101
|
Expression of Nik-related kinase in smooth muscle cells attenuates vascular inflammation and intimal hyperplasia. Aging (Albany NY) 2020; 12:7511-7533. [PMID: 32330120 PMCID: PMC7202544 DOI: 10.18632/aging.103104] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 03/02/2020] [Indexed: 01/18/2023]
Abstract
Inflammation of the vascular microenvironment modulates distinct types of vascular cells, and plays important roles in promoting atherosclerosis, stenosis/restenosis, and vascular-related diseases. Nik-related kinase (Nrk), a member of the Ste20-type kinase family, has been reported to be selectively expressed in embryonic skeletal muscle. However, whether Nrk is expressed in adult vascular smooth muscle, and if it influences intimal hyperplasia is unclear. Here, we found that Nrk is abundantly expressed in cultured vascular smooth muscle cells (VSMC) and mouse arterial intima. Treatment of mouse VSMCs with lipopolysaccharide (LPS) or platelet-derived growth factor significantly reduced Nrk expression. In addition, expression of Nrk was significantly reduced in regions of neointimal formation caused by guide-wire carotid artery injuries in mice, as well as in human atherosclerotic tissues, when compared to normal vessels. We identified that expression of matrix metalloproteinases (MMP3, MMP8 and MMP12) and inflammatory cytokines/chemokines (CCL6, CCL8, CCL11, CXCL1, CXCL3, CXCL5 and CXCL9) are synergistically induced by Nrk siRNA in LPS-treated mouse VSMCs. Moreover, we found that resveratrol significantly impaired LPS- and Nrk siRNA-induced expression of MMP3, CCL8, CCL11, CXCL3 and CXCL5. These results suggested that Nrk may play important roles in regulating pathological progression of atherosclerosis or neointimal- hyperplasia-related vascular diseases.
Collapse
|
102
|
Nicklas JM, Gordon AE, Henke PK. Resolution of Deep Venous Thrombosis: Proposed Immune Paradigms. Int J Mol Sci 2020; 21:E2080. [PMID: 32197363 PMCID: PMC7139924 DOI: 10.3390/ijms21062080] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/14/2020] [Accepted: 03/15/2020] [Indexed: 12/12/2022] Open
Abstract
Venous thromboembolism (VTE) is a pathology encompassing deep vein thrombosis (DVT) and pulmonary embolism (PE) associated with high morbidity and mortality. Because patients often present after a thrombus has already formed, the mechanisms that drive DVT resolution are being investigated in search of treatment. Herein, we review the current literature, including the molecular mechanisms of fibrinolysis and collagenolysis, as well as the critical cellular roles of macrophages, neutrophils, and endothelial cells. We propose two general models for the operation of the immune system in the context of venous thrombosis. In early thrombus resolution, neutrophil influx stabilizes the tissue through NETosis. Meanwhile, macrophages and intact neutrophils recognize the extracellular DNA by the TLR9 receptor and induce fibrosis, a complimentary stabilization method. At later stages of resolution, pro-inflammatory macrophages police the thrombus for pathogens, a role supported by both T-cells and mast cells. Once they verify sterility, these macrophages transform into their pro-resolving phenotype. Endothelial cells both coat the stabilized thrombus, a necessary early step, and can undergo an endothelial-mesenchymal transition, which impedes DVT resolution. Several of these interactions hold promise for future therapy.
Collapse
Affiliation(s)
| | | | - Peter K. Henke
- School of Medicine, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA; (J.M.N.); (A.E.G.)
| |
Collapse
|
103
|
Mo H, Fu C, Wu Z, Liu P, Wen Z, Hong Q, Cai Y, Li G. IL-6-targeted ultrasmall superparamagnetic iron oxide nanoparticles for optimized MRI detection of atherosclerotic vulnerable plaques in rabbits. RSC Adv 2020; 10:15346-15353. [PMID: 35495447 PMCID: PMC9052309 DOI: 10.1039/c9ra10509c] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 04/04/2020] [Indexed: 12/30/2022] Open
Abstract
Vulnerable plaques of atherosclerosis (AS) are the main culprit lesion for the serious risk of acute cardiovascular disease (CVD). Therefore, developing new non-invasive methods to detect vulnerable plaques and to evaluate their stability effectively is of great value in the early diagnosis of CVD. IL-6 plays a vital role in the development and rupture of AS. In this study, IL-6-targeted superparamagnetic iron oxide nanoparticles (Anti-IL-6-USPIO) are synthesized by a chemical condensation reaction. An AS model was established by damaging rabbit abdominal aortic intima with Foley's tube in combination with a high cholesterol diet. The results confirm that Anti-IL-6-USPIO have excellent IL-6-targeting ability and usefulness in detecting vulnerable plaques in vitro and in vivo, which may provide a novel, non-invasive strategy for evaluating acute cardiovascular risk or exploiting anti-atherosclerotic drugs. Herein, we report Anti-IL-6-USPIO for detecting IL-6 in inflammatory macrophages and MR imaging vulnerable plaques of atherosclerosis in rabbit, which would provide a novel non-invasive strategy for evaluating acute cardiovascular risk or exploiting anti-atherosclerotic drugs.![]()
Collapse
Affiliation(s)
- Huaqiang Mo
- Department of Cardiology
- Zhujiang Hospital
- Southern Medical University
- Guangzhou 510280
- People's Republic of China
| | - Chenxing Fu
- Department of Cardiology
- Zhujiang Hospital
- Southern Medical University
- Guangzhou 510280
- People's Republic of China
| | - Zhiye Wu
- Department of Cardiology
- Zhujiang Hospital
- Southern Medical University
- Guangzhou 510280
- People's Republic of China
| | - Peng Liu
- Department of Cardiology
- Zhujiang Hospital
- Southern Medical University
- Guangzhou 510280
- People's Republic of China
| | - Zhibo Wen
- Department of Radiology
- Zhujiang Hospital
- Southern Medical University
- Guangzhou 510280
- People's Republic of China
| | - Qingqing Hong
- Department of Cardiology
- Zhujiang Hospital
- Southern Medical University
- Guangzhou 510280
- People's Republic of China
| | - Yanbin Cai
- Department of Cardiology
- Zhujiang Hospital
- Southern Medical University
- Guangzhou 510280
- People's Republic of China
| | - Gongxin Li
- Department of Cardiology
- Zhujiang Hospital
- Southern Medical University
- Guangzhou 510280
- People's Republic of China
| |
Collapse
|
104
|
Lordan R, Redfern S, Tsoupras A, Zabetakis I. Inflammation and cardiovascular disease: are marine phospholipids the answer? Food Funct 2020; 11:2861-2885. [DOI: 10.1039/c9fo01742a] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
This review presents the latest research on the cardioprotective effects of n-3 fatty acids (FA) and n-3 FA bound to polar lipids (PL). Overall, n-3 PL may have enhanced bioavailability and potentially bioactivityversusfree FA and ester forms of n-3 FA.
Collapse
Affiliation(s)
- Ronan Lordan
- Department of Biological Sciences
- University of Limerick
- Limerick
- Ireland
- Health Research Institute (HRI)
| | - Shane Redfern
- Department of Biological Sciences
- University of Limerick
- Limerick
- Ireland
| | - Alexandros Tsoupras
- Department of Biological Sciences
- University of Limerick
- Limerick
- Ireland
- Health Research Institute (HRI)
| | - Ioannis Zabetakis
- Department of Biological Sciences
- University of Limerick
- Limerick
- Ireland
- Health Research Institute (HRI)
| |
Collapse
|
105
|
Lunde NN, Gregersen I, Ueland T, Shetelig C, Holm S, Kong XY, Michelsen AE, Otterdal K, Yndestad A, Broch K, Gullestad L, Nyman TA, Bendz B, Eritsland J, Hoffmann P, Skagen K, Gonçalves I, Nilsson J, Grenegård M, Poreba M, Drag M, Seljeflot I, Sporsheim B, Espevik T, Skjelland M, Johansen HT, Solberg R, Aukrust P, Björkbacka H, Andersen GØ, Halvorsen B. Legumain is upregulated in acute cardiovascular events and associated with improved outcome - potentially related to anti-inflammatory effects on macrophages. Atherosclerosis 2019; 296:74-82. [PMID: 31870625 DOI: 10.1016/j.atherosclerosis.2019.12.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 11/20/2019] [Accepted: 12/12/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND AIMS We have previously found increased levels of the cysteine protease legumain in plasma and plaques from patients with carotid atherosclerosis. This study further investigated legumain during acute cardiovascular events. METHODS Circulating levels of legumain from patients and legumain released from platelets were assessed by enzyme-linked-immunosorbent assay. Quantitative PCR and immunoblotting were used to study expression, while localization was visualized by immunohistochemistry. RESULTS In the SUMMIT Malmö cohort (n = 339 with or without type 2 diabetes and/or cardiovascular disease [CVD], and 64 healthy controls), the levels of circulating legumain were associated with the presence of CVD in non-diabetics, with no relation to outcome. In symptomatic carotid plaques and in samples from both coronary and intracerebral thrombi obtained during acute cardiovascular events, legumain was co-localized with macrophages in the same regions as platelets. In vitro, legumain was shown to be present in and released from platelets upon activation. In addition, THP-1 macrophages exposed to releasate from activated platelets showed increased legumain expression. Interestingly, primary peripheral blood mononuclear cells stimulated with recombinant legumain promoted anti-inflammatory responses. Finally, in a STEMI population (POSTEMI; n = 272), patients had significantly higher circulating legumain before and immediately after percutaneous coronary intervention compared with healthy controls (n = 67), and high levels were associated with improved outcome. CONCLUSIONS Our data demonstrate for the first time that legumain is upregulated during acute cardiovascular events and is associated with improved outcome.
Collapse
Affiliation(s)
- Ngoc Nguyen Lunde
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway.
| | - Ida Gregersen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway
| | - Christian Shetelig
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway; Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Sverre Holm
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Xiang Yi Kong
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Annika E Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kari Otterdal
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Arne Yndestad
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kaspar Broch
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway; KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Lars Gullestad
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway; KG Jebsen Center for Cardiac Research, University of Oslo, Oslo, Norway and Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Tuula A Nyman
- Proteomics Core Facility, Department of Immunology, Institute of Clinical Medicine, University of Oslo and Rikshospitalet Oslo, Oslo, Norway
| | - Bjørn Bendz
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Jan Eritsland
- Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Pavel Hoffmann
- Section of Interventional Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Karolina Skagen
- Department of Neurology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Isabel Gonçalves
- Experimental Cardiovascular Research Unit, Dept. of Clinical Sciences, Malmö Lund University, Malmö, Sweden; Department of Cardiology, Skåne University Hospital, Sweden
| | - Jan Nilsson
- Experimental Cardiovascular Research Unit, Dept. of Clinical Sciences, Malmö Lund University, Malmö, Sweden
| | | | - Marcin Poreba
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wroclaw University of Technology, Wroclaw, Poland
| | - Marcin Drag
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wroclaw University of Technology, Wroclaw, Poland
| | - Ingebjørg Seljeflot
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway; Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Bjørnar Sporsheim
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Terje Espevik
- Centre of Molecular Inflammation Research, Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Mona Skjelland
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Neurology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Harald Thidemann Johansen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Rigmor Solberg
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway; Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Harry Björkbacka
- Experimental Cardiovascular Research Unit, Dept. of Clinical Sciences, Malmö Lund University, Malmö, Sweden
| | - Geir Øystein Andersen
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway; Department of Cardiology, Oslo University Hospital Ullevål, Oslo, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
106
|
Abstract
Aortic aneurysms are a common vascular disease in Western populations that can involve virtually any portion of the aorta. Abdominal aortic aneurysms are much more common than thoracic aortic aneurysms and combined they account for >25 000 deaths in the United States annually. Although thoracic and abdominal aortic aneurysms share some common characteristics, including the gross anatomic appearance, alterations in extracellular matrix, and loss of smooth muscle cells, they are distinct diseases. In recent years, advances in genetic analysis, robust molecular tools, and increased availability of animal models have greatly enhanced our knowledge of the pathophysiology of aortic aneurysms. This review examines the various proposed cellular mechanisms responsible for aortic aneurysm formation and identifies opportunities for future studies.
Collapse
Affiliation(s)
- Raymundo Alain Quintana
- From the Division of Cardiology, Department of Medicine (R.A.Q., W.R.T.), Emory University School of Medicine, Atlanta, GA
| | - W Robert Taylor
- From the Division of Cardiology, Department of Medicine (R.A.Q., W.R.T.), Emory University School of Medicine, Atlanta, GA.,Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology (W.R.T.), Emory University School of Medicine, Atlanta, GA.,Division of Cardiology, Atlanta VA Medical Center, Decatur, GA (W.R.T.)
| |
Collapse
|
107
|
Holm Nielsen S, Tengryd C, Edsfeldt A, Brix S, Genovese F, Bengtsson E, Karsdal M, Leeming DJ, Nilsson J, Goncalves I. Markers of Basement Membrane Remodeling Are Associated With Higher Mortality in Patients With Known Atherosclerosis. J Am Heart Assoc 2019; 7:e009193. [PMID: 30608207 PMCID: PMC6404182 DOI: 10.1161/jaha.118.009193] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Patients with atherosclerosis have a high risk of cardiovascular events and death. Atherosclerosis is characterized by accumulation of lipids, cells and extracellular matrix proteins in the intima. We hypothesized that dysregulated remodeling of the basement membrane proteins may be associated with clinical outcomes in patients with atherosclerosis. Methods and Results Neoepitope fragments of collagen type IV (C4M) and laminin ( LG 1M) were assessed by ELISA s in serum from 787 endarterectomy patients. Matrix metalloproteinase s were measured using proximity extension assay and correlated to C4M and LG 1M levels using Spearman correlations. A total of 473 patients were followed up for 6 years using national registers, medical charts, and telephone interviews. The incidence of cardiovascular events, cardiovascular mortality, and all-cause mortality were associated to levels of C4M and LG 1M using Kaplan-Meier curves and Cox regression analyses. A total of 101 patients had cardiovascular events, 39 died of cardiovascular mortality, and 64 patients died from all-cause mortality. C4M levels were increased in patients with symptomatic carotid atherosclerotic disease before surgery ( P=0.048). High C4M and LG 1M levels were associated with increased risk of all-cause mortality ( P=0.020 and 0.031, respectively) and predicted all-cause death together with glomerular filtration rate and diabetes mellitus. Conclusions High LG 1M and C4M levels were associated with all-cause mortality, together with glomerular filtration rate and diabetes mellitus. These novel biomarkers need further evaluation but might be tools to identify high-risk patients.
Collapse
Affiliation(s)
- Signe Holm Nielsen
- 1 Nordic Bioscience, Biomarkers and Research Herlev Denmark.,2 Disease Systems Immunology Department of Biotechnology and Biomedicine Technical University of Denmark Kgs. Lyngby Denmark
| | - Christoffer Tengryd
- 3 Experimental Cardiovascular Research Unit Department of Clinical Sciences, Malmö Lund University Malmö Sweden
| | - Andreas Edsfeldt
- 3 Experimental Cardiovascular Research Unit Department of Clinical Sciences, Malmö Lund University Malmö Sweden.,4 Department of Cardiology Skåne University Hospital Malmö Sweden
| | - Susanne Brix
- 2 Disease Systems Immunology Department of Biotechnology and Biomedicine Technical University of Denmark Kgs. Lyngby Denmark
| | | | - Eva Bengtsson
- 3 Experimental Cardiovascular Research Unit Department of Clinical Sciences, Malmö Lund University Malmö Sweden
| | - Morten Karsdal
- 1 Nordic Bioscience, Biomarkers and Research Herlev Denmark
| | | | - Jan Nilsson
- 3 Experimental Cardiovascular Research Unit Department of Clinical Sciences, Malmö Lund University Malmö Sweden
| | - Isabel Goncalves
- 3 Experimental Cardiovascular Research Unit Department of Clinical Sciences, Malmö Lund University Malmö Sweden.,4 Department of Cardiology Skåne University Hospital Malmö Sweden
| |
Collapse
|
108
|
Lunde NN, Bosnjak T, Solberg R, Johansen HT. Mammalian legumain – A lysosomal cysteine protease with extracellular functions? Biochimie 2019; 166:77-83. [DOI: 10.1016/j.biochi.2019.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 06/04/2019] [Indexed: 12/31/2022]
|
109
|
Zhao Y, Wang Z, Zhang W, Zhang L. MicroRNAs play an essential role in autophagy regulation in various disease phenotypes. Biofactors 2019; 45:844-856. [PMID: 31418958 PMCID: PMC6916288 DOI: 10.1002/biof.1555] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 07/31/2019] [Indexed: 12/16/2022]
Abstract
Autophagy is a highly conserved catabolic process and fundamental biological process in eukaryotic cells. It recycles intracellular components to provide nutrients during starvation and maintains quality control of organelles and proteins. In addition, autophagy is a well-organized homeostatic cellular process that is responsible for the removal of damaged organelles and intracellular pathogens. Moreover, it also modulates the innate and adaptive immune systems. Micro ribonucleic acids (microRNAs) are a mature class of post-transcriptional modulators that are widely expressed in tissues and organs. And, it can suppress gene expression by targeting messenger RNAs for translational repression or, at a lesser extent, degradation. Research indicates that microRNAs regulate autophagy through different pathways, playing an essential role in the treatment of various diseases. It is an important regulator of fundamental cellular processes such as proliferation, autophagy, and cell apoptosis. In this review article, we first review the current knowledge of autophagy and the function of microRNAs. Then, we summarize the mechanism of autophagy and the signaling pathways related to autophagy by citing at least the main proteins involved in the different phases of the process. Second, we introduce other members of RNA and report some examples in various pathologies. Finally, we review the current literature regarding microRNA-based therapies for cancer, atherosclerosis, cardiac disease, tuberculosis, and viral diseases. MicroRNAs can cause autophagy upregulation or downregulation by targeting genes or affecting autophagy-related signaling pathways. Therefore, the microRNAs have a huge potential in autophagy regulation, and it is the function as diagnostic and prognostic markers.
Collapse
Affiliation(s)
- Yunyi Zhao
- Laboratory of Pathogenic Microbiology and ImmunologyCollege of Life Science, Jilin Agricultural UniversityChangchunChina
| | - Ze Wang
- Laboratory of Pathogenic Microbiology and ImmunologyCollege of Life Science, Jilin Agricultural UniversityChangchunChina
| | - Wenhui Zhang
- Laboratory of Pathogenic Microbiology and ImmunologyCollege of Life Science, Jilin Agricultural UniversityChangchunChina
- Ministry of Education, Engineering Research Center for Bioreactor and Pharmaceutical DevelopmentJilin Agricultural UniversityChangchunChina
| | - Linbo Zhang
- Laboratory of Pathogenic Microbiology and ImmunologyCollege of Life Science, Jilin Agricultural UniversityChangchunChina
- Ministry of Education, Engineering Research Center for Bioreactor and Pharmaceutical DevelopmentJilin Agricultural UniversityChangchunChina
| |
Collapse
|
110
|
MicroRNA-21 deficiency attenuated atherogenesis and decreased macrophage infiltration by targeting Dusp-8. Atherosclerosis 2019; 291:78-86. [PMID: 31704554 DOI: 10.1016/j.atherosclerosis.2019.10.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 09/10/2019] [Accepted: 10/09/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS Atherosclerosis is a chronic inflammatory disorder mediated by macrophage activation. MicroRNA-21 (miR-21) is a key regulator in the macrophage inflammatory response. However, the functional role of miR-21 in atherogenesis is far from clear. METHODS AND RESULTS Here, we report that miR-21 is significantly upregulated in mouse atherosclerotic plaques and peripheral monocytes from patients with coronary artery disease. Compared with miR-21+/+apoE-/- mice (apoE-/- mice), miR-21-/-apoE-/- (double knockout, DKO) mice showed less atherosclerotic lesions, reduced presence of macrophages, decreased smooth muscle cells(SMC) and collagen content in the aorta. We further explored the role of miR-21 in macrophage activation in vitro. Bone marrow-derived macrophages (BMDMs) from DKO mice not only exhibit impaired function of migration induced by chemokine (C-C motif) ligand 2 (CCL2) but also a weakened macrophage-endothelium interaction activated by tumor necrosis factor-α (TNF-α). However, atherogenic inflammatory cytokine secretion was not affected by miR-21 in vitro or in vivo. Additionally, miR-21 knockdown in BMDMs directly derepressed the expression of dual specificity protein phosphatase 8 (Dusp-8), a previously validated miR-21 target in cardiac fibroblasts, which negatively regulates mitogen-activated protein kinase (MAPK) signaling, particularly the p38-and c-Jun N-terminal kinase (JNK)-related signaling pathways. CONCLUSIONS These data demonstrate that inhibition of miR-21 may restrict the formation of atherosclerotic plaques partly by regulating macrophage migration and adhesion, while, reduced SMCs and collagen content in plaques may lead to a less stable phenotype with the progression of atherosclerosis. Thus, the absence of miR-21 reduces atherosclerotic lesions but may not represent all benefit in atherosclerosis development.
Collapse
|
111
|
Harman JL, Jørgensen HF. The role of smooth muscle cells in plaque stability: Therapeutic targeting potential. Br J Pharmacol 2019; 176:3741-3753. [PMID: 31254285 PMCID: PMC6780045 DOI: 10.1111/bph.14779] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/13/2019] [Accepted: 06/17/2019] [Indexed: 02/02/2023] Open
Abstract
Events responsible for cardiovascular mortality and morbidity are predominantly caused by rupture of "vulnerable" atherosclerotic lesions. Vascular smooth muscle cells (VSMCs) play a key role in atherogenesis and have historically been considered beneficial for plaque stability. VSMCs constitute the main cellular component of the protective fibrous cap within lesions and are responsible for synthesising strength-giving extracellular matrix components. However, lineage-tracing experiments in mouse models of atherosclerosis have shown that, in addition to the fibrous cap, VSMCs also give rise to many of the cell types found within the plaque core. In particular, VSMCs generate a substantial fraction of lipid-laden foam cells, and VSMC-derived cells expressing markers of macrophages, osteochondrocyte, and mesenchymal stem cells have been observed within lesions. Here, we review recent studies that have changed our perspective on VSMC function in atherosclerosis and discuss how VSMCs could be targeted to increase plaque stability.
Collapse
|
112
|
Resveratrol Inhibits MMP3 and MMP9 Expression and Secretion by Suppressing TLR4/NF- κB/STAT3 Activation in Ox-LDL-Treated HUVECs. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9013169. [PMID: 31583048 PMCID: PMC6754947 DOI: 10.1155/2019/9013169] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/09/2019] [Accepted: 06/19/2019] [Indexed: 12/22/2022]
Abstract
Aim Resveratrol is a natural plant polyphenol. The present study investigated the effects of resveratrol on the Toll-like receptor 4- (TLR4-) mediated expression and secretion of matrix metalloproteinases (MMPs) in oxidized low-density lipoprotein- (ox-LDL-) treated human umbilical vein endothelial cells (HUVECs). Methods Protein expression was analyzed by immunoblotting. The secretion of MMPs was measured by an enzyme-linked immunosorbent assay. The animal experiments were performed with and without resveratrol treatment in high-fat chow-fed mice. Results Resveratrol inhibited the expression of TLR4, MMP3, and MMP9 in ox-LDL- and lipopolysaccharide- (LPS-) treated HUVECs. Resveratrol reduced the secretion of MMP3 and MMP9 that was induced by ox-LDL and LPS. The TLR4 inhibitor CLI-095 similarly suppressed the expression and secretion of MMP3 and MMP9 in ox-LDL- and LPS-treated HUVECs. Resveratrol attenuated the phosphorylation of the transcription factors nuclear factor-κB (NF-κB) and signal transducer and activator of transcription 3 (STAT3) that was induced by ox-LDL and LPS. Resveratrol recovered Sirt1 expression. In the animal experiments, resveratrol decreased TLR4 expression in the aorta, MMP9 levels in plasma, and vascular structural changes in high-fat chow-fed mice, with no significant effect on plasma MMP3 levels. Conclusion Resveratrol inhibited the TLR4-mediated expression and secretion of MMP3 and MMP9 in ox-LDL-treated HUVECs. The mechanism of action of resveratrol may be associated with the suppression of NF-κB and STAT3 phosphorylation and restoration of Sirt1 expression. Resveratrol exerts protective effects against vascular structural changes in high-fat chow-fed mice.
Collapse
|
113
|
Tokuhara CK, Santesso MR, Oliveira GSND, Ventura TMDS, Doyama JT, Zambuzzi WF, Oliveira RCD. Updating the role of matrix metalloproteinases in mineralized tissue and related diseases. J Appl Oral Sci 2019; 27:e20180596. [PMID: 31508793 DOI: 10.1590/1678-7757-2018-0596] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 05/30/2019] [Indexed: 02/22/2023] Open
Abstract
Bone development and healing processes involve a complex cascade of biological events requiring well-orchestrated synergism with bone cells, growth factors, and other trophic signaling molecules and cellular structures. Beyond health processes, MMPs play several key roles in the installation of heart and blood vessel related diseases and cancer, ranging from accelerating metastatic cells to ectopic vascular mineralization by smooth muscle cells in complementary manner. The tissue inhibitors of MMPs (TIMPs) have an important role in controlling proteolysis. Paired with the post-transcriptional efficiency of specific miRNAs, they modulate MMP performance. If druggable, these molecules are suggested to be a platform for development of "smart" medications and further clinical trials. Thus, considering the pleiotropic effect of MMPs on mammals, the purpose of this review is to update the role of those multifaceted proteases in mineralized tissues in health, such as bone, and pathophysiological disorders, such as ectopic vascular calcification and cancer.
Collapse
Affiliation(s)
- Cintia Kazuko Tokuhara
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Bauru, São Paulo, Brasil
| | - Mariana Rodrigues Santesso
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Bauru, São Paulo, Brasil
| | - Gabriela Silva Neubern de Oliveira
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Bauru, São Paulo, Brasil
| | - Talita Mendes da Silva Ventura
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Bauru, São Paulo, Brasil
| | - Julio Toshimi Doyama
- Universidade Estadual Paulista Júlio de Mesquita Filho, Campus Botucatu, Rubião Jr, São Paulo, Brasil
| | - Willian Fernando Zambuzzi
- Universidade Estadual Paulista Júlio de Mesquita Filho, Campus Botucatu, Rubião Jr, São Paulo, Brasil
| | - Rodrigo Cardoso de Oliveira
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Bauru, São Paulo, Brasil
| |
Collapse
|
114
|
Immune-Mediated Inflammation in Vulnerable Atherosclerotic Plaques. Molecules 2019; 24:molecules24173072. [PMID: 31450823 PMCID: PMC6749340 DOI: 10.3390/molecules24173072] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/21/2019] [Accepted: 08/22/2019] [Indexed: 01/16/2023] Open
Abstract
Atherosclerosis is a chronic long-lasting vascular disease leading to myocardial infarction and stroke. Vulnerable atherosclerotic (AS) plaques are responsible for these life-threatening clinical endpoints. To more successfully work against atherosclerosis, improvements in early diagnosis and treatment of AS plaque lesions are required. Vulnerable AS plaques are frequently undetectable by conventional imaging because they are non-stenotic. Although blood biomarkers like lipids, C-reactive protein, interleukin-6, troponins, and natriuretic peptides are in pathological ranges, these markers are insufficient in detecting the critical perpetuation of AS anteceding endpoints. Thus, chances to treat the patient in a preventive way are wasted. It is now time to solve this dilemma because clear results indicate a benefit of anti-inflammatory therapy per se without modification of blood lipids (CANTOS Trial, NCT01327846). This fact identifies modulation of immune-mediated inflammation as a new promising point of action for the eradication of fatal atherosclerotic endpoints.
Collapse
|
115
|
Molecular Imaging Probes Based on Matrix Metalloproteinase Inhibitors (MMPIs). Molecules 2019; 24:molecules24162982. [PMID: 31426440 PMCID: PMC6719134 DOI: 10.3390/molecules24162982] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/12/2019] [Accepted: 08/13/2019] [Indexed: 12/12/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc- and calcium-dependent endopeptidases which are secreted or anchored in the cell membrane and are capable of degrading the multiple components of the extracellular matrix (ECM). MMPs are frequently overexpressed or highly activated in numerous human diseases. Owing to the important role of MMPs in human diseases, many MMP inhibitors (MMPIs) have been developed as novel therapeutics, and some of them have entered clinical trials. However, so far, only one MMPI (doxycycline) has been approved by the FDA. Therefore, the evaluation of the activity of a specific subset of MMPs in human diseases using clinically relevant imaging techniques would be a powerful tool for the early diagnosis and assessment of the efficacy of therapy. In recent years, numerous MMPIs labeled imaging agents have emerged. This article begins by providing an overview of the MMP subfamily and its structure and function. The latest advances in the design of subtype selective MMPIs and their biological evaluation are then summarized. Subsequently, the potential use of MMPI-labeled diagnostic agents in clinical imaging techniques are discussed, including positron emission tomography (PET), single-photon emission computed tomography (SPECT) and optical imaging (OI). Finally, this article concludes with future perspectives and clinical utility.
Collapse
|
116
|
YKL-40 promotes the progress of atherosclerosis independent of lipid metabolism in apolipoprotein E -/- mice fed a high-fat diet. Heart Vessels 2019; 34:1874-1881. [PMID: 31114961 DOI: 10.1007/s00380-019-01434-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 05/15/2019] [Indexed: 02/07/2023]
Abstract
YKL-40 is recently regarded as a pro-inflammatory cytokine involved in the pathological process of atherosclerosis and lipid metabolism. However, whether YKL-40 can directly influence the development of atherosclerosis and levels of lipid parameters is unknown. The aim of this study is to explore the effects of YKL-40 on atherosclerotic features, the levels of serum lipids, and biomarkers in apolipoprotein (E)-deficient (ApoE-/-) mice fed a high-fat diet. ApoE-/- mice were injected with a recombinant adenovirus expressing mouse YKL-40 or control adenovirus through the caudal vein. The levels of serum YKL-40, interleukin-6 (IL-6), tumour necrosis factor-alpha (TNF-alpha), matrix metalloproteinase-9 (MMP-9), and soluble vascular cell-adhesion molecule 1 (sVCAM-1) were measured by ELISA. Lipid metabolism parameters were measured using immunoturbidimetric assay. The size of plaque area in aorta was evaluated by Oil Red O and hematoxylin/eosin (HE) staining. The content of collagen fibers was stained with Masson, and the content of macrophages and smooth muscle cells (SMCs) in atherosclerotic lesions was investigated by immunohistochemistry. The serum levels of total cholesterol and triglycerides were similar between these two groups. Compared with the control, the levels of serum YKL-40, IL-6, TNF-alpha, MMP-9, plaque size, and macrophages in plaques were significantly increased in mice with adenovirus overexpressing YKL-40. However, the content of collagen fibers and SMCs was remarkably decreased in mice with adenovirus overexpressing YKL-40 than that in control. YKL-40 prompts the progress of atherosclerosis maybe involved with its role of pro-inflammation, but does not affect lipid metabolism in ApoE-/- mice fed a high-fat diet.
Collapse
|
117
|
Rababa’h AM, Bsoul RW, Alkhatatbeh MJ, Alzoubi KH, Khabour OF. Waterpipe tobacco smoke distresses cardiovascular biomarkers in mice: alterations in protein expression of metalloproteinases, endothelin and myeloperoxidase. Inhal Toxicol 2019; 31:99-106. [DOI: 10.1080/08958378.2019.1606366] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Abeer M. Rababa’h
- Department of Clinical Pharmacy Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Raghad W. Bsoul
- Department of Clinical Pharmacy Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Mohammad J. Alkhatatbeh
- Department of Clinical Pharmacy Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Karem H. Alzoubi
- Department of Clinical Pharmacy Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Omar F. Khabour
- Department of Medical Laboratory Sciences Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
118
|
Wahart A, Hocine T, Albrecht C, Henry A, Sarazin T, Martiny L, El Btaouri H, Maurice P, Bennasroune A, Romier-Crouzet B, Blaise S, Duca L. Role of elastin peptides and elastin receptor complex in metabolic and cardiovascular diseases. FEBS J 2019; 286:2980-2993. [PMID: 30946528 DOI: 10.1111/febs.14836] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/23/2019] [Accepted: 04/02/2019] [Indexed: 12/11/2022]
Abstract
The Cardiovascular Continuum describes a sequence of events from cardiovascular risk factors to end-stage heart disease. It includes conventional pathologies affecting cardiovascular functions such as hypertension, atherosclerosis or thrombosis and was traditionally considered from the metabolic point of view. This Cardiovascular Continuum, originally described by Dzau and Braunwald, was extended by O'Rourke to consider also the crucial role played by degradation of elastic fibers, occurring during aging, in the appearance of vascular stiffness, another deleterious risk factor of the continuum. However, the involvement of the elastin degradation products, named elastin-derived peptides, to the Cardiovascular Continuum progression has not been considered before. Data from our laboratory and others clearly showed that these bioactive peptides are central regulators of this continuum, thereby amplifying appearance and evolution of cardiovascular risk factors such as diabetes or hypertension, of vascular alterations such as atherothrombosis and calcification, but also nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. The Elastin Receptor Complex has been shown to be a crucial actor in these processes. We propose here the participation of these elastin-derived peptides and of the Elastin Receptor Complex in these events, and introduce a revisited Cardiovascular Continuum based on their involvement, for which elastin-based pharmacological strategies could have a strong impact in the future.
Collapse
Affiliation(s)
- Amandine Wahart
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, France
| | - Thinhinane Hocine
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, France
| | - Camille Albrecht
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, France
| | - Auberi Henry
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, France
| | - Thomas Sarazin
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, France
| | - Laurent Martiny
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, France
| | - Hassan El Btaouri
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, France
| | - Pascal Maurice
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, France
| | - Amar Bennasroune
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, France
| | | | - Sébastien Blaise
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, France
| | - Laurent Duca
- UMR CNRS 7369 MEDyC, SFR CAP-Santé, Université de Reims Champagne-Ardenne, France
| |
Collapse
|
119
|
Watanabe Y, Hirao Y, Kasuga K, Tokutake T, Semizu Y, Kitamura K, Ikeuchi T, Nakamura K, Yamamoto T. Molecular Network Analysis of the Urinary Proteome of Alzheimer's Disease Patients. Dement Geriatr Cogn Dis Extra 2019; 9:53-65. [PMID: 31043964 PMCID: PMC6477484 DOI: 10.1159/000496100] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/07/2018] [Indexed: 12/27/2022] Open
Abstract
Background/Aims The identification of predictive biomarkers for Alzheimer's disease (AD) from urine would aid in screening for the disease, but information about biological and pathophysiological changes in the urine of AD patients is limited. This study aimed to explore the comprehensive profile and molecular network relations of urinary proteins in AD patients. Methods Urine samples collected from 18 AD patients and 18 age- and sex-matched cognitively normal controls were analyzed by mass spectrometry and semiquantified with the normalized spectral index method. Bioinformatics analyses were performed on proteins which significantly increased by more than 2-fold or decreased by less than 0.5-fold compared to the control (p < 0.05) using DAVID bioinformatics resources and KeyMolnet software. Results The levels of 109 proteins significantly differed between AD patients and controls. Among these, annotation clusters related to lysosomes, complement activation, and gluconeogenesis were significantly enriched. The molecular relation networks derived from these proteins were mainly associated with pathways of lipoprotein metabolism, heat shock protein 90 signaling, matrix metalloproteinase signaling, and redox regulation by thioredoxin. Conclusion Our findings suggest that changes in the urinary proteome of AD patients reflect systemic changes related to AD pathophysiology.
Collapse
Affiliation(s)
- Yumi Watanabe
- Division of Preventive Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | | | - Kensaku Kasuga
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takayoshi Tokutake
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Yuka Semizu
- Division of Preventive Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kaori Kitamura
- Division of Preventive Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Takeshi Ikeuchi
- Department of Molecular Genetics, Brain Research Institute, Niigata University, Niigata, Japan
| | - Kazutoshi Nakamura
- Division of Preventive Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | | |
Collapse
|
120
|
Zaidi H, Byrkjeland R, Njerve IU, Åkra S, Solheim S, Arnesen H, Seljeflot I, Opstad TB. Effects of exercise training on markers of adipose tissue remodeling in patients with coronary artery disease and type 2 diabetes mellitus: sub study of the randomized controlled EXCADI trial. Diabetol Metab Syndr 2019; 11:109. [PMID: 31890043 PMCID: PMC6923919 DOI: 10.1186/s13098-019-0508-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/13/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Investigate effects of long-term exercise on the remodeling markers MMP-9, TIMP-1, EMMPRIN and Galectin-3 in combined type 2 diabetes mellitus (T2DM) and coronary artery disease (CAD) patients. Any associations between these biomarkers and glucometabolic variables were further assessed at baseline. METHODS 137 patients (age 41-81 years, 17.2% females) were included and randomized to a 12-months exercise program or to a control group. Fasting blood samples and subcutaneous adipose tissue (AT) samples were taken at inclusion and after 12-months. The intervention was a combination of aerobic and strength training for a minimum of 150 min per week. Circulating protein levels were measured by ELISA methods and RNA was extracted from AT and circulating leukocytes. Expression levels were relatively quantified by PCR. RESULTS After 12 months of intervention, both AT-expression and circulating levels of EMMPRIN were increased in the exercise group (p < 0.05, both) with significant difference in change between the two groups (p < 0.05 both). No significant effect was observed on MMP-9, TIMP-1 and Galectin-3. Levels of TIMP-1 (AT-expression and circulating) were significantly correlated to insulin, and HOMA2- after Bonferroni correction (p = 0.001, by 48 performed correlations). CONCLUSION The increase in levels of EMMPRIN after long-term exercise training, might indicate some degree of AT remodeling in these patients after 12-months of exercise, whether beneficial or not. The remodeling markers were to some extent associated with glucometabolic variables in our population with the combined disease.Trial registration clinicaltrials.gov, NCT01232608. Registered 2 November 2010.
Collapse
Affiliation(s)
- Hani Zaidi
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Nydalen, PB 4956, 0424 Oslo, Norway
- Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Rune Byrkjeland
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Nydalen, PB 4956, 0424 Oslo, Norway
- Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ida U. Njerve
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Nydalen, PB 4956, 0424 Oslo, Norway
- Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Sissel Åkra
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Nydalen, PB 4956, 0424 Oslo, Norway
- Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Svein Solheim
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Nydalen, PB 4956, 0424 Oslo, Norway
- Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
| | - Harald Arnesen
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Nydalen, PB 4956, 0424 Oslo, Norway
- Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ingebjørg Seljeflot
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Nydalen, PB 4956, 0424 Oslo, Norway
- Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Trine B. Opstad
- Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevål, Nydalen, PB 4956, 0424 Oslo, Norway
- Center for Heart Failure Research, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
121
|
Holm Nielsen S, Tengryd C, Edsfeldt A, Brix S, Genovese F, Bengtsson E, Karsdal M, Leeming DJ, Nilsson J, Goncalves I. A biomarker of collagen type I degradation is associated with cardiovascular events and mortality in patients with atherosclerosis. J Intern Med 2019; 285:118-123. [PMID: 30156050 DOI: 10.1111/joim.12819] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVE Atherosclerosis is characterized by accumulation of lipids, cells and extracellular matrix (ECM) proteins in the arterial wall. Collagen type I (COL1), a component of the arterial ECM, is cleaved by matrix metalloproteinases (MMPs) and known to be remodelled in atherosclerosis. We explored whether the MMP-mediated COL1 biomarker, C1M, was associated with cardiovascular events, cardiovascular mortality and all-cause mortality in a large prospective cohort of patients with known atherosclerosis. METHODS Serum from 787 patients who underwent a carotid endarterectomy was included. Circulating levels of C1M were measured in serum. A total of 473 patients were followed for 6 years after surgery. Associations between C1M and incidence of cardiovascular events, cardiovascular mortality and all-cause mortality were assessed by Kaplan-Meier curves and Cox regression analysis. RESULTS A total of 101 (21.4%) patients suffered from nonfatal cardiovascular events during the follow-up period, and 64 (13.5%) patients died. Of these, 39 (60.9%) died from cardiovascular diseases. Patients with C1M levels above the median were significantly associated with cardiovascular events, cardiovascular mortality and all-cause mortality (P < 0.001, P = 0.004 and P < 0.001, respectively). C1M was included in the final model for prediction of cardiovascular events (HR 2.15, 95% CI 1.40-3.32, P = 0.001), cardiovascular mortality (HR 2.20, 95% CI 1.07-4.51, P = 0.031) and all-cause mortality (HR 2.98 95% CI 1.67-5.33, P = < 0.001). CONCLUSIONS In patients with atherosclerotic carotid lesions, high levels of C1M predicted cardiovascular events, cardiovascular mortality and all-cause mortality. These findings emphasize the importance of remodelling mechanisms in atherosclerosis that are now becoming more and more explored.
Collapse
Affiliation(s)
- S Holm Nielsen
- Nordic Bioscience, Herlev, Denmark.,Technical University of Denmark, Kgs. Lyngby, Denmark
| | - C Tengryd
- Experimental Cardiovascular Research Unit, Lund University, Malmö, Sweden
| | - A Edsfeldt
- Experimental Cardiovascular Research Unit, Lund University, Malmö, Sweden.,Department of Cardiology, Skåne University Hospital, Malmö, Sweden
| | - S Brix
- Technical University of Denmark, Kgs. Lyngby, Denmark
| | | | - E Bengtsson
- Experimental Cardiovascular Research Unit, Lund University, Malmö, Sweden
| | | | | | - J Nilsson
- Experimental Cardiovascular Research Unit, Lund University, Malmö, Sweden
| | - I Goncalves
- Experimental Cardiovascular Research Unit, Lund University, Malmö, Sweden.,Department of Cardiology, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
122
|
Gargiulo S, Rossin D, Testa G, Gamba P, Staurenghi E, Biasi F, Poli G, Leonarduzzi G. Up-regulation of COX-2 and mPGES-1 by 27-hydroxycholesterol and 4-hydroxynonenal: A crucial role in atherosclerotic plaque instability. Free Radic Biol Med 2018; 129:354-363. [PMID: 30312760 DOI: 10.1016/j.freeradbiomed.2018.09.046] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 09/26/2018] [Accepted: 09/29/2018] [Indexed: 12/12/2022]
Abstract
Atherosclerosis is currently understood to be mainly the consequence of a complicated inflammatory process at the different stages of plaque development. Among the several inflammatory molecules involved, up-regulation of the functional cyclooxygenase 2/membrane-bound prostaglandin E synthase 1 (COX-2/mPGES-1) axis plays a key role in plaque development. Excessive production of oxidized lipids, following low-density lipoprotein (LDL) oxidation, is a characteristic feature of atherosclerosis. Among the oxidized lipids of LDLs, the oxysterol 27-hydroxycholesterol (27-OH) and the aldehyde 4-hydroxynonenal (HNE) substantially accumulate in the atherosclerotic plaque, contributing to its progression and instability through a variety of processes. This study shows that 27-OH and HNE promote up-regulation of both the inducible enzymes COX-2 and mPGES-1, leading to increased production of prostaglandin (PG) E2 and inducible nitric oxide synthase, and the subsequent release of nitric oxide in human promonocytic U937 cells. The study also examined the potential involvement of the functionally coupled COX-2/mPGES-1 in enhancing the production of certain pro-inflammatory cytokines and of matrix metalloproteinase 9 by U937 cells. This enhancement is presumably due to the induction of PGE2 synthesis, as a result of the up-regulation of the COX-2/mPGES-1, stimulated by the two oxidized lipids, 27-OH and HNE. Induction of PGE2 synthesis might thus be a mechanism of plaque instability and eventual rupture, contributing to matrix metalloproteinase production by activated macrophages.
Collapse
Affiliation(s)
- Simona Gargiulo
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Daniela Rossin
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Gabriella Testa
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Paola Gamba
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Erica Staurenghi
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Fiorella Biasi
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Giuseppe Poli
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy
| | - Gabriella Leonarduzzi
- Department of Clinical and Biological Sciences, School of Medicine, University of Turin, Orbassano, Torino, Italy.
| |
Collapse
|
123
|
Xu L, Liu JT, Li K, Wang SY, Xu S. Genistein inhibits Ang II-induced CRP and MMP-9 generations via the ER-p38/ERK1/2-PPARγ-NF-κB signaling pathway in rat vascular smooth muscle cells. Life Sci 2018; 216:140-146. [PMID: 30452971 DOI: 10.1016/j.lfs.2018.11.036] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/15/2018] [Accepted: 11/15/2018] [Indexed: 12/26/2022]
Abstract
AIMS C-reactive protein (CRP) and matrix metalloproteinase (MMP)-9 are involved in the inflammation of atherosclerosis lesions. Genistein (Gen) has been demonstrated to exert beneficial effect on the cardiovascular system. However, it remains unclear whether Gen produces anti-inflammatory effect in vascular smooth muscle cells (VSMCs). Therefore, we investigated the effects of Gen on CRP and MMP-9 expressions induced by angiotensin (Ang) II in VSMCs and the related molecular mechanism. MAIN METHODS Rat VSMCs were cultured, and Ang II was used as a stimulant for CRP and MMP-9 expressions. CRP level was measured by ELISA. The mRNA and protein expressions of related indexes were identified by reverse transcription-polymerase chain reaction and western blot, respectively. KEY FINDINGS Gen inhibited Ang II-stimulated CRP and MMP-9 mRNA and protein expressions in concentration- and time-dependent manners. Additionally, Gen ameliorated Ang II-induced p-ERK1/2, p-p38 and NF-κB expressions, antagonized Ang II-downregulated peroxisome proliferation-activated receptor (PPAR) γ and estrogen receptor (ER) β expressions. After treating the VSMCs with GW9662 or ICI182780 in Gen treated groups, inhibitory effect of Gen on CRP and MMP-9 expressions were antagonized in Ang II-stimulated VSMCs. The treatment of VSMCs with ICI182780 abolished downregulations of p-p38/p-ERK1/2, and antagonized upregulation of PPARγ by Gen in Ang II-stimulated VSMCs. Moreover, the inhibitory effect of Gen on Ang II-stimulated NF-κB expression was abolished after preincubation of VSMCs with GW9662 in Gen treated groups. SIGNIFICANCE Gen exerts anti-inflammatory property via the ER-p38/ERK1/2-PPARγ-NF-κB-CRP/MMP-9 signal pathway in Ang II-stimulated VSMCs.
Collapse
Affiliation(s)
- Li Xu
- Department of Pharmacology, Xi'an Jiaotong University School of Medicine, Xi'an 710061, People's Republic of China; Department of Pharmacy, The First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, People's Republic of China; Hospital Management Institute of Xi'an Medical University, Xi'an 710077, People's Republic of China
| | - Jun-Tian Liu
- Department of Pharmacology, Xi'an Jiaotong University School of Medicine, Xi'an 710061, People's Republic of China.
| | - Kai Li
- Department of Cardiology, Xi'an Medical University, Xi'an 710021, People's Republic of China
| | - Sheng-Yu Wang
- Department of Intensive Care Unit, The First Affiliated Hospital of Xi'an Medical University, Xi'an 710077, People's Republic of China
| | - Shouzhu Xu
- Department of Pharmacology, Xi'an Jiaotong University School of Medicine, Xi'an 710061, People's Republic of China
| |
Collapse
|
124
|
Hohn M, Chang M, Meisel JE, Frost E, Schwegmann K, Hermann S, Schäfers M, Riemann B, Haufe G, Breyholz H, Wagner S. Synthesis and Preliminary In Vitroand In VivoEvaluation of Thiirane‐Based Slow‐Binding MMP Inhibitors as Potential Radiotracers for PET Imaging. ChemistrySelect 2018. [DOI: 10.1002/slct.201803093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Michael Hohn
- Department of Nuclear MedicineUniversity Hospital Münster Albert-Schweitzer-Campus 1 Building A1 D-48149 Münster Germany
- Organic Chemistry InstituteUniversity of Münster Corrensstr. 40 D-48149 Münster Germany
| | - Mayland Chang
- Department of Chemistry and Biochemistry, 354 McCourtney HallUniversity of Notre Dame Notre Dame IN 46556–5710 USA
| | - Jayda E. Meisel
- Chemical, BiologicalRadiological, Nuclearand Explosive DefenseBattelle Memorial Institute 505 King Avenue Columbus Ohio 43201 USA
| | - Emma Frost
- Department of Chemistry and Biochemistry, 354 McCourtney HallUniversity of Notre Dame Notre Dame IN 46556–5710 USA
| | - Katrin Schwegmann
- European Institute for Molecular Imaging (EIMI)University of Münster Waldeyerstraße 15 D-48149 Münster Germany
| | - Sven Hermann
- European Institute for Molecular Imaging (EIMI)University of Münster Waldeyerstraße 15 D-48149 Münster Germany
| | - Michael Schäfers
- Department of Nuclear MedicineUniversity Hospital Münster Albert-Schweitzer-Campus 1 Building A1 D-48149 Münster Germany
- European Institute for Molecular Imaging (EIMI)University of Münster Waldeyerstraße 15 D-48149 Münster Germany
- Cells in Motion (CiM) Cluster of ExcellenceUniversity of Münster D-48149 Münster Germany
| | - Burkhard Riemann
- Department of Nuclear MedicineUniversity Hospital Münster Albert-Schweitzer-Campus 1 Building A1 D-48149 Münster Germany
| | - Günter Haufe
- Organic Chemistry InstituteUniversity of Münster Corrensstr. 40 D-48149 Münster Germany
- Cells in Motion (CiM) Cluster of ExcellenceUniversity of Münster D-48149 Münster Germany
| | - Hans‐Jörg Breyholz
- Department of Nuclear MedicineUniversity Hospital Münster Albert-Schweitzer-Campus 1 Building A1 D-48149 Münster Germany
| | - Stefan Wagner
- Department of Nuclear MedicineUniversity Hospital Münster Albert-Schweitzer-Campus 1 Building A1 D-48149 Münster Germany
| |
Collapse
|
125
|
Administration of hydrogen-rich water prevents vascular aging of the aorta in LDL receptor-deficient mice. Sci Rep 2018; 8:16822. [PMID: 30429524 PMCID: PMC6235982 DOI: 10.1038/s41598-018-35239-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 10/25/2018] [Indexed: 12/14/2022] Open
Abstract
The main cause of arteriosclerosis is atherosclerosis in the aorta. Atherosclerosis is recognized as a chronic inflammatory condition that begins with the dysfunction or activation of arterial endothelium. Low-density lipoprotein (LDL) and especially its oxidized form play a key role in endothelial dysfunction and atherogenesis. Recent studies showed that senescent cells are involved in the development and progression of atherosclerosis, and eliminating senescent cells suppresses the senescence-associated secretory phenotype. We previously reported that molecular hydrogen-rich water (HW) has antioxidant and anti-inflammatory effects in numerous diseases. Here, we used LDL receptor-deficient mice fed a high-fat diet (HFD) for 13 weeks as a model for atherosclerosis and evaluated the effects of continuous administration of HW. The numbers of endothelial cells in the atheroma expressing the senescence factors p16INK4a and p21 decreased in HFD-fed mice given HW compared with HFD-fed mice given control water. Furthermore, macrophage infiltration and Tnfα expression in the atheroma were also suppressed. These results suggest that vascular aging can be suppressed by HW.
Collapse
|
126
|
Gallego-Colon E, Wojakowski W, Francuz T. Incretin drugs as modulators of atherosclerosis. Atherosclerosis 2018; 278:29-38. [DOI: 10.1016/j.atherosclerosis.2018.09.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 07/06/2018] [Accepted: 09/13/2018] [Indexed: 02/06/2023]
|
127
|
Lee AY, Park W, Kang TW, Cha MH, Chun JM. Network pharmacology-based prediction of active compounds and molecular targets in Yijin-Tang acting on hyperlipidaemia and atherosclerosis. JOURNAL OF ETHNOPHARMACOLOGY 2018; 221:151-159. [PMID: 29698773 DOI: 10.1016/j.jep.2018.04.027] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 03/29/2018] [Accepted: 04/18/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Yijin-Tang (YJT) is a traditional prescription for the treatment of hyperlipidaemia, atherosclerosis and other ailments related to dampness phlegm, a typical pathological symptom of abnormal body fluid metabolism in Traditional Korean Medicine. However, a holistic network pharmacology approach to understanding the therapeutic mechanisms underlying hyperlipidaemia and atherosclerosis has not been pursued. AIM OF THE STUDY To examine the network pharmacological potential effects of YJT on hyperlipidaemia and atherosclerosis, we analysed components, performed target prediction and network analysis, and investigated interacting pathways using a network pharmacology approach. MATERIALS AND METHODS Information on compounds in herbal medicines was obtained from public databases, and oral bioavailability and drug-likeness was screened using absorption, distribution, metabolism, and excretion (ADME) criteria. Correlations between compounds and genes were linked using the STITCH database, and genes related to hyperlipidaemia and atherosclerosis were gathered using the GeneCards database. Human genes were identified and subjected to Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. RESULTS Network analysis identified 447 compounds in five herbal medicines that were subjected to ADME screening, and 21 compounds and 57 genes formed the main pathways linked to hyperlipidaemia and atherosclerosis. Among them, 10 compounds (naringenin, nobiletin, hesperidin, galangin, glycyrrhizin, homogentisic acid, stigmasterol, 6-gingerol, quercetin and glabridin) were linked to more than four genes, and are bioactive compounds and key chemicals. Core genes in this network were CASP3, CYP1A1, CYP1A2, MMP2 and MMP9. The compound-target gene network revealed close interactions between multiple components and multiple targets, and facilitates a better understanding of the potential therapeutic effects of YJT. CONCLUSIONS Pharmacological network analysis can help to explain the potential effects of YJT for treating dampness phlegm-related diseases such as hyperlipidaemia and atherosclerosis.
Collapse
Affiliation(s)
- A Yeong Lee
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Republic of Korea
| | - Won Park
- Bioinformatics Group, R&D Center, Insilicogen Corporation, 35, Techno 9-ro, 34027, Republic of Korea
| | - Tae-Wook Kang
- Bioinformatics Group, R&D Center, Insilicogen Corporation, 35, Techno 9-ro, 34027, Republic of Korea
| | - Min Ho Cha
- Clinical Medicine Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Republic of Korea
| | - Jin Mi Chun
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Republic of Korea; Department of Life Systems, Sookmyung Women's University, Cheongpa-ro 47-gil 100, Yongsan-gu, Seoul 04310, Republic of Korea.
| |
Collapse
|
128
|
Montague SJ, Gardiner EE. Matrix metalloproteinase-13 unlucky for the forming thrombus. Res Pract Thromb Haemost 2018; 2:525-528. [PMID: 30046757 PMCID: PMC6046591 DOI: 10.1002/rth2.12105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 04/03/2018] [Indexed: 01/01/2023] Open
Affiliation(s)
- Samantha J. Montague
- ACRF Department of Cancer Biology and TherapeuticsJohn Curtin School of Medical ResearchThe Australian National UniversityCanberraACTAustralia
| | - Elizabeth E. Gardiner
- ACRF Department of Cancer Biology and TherapeuticsJohn Curtin School of Medical ResearchThe Australian National UniversityCanberraACTAustralia
| |
Collapse
|
129
|
Abstract
Over the past decade, studies have repeatedly found single-nucleotide polymorphisms located in the collagen ( COL) 4A1 and COL4A2 genes to be associated with cardiovascular disease (CVD), and the 13q34 locus harboring these genes is one of ~160 genome-wide significant risk loci for coronary artery disease. COL4A1 and COL4A2 encode the α1- and α2-chains of collagen type IV, a major component of basement membranes in various tissues including arteries. Despite the growing body of evidence indicating a role for collagen type IV in CVD, remarkably few studies have aimed to directly investigate such a role. The purpose of this review is to summarize the clinical reports linking 13q34 to coronary artery disease, atherosclerosis, and artery stiffening and to assemble the scattered pieces of evidence from experimental studies based on vascular cells and tissue collectively supporting a role for collagen type IV in atherosclerosis and other macrovascular disease conditions.
Collapse
Affiliation(s)
- L B Steffensen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital , Odense , Denmark.,Centre for Individualized Medicine in Arterial Diseases, Odense University Hospital , Odense , Denmark.,Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark , Odense , Denmark
| | - L M Rasmussen
- Department of Clinical Biochemistry and Pharmacology, Odense University Hospital , Odense , Denmark.,Centre for Individualized Medicine in Arterial Diseases, Odense University Hospital , Odense , Denmark
| |
Collapse
|
130
|
|