1
|
Zisser L, Binder CJ. Extracellular Vesicles as Mediators in Atherosclerotic Cardiovascular Disease. J Lipid Atheroscler 2024; 13:232-261. [PMID: 39355407 PMCID: PMC11439751 DOI: 10.12997/jla.2024.13.3.232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/12/2024] [Accepted: 07/26/2024] [Indexed: 10/03/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial intima, characterized by accumulation of lipoproteins and accompanying inflammation, leading to the formation of plaques that eventually trigger occlusive thrombotic events, such as myocardial infarction and ischemic stroke. Although many aspects of plaque development have been elucidated, the role of extracellular vesicles (EVs), which are lipid bilayer-delimited vesicles released by cells as mediators of intercellular communication, has only recently come into focus of atherosclerosis research. EVs comprise several subtypes that may be differentiated by their size, mode of biogenesis, or surface marker expression and cargo. The functional effects of EVs in atherosclerosis depend on their cellular origin and the specific pathophysiological context. EVs have been suggested to play a role in all stages of plaque formation. In this review, we highlight the known mechanisms by which EVs modulate atherogenesis and outline current limitations and challenges in the field.
Collapse
Affiliation(s)
- Lucia Zisser
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
2
|
Saad MI, Jenkins BJ. The protease ADAM17 at the crossroads of disease: revisiting its significance in inflammation, cancer, and beyond. FEBS J 2024; 291:10-24. [PMID: 37540030 DOI: 10.1111/febs.16923] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/04/2023] [Accepted: 08/02/2023] [Indexed: 08/05/2023]
Abstract
The protease A Disintegrin And Metalloproteinase 17 (ADAM17) plays a central role in the pathophysiology of several diseases. ADAM17 is involved in the cleavage and shedding of at least 80 known membrane-tethered proteins, which subsequently modulate several intracellular signaling pathways, and therefore alter cell behavior. Dysregulated expression and/or activation of ADAM17 has been linked to a wide range of autoimmune and inflammatory diseases, cancer, and cardiovascular disease. In this review, we provide an overview of the current state of knowledge from preclinical models and clinical data on the diverse pathophysiological roles of ADAM17, and discuss the mechanisms underlying ADAM17-mediated protein shedding and the potential therapeutic implications of targeting ADAM17 in these diseases.
Collapse
Affiliation(s)
- Mohamed I Saad
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Vic., Australia
- Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Vic., Australia
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Vic., Australia
- Department of Molecular and Translational Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Vic., Australia
- South Australian immunoGENomics Cancer Institute (SAiGENCI), University of Adelaide, SA, Australia
| |
Collapse
|
3
|
Zhou G, Liu J. Prognostic value of elevated plasma angiotensin-converting enzyme 2 in cardiometabolic diseases: A review. Medicine (Baltimore) 2023; 102:e33251. [PMID: 36897667 PMCID: PMC9997766 DOI: 10.1097/md.0000000000033251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 02/22/2023] [Indexed: 03/11/2023] Open
Abstract
Angiotensin-converting enzyme 2, as an internal anti regulator of the renin-angiotensin hormone cascade reaction, plays a protective role in vasodilation, inhibition of fibrosis, and initiation of anti-inflammatory and antioxidative stress by degrading angiotensin II and generating angiotensin (1-7). Multiple studies have shown that plasma angiotensin-converting enzyme 2 activity is low in healthy populations without significant cardiometabolic disease, and elevated plasma angiotensin-converting enzyme 2 levels can be used as a novel biomarker of abnormal myocardial structure and/or adverse events in cardiometabolic diseases. This article aims to elaborate the determinants of plasma angiotensin-converting enzyme 2 concentration, the relevance between angiotensin-converting enzyme 2 and cardiometabolic disease risk markers, and its relative importance compared with known cardiovascular disease risk factors. Confronted with the known cardiovascular risk factors, plasma angiotensin-converting enzyme 2 (ACE2) concentration uniformly emerged as a firm predictor of abnormal myocardial structure and/or adverse events in cardiometabolic diseases and may improve the risk prediction of cardiometabolic diseases when combined with other conventional risk factors. Cardiovascular disease is the leading cause of death worldwide, while the renin-angiotensin system is the main hormone cascade system involved in the pathophysiology of cardiovascular disease. A multi-ancestry global cohort study from the general population by Narula et al revealed that plasma ACE2 concentration was strongly associated with cardiometabolic disease and might be an easily measurable indicator of renin-angiotensin system disorder. The association between this atypical hormone disorder marker and cardiometabolic disease is isolated from conventional cardiac risk factors and brain natriuretic peptide, suggesting that a clearer comprehending of the changes in plasma ACE2 concentration and activity may help us to improve the risk prediction of cardiometabolic disease, guide early diagnosis and feasible therapies, and develop and test new therapeutic targets.
Collapse
Affiliation(s)
- Gang Zhou
- Department of First Clinical Medical College, Guangxi Medical University, Nanning, Guangxi, China
| | - Jingchen Liu
- Department of Anesthesiology, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
4
|
Sharma D, Singh NK. The Biochemistry and Physiology of A Disintegrin and Metalloproteinases (ADAMs and ADAM-TSs) in Human Pathologies. Rev Physiol Biochem Pharmacol 2023; 184:69-120. [PMID: 35061104 DOI: 10.1007/112_2021_67] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Metalloproteinases are a group of proteinases that plays a substantial role in extracellular matrix remodeling and its molecular signaling. Among these metalloproteinases, ADAMs (a disintegrin and metalloproteinases) and ADAM-TSs (ADAMs with thrombospondin domains) have emerged as highly efficient contributors mediating proteolytic processing of various signaling molecules. ADAMs are transmembrane metalloenzymes that facilitate the extracellular domain shedding of membrane-anchored proteins, cytokines, growth factors, ligands, and their receptors and therefore modulate their biological functions. ADAM-TSs are secretory, and soluble extracellular proteinases that mediate the cleavage of non-fibrillar extracellular matrix proteins. ADAMs and ADAM-TSs possess pro-domain, metalloproteinase, disintegrin, and cysteine-rich domains in common, but ADAM-TSs have characteristic thrombospondin motifs instead of the transmembrane domain. Most ADAMs and ADAM-TSs are activated by cleavage of pro-domain via pro-protein convertases at their N-terminus, hence directing them to various signaling pathways. In this article, we are discussing not only the structure and regulation of ADAMs and ADAM-TSs, but also the importance of these metalloproteinases in various human pathophysiological conditions like cardiovascular diseases, colorectal cancer, autoinflammatory diseases (sepsis/rheumatoid arthritis), Alzheimer's disease, proliferative retinopathies, and infectious diseases. Therefore, based on the emerging role of ADAMs and ADAM-TSs in various human pathologies, as summarized in this review, these metalloproteases can be considered as critical therapeutic targets and diagnostic biomarkers.
Collapse
Affiliation(s)
- Deepti Sharma
- Department of Ophthalmology, Visual and Anatomical Sciences, Integrative Biosciences Center (IBio), Wayne State University School of Medicine, Detroit, MI, USA
| | - Nikhlesh K Singh
- Department of Ophthalmology, Visual and Anatomical Sciences, Integrative Biosciences Center (IBio), Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
5
|
Tang BY, Ge J, Wu Y, Wen J, Tang XH. The Role of ADAM17 in Inflammation-Related Atherosclerosis. J Cardiovasc Transl Res 2022; 15:1283-1296. [PMID: 35648358 DOI: 10.1007/s12265-022-10275-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 05/06/2022] [Indexed: 10/18/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease that poses a huge economic burden due to its extremely poor prognosis. Therefore, it is necessary to explore potential mechanisms to improve the prevention and treatment of atherosclerosis. A disintegrin and metalloprotease 17 (ADAM17) is a cell membrane-bound protein that performs a range of functions through membrane protein shedding and intracellular signaling. ADAM17-mediated inflammation has been identified to be an important contributor to atherosclerosis; however, the specific relationship between its multiple regulatory roles and the pathogenesis of atherosclerosis remains unclear. Here, we reviewed the activation, function, and regulation of ADAM17, described in detail the role of ADAM17-mediated inflammatory damage in atherosclerosis, and discussed several controversial points. We hope that these insights into ADAM17 biology will lead to rational management of atherosclerosis. ADAM17 promotes vascular inflammation in endothelial cells, smooth muscle cells, and macrophages, and regulates the occurrence and development of atherosclerosis.
Collapse
Affiliation(s)
- Bai-Yi Tang
- Department of Cardiology, Third Xiang-Ya Hospital, Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Jin Ge
- Department of Cardiology, Third Xiang-Ya Hospital, Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, China
| | - Yang Wu
- Department of Cardiology, Third Hospital of Changsha, 176 W. Laodong Road, Changsha, 410015, Hunan, China
| | - Juan Wen
- Department of Cardiology, Third Xiang-Ya Hospital, Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, China.
| | - Xiao-Hong Tang
- Department of Cardiology, Third Xiang-Ya Hospital, Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, China.
| |
Collapse
|
6
|
ADAM17: A novel treatment target for aneurysms. Biomed Pharmacother 2022; 148:112712. [DOI: 10.1016/j.biopha.2022.112712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/02/2022] [Accepted: 02/08/2022] [Indexed: 12/20/2022] Open
|
7
|
Li L, Liu S, Tan J, Wei L, Wu D, Gao S, Weng Y, Chen J. Recent advance in treatment of atherosclerosis: Key targets and plaque-positioned delivery strategies. J Tissue Eng 2022; 13:20417314221088509. [PMID: 35356091 PMCID: PMC8958685 DOI: 10.1177/20417314221088509] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Atherosclerosis, a chronic inflammatory disease of vascular wall, is a progressive pathophysiological process with lipids oxidation/depositing initiation and innate/adaptive immune responses. The coordination of multi systems covering oxidative stress, dysfunctional endothelium, diseased lipid uptake, cell apoptosis, thrombotic and pro-inflammatory responding as well as switched SMCs contributes to plaque growth. In this circumstance, inevitably, targeting these processes is considered to be effective for treating atherosclerosis. Arriving, retention and working of payload candidates mediated by targets in lesion direct ultimate therapeutic outcomes. Accumulating a series of scientific studies and clinical practice in the past decades, lesion homing delivery strategies including stent/balloon/nanoparticle-based transportation worked as the potent promotor to ensure a therapeutic effect. The objective of this review is to achieve a very brief summary about the effective therapeutic methods cooperating specifical targets and positioning-delivery strategies in atherosclerosis for better outcomes.
Collapse
Affiliation(s)
- Li Li
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| | - Sainan Liu
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| | - Jianying Tan
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| | - Lai Wei
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| | - Dimeng Wu
- Chengdu Daxan Innovative Medical Tech. Co., Ltd., Chengdu, PR China
| | - Shuai Gao
- Chengdu Daxan Innovative Medical Tech. Co., Ltd., Chengdu, PR China
| | - Yajun Weng
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| | - Junying Chen
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu, PR China
| |
Collapse
|
8
|
Kawai T, Elliott KJ, Scalia R, Eguchi S. Contribution of ADAM17 and related ADAMs in cardiovascular diseases. Cell Mol Life Sci 2021; 78:4161-4187. [PMID: 33575814 PMCID: PMC9301870 DOI: 10.1007/s00018-021-03779-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/23/2020] [Accepted: 01/27/2021] [Indexed: 02/06/2023]
Abstract
A disintegrin and metalloproteases (ADAMs) are key mediators of cell signaling by ectodomain shedding of various growth factors, cytokines, receptors and adhesion molecules at the cellular membrane. ADAMs regulate cell proliferation, cell growth, inflammation, and other regular cellular processes. ADAM17, the most extensively studied ADAM family member, is also known as tumor necrosis factor (TNF)-α converting enzyme (TACE). ADAMs-mediated shedding of cytokines such as TNF-α orchestrates immune system or inflammatory cascades and ADAMs-mediated shedding of growth factors causes cell growth or proliferation by transactivation of the growth factor receptors including epidermal growth factor receptor. Therefore, increased ADAMs-mediated shedding can induce inflammation, tissue remodeling and dysfunction associated with various cardiovascular diseases such as hypertension and atherosclerosis, and ADAMs can be a potential therapeutic target in these diseases. In this review, we focus on the role of ADAMs in cardiovascular pathophysiology and cardiovascular diseases. The main aim of this review is to stimulate new interest in this area by highlighting remarkable evidence.
Collapse
Affiliation(s)
- Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine At Temple University, Philadelphia, PA, USA
| | - Katherine J Elliott
- Cardiovascular Research Center, Lewis Katz School of Medicine At Temple University, Philadelphia, PA, USA
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine At Temple University, Philadelphia, PA, USA
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine At Temple University, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Study on the Potential Mechanism of Fructus Tribuli in the Treatment of Hypertensive Vascular Remodeling Based on Network Pharmacology and Molecular Docking. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:8862176. [PMID: 33505509 PMCID: PMC7810546 DOI: 10.1155/2021/8862176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 12/09/2020] [Accepted: 12/23/2020] [Indexed: 01/07/2023]
Abstract
Background Hypertensive vascular remodeling (HVR) is the pathophysiological basis of hypertension, which is also an important cause of vascular disease and target organ damage. Treatment with Fructus Tribuli (FT), a traditional Chinese medicine, has a positive effect on HVR. However, the pharmacological mechanisms of FT are still unclear. Therefore, this study aimed to reveal the potential mechanisms involved in the effects of FT on HVR based on network pharmacology and molecular docking. Materials and Methods We selected the active compounds and targets of FT according to the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP) and the Swiss Target Prediction database, and the targets of HVR were collected from the Online Mendelian Inheritance in Man (OMIM), GeneCards, and DrugBank databases. The protein-protein interaction network (PPI) was established using the STRING database. Moreover, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses and network analysis were performed to further explore the potential mechanisms. Finally, molecular docking methods were used to evaluate the affinity between the active compounds and the main target. Results Seventeen active compounds of FT and 164 potential targets for the treatment of HVR were identified. Component-target and PPI networks were constructed, and 12 main active components and 33 main targets were identified by analyzing the topological parameters. Additionally, GO analysis indicated that the potential targets were enriched in 483 biological processes, 52 cellular components, and 110 molecular functions. KEGG analysis revealed that the potential targets were correlated with 122 pathways, such as the HIF-1 signaling pathway, ErbB signaling pathway, and VEGF signaling pathway. Finally, molecular docking showed that the 12 main active components had a good affinity for the top five main targets. Conclusion This study demonstrated the multiple compounds, targets, and pathway characteristics of FT in the treatment of HVR. The network pharmacology method provided a novel research approach to analyze potential mechanisms.
Collapse
|
10
|
Hernandez AA, Foster GA, Soderberg SR, Fernandez A, Reynolds MB, Orser MK, Bailey KA, Rogers JH, Singh GD, Wu H, Passerini AG, Simon SI. An Allosteric Shift in CD11c Affinity Activates a Proatherogenic State in Arrested Intermediate Monocytes. THE JOURNAL OF IMMUNOLOGY 2020; 205:2806-2820. [PMID: 33055281 DOI: 10.4049/jimmunol.2000485] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 09/10/2020] [Indexed: 11/19/2022]
Abstract
Intermediate monocytes (iMo; CD14+CD16+) increase in number in the circulation of patients with unstable coronary artery disease (CAD), and their recruitment to inflamed arteries is implicated in events leading to mortality following MI. Monocyte recruitment to inflamed coronary arteries is initiated by high affinity β2-integrin (CD11c/CD18) that activates β1-integrin (VLA-4) to bind endothelial VCAM-1. How integrin binding under shear stress mechanosignals a functional shift in iMo toward an inflammatory phenotype associated with CAD progression is unknown. Whole blood samples from patients treated for symptomatic CAD including non-ST elevation MI, along with healthy age-matched subjects, were collected to assess chemokine and integrin receptor levels on monocytes. Recruitment on inflamed human aortic endothelium or rVCAM-1 under fluid shear stress was assessed using a microfluidic-based artery on a chip (A-Chip). Membrane upregulation of high affinity CD11c correlated with concomitant activation of VLA-4 within focal adhesive contacts was required for arrest and diapedesis across inflamed arterial endothelium to a greater extent in non-ST elevation MI compared with stable CAD patients. The subsequent conversion of CD11c from a high to low affinity state under fluid shear activated phospho-Syk- and ADAM17-mediated proteolytic cleavage of CD16. This marked the conversion of iMo to an inflammatory phenotype associated with nuclear translocation of NF-κB and production of IL-1β+ We conclude that CD11c functions as a mechanoregulator that activates an inflammatory state preferentially in a majority of iMo from cardiac patients but not healthy patients.
Collapse
Affiliation(s)
- Alfredo A Hernandez
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616
| | - Greg A Foster
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616
| | - Stephanie R Soderberg
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616
| | - Andrea Fernandez
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616
| | - Mack B Reynolds
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616
| | - Mable K Orser
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616
| | - Keith A Bailey
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616
| | - Jason H Rogers
- Department of Cardiovascular and Internal Medicine, University of California, Davis Medical Center, Sacramento, CA 95817; and
| | - Gagan D Singh
- Department of Cardiovascular and Internal Medicine, University of California, Davis Medical Center, Sacramento, CA 95817; and
| | - Huaizhu Wu
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX 77030
| | - Anthony G Passerini
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616
| | - Scott I Simon
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616;
| |
Collapse
|
11
|
Meka IA, Anyim OB, Enebe JT, Ukwaja KN, Ugonabo MC. Association of MiRNA122 & ADAM17 with Lipids among Hypertensives in Nigeria. Open Med (Wars) 2018; 13:350-358. [PMID: 30211317 PMCID: PMC6132086 DOI: 10.1515/med-2018-0053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 06/25/2018] [Indexed: 11/15/2022] Open
Abstract
Background Dyslipidaemia and hypertension are established major risk factors for cardiovascular diseases. The suggested roles of miRNA-122 and ADAM17 in lipid metabolism can therefore be applied in the management of metabolic disorders. The authors’ aim was to determine the association between miRNA-122 and ADAM17, as well as the association between miRNA-122 and lipid fractions, in the study participants. Method A comparative cross-sectional study was conducted among 200 hypertensive patients and 100 non-hypertensive adult controls between May, 2015, and June, 2016, in Nigeria. Lipids were analysed with spectrophotometric methods whereas ADAM17 and miRNA-122 were analysed with enzyme linked immunosorbent assay and quantitative polymerase chain reaction, respectively. Results The mean (standard deviation [SD]) ages of 200 hypertensives and 100 controls were 56.3 (6.9) and 54.9 (8.3) years, respectively. miRNA-112 and ADAM17 had significantly higher values among dyslipidaemic individuvals compared with non-dyslipidaemic participants. The correlation between miRNA-122 and ADAM17 levels was strongly positive, r=0.82, p<0.05. LDL-cholesterol and total cholesterol also showed statistically significant positive correlation with miRNA-122, r=0.53, r=0.51, (p< 0.001) respectively. Conclusion In this study, miRNA-122 showed a strong correlation with ADAM17 and a positive correlation with LDL-cholesterol and total cholesterol. These findings support the stimulant roles of miRNA-122 and ADAM17 in lipid metabolism and thus could be used in the management of dyslipidaemia.
Collapse
Affiliation(s)
- Ijeoma A Meka
- Department of Chemical Pathology, University of Nigeria Enugu Campus, Enugu, Nigeria
| | - Obumneme B Anyim
- Department of Internal Medicine, University of Nigeria Teaching Hospital, Ituku-Ozalla, Enugu State, Enugu, Nigeria
| | - Joseph T Enebe
- Department of Obstetrics and Gynaecology, Enugu State University of Science and Technology, Enugu State, Enugu, Nigeria
| | - Kingsley N Ukwaja
- Department of Internal Medicine, Federal Teaching Hospital Abakaliki, Ebonyi State, Enugu, Nigeria
| | - Martin C Ugonabo
- Department of Chemical Pathology, University of Nigeria Enugu Campus, Enugu State, Enugu, Nigeria
| |
Collapse
|
12
|
Elevated plasma angiotensin converting enzyme 2 activity is an independent predictor of major adverse cardiac events in patients with obstructive coronary artery disease. PLoS One 2018; 13:e0198144. [PMID: 29897923 PMCID: PMC5999069 DOI: 10.1371/journal.pone.0198144] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 05/14/2018] [Indexed: 12/16/2022] Open
Abstract
Background Angiotensin converting enzyme 2 (ACE2) is an endogenous regulator of the renin angiotensin system. Increased circulating ACE2 predicts adverse outcomes in patients with heart failure (HF), but it is unknown if elevated plasma ACE2 activity predicts major adverse cardiovascular events (MACE) in patients with obstructive coronary artery disease (CAD). Methods We prospectively recruited patients with obstructive CAD (defined as ≥50% stenosis of the left main coronary artery and/or ≥70% stenosis in ≥ 1 other major epicardial vessel on invasive coronary angiography) and measured plasma ACE2 activity. Patients were followed up to determine if circulating ACE2 activity levels predicted the primary endpoint of MACE (cardiovascular mortality, HF or myocardial infarction). Results We recruited 79 patients with obstructive coronary artery disease. The median (IQR) plasma ACE2 activity was 29.3 pmol/ml/min [21.2–41.2]. Over a median follow up of 10.5 years [9.6–10.8years], MACE occurred in 46% of patients (36 events). On Kaplan-Meier analysis, above-median plasma ACE2 activity was associated with MACE (log-rank test, p = 0.035) and HF hospitalisation (p = 0.01). After Cox multivariable adjustment, log ACE2 activity remained an independent predictor of MACE (hazard ratio (HR) 2.4, 95% confidence interval (CI) 1.24–4.72, p = 0.009) and HF hospitalisation (HR: 4.03, 95% CI: 1.42–11.5, p = 0.009). Conclusions Plasma ACE2 activity independently increased the hazard of adverse long-term cardiovascular outcomes in patients with obstructive CAD.
Collapse
|
13
|
Tawfik MK, El-Kherbetawy MK, Makary S. Cardioprotective and Anti-Aggregatory Effects of Levosimendan on Isoproterenol-Induced Myocardial Injury in High-Fat-Fed Rats Involves Modulation of PI3K/Akt/mTOR Signaling Pathway and Inhibition of Apoptosis. J Cardiovasc Pharmacol Ther 2018; 23:456-471. [DOI: 10.1177/1074248418763957] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hyperlipidemia and hypercoagulability states are linked with the increased risks of myocardial infarction (MI). Levosimendan has vasorelaxant and anti-aggregatory properties. The present study evaluated the anti-aggregatory and cardioprotective effects of levosimendan versus cilostazol in high-fat diet (HFD)-fed rats subjected to isoproterenol-induced MI. Rats were assigned to normal, HFD, HFD + isoproterenol, HFD + isoproterenol + cilostazol, and HFD + isoproterenol + levosimendan. The present study investigated the anti-aggregatory effect of both levosimendan and cilostazol and revealed that both drugs attenuated the severity of platelet aggregation. Moreover, both levosimendan and cilostazol revealed effectiveness in attenuating the severity of HFD/isoproterenol-induced myocardial injury as revealed by electrocardiogram signs, apoptotic markers, and histopathological score via counteracting the oxidative stress burden, increments in the expression of inflammatory mediators, and modulating nuclear factor kappa-B (NF-κB) and phosphatidylinositide 3-kinases (PI3K)/protein kinase B (Akt)/ mechanistic target of rapamycin (mTOR) pathway. It was obvious that levosimendan offered more cardioprotective properties than cilostazol. The study showed the relations between hyperlipedemia, hyperaggregability state, and myocardial injury with the modulation of NF-κB and PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Mona K. Tawfik
- Department of Pharmacology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | | | - Samy Makary
- Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
14
|
Chemaly M, McGilligan V, Gibson M, Clauss M, Watterson S, Alexander HD, Bjourson AJ, Peace A. Role of tumour necrosis factor alpha converting enzyme (TACE/ADAM17) and associated proteins in coronary artery disease and cardiac events. Arch Cardiovasc Dis 2017; 110:700-711. [DOI: 10.1016/j.acvd.2017.08.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/13/2017] [Accepted: 08/16/2017] [Indexed: 02/07/2023]
|
15
|
Yadav N, Chandra H. Suppression of inflammatory and infection responses in lung macrophages by eucalyptus oil and its constituent 1,8-cineole: Role of pattern recognition receptors TREM-1 and NLRP3, the MAP kinase regulator MKP-1, and NFκB. PLoS One 2017; 12:e0188232. [PMID: 29141025 PMCID: PMC5687727 DOI: 10.1371/journal.pone.0188232] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 11/02/2017] [Indexed: 01/09/2023] Open
Abstract
Eucalyptus oil (EO) used in traditional medicine continues to prove useful for aroma therapy in respiratory ailments; however, there is a paucity of information on its mechanism of action and active components. In this direction, we investigated EO and its dominant constituent 1,8–cineole (eucalyptol) using the murine lung alveolar macrophage (AM) cell line MH-S. In an LPS-induced AM inflammation model, pre-treatment with EO significantly reduced (P ≤0.01or 0.05) the pro-inflammatory mediators TNF-α, IL-1 (α and β), and NO, albeit at a variable rate and extent; 1,8-cineole diminished IL-1 and IL-6. In a mycobacterial-infection AM model, EO pre-treatment or post-treatment significantly enhanced (P ≤0.01) the phagocytic activity and pathogen clearance. 1,8-cineole also significantly enhanced the pathogen clearance though the phagocytic activity was not significantly altered. EO or 1,8-cineole pre-treatment attenuated LPS-induced inflammatory signaling pathways at various levels accompanied by diminished inflammatory response. Among the pattern recognition receptors (PRRs) involved in LPS signaling, the TREM pathway surface receptor (TREM-1) was significantly downregulated. Importantly, the pre-treatments significantly downregulated (P ≤0.01) the intracellular PRR receptor NLRP3 of the inflammasome, which is consistent with the decrease in IL-1β secretion. Of the shared downstream signaling cascade for these PRR pathways, there was significant attenuation of phosphorylation of the transcription factor NF-κB and p38 (but increased phosphorylation of the other two MAP kinases, ERK1/2 and JNK1/2). 1,8-cineole showed a similar general trend except for an opposite effect on NF-κB and JNK1/2. In this context, either pre-treatment caused a significant downregulation of MKP-1 phosphatase, a negative regulator of MAPKs. Collectively, our results demonstrate that the anti-inflammatory activity of EO and 1,8-cineole is modulated via selective downregulation of the PRR pathways, including PRR receptors (TREM-1 and NLRP3) and common downstream signaling cascade partners (NF-κB, MAPKs, MKP-1). To our knowledge, this is the first report on the modulatory role of TREM-1 and NLRP3 inflammasome pathways and the MAPK negative regulator MKP-1 in context of the anti-inflammatory potential of EO and its constituent 1,8-cineole.
Collapse
Affiliation(s)
- Niket Yadav
- Microbial Pathogenesis and Immunotoxicology Laboratory, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Harish Chandra
- Microbial Pathogenesis and Immunotoxicology Laboratory, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
16
|
Anwaier G, Chen C, Cao Y, Qi R. A review of molecular imaging of atherosclerosis and the potential application of dendrimer in imaging of plaque. Int J Nanomedicine 2017; 12:7681-7693. [PMID: 29089763 PMCID: PMC5656339 DOI: 10.2147/ijn.s142385] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Despite the fact that technological advancements have been made in diagnosis and treatment, cardiovascular diseases (CVDs) remain the leading cause of mortality and morbidity worldwide. Early detection of atherosclerosis (AS), especially vulnerable plaques, plays a crucial role in the prevention of acute coronary syndrome (ACS). Targeting the critical cytokines and molecules that are upregulated during the biological process of AS by in vivo molecular imaging has been widely used in plaque imaging. With their three-dimensional architecture, composition, and abundant terminal functional groups, dendrimers provide a platform for multitargeting and multimodal imaging. Thus, modified dendrimers with the key molecules upregulated in AS plaques will be an innovative attempt to achieve targeted imaging of AS plaques specifically and efficiently. This review was aimed to address some recent works on imaging of AS plaques using various types of image technology and further discuss the applications of dendrimers, an innovative yet seldom used method in imaging of AS plaques due to some limitations and challenges, and we highlight the bright future of the modified dendrimers in characterizing AS plaques.
Collapse
Affiliation(s)
- Gulinigaer Anwaier
- Peking University Institute of Cardiovascular Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of education, Peking University Health Science Center.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing.,School of Basic Medical Science, Shihezi University, Shihezi, Xinjiang, People's Republic of China
| | - Cong Chen
- Peking University Institute of Cardiovascular Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of education, Peking University Health Science Center.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing
| | - Yini Cao
- Peking University Institute of Cardiovascular Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of education, Peking University Health Science Center.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing
| | - Rong Qi
- Peking University Institute of Cardiovascular Sciences, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of education, Peking University Health Science Center.,Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Beijing.,School of Basic Medical Science, Shihezi University, Shihezi, Xinjiang, People's Republic of China
| |
Collapse
|
17
|
Lind M, Hayes A, Caprnda M, Petrovic D, Rodrigo L, Kruzliak P, Zulli A. Inducible nitric oxide synthase: Good or bad? Biomed Pharmacother 2017. [PMID: 28651238 DOI: 10.1016/j.biopha.2017.06.036] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Nitric oxide synthases (NOS) are a family of isoforms responsible for the synthesis of the potent dilator nitric oxide (NO). Expression of inducible NOS (iNOS) occurs in conditions of inflammation, and produces large amounts of NO. In pathological conditions iNOS is regarded as a harmful enzyme and is proposed to be a major contributor to diseases of the cardiovascular system such as atherosclerosis. In this review, we address the notion that iNOS is a detrimental enzyme in disease and discuss its potentially beneficial roles. Additionally, we describe other molecules associated with iNOS in diseases such as atherosclerosis, and current research on therapeutic inhibitors tested to reduced pathology associated with cardiovascular diseases (CVD).
Collapse
Affiliation(s)
- Maggie Lind
- Centre for Chronic Disease (CCD), College of Health & Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Alan Hayes
- Centre for Chronic Disease (CCD), College of Health & Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Martin Caprnda
- 1st Department of Internal Medicine, Faculty of Medicine, Comenius University and University Hospital, Bratislava, Slovakia
| | - Daniel Petrovic
- Institute of Histology and Embryology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Luis Rodrigo
- Faculty of Medicine, University of Oviedo, Central University Hospital of Asturias (HUCA), Oviedo, Spain
| | - Peter Kruzliak
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic; 2nd Department of Surgery, Centre of Vascular Diseases, Faculty of Medicine, Masaryk University and St. Anne´s Faculty Hospital, Brno, Czech Republic.
| | - Anthony Zulli
- Centre for Chronic Disease (CCD), College of Health & Biomedicine, Victoria University, Melbourne, Victoria, Australia.
| |
Collapse
|
18
|
Zhang H, Du J, Wang H, Wang H, Jiang J, Zhao J, Lu H. Comparison of diagnostic values of ultrasound micro-flow imaging and contrast-enhanced ultrasound for neovascularization in carotid plaques. Exp Ther Med 2017; 14:680-688. [PMID: 28672985 PMCID: PMC5488622 DOI: 10.3892/etm.2017.4525] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 05/17/2017] [Indexed: 12/12/2022] Open
Abstract
The aim of the present study was to compare the diagnostic values of ultrasound micro-flow imaging (SMI) and contrast-enhanced ultrasound (CEUS) for neovascularization in carotid plaques, and to investigate their capacities for predicting the risks of cerebral stroke. A total of 39 patients (64 carotid plaques) with severe carotid artery stenosis undergoing carotid endarterectomy were selected between February 2015 and February 2016, and SMI and CEUS were used to detect neovascularization in plaques. According to the CEUS dynamic graph of plaques, the enhanced intensity visual scales and contrast parameters were obtained. Carotid atherosclerotic plaques were divided into 4 groups. The differences in the enhanced intensity visual scales, contrast parameters, and gray-scale median (GSM) values among the 4 groups were analyzed. Carotid plaque tissue samples from patients were stained for CD34, and the consistency of the methods for the diagnosis of neovascularization in plaques was analyzed. The differences in GSM values, enhanced intensities, and enhanced densities among the 4 groups of plaques were statistically significant (F=29.365, χ2=29.025, χ2=30.871, P<0.001); the differences in enhanced intensities of carotid atherosclerotic plaques with different echo types were statistically significant (χ2=17.951, P<0.001). The enhanced intensity of plaques was negatively correlated with the GSM value (r=−0.376, P<0.01), and the enhanced density of plaques was negatively correlated with the GSM value (r=−0.252, P<0.01). SMI and CEUS grading had good consistency (κ=0.860>0), there were statistically significant differences in new vessel densities with different SMI gradings (P<0.001), and the clinical symptoms and severity were positively correlated with SMI grading (rs=0.592>0). In conclusion, SMI and CEUS have good consistency for evaluating neovascularization in carotid plaques, and have good clinical value for evaluating neovascularization in carotid plaques.
Collapse
Affiliation(s)
- Hongxue Zhang
- Department of UItrasonic Diagnosis, The Second Clinic, Institute of the Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Jianwen Du
- Department of UItrasonic Diagnosis, The Second Clinic, Institute of the Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Hong Wang
- Department of UItrasonic Diagnosis, The Second Clinic, Institute of the Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Haili Wang
- Department of UItrasonic Diagnosis, The Second Clinic, Institute of the Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Jianhui Jiang
- Department of UItrasonic Diagnosis, The Second Clinic, Institute of the Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Jingjie Zhao
- Department of Pathology, The Second Clinic, Institute of the Chengde Medical College, Chengde, Hebei 067000, P.R. China
| | - Huan Lu
- Department of UItrasonic Diagnosis, The Second Clinic, Institute of the Chengde Medical College, Chengde, Hebei 067000, P.R. China
| |
Collapse
|
19
|
Willcox BJ, Morris BJ, Tranah GJ, Chen R, Masaki KH, He Q, Willcox DC, Allsopp RC, Moisyadi S, Gerschenson M, Davy PMC, Poon LW, Rodriguez B, Newman AB, Harris TB, Cummings SR, Liu Y, Parimi N, Evans DS, Donlon TA. Longevity-Associated FOXO3 Genotype and its Impact on Coronary Artery Disease Mortality in Japanese, Whites, and Blacks: A Prospective Study of Three American Populations. J Gerontol A Biol Sci Med Sci 2017; 72:724-728. [PMID: 27694344 PMCID: PMC5964743 DOI: 10.1093/gerona/glw196] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 09/13/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND We recently reported that protection against coronary artery disease (CAD) mortality is the major contributor to longer life associated with FOXO3 genotype. The present study examined this relation in more detail. METHODS We performed a 15-year observational study of 3,584 older American men of Japanese ancestry from the Kuakini Honolulu Heart Program cohort and 1,595 White and 1,067 Black elderly individuals from the Health Aging and Body Composition study. RESULTS Multivariate Cox regression models demonstrated that carriage of the longevity-associated G allele of FOXO3 single nucleotide polymorphisms rs2802292 was a protective factor against CAD mortality in all three populations. In Japanese and Whites, but not in Blacks, the protective effect of the G allele was little changed in models adjusted for other major risk factors. Population-attributable risk (PAR) models found that the nonprotective TT genotype contributed 15%, 9%, and 3% to CAD mortality risk in Japanese, White, and Black Americans, respectively, and was one of the top three contributing factors to CAD mortality. In Japanese, this effect size was comparable with hypertension (15%), but in Whites and Blacks PAR for hypertension was higher (29% and 26%, respectively). G-allele carriers had lower plasma TNF-α than noncarriers, suggesting inflammation as a potential mediating factor for CAD mortality risk. CONCLUSION FOXO3 genotype is an important risk factor for CAD mortality in older populations. More research is needed to identify potential mechanisms and targets for intervention.
Collapse
Affiliation(s)
- Bradley J Willcox
- Department of Research, Kuakini Medical Center, Honolulu, Hawaii
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu
| | - Brian J Morris
- Department of Research, Kuakini Medical Center, Honolulu, Hawaii
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu
- School of Medical Sciences and Bosch Institute, University of Sydney, New South Wales, Australia
| | - Gregory J Tranah
- California Pacific Medical Center Research Institute, San Francisco
| | - Randi Chen
- Department of Research, Kuakini Medical Center, Honolulu, Hawaii
| | - Kamal H Masaki
- Department of Research, Kuakini Medical Center, Honolulu, Hawaii
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu
| | - Qimei He
- Department of Research, Kuakini Medical Center, Honolulu, Hawaii
| | - D Craig Willcox
- Department of Research, Kuakini Medical Center, Honolulu, Hawaii
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu
- Department of Human Welfare, Okinawa International University, Ginowan, Japan
| | | | - Stefan Moisyadi
- Institute for Biogenesis Research, University of Hawaii, Honolulu
| | - Mariana Gerschenson
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu
| | - Philip M C Davy
- Institute for Biogenesis Research, University of Hawaii, Honolulu
| | | | - Beatriz Rodriguez
- Department of Research, Kuakini Medical Center, Honolulu, Hawaii
- Department of Geriatric Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu
| | - Anne B Newman
- Department of Epidemiology, University of Pittsburgh, Pennsylvania
| | - Tamara B Harris
- Laboratory of Neurogenetics, Intramural Research Program, National Institute on Aging, Bethesda, Maryland
| | | | - Yongmei Liu
- Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Neeta Parimi
- California Pacific Medical Center Research Institute, San Francisco
| | - Daniel S Evans
- California Pacific Medical Center Research Institute, San Francisco
| | - Timothy A Donlon
- Department of Research, Kuakini Medical Center, Honolulu, Hawaii
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu
- Department of Pathology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii
| |
Collapse
|
20
|
Wang L, Chen Q, Ke D, Li G. Ghrelin inhibits atherosclerotic plaque angiogenesis and promotes plaque stability in a rabbit atherosclerotic model. Peptides 2017; 90:17-26. [PMID: 28189525 DOI: 10.1016/j.peptides.2017.01.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 01/16/2017] [Accepted: 01/27/2017] [Indexed: 12/17/2022]
Abstract
Intraplaque angiogenesis associates with the instability of atherosclerotic plaques. In the present study, we investigated the effects of ghrelin on intraplaque angiogenesis and plaque instability in a rabbit model of atherosclerosis. The rabbits were randomly divided into three groups, namely, the control group, atherosclerotic model group, and ghrelin-treated group, with treatments lasting for 4 weeks. We found that the thickness ratio of the intima to media in rabbits of the ghrelin-treated group was significantly lower than that in rabbits of the atherosclerotic model group. The number of neovessels and the levels of vascular endothelial growth factor (VEGF) and vascular endothelial growth factor receptor 2 (VEGFR2) decreased dramatically in rabbits of the ghrelin-treated group compared to those of the atherosclerotic model group. Ghrelin significantly decreased the plaque content of macrophages, matrix metalloproteinase (MMP)-2, and MMP-9, in a rabbit model of atherosclerosis. In addition, the level of the pro-inflammatory factor monocyte chemoattractant protein (MCP)-1 was significantly lower in rabbits of the ghrelin-treated group than in rabbits of the atherosclerotic model group. In summary, ghrelin can inhibit intraplaque angiogenesis and promote plaque stability by down-regulating VEGF and VEGFR2 expression, inhibiting the plaque content of macrophages, and reducing MCP-1 expression at an advanced stage of atherosclerosis in rabbits.
Collapse
Affiliation(s)
- Li Wang
- Department of Geriatrics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Qingwei Chen
- Department of Geriatrics, The Second Affiliated Hospital, Chongqing University of Medical Sciences, Chongqing, 400010, China.
| | - Dazhi Ke
- Department of Geriatrics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Guiqiong Li
- Department of Geriatrics, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| |
Collapse
|
21
|
Xu T, Liu S, Ma T, Jia Z, Zhang Z, Wang A. Aldehyde dehydrogenase 2 protects against oxidative stress associated with pulmonary arterial hypertension. Redox Biol 2016; 11:286-296. [PMID: 28030785 PMCID: PMC5192477 DOI: 10.1016/j.redox.2016.12.019] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 12/09/2016] [Accepted: 12/17/2016] [Indexed: 12/18/2022] Open
Abstract
The cardioprotective benefits of aldehyde dehydrogenase 2 (ALDH2) are well established, although the regulatory role of ALDH2 in vascular remodeling in pulmonary arterial hypertension (PAH) is largely unknown. ALDH2 potently regulates the metabolism of aldehydes such as 4-hydroxynonenal (4-HNE), the endogenous product of lipid peroxidation. Thus, we hypothesized that ALDH2 ameliorates the proliferation and migration of human pulmonary artery smooth muscle cells (HPASMCs) by inhibiting 4-HNE accumulation and regulating downstream signaling pathways, thereby ameliorating pulmonary vascular remodeling. We found that low concentrations of 4-HNE (0.1 and 1μM) stimulated cell proliferation by enhancing cyclin D1 and c-Myc expression in primary HPASMCs. Low 4-HNE concentrations also enhanced cell migration by activating the nuclear factor kappa B (NF-κB) signaling pathway, thereby regulating matrix metalloprotein (MMP)-9 and MMP2 expression in vitro. In vivo, Alda-1, an ALDH2 agonist, significantly stimulated ALDH2 activity, reducing elevated 4-HNE and malondialdehyde levels and right ventricular systolic pressure in a monocrotaline-induced PAH animal model to the level of control animals. Our findings indicate that 4-HNE plays an important role in the abnormal proliferation and migration of HPASMCs, and that ALDH2 activation can attenuate 4-HNE-induced PASMC proliferation and migration, possibly by regulating NF-κB activation, in turn ameliorating vascular remodeling in PAH. This mechanism might reflect a new molecular target for treating PAH.
Collapse
Affiliation(s)
- Tao Xu
- Life Science Institute, Jinzhou Medical University, Jinzhou, Liaoning 121000, PR China.
| | - Shuangyue Liu
- Department of Physiology, Jinzhou Medical University, Jinzhou, Liaoning 121000, PR China
| | - Tingting Ma
- Department of Physiology, Jinzhou Medical University, Jinzhou, Liaoning 121000, PR China
| | - Ziyi Jia
- College of Economics and Management, Huazhong Agricultural University, Wuhan, Hubei 430070, PR China
| | - Zhifei Zhang
- Department of Physiology and Pathophysiology, Capital Medical University, School of Basic Medical Sciences, Beijing 100069, PR China
| | - Aimei Wang
- Department of Physiology, Jinzhou Medical University, Jinzhou, Liaoning 121000, PR China.
| |
Collapse
|
22
|
Li W, Li J, Hao P, Chen W, Meng X, Li H, Zhang Y, Zhang C, Yang J. Imbalance between angiotensin II and angiotensin-(1-7) in human coronary atherosclerosis. J Renin Angiotensin Aldosterone Syst 2016; 17:17/3/1470320316659618. [PMID: 27432541 PMCID: PMC5843867 DOI: 10.1177/1470320316659618] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 05/24/2016] [Indexed: 11/24/2022] Open
Abstract
Objective: Our previous studies found that angiotensin-(1–7) (Ang-(1–7)) is an endogenous counter-factor of angiotensin II (Ang-II). However, the balance between Ang-II and Ang-(1–7) in the development of human coronary atherosclerosis is not determined. Methods and results: The plasma levels of Ang-II and Ang-(1–7) were detected by enzyme-linked immunosorbent assay (ELISA) in 112 patients with known or suspected coronary artery disease (CAD) undergoing coronary angiography. Patients were divided into three groups based on the coronary angiography as follows: (1) normal (n = 13); (2) noncritical CAD (<50% stenosis, n = 17); and (3) critical CAD (⩾50% stenosis, n = 82). The plasma levels of Ang-II, Ang-(1–7) and the ratio of Ang-II and Ang-(1–7) (Ang-II/Ang-(1–7) were comparable between the normal and noncritical CAD groups. However, Ang-II, Ang-(1–7), and especially Ang-II/Ang-(1–7), were elevated in patients with critical CAD, compared with patients with normal or noncritical CAD. The level of Ang-II/Ang-(1–7) was positively associated with serious coronary stenosis, and correlated with tumor necrosis factor-alpha (TNF-α) level. Conclusion: Both Ang-II and Ang-(1–7) expression are significantly increased in patients with critical CAD. However, increased Ang-II/Ang-(1–7) ratios may lead to Ang-II over-activation and aggravate atherosclerosis progression.
Collapse
Affiliation(s)
- Wenjing Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, P.R. China Fine Arts School of Shandong University, P.R. China
| | - Jifu Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, P.R. China
| | - Panpan Hao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, P.R. China
| | - Wenqiang Chen
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, P.R. China
| | - Xiao Meng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, P.R. China
| | - Hongxuan Li
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, P.R. China
| | - Yun Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, P.R. China
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, P.R. China
| | - Jianmin Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, P.R. China
| |
Collapse
|