaier M. Correlation between optical coherence tomography-derived intraluminal parameters and fractional flow reserve measurements in intermediate grade coronary lesions: a comparison between diabetic and non-diabetic patients. Clin Res Cardiol 2014; 104:59-70. [DOI: 10.1007/s00392-014-0759-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 08/11/2014] [Indexed: 10/24/2022]
|
185
|
de Ferranti SD, de Boer IH, Fonseca V, Fox CS, Golden SH, Lavie CJ, Magge SN, Marx N, McGuire DK, Orchard TJ, Zinman B, Eckel RH. Type 1 diabetes mellitus and cardiovascular disease: a scientific statement from the American Heart Association and American Diabetes Association. Circulation 2014; 130:1110-30. [PMID: 25114208 DOI: 10.1161/cir.0000000000000034] [Citation(s) in RCA: 239] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
186
|
Chaabane C, Heizmann CW, Bochaton-Piallat ML. Extracellular S100A4 induces smooth muscle cell phenotypic transition mediated by RAGE. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:2144-57. [PMID: 25110349 DOI: 10.1016/j.bbamcr.2014.07.022] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 07/20/2014] [Accepted: 07/29/2014] [Indexed: 01/11/2023]
Abstract
We identified S100A4 as a marker of rhomboid (R) smooth muscle cells (SMCs) in vitro (the synthetic phenotype, typical of intimal SMCs) in the porcine coronary artery and of intimal SMCs in vivo in both pigs and humans. S100A4 is an intracellular Ca²⁺ signaling protein and can be secreted; it has extracellular functions via the receptor for advanced glycation end products (RAGE). Our objective was to explore the role of S100A4 in SMC phenotypic change, a phenomenon characteristic of atherosclerotic plaque formation. Transfection of a human S100A4-containing plasmid in spindle-shaped (S) SMCs (devoid of S100A4) led to approximately 10% of S100A4-overexpressing SMCs, S100A4 release, and a transition towards a R-phenotype of the whole SMC population. Furthermore treatment of S-SMCs with S100A4-rich conditioned medium collected from S100A4-transfected S-SMCs induced a transition towards a R-phenotype, which was associated with decreased SMC differentiation markers and increased proliferation and migration by activating the urokinase-type plasminogen activator (uPA), matrix metalloproteinases (MMPs) and their inhibitors (TIMPs). It yielded NF-κB activation in a RAGE-dependent manner. Blockade of extracellular S100A4 in R-SMCs with S100A4 neutralizing antibody induced a transition from R- to S-phenotype, decreased proliferative activity and upregulation of SMC differentiation markers. By contrast, silencing of S100A4 mRNA in R-SMCs did not change the level of extracellular S100A4 or SMC morphology in spite of decreased proliferative activity. Our results show that extracellular S100A4 plays a pivotal role in SMC phenotypic changes. It could be a new target to prevent SMC accumulation during atherosclerosis and restenosis. This article is part of a Special Issue entitled: 13th European Symposium on Calcium.
Collapse
Affiliation(s)
- Chiraz Chaabane
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Claus W Heizmann
- Department of Pediatrics, Division of Clinical Chemistry and Biochemistry, University of Zürich, Zürich, Switzerland
| | | |
Collapse
|
187
|
Scholtes VP, Peeters W, van Lammeren GW, Howard DP, de Vries JPP, de Borst GJ, Redgrave JN, Kemperman H, Schalkwijk CG, den Ruijter HM, de Kleijn DP, Moll FL, Rothwell PM, Pasterkamp G. Type 2 diabetes is not associated with an altered plaque phenotype among patients undergoing carotid revascularization. A histological analysis of 1455 carotid plaques. Atherosclerosis 2014; 235:418-23. [DOI: 10.1016/j.atherosclerosis.2014.05.941] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 05/13/2014] [Accepted: 05/21/2014] [Indexed: 10/25/2022]
|
188
|
Histologic and immunohistochemical analysis of the antiatherogenic effects of myocardial bridging in the adult human heart. Cardiovasc Pathol 2014; 23:198-203. [DOI: 10.1016/j.carpath.2014.03.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Revised: 03/12/2014] [Accepted: 03/13/2014] [Indexed: 12/30/2022] Open
|
189
|
Lipidomics: Potential role in risk prediction and therapeutic monitoring for diabetes and cardiovascular disease. Pharmacol Ther 2014; 143:12-23. [DOI: 10.1016/j.pharmthera.2014.02.001] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 01/27/2014] [Indexed: 01/07/2023]
|
190
|
Choi YH, Hong YJ, Ahn Y, Park IH, Jeong MH. Relationship between neutrophil-to-lymphocyte ratio and plaque components in patients with coronary artery disease: virtual histology intravascular ultrasound analysis. J Korean Med Sci 2014; 29:950-6. [PMID: 25045227 PMCID: PMC4101783 DOI: 10.3346/jkms.2014.29.7.950] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Accepted: 04/14/2014] [Indexed: 11/20/2022] Open
Abstract
The aim of this study was to evaluate the relation between neutrophil-to-lymphocyte ratio (NLR) and plaque components assessed by virtual histology-intravascular ultrasound in 399 coronary artery disease (CAD) patients with 471 coronary lesions. We classified the lesions into two groups according to the NLR on admission {low NLR group (NLR≤2.73 [n=370]) vs. high NLR group (NLR>2.73 [n=101])}. By volumetric analysis, total atheroma and the absolute necrotic core (NC) volumes were significantly greater in high NLR group (249.9±149.7 µL vs. 192.5±127.7 µL, P=0.001, and 32.7±26.8 µL vs. 22.8±19.4 µL, P=0.001, respectively) and thin-cap fibroatheroma (TCFA) was observed more frequently in high NLR group (33% vs. 18%, P=0.001). ST segment elevation myocardial infarction (odds ratio [OR], 2.159; 95% CI, 1.000-4.660, P=0.050) and NLR>2.73 (OR, 1.848; 95% CI, 1.016-3.360, P=0.044) and total atheroma volume (OR, 1.003; 95% CI, 1.001-1.004, P=0.004) were the independent predictors of TCFA. CAD patients with high NLR had more vulnerable plaque components (greater NC-containing plaques) than those with low NLR.
Collapse
Affiliation(s)
- Yun Ha Choi
- The Division of Cardiology, Chonnam National University Hospital, Gwangju, Korea
- College of Nursing, Chonnam National University, Gwangju, Korea
| | - Young Joon Hong
- The Division of Cardiology, Chonnam National University Hospital, Gwangju, Korea
| | - Youngkeun Ahn
- The Division of Cardiology, Chonnam National University Hospital, Gwangju, Korea
| | - In Hyae Park
- College of Nursing, Chonnam National University, Gwangju, Korea
| | - Myung Ho Jeong
- The Division of Cardiology, Chonnam National University Hospital, Gwangju, Korea
| |
Collapse
|
191
|
Johnson LL, Tekabe Y, Kollaros M, Eng G, Bhatia K, Li C, Krueger CG, Shanmuganayagam D, Schmidt AM. Imaging RAGE expression in atherosclerotic plaques in hyperlipidemic pigs. EJNMMI Res 2014; 4:26. [PMID: 25006545 PMCID: PMC4078320 DOI: 10.1186/s13550-014-0026-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 04/29/2014] [Indexed: 02/02/2023] Open
Abstract
Background Receptor for advanced glycated end product (RAGE) expression is a prominent feature of atherosclerosis. We have previously shown in apoE null mice uptake of a radiolabeled anti-RAGE antibody in atherosclerotic plaque and now evaluate RAGE-directed imaging to identify advanced plaques in a large animal model. Methods Nine hyperlipidemic (HL) pigs were injected with 603.1 ± 129.5 MBq of 99mTc-anti-RAGE F(ab′)2, and after 6 h (blood pool clearance), they underwent single-photon emission computed tomography/computed tomography (SPECT/CT) imaging of the neck, thorax, and hind limbs. Two HL pigs received 99mTc non-immune IgG F(ab′)2, and three farm pigs were injected with 99mTc-anti-RAGE F(ab′)2. After imaging, the pigs were euthanized. The aorta from the root to bifurcation was dissected, and the innominates, proximal carotids, and coronaries were dissected and counted, stained for H&E and RAGE, and AHA-classified. Results On pathology, 24% of the arterial segments showed AHA class III or IV lesions, and these lesions were confined almost exclusively to coronaries and carotids with % stenosis from 15% to 65%. Scatter plots of %ID/g for class III/IV vs. I/II lesions showed almost complete separation. Focal vascular uptake of tracer visualized on SPECT scans corresponded to class III/IV lesions in the coronary and carotid vessels. In addition, uptake in the hind limbs was noted in the HL pigs and corresponded to RAGE staining of small arteries in the muscle sections. Correlations for the vascular lesions were r = 0.747, P = 0.001 for %ID vs. %ID/g and r = 0.83, P = 0.002 for %ID/g vs. % RAGE staining. Conclusions Uptake of radiolabeled anti-RAGE antibody in coronary and carotid fibroatheroma and in the small arteries of the hind limbs in a relevant large animal model of atherosclerosis supports the important role of RAGE in atherosclerosis and peripheral artery disease as a target for imaging and treatment.
Collapse
Affiliation(s)
- Lynne L Johnson
- Department of Medicine, Columbia University Medical Center, 622 West 168 St, New York 10032, NY, USA
| | - Yared Tekabe
- Department of Medicine, Columbia University Medical Center, 622 West 168 St, New York 10032, NY, USA
| | - Maria Kollaros
- Department of Medicine, Columbia University Medical Center, 622 West 168 St, New York 10032, NY, USA
| | - George Eng
- Department of Medicine, Columbia University Medical Center, 622 West 168 St, New York 10032, NY, USA
| | - Ketan Bhatia
- Department of Medicine, Columbia University Medical Center, 622 West 168 St, New York 10032, NY, USA
| | - Chong Li
- Department of Medicine, Columbia University Medical Center, 622 West 168 St, New York 10032, NY, USA
| | - Christian G Krueger
- Department of Veterinary Medicine, University of Wisconsin, Madison 53706, WI, USA
| | | | | |
Collapse
|
192
|
Watson AMD, Li J, Samijono D, Bierhaus A, Thomas MC, Jandeleit-Dahm KAM, Cooper ME. Quinapril treatment abolishes diabetes-associated atherosclerosis in RAGE/apolipoprotein E double knockout mice. Atherosclerosis 2014; 235:444-8. [PMID: 24945577 DOI: 10.1016/j.atherosclerosis.2014.05.945] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 05/19/2014] [Accepted: 05/22/2014] [Indexed: 11/18/2022]
Abstract
OBJECTIVE/RATIONALE Both the renin-angiotensin system (RAS) and the receptor for advanced glycation end products (RAGE) potentiate diabetes-associated atherosclerosis (DAA). We assessed the effectiveness of concomitant RAS and RAGE inhibition on DAA. METHODS Diabetic (5 × 55 mg/kg streptozotocin daily) and non-diabetic male RAGE/apolipoprotein E double knockout (RAGE/apoE DKO) mice were treated with quinapril (30 mg/kg/day) for 20 weeks. At the end of the study aortic plaques were assessed. RESULTS Diabetic RAGE/apoE DKO showed significantly less plaque area than diabetic apoE KO mice. Plaque deposition was almost abolished in quinapril treated diabetic RAGE/apoE DKOs, with significant attenuation of vascular collagen deposition, nitrotyrosine staining, and reduced macrophage infiltration. Expression of the advanced glycation end product receptor 3 (galectin 3) was also significantly reduced. CONCLUSION Concomitant inhibition of RAS and RAGE signalling almost completely inhibited the development of experimental DAA. A dual therapeutic approach may be a superior strategy for the treatment of diabetic macrovascular disease..
Collapse
Affiliation(s)
- Anna M D Watson
- Diabetes Complications-Diabetes and The Kidney Laboratory, Baker IDI Heart and Diabetes Research Institute, Melbourne, Victoria, Australia.
| | - Jiaze Li
- Diabetes Complications-Diabetes and The Kidney Laboratory, Baker IDI Heart and Diabetes Research Institute, Melbourne, Victoria, Australia
| | - Dian Samijono
- Diabetes Complications-Diabetes and The Kidney Laboratory, Baker IDI Heart and Diabetes Research Institute, Melbourne, Victoria, Australia
| | - Angelika Bierhaus
- Department of Medicine and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
| | - Merlin C Thomas
- Diabetes Complications-Diabetes and The Kidney Laboratory, Baker IDI Heart and Diabetes Research Institute, Melbourne, Victoria, Australia; Department of Medicine, Central Clinical School, Monash University, Australia
| | - Karin A M Jandeleit-Dahm
- Diabetes Complications-Diabetes and The Kidney Laboratory, Baker IDI Heart and Diabetes Research Institute, Melbourne, Victoria, Australia; Department of Medicine, Central Clinical School, Monash University, Australia.
| | - Mark E Cooper
- Diabetes Complications-Diabetes and The Kidney Laboratory, Baker IDI Heart and Diabetes Research Institute, Melbourne, Victoria, Australia; Department of Medicine, Central Clinical School, Monash University, Australia
| |
Collapse
|
193
|
Prognostic Value of Stress Myocardial Perfusion Imaging in Asymptomatic Diabetic Patients. CURRENT CARDIOVASCULAR IMAGING REPORTS 2014. [DOI: 10.1007/s12410-014-9268-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
194
|
Yan L, Mathew L, Chellan B, Gardner B, Earley J, Puri TS, Hofmann Bowman MA. S100/Calgranulin-mediated inflammation accelerates left ventricular hypertrophy and aortic valve sclerosis in chronic kidney disease in a receptor for advanced glycation end products-dependent manner. Arterioscler Thromb Vasc Biol 2014; 34:1399-411. [PMID: 24855059 DOI: 10.1161/atvbaha.114.303508] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE S100A12 and fibroblast growth factor 23 are biomarkers of cardiovascular morbidity and mortality in patients with chronic kidney disease (CKD). We tested the hypothesis that human S100/calgranulin would accelerate cardiovascular disease in mice subjected to CKD. APPROACH AND RESULTS A bacterial artificial chromosome of the human S100/calgranulin gene cluster containing the genes and regulatory elements for S100A8, S100A9, and S100A12 was expressed in C57BL/6J mouse (hBAC-S100) to generate a novel humanized mouse model. CKD was induced by ureteral ligation, and hBAC-S100 mice and wild-type mice were studied after 10 weeks of chronic uremia. hBAC-S100 mice with CKD showed increased fibroblast growth factor 23 in the hearts, left ventricular hypertrophy, diastolic dysfunction, focal cartilaginous metaplasia, and calcification of the mitral and aortic valve annulus together with aortic valve sclerosis. This phenotype was not observed in wild-type mice with CKD or in hBAC-S100 mice lacking the receptor for advanced glycation end products with CKD, suggesting that the inflammatory milieu mediated by S100/receptor for advanced glycation end products promotes pathological cardiac hypertrophy in CKD. In vitro, inflammatory stimuli including interleukin-6, tumor necrosis factor-α, lipopolysaccarides, or serum from hBAC-S100 mice upregulated fibroblast growth factor 23 mRNA and protein in primary murine neonatal and adult cardiac fibroblasts. CONCLUSIONS Myeloid-derived human S100/calgranulin is associated with the development of cardiac hypertrophy and ectopic cardiac calcification in a receptor for advanced glycation end products-dependent manner in a mouse model of CKD. We speculate that fibroblast growth factor 23 produced by cardiac fibroblasts in response to cytokines may act in a paracrine manner to accelerate left ventricular hypertrophy and diastolic dysfunction in hBAC-S100 mice with CKD.
Collapse
Affiliation(s)
- Ling Yan
- From the Department of Medicine, Cardiology (L.Y., B.C., B.G., J.E., M.A.H.B.) and Medicine, Nephrology (L.M., T.S.P.), The University of Chicago, IL
| | - Liby Mathew
- From the Department of Medicine, Cardiology (L.Y., B.C., B.G., J.E., M.A.H.B.) and Medicine, Nephrology (L.M., T.S.P.), The University of Chicago, IL
| | - Bijoy Chellan
- From the Department of Medicine, Cardiology (L.Y., B.C., B.G., J.E., M.A.H.B.) and Medicine, Nephrology (L.M., T.S.P.), The University of Chicago, IL
| | - Brandon Gardner
- From the Department of Medicine, Cardiology (L.Y., B.C., B.G., J.E., M.A.H.B.) and Medicine, Nephrology (L.M., T.S.P.), The University of Chicago, IL
| | - Judy Earley
- From the Department of Medicine, Cardiology (L.Y., B.C., B.G., J.E., M.A.H.B.) and Medicine, Nephrology (L.M., T.S.P.), The University of Chicago, IL
| | - Tipu S Puri
- From the Department of Medicine, Cardiology (L.Y., B.C., B.G., J.E., M.A.H.B.) and Medicine, Nephrology (L.M., T.S.P.), The University of Chicago, IL
| | - Marion A Hofmann Bowman
- From the Department of Medicine, Cardiology (L.Y., B.C., B.G., J.E., M.A.H.B.) and Medicine, Nephrology (L.M., T.S.P.), The University of Chicago, IL.
| |
Collapse
|
195
|
Barmperis D, Bouki K, Apostolou T, Chalkias A, Xanthos T. Comparison of coronary calcification of the culprit lesion between diabetic and non-diabetic patients with acute coronary syndrome. Am J Emerg Med 2014; 32:480-2. [DOI: 10.1016/j.ajem.2014.01.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 01/30/2014] [Indexed: 11/24/2022] Open
|
196
|
Thomas MC. Glycemic exposure, glycemic control, and metabolic karma in diabetic complications. Adv Chronic Kidney Dis 2014; 21:311-7. [PMID: 24780460 DOI: 10.1053/j.ackd.2014.03.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 03/10/2014] [Indexed: 12/21/2022]
Abstract
Diabetes continues to cast a long shadow over the lives of many people. It is now clear that even transient hyper- or hypoglycemia or increased glycemic variability around healthy mean glucose levels can have long-lasting and long-term effects on the development and progression of diabetic complications, including cardiovascular disease, kidney disease, retinopathy, and neuropathy. Even after glycemic control has been achieved and maintained for many years, it appears hard to undo the changes that are instilled, including epigenetic programming, compositional changes, post-translational modifications, or simply lead time toward an inevitable fate. This phenomenon has become known as "metabolic memory" or the "legacy effect," but it may be better characterized as "metabolic karma," in which the intent and actions of an individual (with respect to metabolic control) influence the future health of that individual. This "bad karma" has been used to explain many clinical observations surrounding diabetes and its management, including the lack of benefits in many short- and intermediate-term trials, and the potential utility of early intensive glycemic control. Further understanding the molecular basis of a metabolic legacy in diabetes will certainly provide new targets for intervention.
Collapse
|
197
|
Tekabe Y, Kollaros M, Li Q, Zhang G, Li C, Schmidt AM, Johnson LL. Beneficial Effect of Glucose Control on Atherosclerosis Progression in Diabetic ApoE(-/-) Mice: Shown by Rage Directed Imaging. INTERNATIONAL JOURNAL OF MOLECULAR IMAGING 2014; 2014:695391. [PMID: 24829796 PMCID: PMC4009322 DOI: 10.1155/2014/695391] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 03/14/2014] [Indexed: 12/31/2022]
Abstract
Objective. Receptor for advanced glycated endproducts (RAGE) plays an important role in atherogenesis in diabetes. We imaged RAGE to investigate the effect of glucose control to suppress RAGE and reduce atherosclerosis in apolipoprotein E null (apoE(-/-)) diabetic mice. Methods and Results. Thirty-three apoE(-/-) mice received streptozotocin and 6 weeks later 15 began treatment with insulin implants. Blood glucose measurements during study averaged: 140 ± 23 mg/dL (treated) and 354 ± 14 mg/dL (untreated). After 15 wk 30 mice were injected with (99m)Tc-anti-RAGE F(ab')2, 3 with (99m)Tc-nonimmune IgG F(ab')2, and all with CT contrast agent and underwent SPECT/CT imaging. At necropsy, the proximal aorta was weighed, counted, and sectioned and the % injected dose per gram (%ID/g) was calculated. From the merged SPECT/CT scans, tracer uptake localized to arteries was lower in the treated mice: 3.15 ± 1.82 × 10(-3) versus 8.69 ± 4.58 × 10(-3)%ID (P = 0.001). Percent cross-sectional lesion area was smaller in the treated (14.3 ± 7.8% versus 29.5 ± 10.9%) (P = 0.03). RAGE uptake on scans (%ID) correlated with quantitative RAGE staining in the atheroma and with %ID/g (R = 0.6887; P = 0.01). Lesion size as percent cross-sectional area was smaller in the treated (14.3 ± 7.8% versus 29.5 ± 10.9%) (P = 0.03). RAGE uptake on scans (%ID) correlated with quantitative RAGE staining in the atheroma and with %ID/g (R = 0.6887; P = 0.01). Conclusions. These results support the importance of suppressing RAGE to reduce atherosclerotic complications of diabetes and value of molecular imaging to assess treatment effect.
Collapse
Affiliation(s)
- Yared Tekabe
- Department of Medicine, Columbia University Medical Center, 650 West 168 Street, New York, NY 10032, USA
| | - Maria Kollaros
- Department of Medicine, Columbia University Medical Center, 650 West 168 Street, New York, NY 10032, USA
| | - Qing Li
- Department of Medicine, Columbia University Medical Center, 650 West 168 Street, New York, NY 10032, USA
| | - Geping Zhang
- Department of Medicine, Columbia University Medical Center, 650 West 168 Street, New York, NY 10032, USA
| | - Chong Li
- Department of Medicine, Columbia University Medical Center, 650 West 168 Street, New York, NY 10032, USA
| | - Ann Marie Schmidt
- Department of Medicine, New York University Medical Center, New York, NY 10016, USA
| | - Lynne L. Johnson
- Department of Medicine, Columbia University Medical Center, 650 West 168 Street, New York, NY 10032, USA
| |
Collapse
|
198
|
Bornfeldt KE. 2013 Russell Ross memorial lecture in vascular biology: cellular and molecular mechanisms of diabetes mellitus-accelerated atherosclerosis. Arterioscler Thromb Vasc Biol 2014; 34:705-14. [PMID: 24665124 PMCID: PMC3967130 DOI: 10.1161/atvbaha.113.301928] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 11/22/2013] [Indexed: 01/12/2023]
Abstract
Adults with diabetes mellitus are much more likely to have cardiovascular disease than those without diabetes mellitus. Genetically engineered mouse models have started to provide important insight into the mechanisms whereby diabetes mellitus promotes atherosclerosis. Such models have demonstrated that diabetes mellitus promotes formation of atherosclerotic lesions, progression of lesions into advanced hemorrhaged lesions, and that it prevents lesion regression. The proatherosclerotic effects of diabetes mellitus are driven in part by the altered function of myeloid cells. The protein S100A9 and the receptor for advanced glycation end-products are important modulators of the effect of diabetes mellitus on myelopoiesis, which might promote monocyte accumulation in lesions. Furthermore, myeloid cell expression of the enzyme acyl-CoA synthetase 1 (ACSL1), which converts long-chain fatty acids into their acyl-CoA derivatives, has emerged as causal to diabetes mellitus-induced lesion initiation. The protective effects of myeloid ACSL1-deficiency in diabetic mice, but not in nondiabetic mice, indicate that myeloid cells are activated by diabetes mellitus through mechanisms that play minor roles in the absence of diabetes mellitus. The roles of reactive oxygen species and insulin resistance in diabetes mellitus-accelerated atherosclerosis are also discussed, primarily in relation to endothelial cells. Translational studies addressing whether the mechanisms identified in mouse models are equally important in humans with diabetes mellitus will be paramount.
Collapse
Affiliation(s)
- Karin E Bornfeldt
- From the Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, and Department of Pathology, Diabetes and Obesity Center of Excellence, University of Washington, Seattle, WA
| |
Collapse
|
199
|
Labonte AC, Tosello-Trampont AC, Hahn YS. The role of macrophage polarization in infectious and inflammatory diseases. Mol Cells 2014; 37:275-85. [PMID: 24625576 PMCID: PMC4012075 DOI: 10.14348/molcells.2014.2374] [Citation(s) in RCA: 272] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2013] [Accepted: 12/16/2013] [Indexed: 02/08/2023] Open
Abstract
Macrophages, found in circulating blood as well as integrated into several tissues and organs throughout the body, represent an important first line of defense against disease and a necessary component of healthy tissue homeostasis. Additionally, macrophages that arise from the differentiation of monocytes recruited from the blood to inflamed tissues play a central role in regulating local inflammation. Studies of macrophage activation in the last decade or so have revealed that these cells adopt a staggering range of phenotypes that are finely tuned responses to a variety of different stimuli, and that the resulting subsets of activated macrophages play critical roles in both progression and resolution of disease. This review summarizes the current understanding of the contributions of differentially polarized macrophages to various infectious and inflammatory diseases and the ongoing effort to develop novel therapies that target this key aspect of macrophage biology.
Collapse
Affiliation(s)
- Adam C. Labonte
- Department of Microbiology, Beirne B. Carter Center for Immunology Research, University of Virginia,
USA
| | | | - Young S. Hahn
- Department of Microbiology, Beirne B. Carter Center for Immunology Research, University of Virginia,
USA
| |
Collapse
|
200
|
Highly expressed S100A12 in aortic wall of patients with DeBakey type I aortic dissection could be a promising marker to predict perioperative complications. Ann Vasc Surg 2014; 28:1556-62. [PMID: 24691129 DOI: 10.1016/j.avsg.2014.03.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 02/12/2014] [Accepted: 03/21/2014] [Indexed: 11/22/2022]
Abstract
BACKGROUND Thoracic aortic dissection (TAD) is a catastrophic acute disease with a high postoperative mortality and few biochemical factors are known to predict outcomes. This study evaluated whether S100A12 could be a promising marker for TAD. METHODS A total of 72 patients with DeBakey Type I TAD and 18 heart donors as control group were studied. Immunohistochemistry of TAD tissue for S100A12 and hematoxylin-eosin staining, and alizarin red staining were examined. The expression of S100A12, proinflammatory protein specific for early recruited phagocytes, was studied by Western blotting of biopsies. In addition, S100A12 was further detected in serum samples from the same groups. RESULTS S100A12 was markedly expressed in the tissue of patients with TAD in comparison with healthy control subjects (48; 66.7% vs. 0%). Serum concentrations of S100A12 in patients with TAD were significantly higher than in healthy controls (27.5 ± 2.2 vs. 16.0 ± 1.9 μg/L; P < 0.001). The upward trend of serum was consistent with that of tissue. The length of hospitalization differed significantly among S100A12 immunohistochemical groups (P < 0.001). Increased S100A12 serum levels correlated significantly with postoperative stay in hospital (r = 0.457; P < 0.001). CONCLUSIONS Our findings suggest that an elevated S100A12 level could play a crucial role in systemic inflammation and may be a promising biomarker for predicting cardiovascular events and perioperative complications in patients with TAD.
Collapse
|