1
|
Tufaro V, Serruys PW, Räber L, Bennett MR, Torii R, Gu SZ, Onuma Y, Mathur A, Baumbach A, Bourantas CV. Intravascular imaging assessment of pharmacotherapies targeting atherosclerosis: advantages and limitations in predicting their prognostic implications. Cardiovasc Res 2023; 119:121-135. [PMID: 35394014 DOI: 10.1093/cvr/cvac051] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 02/17/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Intravascular imaging has been often used over the recent years to examine the efficacy of emerging therapies targeting plaque evolution. Serial intravascular ultrasound, optical coherence tomography, or near-infrared spectroscopy-intravascular ultrasound studies have allowed us to evaluate the effects of different therapies on plaque burden and morphology, providing unique mechanistic insights about the mode of action of these treatments. Plaque burden reduction, a decrease in necrotic core component or macrophage accumulation-which has been associated with inflammation-and an increase in fibrous cap thickness over fibroatheromas have been used as surrogate endpoints to assess the value of several drugs in inhibiting plaque evolution and improving clinical outcomes. However, some reports have demonstrated weak associations between the effects of novel treatments on coronary atheroma and composition and their prognostic implications. This review examines the value of invasive imaging in assessing pharmacotherapies targeting atherosclerosis. It summarizes the findings of serial intravascular imaging studies assessing the effects of different drugs on atheroma burden and morphology and compares them with the results of large-scale trials evaluating their impact on clinical outcome. Furthermore, it highlights the limited efficacy of established intravascular imaging surrogate endpoints in predicting the prognostic value of these pharmacotherapies and introduces alternative imaging endpoints based on multimodality/hybrid intravascular imaging that may enable more accurate assessment of the athero-protective and prognostic effects of emerging therapies.
Collapse
Affiliation(s)
- Vincenzo Tufaro
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | | | - Lorenz Räber
- Department of Cardiology, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Ryo Torii
- Department of Mechanical Engineering, University College London, London, UK
| | - Sophie Zhaotao Gu
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Yoshinobu Onuma
- Department of Cardiology, National University of Ireland, Galway, Ireland
| | - Anthony Mathur
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Andreas Baumbach
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
- Yale University School of Medicine, New Haven, CT, USA
| | - Christos Vasileios Bourantas
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
- Institute of Cardiovascular Sciences, University College London, London, UK
| |
Collapse
|
2
|
Chen J, Zhang H, Li L, Zhang X, Zhao D, Wang L, Wang J, Yang P, Sun H, Liu K, Chen W, Li L, Lin F, Li Z, Chen YE, Zhang J, Pang D, Ouyang H, He Y, Fan J, Tang X. Lp-PLA 2 (Lipoprotein-Associated Phospholipase A 2) Deficiency Lowers Cholesterol Levels and Protects Against Atherosclerosis in Rabbits. Arterioscler Thromb Vasc Biol 2023; 43:e11-e28. [PMID: 36412196 DOI: 10.1161/atvbaha.122.317898] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND Elevated plasma Lp-PLA2 (lipoprotein-associated phospholipase A2) activity is closely associated with an increased risk of cardiovascular events. However, whether and how Lp-PLA2 is directly involved in the pathogenesis of atherosclerosis is still unclear. To examine the hypothesis that Lp-PLA2 could be a potential preventative target of atherosclerosis, we generated Lp-PLA2 knockout rabbits and investigated the pathophysiological functions of Lp-PLA2. METHODS Lp-PLA2 knockout rabbits were generated using CRISPR/Cas9 system to assess the role of Lp-PLA2 in plasma lipids regulation and identify its underlying molecular mechanisms. Homozygous knockout rabbits along with wild-type rabbits were fed a cholesterol-rich diet for up to 14 weeks and their atherosclerotic lesions were compared. Moreover, the effects of Lp-PLA2 deficiency on the key cellular behaviors in atherosclerosis were assessed in vitro. RESULTS When rabbits were fed a standard diet, Lp-PLA2 deficiency led to a significant reduction in plasma lipids. The decreased protein levels of SREBP2 (sterol regulatory element-binding protein 2) and HMGCR (3-hydroxy-3-methylglutaryl coenzyme A reductase) in livers of homozygous knockout rabbits indicated that the cholesterol biosynthetic pathway was impaired with Lp-PLA2 deficiency. In vitro experiments further demonstrated that intracellular Lp-PLA2 efficiently enhanced SREBP2-related cholesterol biosynthesis signaling independently of INSIGs (insulin-induced genes). When fed a cholesterol-rich diet, homozygous knockout rabbits exhibited consistently lower level of hypercholesterolemia, and their aortic atherosclerosis lesions were significantly reduced by 60.2% compared with those of wild-type rabbits. The lesions of homozygous knockout rabbits were characterized by reduced macrophages and the expression of inflammatory cytokines. Macrophages of homozygous knockout rabbits were insensitive to M1 polarization and showed reduced DiI-labeled lipoprotein uptake capacity compared with wild-type macrophages. Lp-PLA2 deficiency also inhibited the adhesion between monocytes and endothelial cells. CONCLUSIONS These results demonstrate that Lp-PLA2 plays a causal role in regulating blood lipid homeostasis and Lp-PLA2 deficiency protects against dietary cholesterol-induced atherosclerosis in rabbits. Lp-PLA2 could be a potential target for the prevention of atherosclerosis.
Collapse
Affiliation(s)
- Jiahuan Chen
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Huanyu Zhang
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Linquan Li
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Xinwei Zhang
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Dazhong Zhao
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Lingyu Wang
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Jiaqi Wang
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Ping Yang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China (P.Y., H.S., K.L., W.C., Y.H.)
| | - Huan Sun
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China (P.Y., H.S., K.L., W.C., Y.H.)
| | - Kun Liu
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China (P.Y., H.S., K.L., W.C., Y.H.)
| | - Weiwei Chen
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China (P.Y., H.S., K.L., W.C., Y.H.)
| | - Lin Li
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Feng Lin
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Zhanjun Li
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.)
| | - Y Eugene Chen
- Department of Internal Medicine, Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor (Y.E.C., J.Z.)
| | - Jifeng Zhang
- Department of Internal Medicine, Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor (Y.E.C., J.Z.)
| | - Daxin Pang
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.).,Chongqing Research Institute, Jilin University, Chongqing, China (D.P., H.O., X.T.)
| | - Hongsheng Ouyang
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.).,Chongqing Research Institute, Jilin University, Chongqing, China (D.P., H.O., X.T.)
| | - Yuquan He
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China (P.Y., H.S., K.L., W.C., Y.H.)
| | - Jianglin Fan
- Department of Molecular Pathology, Faculty of Medicine, Graduate School of Medical Sciences, University of Yamanashi, Japan (J.F.)
| | - Xiaochun Tang
- College of Animal Sciences, Jilin University, Changchun, Jilin Province, China (J.C., H.Z., Linquan Li, X.Z., D.Z., L.W., J.W., Lin Li, F.L., Z.L., D.P., H.O., X.T.).,Chongqing Research Institute, Jilin University, Chongqing, China (D.P., H.O., X.T.)
| |
Collapse
|
3
|
Pantazi D, Tellis C, Tselepis AD. Oxidized phospholipids and lipoprotein-associated phospholipase A 2 (Lp-PLA 2 ) in atherosclerotic cardiovascular disease: An update. Biofactors 2022; 48:1257-1270. [PMID: 36192834 DOI: 10.1002/biof.1890] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/06/2022] [Indexed: 12/24/2022]
Abstract
Inflammation and oxidative stress conditions lead to a variety of oxidative modifications of lipoprotein phospholipids implicated in the occurrence and development of atherosclerotic lesions. Lipoprotein-associated phospholipase A2 (Lp-PLA2 ) is established as an independent risk biomarker of atherosclerosis-related cardiovascular disease (ASCVD) and mediates vascular inflammation through the regulation of lipid metabolism in the blood and in atherosclerotic lesions. Lp-PLA2 is associated with low- and high-density lipoproteins and Lipoprotein (a) in human plasma and specifically hydrolyzes oxidized phospholipids involved in oxidative stress modification. Several oxidized phospholipids (OxPLs) subspecies can be detoxified through enzymatic degradation by Lp-PLA2 activation, forming lysophospholipids and oxidized non-esterified fatty acids (OxNEFAs). Lysophospholipids promote the expression of adhesion molecules, stimulate cytokines production (TNF-α, IL-6), and attract macrophages to the arterial intima. The present review article discusses new data on the functional roles of OxPLs and Lp-PLA2 associated with lipoproteins.
Collapse
Affiliation(s)
- Despoina Pantazi
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Constantinos Tellis
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| | - Alexandros D Tselepis
- Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, Ioannina, Greece
| |
Collapse
|
4
|
Wang J, Jiang M, Xiong A, Zhang L, Luo L, Liu Y, Liu S, Ran Q, Wu D, Xiong Y, He X, Leung ELH, Li G. Integrated analysis of single-cell and bulk RNA sequencing reveals pro-fibrotic PLA2G7 high macrophages in pulmonary fibrosis. Pharmacol Res 2022; 182:106286. [PMID: 35662628 DOI: 10.1016/j.phrs.2022.106286] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/09/2022] [Accepted: 05/29/2022] [Indexed: 10/18/2022]
Abstract
Pulmonary fibrosis (PF) is the pathological change of end-stage interstitial lung diseases with high mortality and limited therapeutic options. Lung macrophages have distinct subsets with divergent functions, and play critical roles in the pathogenesis of PF. In this study, integrative analysis of lung single-cell and bulk RNA-seq data from patients with fibrotic hypersensitivity pneumonitis and idiopathic pulmonary fibrosis was utilized to identify particular macrophage subsets during the development of PF. We find a specific macrophage subpopulation highly expressing PLA2G7 in fibrotic lungs. We performed additional single-cell RNA-seq analysis to identify analogous macrophage population in bleomycin (BLM)-induced mouse pulmonary fibrosis models. By in vitro and in vivo experiments, we further reveal the pro-fibrotic role for this PLA2G7high macrophage subset in fibroblast-to-myofibroblast transition (FMT) during pulmonary fibrosis. PLA2G7 promotes FMT via LPC/ATX/LPA/LPA2 axis in macrophages. Moreover, PLA2G7 is regulated by STAT1, and pharmacological inhibition of PLA2G7 by Darapladib ameliorates pulmonary fibrosis in BLM-induced mice. The results of this study support the view that PLA2G7high macrophage subpopulation contributes importantly to the pathogenesis of PF, which provides a potential way for targeted therapy.
Collapse
Affiliation(s)
- Junyi Wang
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China; Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Chengdu, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science & Technology, Taipa, Macao Special Administrative Region of China
| | - Manling Jiang
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China; Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Chengdu, China
| | - Anying Xiong
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China; Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Chengdu, China
| | - Lei Zhang
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China; Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Chengdu, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science & Technology, Taipa, Macao Special Administrative Region of China
| | - Li Luo
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Yao Liu
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Shengbin Liu
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Qin Ran
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China; Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Chengdu, China
| | - Dehong Wu
- Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Chengdu, China
| | - Ying Xiong
- Department of Pulmonary and Critical Care Medicine, Sichuan Friendship Hospital, Chengdu, China
| | - Xiang He
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China; Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Chengdu, China.
| | - Elaine Lai-Han Leung
- Faculty of Health Sciences, University of Macau, Taipa, Macao Special Administrative Region of China.
| | - Guoping Li
- Laboratory of Allergy and Precision Medicine, Chengdu Institute of Respiratory Health, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu, China; Department of Pulmonary and Critical Care Medicine, Chengdu Institute of Respiratory Health, Chengdu Third People's Hospital Branch of National Clinical Research Center for Respiratory Disease, Chengdu, China.
| |
Collapse
|
5
|
Liu X, Zhou W, Fan W, Li A, Pang J, Chen Z, Li X, Hu X, Zeng Y, Tang L. The benefit of exercise rehabilitation guided by 6-minute walk test on lipoprotein-associated phospholipase A2 in patients with coronary heart disease undergoing percutaneous coronary intervention: a prospective randomized controlled study. BMC Cardiovasc Disord 2022; 22:177. [PMID: 35430800 PMCID: PMC9014591 DOI: 10.1186/s12872-021-02430-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 12/14/2021] [Indexed: 11/22/2022] Open
Abstract
Background Lipoprotein-associated phospholipase A2 (Lp-PLA2) has been taken as a biomarker of inflammation in patients with acute coronary diseases. Regular exercise rehabilitation could attenuate inflammation and promote the rehabilitation of coronary heart disease (CHD). The level of Lp-PLA2 is negatively correlated with 6-min walk test (6-MWT). The exercise prescription of appropriate intensity is the basis of exercise rehabilitation. 6-MWT is associated with maximal oxygen consumption, and can be used to determine the intensity of exercise prescription guiding patients how to do exercise rehabilitation. The aim of this study was to observe the benefit of 6-MWT guided exercise rehabilitation on the level of Lp-PLA2 in patients with CHD undergoing percutaneous coronary intervention (PCI). Methods We prospectively, consecutively enrolled 100 patients between Dec 2018 and Dec 2020 in the fourth ward of the Department of Cardiology, Yuebei People's Hospital Affiliated to Shantou University. Eligible patients were 1:1 divided into Group A, with no exercise rehabilitation, and Group B, with regular exercise rehabilitation, using random number table method of simple randomization allocation. Clinical data such as general information, the profile of lipids and the level of Lp-PLA2 were collected at baseline and at 12-week follow-up. Results There were no statistically significant differences of the percentages of gender, hypertension, type-2 diabetes mellitus (T2DM), the profile of lipids and level of Lp-PLA2 between the groups at baseline (P > 0.05). The level of Lp-PLA2 decreased at 12-week follow-up, moreover, the decline of the Lp-PLA2 level in Group B was more significant than that in Group A (t = 2.875, P = 0.005). Multivariate linear regression analysis indicated that exercise rehabilitation was independently correlated with the level of Lp-PLA2 (β′ = − 0.258, t = − 2.542, P = 0.013). Conclusion Exercise rehabilitation for 12 weeks guided by 6-MWT can further reduce the level of LP-PLA2 in patients with CHD undergoing PCI. Trial registration This trial was registered on the Chinese Clinical Trial Registry: ChiCTR2100048124, registered 3 July 2021- Retrospectively registered. The study protocol adheres to the CONSORT guidelines.
Collapse
|
6
|
Koutsogianni AD, Liberopoulos E, Tellis K, Tselepis AD. Oxidized phospholipids and lipoprotein(a): An update. Eur J Clin Invest 2022; 52:e13710. [PMID: 34837383 DOI: 10.1111/eci.13710] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/21/2021] [Accepted: 10/21/2021] [Indexed: 12/17/2022]
Abstract
Over the past few years, there has been an undiminished interest in lipoprotein(a) [Lp(a)] and oxidized phospholipids (OxPLs), mainly carried on this lipoprotein. Elevated Lp(a) has been established as an independent causal risk factor for cardiovascular disease. OxPLs play an important role in atherosclerosis. The main questions that remain to be answered, however, is to what extent OxPLs contribute to the atherogenicity of Lp(a), what effect hypolipidaemic medications may have on their levels and the potential clinical benefit of their reduction. This narrative review aimed to summarize currently available data on OxPLs and cardiovascular risk, as well as the effect of established and emerging hypolipidaemic medications on Lp(a)-OxPLs.
Collapse
Affiliation(s)
| | - Evangelos Liberopoulos
- Department of Internal Medicine, Faculty of Medicine, University of Ioannina, Ioannina, Greece
| | - Konstantinos Tellis
- Department of Chemistry, Atherothrombosis Research Centre/Laboratory of Biochemistry, University of Ioannina, Ioannina, Greece
| | - Alexandros D Tselepis
- Department of Chemistry, Atherothrombosis Research Centre/Laboratory of Biochemistry, University of Ioannina, Ioannina, Greece
| |
Collapse
|
7
|
Patoulias D, Stavropoulos K, Imprialos K, Athyros V, Grassos H, Doumas M, Faselis C. Inflammatory Markers in Cardiovascular Disease; Lessons Learned and Future Perspectives. Curr Vasc Pharmacol 2021; 19:323-342. [PMID: 32188386 DOI: 10.2174/1570161118666200318104434] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 02/24/2020] [Accepted: 02/24/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cardiovascular disease (CVD) still remains the leading cause of morbidity and mortality worldwide. It is now established that inflammation plays a crucial role in atherosclerosis and atherothrombosis, and thus, it is closely linked to cardiovascular disease. OBJECTIVE The aim of the present review is to summarize and critically appraise the most relevant evidence regarding the potential use of inflammatory markers in the field of CVD. METHODS We conducted a comprehensive research of the relevant literature, searching MEDLINE from its inception until November 2018, primarily for meta-analyses, randomized controlled trials and observational studies. RESULTS Established markers of inflammation, mainly C-reactive protein, have yielded significant results both for primary and secondary prevention of CVD. Newer markers, such as lipoprotein-associated phospholipase A2, lectin-like oxidized low-density lipoprotein receptor-1, cytokines, myeloperoxidase, cell adhesion molecules, matrix metalloproteinases, and the CD40/CD40 ligand system, have been largely evaluated in human studies, enrolling both individuals from the general population and patients with established CVD. Some markers have yielded conflicting results; however, others are now recognized not only as promising biomarkers of CVD, but also as potential therapeutic targets, establishing the role of anti-inflammatory and pleiotropic drugs in CVD. CONCLUSION There is significant evidence regarding the role of consolidated and novel inflammatory markers in the field of diagnosis and prognosis of CVD. However, multimarker model assessment, validation of cut-off values and cost-effectiveness analyses are required in order for those markers to be integrated into daily clinical practice.
Collapse
Affiliation(s)
- Dimitrios Patoulias
- Second Propedeutic Department of Internal Medicine, Aristotle University, Thessaloniki, Greece
| | | | - Konstantinos Imprialos
- Second Propedeutic Department of Internal Medicine, Aristotle University, Thessaloniki, Greece
| | - Vasilios Athyros
- Second Propedeutic Department of Internal Medicine, Aristotle University, Thessaloniki, Greece
| | | | - Michael Doumas
- Second Propedeutic Department of Internal Medicine, Aristotle University, Thessaloniki, Greece
| | - Charles Faselis
- VA Medical Center, and George Washington University, Washington, DC 20422, United States
| |
Collapse
|
8
|
Yeo KP, Lim HY, Angeli V. Leukocyte Trafficking via Lymphatic Vessels in Atherosclerosis. Cells 2021; 10:cells10061344. [PMID: 34072313 PMCID: PMC8229118 DOI: 10.3390/cells10061344] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 02/03/2023] Open
Abstract
In recent years, lymphatic vessels have received increasing attention and our understanding of their development and functional roles in health and diseases has greatly improved. It has become clear that lymphatic vessels are critically involved in acute and chronic inflammation and its resolution by supporting the transport of immune cells, fluid, and macromolecules. As we will discuss in this review, the involvement of lymphatic vessels has been uncovered in atherosclerosis, a chronic inflammatory disease of medium- and large-sized arteries causing deadly cardiovascular complications worldwide. The progression of atherosclerosis is associated with morphological and functional alterations in lymphatic vessels draining the diseased artery. These defects in the lymphatic vasculature impact the inflammatory response in atherosclerosis by affecting immune cell trafficking, lymphoid neogenesis, and clearance of macromolecules in the arterial wall. Based on these new findings, we propose that targeting lymphatic function could be considered in conjunction with existing drugs as a treatment option for atherosclerosis.
Collapse
|
9
|
Abstract
Significance: Coronary artery disease (CAD) continues to be a leading cause of morbidity and mortality across the world despite significant progress in the prevention, diagnosis, and treatment of atherosclerotic disease. Recent Advances: The focus of the cardiovascular community has shifted toward seeking a better understanding of the inflammatory mechanisms driving residual CAD risk that is not modulated by current therapies. Significant progress has been achieved in revealing both proinflammatory and anti-inflammatory mechanisms, and how shift of the balance in favor of the former can drive the development of disease. Critical Issues: Advances in the noninvasive detection of coronary artery inflammation have been forthcoming. These advances include multiple imaging modalities, with novel applications of computed tomography both with and without positron emission tomography, and experimental ultrasound techniques. These advances will enable better selection of patients for anti-inflammatory treatments and assessment of treatment response. The rapid advancement in pharmaceutical design has enabled the production of specific antibodies against inflammatory pathways of atherosclerosis, with modest success to date. The pursuit of demonstrating the efficacy and safety of novel anti-inflammatory and/or proinflammatory resolution therapies for atherosclerotic CAD has become a major focus. Future Directions: This review seeks to provide an update of the latest evidence of all three of these highly related but disparate areas of inquiry: Our current understanding of the key mechanisms by which inflammation contributes to coronary artery atherosclerosis, the evidence for noninvasive assessment of coronary artery inflammation, and finally, the evidence for targeted therapies to treat coronary inflammation for the reduction of CAD risk. Antioxid. Redox Signal. 34, 1217-1243.
Collapse
Affiliation(s)
- Henry W West
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
10
|
Kreutzer FP, Meinecke A, Schmidt K, Fiedler J, Thum T. Alternative strategies in cardiac preclinical research and new clinical trial formats. Cardiovasc Res 2021; 118:746-762. [PMID: 33693475 PMCID: PMC7989574 DOI: 10.1093/cvr/cvab075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 03/03/2021] [Indexed: 02/07/2023] Open
Abstract
An efficient and safe drug development process is crucial for the establishment of new drugs on the market aiming to increase quality of life and life-span of our patients. Despite technological advances in the past decade, successful launches of drug candidates per year remain low. We here give an overview about some of these advances and suggest improvements for implementation to boost preclinical and clinical drug development with a focus on the cardiovascular field. We highlight advantages and disadvantages of animal experimentation and thoroughly review alternatives in the field of three-dimensional cell culture as well as preclinical use of spheroids and organoids. Microfluidic devices and their potential as organ-on-a-chip systems, as well as the use of living animal and human cardiac tissues are additionally introduced. In the second part, we examine recent gold standard randomized clinical trials and present possible modifications to increase lead candidate throughput: adaptive designs, master protocols, and drug repurposing. In silico and N-of-1 trials have the potential to redefine clinical drug candidate evaluation. Finally, we briefly discuss clinical trial designs during pandemic times.
Collapse
Affiliation(s)
- Fabian Philipp Kreutzer
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Anna Meinecke
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Kevin Schmidt
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Jan Fiedler
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.,Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany.,Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| |
Collapse
|
11
|
Mourouzis K, Siasos G, Oikonomou E, Zaromitidou M, Tsigkou V, Antonopoulos A, Bletsa E, Stampouloglou P, Vlasis K, Vavuranakis M, Tousoulis D. Lipoprotein-associated phospholipase A2 levels, endothelial dysfunction and arterial stiffness in patients with stable coronary artery disease. Lipids Health Dis 2021; 20:12. [PMID: 33583415 PMCID: PMC7883455 DOI: 10.1186/s12944-021-01438-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/27/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Lipoprotein-associated Phospholipase A2 (Lp-PLA2), can exert proinflammatory as well as proatherogenic properties on the vascular wall. The current study sought to evaluate the influence of high Lp-PLA2 levels on indices of arterial wall properties in patients with stable coronary artery disease (CAD). METHODS Three hundred seventy-four consecutive patients with stable CAD (mean age 61 ± 11 years, 89% males) were enrolled in this single-center cross-sectional study. Flow-mediated dilation (FMD) was used to assess endothelial function and augmentation index (AIx) of the central aortic pressure was used to assess reflected waves. ELISA was used to determine Lp-PLA2 serum levels. RESULTS After dividing the participants in 3 equal groups based on the tertiles of circulating Lp-PLA2 values, no significant differences were demonstrated between those in the 3rd tertile with Lp-PLA2 values > 138 μg/L, in the 2nd tertile with Lp-PLA2 values between 101 and 138 μg/L and in the 1st tertile (Lp-PLA2 values < 101 μg/L) regarding age, male gender, smoking habits, family history of CAD or history of a previous myocardial infarction, diabetes mellitus, arterial hypertension, hyperlipidemia, duration of CAD and treatment with relevant medication. Importantly, subjects with Lp-PLA2 values in the highest tertile, had significantly reduced FMD values compared to the middle and lower tertile (4.43 ± 2.37% vs. 4.61 ± 1.97% vs. 5.20 ± 2.52% respectively, P = 0.03). Patients in the highest tertile of Lp-PLA2 values had significantly higher AIx values (24.65 ± 8.69% vs. 23.33 ± 9.65%, P = 0.03), in comparison to the lowest tertile, with Lp-PLA2 values < 101 μg/L. A linear regression analysis showed that Lp-PLA2 values > 138 μg/L negatively correlated to FMD [b = - 0.45 (95% CI: - 0.79 - -0.11), P = 0.01] and AIx values [b = 1.81 (95% CI: 0.57-3.05), P < 0.001] independently of cofounders like gender, age, diabetes mellitus, arterial hypertension, dyslipidemia, smoking habits, family history of CAD, history of previous myocardial infarction, serum glucose, circulating lipid levels, duration of CAD, antihypertensive medication, antidiabetic drugs, statin therapy and treatment with β-blockers. CONCLUSIONS Elevated Lp-PLA2 levels relate to endothelial dysfunction and arterial stiffness in patients with stable CAD independently from classical risk factors for CAD, statin use, antihypertensive treatment, and duration of the disease.
Collapse
Affiliation(s)
- Konstantinos Mourouzis
- 1st Cardiology Department, Hippokration Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece.,Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Munich, Germany
| | - Gerasimos Siasos
- 1st Cardiology Department, Hippokration Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece. .,Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Evangelos Oikonomou
- 1st Cardiology Department, Hippokration Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Marina Zaromitidou
- 1st Cardiology Department, Hippokration Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Vicky Tsigkou
- 1st Cardiology Department, Hippokration Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Alexis Antonopoulos
- 1st Cardiology Department, Hippokration Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Evanthia Bletsa
- 1st Cardiology Department, Hippokration Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiota Stampouloglou
- 1st Cardiology Department, Hippokration Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Vlasis
- Department of Anatomy, Laiko General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Manolis Vavuranakis
- 1st Cardiology Department, Hippokration Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitris Tousoulis
- 1st Cardiology Department, Hippokration Hospital, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
12
|
Fras Z, Tršan J, Banach M. On the present and future role of Lp-PLA 2 in atherosclerosis-related cardiovascular risk prediction and management. Arch Med Sci 2021; 17:954-964. [PMID: 34336025 PMCID: PMC8314407 DOI: 10.5114/aoms.2020.98195] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 02/02/2020] [Indexed: 12/23/2022] Open
Abstract
Circulating concentration and activity of secretory phospholipase A2 (sPLA2) and lipoprotein-associated phospholipase A2 (Lp-PLA2) have been proven as biomarkers of increased risk of atherosclerosis-related cardiovascular disease (ASCVD). Lp-PLA2 might be part of the atherosclerotic process and may contribute to plaque destabilisation through inflammatory activity within atherosclerotic lesions. However, all attempts to translate the inhibition of phospholipase into clinically beneficial ASCVD risk reduction, including in randomised studies, by either non-specific inhibition of sPLA2 (by varespladib) or specific Lp-PLA2 inhibition by darapladib, unexpectedly failed. This gives us a strong imperative to continue research aimed at a better understanding of how Lp-PLA2 and sPLA2 regulate vascular inflammation and atherosclerotic plaque development. From the clinical viewpoint there is a need to establish and validate the existing and emerging novel anti-inflammatory therapeutic strategies to fight against ASCVD development, by using potentially better animal models and differently designed clinical trials in humans.
Collapse
Affiliation(s)
- Zlatko Fras
- Centre for Preventive Cardiology, Department of Vascular Medicine, Division of Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Chair of Internal Medicine, Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Jure Tršan
- Centre for Preventive Cardiology, Department of Vascular Medicine, Division of Medicine, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Maciej Banach
- Department of Hypertension, Medical University of Lodz, Poland
- Polish Mother’s Memorial Hospital Research Institute, Lodz, Poland
| |
Collapse
|
13
|
Liu X, Tang L, Fan W, Li A, Pang J, Feng Y. The recent effects of small dose of folic acid on lipoprotein-associated phospholipase A2 and systolic blood pressure variability in coronary heart disease patients with hyperhomocysteinemia: A single-center prospective cohort study. Medicine (Baltimore) 2020; 99:e23573. [PMID: 33371087 PMCID: PMC7748170 DOI: 10.1097/md.0000000000023573] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 11/05/2020] [Indexed: 11/26/2022] Open
Abstract
To Investigate the recent effects of small dose of folic acid on lipoprotein-associated phospholipase A2 (LP-PLA2) and systolic blood pressure variability in coronary heart disease (CHD) patients with hyperhomocysteinemia.In this prospective cohort study, a total of 167 CHD patients with hyperhomocysteinemia were consecutively enrolled, and they were divided into Group A (without folic acid intervention, n = 99), Group B (with 0.4 mg of folic acid intervention, n = 34), Group C (0.8 mg of folic acid intervention, n = 34). General information, fasting blood glucose, and blood lipid, folic acid, homocysteine, Lp-PLA2, and blood pressure variability were compared among 3 groups. The above indicators were reviewed after 3 months of treatment.There were no statistically significant differences of age, gender, blood pressure, incidence of type 2 diabetes mellitus, fasting blood glucose, folic acid, homocysteine, Lp-PLA2, total cholesterol, 3 acyl glycerin, apolipoprotein B, lipoprotein (a), high density lipoprotein cholesterol, and low density lipoprotein cholesterol were found among 3 groups (P > .05); however, after being treated for 3 months, there was statistically significant difference in folic acid among 3 groups (P < .05), there was statistically significant difference in apolipoprotein A between Group A and Group B (t = 0.505, P = .039), and also between Group A and Group C (t = 0.052, P = .017). There were statistically significant differences in Lp-PLA2 (t = 24.320, P = .016) and systolic blood pressure variability (t = 0.154, P = .018) between Group A and Group C.For CHD patients with hyperhomocysteinemia, the higher dose (0.8 mg) of folic acid supplement was beneficial for increasing the apolipoprotein A, reducing the Lp-PLA2, and improving the systolic blood pressure variation, which might help to improve the prognosis in these patients.
Collapse
Affiliation(s)
| | | | | | | | | | - Yingjun Feng
- Department of Physical Diagnostics, Yue Bei People's Hospital Affiliated to Shantou University, Shaoguan, Guangdong, China
| |
Collapse
|
14
|
Zhuo S, Yuan C. Active site competition is the mechanism for the inhibition of lipoprotein-associated phospholipase A 2 by detergent micelles or lipoproteins and for the efficacy reduction of darapladib. Sci Rep 2020; 10:17232. [PMID: 33057060 PMCID: PMC7560626 DOI: 10.1038/s41598-020-74236-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 09/18/2020] [Indexed: 01/27/2023] Open
Abstract
Lipoprotein associated phospholipase A2 (Lp-PLA2) has been characterized for its interfacial activation as well as inhibition by detergent micelles and lipoprotein particles. The enzyme has been shown to bind on the surfaces of hydrophobic aggregates, such as detergent micelles, lipoprotein particles and even polystyrene latex nanobeads. Binding to hydrophobic aggregates stimulates the activity of Lp-PLA2 but may not be the necessary step for catalysis. However, at higher concentrations, detergent micelles, latex nanobeads or lipoprotein particles inhibit Lp-PLA2 possibly by blocking the access of substrates to the active site. The competition mechanism also blocks inhibitors such as darapladib binding to Lp-PLA2 and reduces the efficacy of the drug. Darapladib has very low solubility and mainly exists in solutions as complexes with detergents or lipoprotein particles. The inhibition of Lp-PLA2 by darapladib is dependent on many factors such as concentrations of detergents or lipoproteins, incubation time, as well as the order of mixing reaction components. The in vitro Lp-PLA2 activity assays used in clinical studies may not accurately reflect the residual Lp-PLA2 activity in vivo. Darapladib has been found mainly bound on HDL and albumin when it is incubated with human serum. However, Lp-PLA2 is more sensitive to darapladib when bound on LDL and relatively resistant to darapladib when bound on HDL. Therefore, high cholesterol levels may decrease the efficacy of darapladip and cause the drug to be less effective in high risk patients. Our study will help to design better inhibitors for Lp-PLA2. The discoveries also contribute to understanding the mechanism of interfacial activation and inhibition for Lp-PLA2 and provide a new concept for researchers in building better kinetic model for interfacial enzymes.
Collapse
Affiliation(s)
- Shaoqiu Zhuo
- Diazyme Laboratories, Inc, 12889 Gregg Ct., Poway, CA, 92064, USA. .,Bayer HealthCare, 800 Dwight Way, Berkeley, CA, 94710, USA.
| | - Chong Yuan
- Diazyme Laboratories, Inc, 12889 Gregg Ct., Poway, CA, 92064, USA
| |
Collapse
|
15
|
Shields GS, Spahr CM, Slavich GM. Psychosocial Interventions and Immune System Function: A Systematic Review and Meta-analysis of Randomized Clinical Trials. JAMA Psychiatry 2020; 77:1031-1043. [PMID: 32492090 PMCID: PMC7272116 DOI: 10.1001/jamapsychiatry.2020.0431] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
IMPORTANCE Recent estimates suggest that more than 50% of all deaths worldwide are currently attributable to inflammation-related diseases. Psychosocial interventions may represent a potentially useful strategy for addressing this global public health problem, but which types of interventions reliably improve immune system function, under what conditions, and for whom are unknown. OBJECTIVE To address this issue, we conducted a systematic review and meta-analysis of randomized clinical trials (RCTs) in which we estimated associations between 8 different psychosocial interventions and 7 markers of immune system function, and examined 9 potential moderating factors. DATA SOURCES PubMed, Scopus, PsycInfo, and ClinicalTrials.gov databases were systematically searched from February 1, 2017, to December 31, 2018, for all relevant RCTs published through December 31, 2018. STUDY SELECTION Eligible RCTs included a psychosocial intervention, immune outcome, and preintervention and postintervention immunologic assessments. Studies were independently examined by 2 investigators. Of 4621 studies identified, 62 were eligible and 56 included. DATA EXTRACTION AND SYNTHESIS Data were extracted and analyzed from January 1, 2019, to July 29, 2019. The Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guideline was followed. Data were extracted by 2 investigators who were blind to study hypotheses and analyses, and were then analyzed using robust variance estimation. Analysis included 8 psychosocial interventions (behavior therapy, cognitive therapy, cognitive behavior therapy [CBT], CBT plus additive treatment or mode of delivery that augmented the CBT, bereavement or supportive therapy, multiple or combined interventions, other psychotherapy, and psychoeducation), 7 immune outcomes (proinflammatory cytokine or marker levels, anti-inflammatory cytokine levels, antibody levels, immune cell counts, natural killer cell activity, viral load, and other immune outcomes), and 9 moderating factors (intervention type, intervention format, intervention length, immune marker type, basal vs stimulated markers, immune marker measurement timing, disease state or reason for treatment, age, and sex). MAIN OUTCOMES AND MEASURES The primary a priori outcomes were pretest-posttest-control (ppc) group effect sizes (ppc g) for the 7 immunologic outcomes investigated. RESULTS Across 56 RCTs and 4060 participants, psychosocial interventions were associated with enhanced immune system function (ppc g = 0.30, 95% CI, 0.21-0.40; t50.9 = 6.22; P < .001). Overall, being randomly assigned to a psychosocial intervention condition vs a control condition was associated with a 14.7% (95% CI, 5.7%-23.8%) improvement in beneficial immune system function and an 18.0% (95% CI, 7.2%-28.8%) decrease in harmful immune system function over time. These associations persisted for at least 6 months following treatment and were robust across age, sex, and intervention duration. These associations were most reliable for CBT (ppc g = 0.33, 95% CI, 0.19-0.47; t27.2 = 4.82; P < .001) and multiple or combined interventions (ppc g = 0.52, 95% CI, 0.17-0.88; t5.7 = 3.63; P = .01), and for studies that assessed proinflammatory cytokines or markers (ppc g = 0.33, 95% CI, 0.19-0.48; t25.6 = 4.70; P < .001). CONCLUSIONS AND RELEVANCE These findings suggest that psychosocial interventions are reliably associated with enhanced immune system function and may therefore represent a viable strategy for improving immune-related health.
Collapse
Affiliation(s)
| | - Chandler M. Spahr
- Department of Psychology, San Diego State University, San Diego, California
| | - George M. Slavich
- Cousins Center for Psychoneuroimmunology and Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles
| |
Collapse
|
16
|
Liberale L, Montecucco F, Schwarz L, Lüscher TF, Camici GG. Inflammation and cardiovascular diseases: lessons from seminal clinical trials. Cardiovasc Res 2020; 117:411-422. [PMID: 32666079 DOI: 10.1093/cvr/cvaa211] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/29/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
Inflammation has been long regarded as a key contributor to atherosclerosis. Inflammatory cells and soluble mediators play critical roles throughout arterial plaque development and accordingly, targeting inflammatory pathways effectively reduces atherosclerotic burden in animal models of cardiovascular (CV) diseases. Yet, clinical translation often led to inconclusive or even contradictory results. The Canakinumab Anti-inflammatory Thrombosis Outcome Study (CANTOS) followed by the Colchicine Cardiovascular Outcomes Trial (COLCOT) were the first two randomized clinical trials to convincingly demonstrate the effectiveness of specific anti-inflammatory treatments in the field of CV prevention, while other phase III trials-including the Cardiovascular Inflammation Reduction Trial one using methotrexate-were futile. This manuscript reviews the main characteristics and findings of recent anti-inflammatory Phase III trials in cardiology and discusses their similarities and differences in order to get further insights into the contribution of specific inflammatory pathways on CV outcomes. CANTOS and COLCOT demonstrated efficacy of two anti-inflammatory drugs (canakinumab and colchicine, respectively) in the secondary prevention of major adverse CV events (MACE) thus providing the first confirmation of the involvement of a specific inflammatory pathway in human atherosclerotic CV disease (ASCVD). Also, they highlighted the NOD-, LRR-, and pyrin domain-containing protein 3 inflammasome-related pathway as an effective therapeutic target to blunt ASCVD. In contrast, other trials interfering with a number of inflammasome-independent pathways failed to provide benefit. Lastly, all anti-inflammatory trials underscored the importance of balancing the risk of impaired host defence with an increase in infections and the prevention of MACE in CV patients with residual inflammatory risk.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular Cardiology, University of Zurich, 12 Wagistrasse, 8952 Schlieren, Switzerland.,First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, 10 Largo Benzi, 16132 Genoa, Italy.,First Clinic of Internal Medicine, Department of Internal Medicine, Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 6 viale Benedetto XV, 16132 Genoa, Italy
| | - Lena Schwarz
- Center for Molecular Cardiology, University of Zurich, 12 Wagistrasse, 8952 Schlieren, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, 12 Wagistrasse, 8952 Schlieren, Switzerland.,Royal Brompton and Harefield Hospitals and Imperial College, London, UK
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, 12 Wagistrasse, 8952 Schlieren, Switzerland.,Department of Cardiology, University Heart Center, University Hospital Zurich, Rämistrasse 100, 8092 Zurich, Switzerland.,Department of Research and Education, University Hospital Zurich, Rämistrasse 100, 8092, Zurich, Switzerland
| |
Collapse
|
17
|
Amadi PU, Agomuo EN, Adumekwe CW. Modulatory properties of cardiac and quercetin glycosides from Dacryodes edulis seeds during L-NAME-induced vascular perturbation. J Basic Clin Physiol Pharmacol 2020; 31:/j/jbcpp.ahead-of-print/jbcpp-2019-0116/jbcpp-2019-0116.xml. [PMID: 32653874 DOI: 10.1515/jbcpp-2019-0116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 03/06/2020] [Indexed: 02/06/2023]
Abstract
Background Numerous food wastes have been identified to possess potent bioactive compounds used for the treatment of several diseases. Therefore this study evaluated the potentials of cardiac and quercetin glycosides extracted from Dacryodes edulis seeds to reverse vascular and endothelial damage (VAED). Methods The glycoside composition of the seeds was extracted using standard methods and characterized by gas chromatography. We then recruited rats with L-NAME-induced VAED based on confirmatory biomarkers cardiac troponin (CnT), cellular adhesion molecule (VCAM-1), lipoprotein associated phospholipase A2 (Lp-PLA2), RAAS, VWF, endothelin, eNOx, and homocysteine. Only rats that showed total alterations of all biomarkers were recruited into the respective experimental groups and treated with either metaprolol succinate (met.su) + losartan or glycoside extracts of D. edulis seeds (NPSG). Results Chromatographic isolation of glycosides in the seed showed predominance of artemetin (1.59 mg/100 g), amygdalin (3.68 mg/100 g), digitoxin (19.21 mg/100 g), digoxin (27.23 mg/100 g), avicularin (133.59 mg/100 g), and hyperoside (481.76 mg/100 g). We observed decreased water intake and higher heart beats under vascular damage as the experiment progressed up to the fourth week. The met.su + losartan and H.D NPSG proved effective in restoring troponin, but both doses of NPSG normalized the VCAM-1 and RAAS activities excluding aldosterone and Lp-PLA2. Among the endothelial dysfunction biomarkers, H.D NPSG produced equivalent effects to met.su + losartan towards restoring the eNOx and VWF activities, but showed higher potency in normalizing the endothelin and Hcy levels. Conclusions We thus propose that the synergistic effect of the isolated glycosides from D. edulis shown in our study proved potent enough at high doses in treatment of vascular and endothelial dysfunction.
Collapse
Affiliation(s)
- Peter Uchenna Amadi
- Department of Biochemistry, University of Port Harcourt, Choba, Rivers State, Nigeria.,Department of Biochemistry, Imo State, University, Owerri, Imo State, Nigeria
| | | | | |
Collapse
|
18
|
Zhang H, Gao Y, Wu D, Zhang D. The relationship of lipoprotein-associated phospholipase A2 activity with the seriousness of coronary artery disease. BMC Cardiovasc Disord 2020; 20:295. [PMID: 32546193 PMCID: PMC7298745 DOI: 10.1186/s12872-020-01580-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 06/10/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The level of lipoprotein-associated phospholipase A2 (LP-PLA2) in serum is independently correlated to coronary artery diseases (CAD). The aim of the study was to determine whether LP-PLA2 activity is positively associated with the seriousness of CAD. METHODS Amount to 1056 patients suspected of having CAD underwent coronary angiography (CAG) to determine the seriousness of CAD. According to the amount of diseased coronary branches, the 1056 patients were split into three groups: single-vessel stenosis group, multiple-vessels stenosis group (> or = 2 diseased coronary branches),and control group (no diseased coronary branches). According to CAG results, electrocardiography, cardiac biomarker, and clinical presentation, all patients were split into four groups: acute myocardial infarction (AMI), unstable angina (UA), stable angina (SA), and control groups (excluding CAD). The activity of LP-PLA2 was compared statistically among the subgroups. Receiver operating characteristic analysis was applied to investigate the role of LP-PLA2 in evaluating the presence and seriousness of CAD. RESULTS The level of LP-PLA2 increased in line with the number of diseased coronary branches. The levels of LP-PLA2 in the AMI and UA groups were observably higher when compared with the control and SA groups. LP-PLA2 had 75.6% sensitivity and 67.3% specificity for recognizing CAD, and 53.0% sensitivity and 80.3% specificity for recognizing severe coronary artery lesions. CONCLUSION The activity of LP-PLA2 is positively correlated to the seriousness of CAD.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, 41# Taizhou road, Yangzhou, 225000, Jiangsu Province, China
| | - Yang Gao
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, 41# Taizhou road, Yangzhou, 225000, Jiangsu Province, China
| | - Dan Wu
- Department of Cardiology, The Affiliated Hospital of Yangzhou University, 41# Taizhou road, Yangzhou, 225000, Jiangsu Province, China
| | - Dingguo Zhang
- Department of Cardiology, First Affiliated Hospital of Nanjing Medical University, 300# Guangzhou road, Nanjing, 210029, Jiangsu Province, China.
| |
Collapse
|
19
|
Lipoprotein(a) and calcific aortic valve stenosis: A systematic review. Prog Cardiovasc Dis 2020; 63:496-502. [PMID: 32526213 DOI: 10.1016/j.pcad.2020.06.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 06/01/2020] [Indexed: 02/06/2023]
Abstract
Calcific aortic valve stenosis (AS) is the most common form of acquired valvular heart disease needing intervention and our understanding of this disease has evolved from one of degenerative calcification to that of an active process driven by the interplay of genetic factors and chronic inflammation modulated by risk factors such as smoking, hypertension and elevated cholesterol. Lipoprotein(a) [Lp (a)] is a cholesterol rich particle secreted by the liver which functions as the major lipoprotein carrier of phosphocholine-containing oxidized phospholipids. Lp(a) levels are largely genetically determined by polymorphisms in the LPA gene. While there is an extensive body of evidence linking Lp(a) to atherosclerotic cardiovascular disease, emerging evidence now suggests a similar association of Lp(a) to calcific AS. In this article, we performed a systematic review of all published literature to assess the association between Lp(a) and calcific aortic valve (AV) disease. In addition, we review the potential mechanisms by which Lp(a) influences the progression of valve disease. Our review identified a total of 21 studies, varying from case-control studies, prospective or retrospective observational cohort studies to Mendelian randomized studies that assessed the association between Lp(a) and calcific AS. All but one of the above studies demonstrated significant association between elevated Lp(a) and calcific AS. We conclude that there is convincing evidence supporting a causal association between elevated Lp(a) and calcific AS. In addition, elevated Lp(a) predicts a faster hemodynamic progression of AS, and increased risk of AV replacement, especially in younger patients. Further research into the clinical utility of Lp(a) as a marker for predicting the incidence, progression, and outcomes of sclerodegenerative AV disease is needed.
Collapse
|
20
|
Yin YJ, Chen YC, Xu L, Zhao XH, Song Yang. Relationship of lipoprotein-associated phospholipase A2(Lp-PLA2) and periprocedural myocardial injury in patients undergoing elective percutaneous coronary intervention. IJC HEART & VASCULATURE 2020; 28:100541. [PMID: 32490148 PMCID: PMC7256635 DOI: 10.1016/j.ijcha.2020.100541] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/12/2020] [Accepted: 05/17/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Percutaneous coronary intervention (PCI) is one of the dominant methods for revascularization in patients with coronary heart disease (CHD). However, periprocedural myocardial injury (PMI) is a frequent complication following PCI and is known to be a predictor of postprocedural cardiovascular morbidity and mortality. Although several studies try to identify serum markers to predict the PMI, there is a little information about the role of lipoprotein-associated phospholipase A2 (Lp-PLA2) as a predictor of PMI. Therefore, we aimed to investigate the relationship of Lp-PLA2 levels and PMI in patients undergoing elective PCI. METHODS This study included 265 consecutive patients with normal preprocedural cardiac troponin T(cTNT) who received elective PCI. The samples for cTNT were collected at 8, 16, and 24 h after PCI to assess perioperative myocardial injury. The Lp-PLA2 and other serum lipid parameters were measured after 12 fasting hours before PCI. RESULTS The data suggested that the patients with preprocedural high Lp-PLA2 were strongly and independently correlated with the risk of PMI. Pearson correlation analysis showed that preprocedural Lp-PLA2 was significantly positively correlated with postprocedural cTnT elevation (r = 0.694, p < 0.05). Binary logistic regression analysis was used to analyze the risk factors of PMI, we found that Lp-PLA2 was independent risk factor for postprocedural cTnT elevation. The area under Receiver Operating Characteristic curve of Lp-PLA2 was 0.757 (95%CI 0.692 ~ 0.821, p < 0.001), the best cut-off point was 185 ng/ml, sensitivity and specificity were 65.33% and 76.32%. CONCLUSION Our study demonstrated that preprocedural Lp-PLA2 was associated with postprocedural cTnT elevation and was the independent risk factor of PMI.
Collapse
Affiliation(s)
| | | | - Liang Xu
- Department of Cardiology, Yixing People’s Hospital, Yixing, Jiangsu Province 214200, PR China
| | - Xiang-hai Zhao
- Department of Cardiology, Yixing People’s Hospital, Yixing, Jiangsu Province 214200, PR China
| | - Song Yang
- Department of Cardiology, Yixing People’s Hospital, Yixing, Jiangsu Province 214200, PR China
| |
Collapse
|
21
|
Forty Years Since the Structural Elucidation of Platelet-Activating Factor (PAF): Historical, Current, and Future Research Perspectives. Molecules 2019; 24:molecules24234414. [PMID: 31816871 PMCID: PMC6930554 DOI: 10.3390/molecules24234414] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 12/11/2022] Open
Abstract
In the late 1960s, Barbaro and Zvaifler described a substance that caused antigen induced histamine release from rabbit platelets producing antibodies in passive cutaneous anaphylaxis. Henson described a ‘soluble factor’ released from leukocytes that induced vasoactive amine release in platelets. Later observations by Siraganuan and Osler observed the existence of a diluted substance that had the capacity to cause platelet activation. In 1972, the term platelet-activating factor (PAF) was coined by Benveniste, Henson, and Cochrane. The structure of PAF was later elucidated by Demopoulos, Pinckard, and Hanahan in 1979. These studies introduced the research world to PAF, which is now recognised as a potent phospholipid mediator. Since its introduction to the literature, research on PAF has grown due to interest in its vital cell signalling functions and more sinisterly its role as a pro-inflammatory molecule in several chronic diseases including cardiovascular disease and cancer. As it is forty years since the structural elucidation of PAF, the aim of this review is to provide a historical account of the discovery of PAF and to provide a general overview of current and future perspectives on PAF research in physiology and pathophysiology.
Collapse
|
22
|
Wihastuti TA, Aini FN, Tjahjono CT, Sulfia YH, Sholichah Z, Heriansyah T. Lp-PLA 2 Selective Inhibitor (Darapladib) Effect In Lowering The Expression Level Of IL-1B And IL-6 In The Renal At Type 2 Diabetes Mellitus. Vasc Health Risk Manag 2019; 15:503-508. [PMID: 31802883 PMCID: PMC6830380 DOI: 10.2147/vhrm.s217904] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/11/2019] [Indexed: 12/21/2022] Open
Abstract
PURPOSE The aim of this study is to prove that type 2 diabetes mellitus can induce increasing inflammation marker in renal and that the provision of darapladib as Lp-LA2 Inhibitor agents can inhibit inflammation that were measured from the expression of IL-1B and IL-6- type cytokine in renal. This study also discusses the correlation between IL-1B and IL-6- type cytokine expression in renal. METHODS Thirty Sprague-Dawley (SD) rats were divided into three main groups; those are negative control group (NC), Type 2 Diabetes Mellitus group (T2DM) given high fat diet (HFD) with streptozotocin intraperitoneal injection (35mg/kg BW) and diabetes mellitus + darapladib group (DM + DP). Each group was treated within two serial treatment time: 8 weeks and 16 weeks. Expressions of IL-1B and IL-6- type cytokine in renal were the markers that we measured by immunofluorosense method. RESULTS The administration of darapladib can significantly decrease the expression of IL-1B- type cytokine (p ANOVA = 0.029, p < 0.005) measured in rats' renal both at weeks 8 and 16 in the T2DM group. The Expression of IL-6- type cytokine also showed a significant difference after treated with darapladib both at weeks 8 and 16 in T2DM group with p-value of ANOVA = 0.033, p < 0.005. The Pearson correlation showed a strong correlation (linear regression value was r2 = 0.743). CONCLUSION Our results show that atherosclerosis caused by inflammation in renal T2DM SD rats could be inhibited by the administration of darapladib.
Collapse
Affiliation(s)
- Titin Andri Wihastuti
- Department of Basic Science in Nursing, Faculty of Medicine, University of Brawijaya, Malang, East Java, Indonesia
| | - Fitria Nugraha Aini
- Faculty of Medicine, University of Islam Malang, Malang, East Java, Indonesia
- Master Programme of Biomedicine, Faculty of Medicine, Brawijaya University, Malang, East Java, Indonesia
| | - Cholid Tri Tjahjono
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, Brawijaya University, Malang, East Java, Indonesia
| | - Yuni Hendrati Sulfia
- Master Programme of Biomedicine, Faculty of Medicine, Brawijaya University, Malang, East Java, Indonesia
| | - Zuhrotus Sholichah
- Master Programme of Biomedicine, Faculty of Medicine, Brawijaya University, Malang, East Java, Indonesia
| | - Teuku Heriansyah
- Department of Cardiology and Vascular Medicine, Faculty of Medicine, University of Syiah Kuala, Banda Aceh, Aceh, Indonesia
| |
Collapse
|
23
|
Pharmacokinetics and exploratory efficacy biomarkers of bococizumab, an anti-PCSK9 monoclonal antibody, in hypercholesterolemic Japanese subjects
. Int J Clin Pharmacol Ther 2019; 57:575-589. [PMID: 31549625 PMCID: PMC6862531 DOI: 10.5414/cp203418] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2019] [Indexed: 12/21/2022] Open
Abstract
Objective: Bococizumab, a monoclonal antibody targeting proprotein convertase subtilisin/kexin type 9, has been shown to reduce low-density lipoprotein cholesterol (LDL-C). Here, we describe the pharmacokinetics and pharmacodynamics of bococizumab and its effect on lipoprotein particle composition and other biomarkers, based on a double-blind, placebo-controlled, randomized, dose-ranging study. Materials and methods: The study consisted of two populations: Japanese subjects with uncontrolled LDL-C (LDL-C ≥ 100 mg/dL) despite treatment with atorvastatin (n = 121) and Japanese subjects naïve to lipid-lowering agents with LDL-C ≥ 130 mg/dL (n = 97). Subjects were randomized to receive either bococizumab 50, 100, or 150 mg or placebo, every 2 weeks. One arm of subjects in the atorvastatin-treated population received ezetimibe 10 mg instead of bococizumab. Results: In both populations, bococizumab exposure increased with increasing dose, and subjects with lower body weights tended to have higher exposures. Bococizumab treatment was associated with a dose-dependent reduction in LDL particles and a small increase in total high-density lipoprotein (HDL) particles. Significant reductions in lipoprotein-associated phospholipase A2 (Lp-PLA2) were observed for bococizumab-treated subjects but not for subjects treated with placebo or ezetimibe. Conclusion: Increased bococizumab dosage resulted in increased exposure. Levels of LDL and HDL particles and biomarkers such as Lp-PLA2 were also altered with bococizumab treatment. (ClinicalTrials.gov identifier: NCT02055976).
Collapse
|
24
|
Yi M, Li Q, Zhao Y, Nie S, Wu N, Wang D. Metabolomics study on the therapeutic effect of traditional Chinese medicine Xue-Fu-Zhu-Yu decoction in coronary heart disease based on LC-Q-TOF/MS and GC-MS analysis. Drug Metab Pharmacokinet 2019; 34:340-349. [PMID: 31474470 DOI: 10.1016/j.dmpk.2019.07.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 07/07/2019] [Accepted: 07/31/2019] [Indexed: 11/29/2022]
Abstract
The present study aims is to investigate the metabolic mechanism of Xue-Fu-Zhu-Yu decoction (XFZYD) in the treatment of blood-stasis syndrome in Coronary Heart Disease (CHD). To that end, 30 CHD patients with Blood-Stasis Syndrome (BSS) and 20 healthy subjects were enrolled. LC-Q-TOF/MS analysis determined that in comparison between CHD with BSS patients (Group A) and healthy subjects (Group C), 59 significantly differential metabolites in the positive mode and 18 significantly differential metabolites in the negative mode. The metabolite constituents in the plasma of 30 CHD with BSS patients before (group A) and after 30 days of treatment (Group B), and 20 healthy subjects (Group C) were analyzed using LC-Q-TOF/MS and GC-MS. Based on multivariate statistical analysis (PCA, PLS-DA and OPLS-DA), we determined 69 differential metabolites. The levels of hemorheology indexes were significantly down-regulated after treatment. Metabolic pathway attribution analysis showed that lipid metabolism, amino acid metabolism and bile acid metabolism pathways are involved. Our study identifies the metabolic networks of CHD and demonstrates the efficacy of this metabolomics approach to systematically study the therapeutic effect of XFZYC on CHD.
Collapse
Affiliation(s)
- Min Yi
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Kaifu District, Changsha, Hunan, 410008, China
| | - Qiuxia Li
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Kaifu District, Changsha, Hunan, 410008, China
| | - Yuhang Zhao
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Kaifu District, Changsha, Hunan, 410008, China
| | - Shanshan Nie
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Kaifu District, Changsha, Hunan, 410008, China
| | - Ning Wu
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Kaifu District, Changsha, Hunan, 410008, China
| | - Dongsheng Wang
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Kaifu District, Changsha, Hunan, 410008, China.
| |
Collapse
|
25
|
Lyubarova R, Albers JJ, Marcovina SM, Yao Y, McBride R, Topliceanu A, Anderson T, Fleg JL, Desvigne-Nickens P, Kashyap ML, McGovern ME, Boden WE. Effects of Extended-Release Niacin on Quartile Lp-PLA2 Levels and Clinical Outcomes in Statin-treated Patients with Established Cardiovascular Disease and Low Baseline Levels of HDL-Cholesterol: Post Hoc Analysis of the AIM HIGH Trial. J Cardiovasc Pharmacol Ther 2019; 24:534-541. [DOI: 10.1177/1074248419852955] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Lipoprotein-associated phospholipase A2 (LpPLA2) is an inflammatory marker that has been associated with the presence of vulnerable plaque and increased risk of cardiovascular (CV) events. Objective: To assess the effect of extended-release niacin (ERN) on Lp-PLA2 activity and clinical outcomes. Methods: We performed a post hoc analysis in 3196 AIM-HIGH patients with established CV disease and low baseline levels of high-density lipoprotein cholesterol (HDL-C) who were randomized to ERN versus placebo on a background of simvastatin therapy (with or without ezetimibe) to assess the association between baseline Lp-PLA2 activity and the rate of the composite primary end point (CV death, myocardial infarction, stroke, hospitalization for unstable angina, and symptom-driven revascularization). Results: Participants randomized to ERN, but not those randomized to placebo, experienced a significant 8.9% decrease in LpPLA2. In univariate analysis, the highest quartile of LpPLA2 activity (>208 nmol/min/mL, Q4) was associated with higher event rates compared to the lower quartiles in the placebo group (log rank P = .032), but not in the ERN treated participants (log rank P = .718). However, in multivariate analysis, adjusting for sex, diabetes, baseline LDL-C, HDL-C, and triglycerides, there was no significant difference in outcomes between the highest Lp-PLA2 activity quartile versus the lower quartiles in both the placebo and the ERN groups. Conclusion: Among participants with stable CV disease on optimal medical therapy, elevated Lp-PLA2 was associated with higher CV events; however, addition of ERN mitigates this effect. This association in the placebo group was attenuated after multivariable adjustment, which suggests that Lp-PLA2 does not improve risk assessment beyond traditional risk factors.
Collapse
Affiliation(s)
- Radmila Lyubarova
- Division of Cardiology, Albany Medical Center, Albany Medical College, Albany, NY, USA
| | - John J. Albers
- University of Washington, Northwest Lipid Metabolism, And Diabetes Research Laboratories, Seattle, WA, USA
| | - Santica M. Marcovina
- University of Washington, Northwest Lipid Metabolism, And Diabetes Research Laboratories, Seattle, WA, USA
| | - Yao Yao
- Axio Research LLC, Seattle, WA, USA
| | | | - Alexandru Topliceanu
- Division of Cardiology, Albany Medical Center, Albany Medical College, Albany, NY, USA
| | - Todd Anderson
- University of Calgary and Libin Cardiovascular Institute, Foothills Medical Centre, Calgary, Canada
| | - Jerome L. Fleg
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | | |
Collapse
|
26
|
Zhao TX, Mallat Z. Targeting the Immune System in Atherosclerosis. J Am Coll Cardiol 2019; 73:1691-1706. [DOI: 10.1016/j.jacc.2018.12.083] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/20/2018] [Accepted: 12/30/2018] [Indexed: 02/08/2023]
|
27
|
Park JG, Oh GT. Current pharmacotherapies for atherosclerotic cardiovascular diseases. Arch Pharm Res 2019; 42:206-223. [DOI: 10.1007/s12272-019-01116-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 01/11/2019] [Indexed: 12/19/2022]
|
28
|
Alkhalil M, Choudhury RP. Current concepts in atherosclerosis. Indian J Thorac Cardiovasc Surg 2018; 34:198-205. [PMID: 33060939 PMCID: PMC7525593 DOI: 10.1007/s12055-018-0699-y] [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: 04/25/2018] [Revised: 07/06/2018] [Accepted: 07/12/2018] [Indexed: 11/30/2022] Open
Abstract
Atherosclerosis is a complex disease process. It is increasingly recognised that both lipoprotein retention and inflammatory cellular components are intricately related in the initiation and development of atherosclerotic plaque. LDL-c (cholesterol) has been long established as a cause for atherosclerosis; additionally, inflammatory cells such as monocytes and subsequently foam cells have also been directly linked to the progression of atherosclerotic disease. Emerging data suggest that structures outside vascular intima and media are also closely related to atherosclerosis. Perivascular adipose tissue (PVAT) may be a determinant of the inflammatory status of the atherosclerotic plaque. All these features are becoming extremely relevant as therapies against atherosclerosis are targeting both lipid retention and inflammation. Recently, there has been some success in these novel therapies, such as the proprotein convertase subtilisin-kexin type 9 (PCSK-9) inhibitor evolocumab and the interleukin-1ß neutralising antibody, canakinumab, in reducing cardiovascular events when added to standard therapy such as statin. This review will discuss the pathogenesis of atherosclerosis, including some novel features, and its management using new anti-atherosclerotic drugs.
Collapse
Affiliation(s)
- Mohammad Alkhalil
- Acute Vascular Imaging Centre, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU UK.,Cardiology Department, Royal Victoria Hospital, Belfast, UK
| | - Robin P Choudhury
- Acute Vascular Imaging Centre, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, OX3 9DU UK
| |
Collapse
|
29
|
Wang W, Xu Z, Zhu X, Chang X. Mining the potential therapeutic targets for coronary artery disease by bioinformatics analysis. Mol Med Rep 2018; 18:5069-5075. [PMID: 30320387 PMCID: PMC6236289 DOI: 10.3892/mmr.2018.9551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 09/17/2018] [Indexed: 12/11/2022] Open
Abstract
The present study aimed to mine therapeutic molecular targets that play an important part in the progression of coronary artery disease (CAD). The gene expression profile GSE28829 dataset and the microRNA (miRNA) expression profile GSE59421 dataset were downloaded from the Gene Expression Omnibus (GEO) database. The GEO2R online analytical tool was used to identify differentially expressed genes (DEGs) and miRNAs (DEMs). The target genes of DEMs were identified using the miRWalk2.0 web-based tool and 2 miRNA-gene regulatory networks were constructed using Cytoscape software. Subsequently, enriched Gene Ontology (GO) terms of miRNA-target DEGs were obtained using the Database for Visualization, Annotation and Integrated Analysis, and locations of these genes in the chromosomes were determined by Map Viewer. In the present study, 350 DEGs and 66 DEMs were screened. A total of 3,588 target genes were identified from the DEMs, and 57 of these target genes and established DEGs were identified to overlap. GO terms associated with 5 processes, and 4 types of composition were identified to be enriched in the miRNA-target DEGs. Furthermore, 26 miRNA-gene regulatory pairs were obtained between the 57 target genes and DEMs. The 26 miRNA-target DEGs were unevenly distributed, and no genes were located on the sex chromosomes. As a result of the present study, potential therapeutic targets for CAD were identified through bioinformatics analysis.
Collapse
Affiliation(s)
- Wendong Wang
- Department of Biochemistry, College of Laboratory Medicine, Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Zhiwei Xu
- Department of Biochemistry, College of Laboratory Medicine, Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Xiaobo Zhu
- Department of Biochemistry, College of Laboratory Medicine, Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| | - Xiaotong Chang
- Department of Biochemistry, College of Laboratory Medicine, Hebei North University, Zhangjiakou, Hebei 075000, P.R. China
| |
Collapse
|
30
|
Prasad M, Lennon R, Barsness GW, Prasad A, Gulati R, Lerman LO, Lerman A. Chronic inhibition of lipoprotein-associated phospholipase A 2 does not improve coronary endothelial function: A prospective, randomized-controlled trial. Int J Cardiol 2018; 253:7-13. [PMID: 29306475 DOI: 10.1016/j.ijcard.2017.09.171] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 09/07/2017] [Accepted: 09/18/2017] [Indexed: 12/14/2022]
Abstract
AIMS Lipoprotein-associated phospholipase A2 (Lp-PLA2), a novel biomarker for vascular inflammation, is associated with coronary endothelial dysfunction (CED) and independently predicts cardiovascular events. The current study aimed to determine whether darapladib, an orally administered Lp-PLA2 inhibitor, improved CED. METHODS AND RESULTS Fifty-four patients with CED were enrolled in a double-blinded randomized placebo-controlled trial, and were randomized to receive oral darapladib, 160mg daily, or placebo. Coronary angiography and invasive coronary endothelial function assessment were performed at baseline and post-6months of treatment. Primary endpoints were change in coronary artery diameter and coronary blood flow in response to acetylcholine. Additionally, Lp-PLA2 activity was measured at baseline and on follow-up to evaluate for adherence and drug effect. Fifty-four patients were randomized to placebo (n=29) and darapladib (n=25). Mean age in darapladib group was 55.2.±11.7years vs. 54.0±10.5years (p=0.11). On follow-up, there was no significant difference in the percent response to acetylcholine of coronary artery diameter in treatment vs. placebo group (+3 (IQR -9, 15) vs. +3 (-12, 19); p=0.87) or coronary blood flow (-5 (IQR -24, 54) vs. 39 (IQR -26, 67); p=0.41). There was significant reduction in Lp-PLA2 activity in the treatment arm vs. placebo (-76 (IQR -113, -52) vs. -7(-21, -7); p<0.001). DISCUSSION Lp-PLA2 inhibition with darapladib did not improve coronary endothelial function, despite significantly reduced Lp-PLA2 activity with darapladib. This study suggests endogenous Lp-PLA2 may not play a primary role in coronary endothelial function in humans. CLINICALTRIALS. GOV IDENTIFIER NCT01067339.
Collapse
Affiliation(s)
- Megha Prasad
- Mayo Clinic, Department of Cardiovascular Diseases, Rochester, MN, United States
| | - Ryan Lennon
- Mayo Clinic, Department of Health Sciences Research, Rochester, MN, United States
| | - Gregory W Barsness
- Mayo Clinic, Department of Cardiovascular Diseases, Rochester, MN, United States
| | - Abhiram Prasad
- Mayo Clinic, Department of Cardiovascular Diseases, Rochester, MN, United States
| | - Rajiv Gulati
- Mayo Clinic, Department of Cardiovascular Diseases, Rochester, MN, United States
| | - Lilach O Lerman
- Mayo Clinic, Department of Cardiovascular Diseases, Rochester, MN, United States
| | - Amir Lerman
- Mayo Clinic, Department of Cardiovascular Diseases, Rochester, MN, United States.
| |
Collapse
|
31
|
Relationship between atherosclerosis risks and lipoprotein-dependent phospholipase a2 activity in type 2 diabetic patients. JOURNAL OF SURGERY AND MEDICINE 2018. [DOI: 10.28982/josam.440016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
32
|
Katan M, Elkind MSV. The potential role of blood biomarkers in patients with ischemic stroke. CLINICAL AND TRANSLATIONAL NEUROSCIENCE 2018. [DOI: 10.1177/2514183x18768050] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Mira Katan
- Department of Neurology, Universitiy Hospital of Zurich, Zürich, Switzerland
| | - Mitchell SV Elkind
- Department of Neurology, Division of Stroke, Columbia University, New York, NY, USA
| |
Collapse
|
33
|
Li B, Li W, Li X, Zhou H. Inflammation: A Novel Therapeutic Target/Direction in Atherosclerosis. Curr Pharm Des 2018; 23:1216-1227. [PMID: 28034355 PMCID: PMC6302344 DOI: 10.2174/1381612822666161230142931] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 12/27/2016] [Indexed: 12/27/2022]
Abstract
Over the past two decades, the viewpoint of atherosclerosis has been replaced gradually by a lipid-driven, chronic, low-grade inflammatory disease of the arterial wall. Current treatment of atherosclerosis is focused on limiting its risk factors, such as hyperlipidemia or hypertension. However, treatment targeting the inflammatory nature of atherosclerosis is still very limited and deserves further attention to fight atherosclerosis successfully. Here, we review the current development of inflammation and atherosclerosis to discuss novel insights and potential targets in atherosclerosis, and to address drug discovery based on anti-inflammatory strategy in atherosclerotic disease.
Collapse
Affiliation(s)
- Bin Li
- Department of Pharmacology, College of Pharmacy, Third Military Medical University, Chongqing 400038. China
| | - Weihong Li
- Assisted Reproductive Center, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016. China
| | - Xiaoli Li
- Department of Pharmacology, College of Pharmacy, Third Military Medical University, Chongqing 400038. China
| | - Hong Zhou
- Department of Pharmacology, College of Pharamacy, The Third Military Medical University, P.O. Box: 400038, Chongqing. China
| |
Collapse
|
34
|
Chistiakov DA, Melnichenko AA, Grechko AV, Myasoedova VA, Orekhov AN. Potential of anti-inflammatory agents for treatment of atherosclerosis. Exp Mol Pathol 2018; 104:114-124. [PMID: 29378168 DOI: 10.1016/j.yexmp.2018.01.008] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 12/30/2017] [Accepted: 01/17/2018] [Indexed: 12/23/2022]
Abstract
Chronic inflammation is a central pathogenic mechanism of atherosclerosis induction and progression. Vascular inflammation is associated with accelerated onset of late atherosclerosis complications. Atherosclerosis-related inflammation is mediated by a complex cocktail of pro-inflammatory cytokines, chemokines, bioactive lipids, and adhesion molecules, and blocking the key pro-atherogenic inflammatory mechanisms can be beneficial for treatment of atherosclerosis. Therapeutic agents that specifically target some of the atherosclerosis-related inflammatory mechanisms have been evaluated in preclinical and clinical studies. The most promising anti-inflammatory compounds for treatment of atherosclerosis include non-specific anti-inflammatory drugs, phospholipase inhibitors, blockers of major inflammatory cytokines, leukotrienes, adhesion molecules, and pro-inflammatory signaling pathways, such as CCL2-CCR2 axis or p38 MAPK pathway. Ongoing studies attempt evaluating therapeutic utility of these anti-inflammatory drugs for treatment of atherosclerosis. The obtained results are important for our understanding of atherosclerosis-related inflammatory mechanisms and for designing randomized controlled studies assessing the effect of specific anti-inflammatory strategies on cardiovascular outcomes.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- Department of Neurochemistry, Division of Basic and Applied Neurobiology, Serbsky Federal Medical Research Center of Psychiatry and Narcology, Moscow 119991, Russia
| | - Alexandra A Melnichenko
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow 125315, Russia
| | - Andrey V Grechko
- Federal Scientific Clinical Center for Resuscitation and Rehabilitation, Moscow 109240, Russia
| | - Veronika A Myasoedova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow 125315, Russia
| | - Alexander N Orekhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow 125315, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow 121609, Russia.
| |
Collapse
|
35
|
Tselepis AD. Oxidized phospholipids and lipoprotein-associated phospholipase A 2 as important determinants of Lp(a) functionality and pathophysiological role. J Biomed Res 2018; 31. [PMID: 27346583 PMCID: PMC5956253 DOI: 10.7555/jbr.31.20160009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 01/29/2016] [Accepted: 02/12/2016] [Indexed: 12/23/2022] Open
Abstract
Lipoprotein(a) [Lp(a)] is composed of a low density lipoprotein (LDL)-like particle to which apolipoprotein (a) [apo(a)] is linked by a single disulfide bridge. Lp(a) is considered a causal risk factor for ischemic cardiovascular disease (CVD) and calcific aortic valve stenosis (CAVS). The evidence for a causal role of Lp(a) in CVD and CAVS is based on data from large epidemiological databases, mendelian randomization studies, and genome-wide association studies. Despite the well-established role of Lp(a) as a causal risk factor for CVD and CAVS, the underlying mechanisms are not well understood. A key role in the Lp(a) functionality may be played by its oxidized phospholipids (OxPL) content. Importantly, most of circulating OxPL are associated with Lp(a); however, the underlying mechanisms leading to this preferential sequestration of OxPL on Lp(a) over the other lipoproteins, are mostly unknown. Several studies support the hypothesis that the risk of Lp(a) is primarily driven by its OxPL content. An important role in Lp(a) functionality may be played by the lipoprotein-associated phospholipase A2 (Lp-PLA2), an enzyme that catalyzes the degradation of OxPL and is bound to plasma lipoproteins including Lp(a). The present review article discusses new data on the pathophysiological role of Lp(a) and particularly focuses on the functional role of OxPL and Lp-PLA2 associated with Lp(a).
Collapse
Affiliation(s)
- Alexandros D Tselepis
- Atherothrombosis Research Centre / Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, 45110 Ioannina, Greece.
| |
Collapse
|
36
|
Chen AD, Wang CL, Qin Y, Tian L, Chen LB, Yuan XM, Ma LX, Wang YF, Sun JR, Wang HS, Dai N. The effect of Danshen extract on lipoprotein-associated phospholipase A 2 levels in patients with stable angina pectoris: study protocol for a randomized controlled trial - the DOLPHIN study. Trials 2017; 18:606. [PMID: 29262859 PMCID: PMC5738193 DOI: 10.1186/s13063-017-2336-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 10/28/2017] [Indexed: 01/27/2023] Open
Abstract
Background Lipoprotein-associated phospholipase A2 (Lp-PLA2), a biomarker of oxidation and inflammation, has been associated with increased coronary artery disease risk. To date, very few studies have examined the Chinese herbal drug Danshen or its extract on Lp-PLA2 in patients with stable angina pectoris. In this study, we aim to investigate the effect of Danshen extract on Lp-PLA2 level in patients with stable angina. Methods/design This is a randomized, single-blind, placebo-controlled, adaptive clinical trial. A total of 156 patients meeting the eligibility criteria will be randomly assigned to either the Danshen extract (DanshenDuofensuanyan injection and Danshen drop spill) group or the placebo group in a 1:1 ratio. Participants will then undergo treatment with DanshenDuofensuanyan injection or placebo (glucose) during hospitalization, followed by open-label Danshen drop spill (30 pills/day) in Danshen extract group for 60 days after discharge. Because this is an adaptive trial, two interim analyses are prospectively planned. These will be performed after one-third and two-thirds of the patients, respectively, have completed the trial. On the basis of the results of these interim analyses, a data monitoring committee will determine how to modify aspects of the study without undermining the validity and integrity of the trial. The primary outcome measure is the serum level of Lp-PLA2 in the Danshen extract group and the placebo group. The secondary outcomes include the proportion of patients who show a clinically significant change, which is defined as at least a 20-point improvement in angina frequency score on the Seattle Angina Questionnaire and the carotid intima-media thickness, which will be measured using ultrasound. Other secondary efficacy and safety outcomes will also be assessed. Discussion This study will provide evidence that Danshen extract is beneficial for stable angina and may establish a possible mechanism of Danshen treatment effects on cardiovascular disease. This study may also validate an objective blood test (LP-PLA2 level) for assessing the effectiveness of Danshen therapy in patients with stable angina pectoris. Trial registration ClinicalTrials.gov, NCT02870764. Registered on 13 August 2016. Electronic supplementary material The online version of this article (doi:10.1186/s13063-017-2336-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- A-Di Chen
- Cardiology Department, Taizhou Fourth People's Hospital, Taizhou, Jiangsu Province, 225300, China
| | - Chun-Ling Wang
- Cardiology Department, Taizhou Fourth People's Hospital, Taizhou, Jiangsu Province, 225300, China
| | - Yang Qin
- Cardiology Department, Taizhou Fourth People's Hospital, Taizhou, Jiangsu Province, 225300, China
| | - Liang Tian
- Cardiology Department, Taizhou Fourth People's Hospital, Taizhou, Jiangsu Province, 225300, China
| | - Li-Bin Chen
- Cardiology Department, Taizhou Fourth People's Hospital, Taizhou, Jiangsu Province, 225300, China
| | - Xiao-Ming Yuan
- Cardiology Department, Taizhou Fourth People's Hospital, Taizhou, Jiangsu Province, 225300, China
| | - Lin-Xiu Ma
- Cardiology Department, Taizhou Fourth People's Hospital, Taizhou, Jiangsu Province, 225300, China
| | - Yu-Feng Wang
- Cardiology Department, Taizhou Fourth People's Hospital, Taizhou, Jiangsu Province, 225300, China
| | - Ji-Rong Sun
- Cardiology Department, Taizhou Fourth People's Hospital, Taizhou, Jiangsu Province, 225300, China
| | - Hao-Sen Wang
- Department of Science and Education, Taizhou Fourth People's Hospital, Taizhou, Jiangsu Province, 225300, China
| | - Neng Dai
- Cardiology Department, Tenth People's Hospital of Tongji University, Shanghai, 200072, China. .,Cardiology Department, Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, China.
| | | |
Collapse
|
37
|
Mohler ER. From the Masters: Research lessons I've learned over a career in vascular medicine. Vasc Med 2017; 22:461-464. [PMID: 28976248 DOI: 10.1177/1358863x17728332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Emile R Mohler
- Vascular Medicine Section, Cardiovascular Division, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
38
|
Sun S, Zhang M, Yang Q, Shen Z, Chen J, Yu B, Wang H, Qu J, Pang D, Ren W, Ouyang H, Tang X. Resveratrol suppresses lipoprotein-associated phospholipase A2
expression by reducing oxidative stress in macrophages and animal models. Mol Nutr Food Res 2017; 61. [DOI: 10.1002/mnfr.201601112] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 06/04/2017] [Accepted: 06/06/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Shengnan Sun
- Jilin Provincial Model Animal Engineering Research Center; College of Animal Sciences,; Jilin University; Changchun China
| | - Mingjun Zhang
- Jilin Provincial Model Animal Engineering Research Center; College of Animal Sciences,; Jilin University; Changchun China
| | - Qiangbing Yang
- Jilin Provincial Model Animal Engineering Research Center; College of Animal Sciences,; Jilin University; Changchun China
| | - Ziying Shen
- Jilin Provincial Model Animal Engineering Research Center; College of Animal Sciences,; Jilin University; Changchun China
| | - Jiahuan Chen
- Jilin Provincial Model Animal Engineering Research Center; College of Animal Sciences,; Jilin University; Changchun China
| | - Biao Yu
- Jilin Provincial Model Animal Engineering Research Center; College of Animal Sciences,; Jilin University; Changchun China
| | - He Wang
- Jilin Provincial Model Animal Engineering Research Center; College of Animal Sciences,; Jilin University; Changchun China
| | - Jiali Qu
- Jilin Provincial Model Animal Engineering Research Center; College of Animal Sciences,; Jilin University; Changchun China
| | - Daxin Pang
- Jilin Provincial Model Animal Engineering Research Center; College of Animal Sciences,; Jilin University; Changchun China
| | - Wenzhi Ren
- Jilin Provincial Model Animal Engineering Research Center; College of Animal Sciences,; Jilin University; Changchun China
| | - Hongsheng Ouyang
- Jilin Provincial Model Animal Engineering Research Center; College of Animal Sciences,; Jilin University; Changchun China
| | - Xiaochun Tang
- Jilin Provincial Model Animal Engineering Research Center; College of Animal Sciences,; Jilin University; Changchun China
| |
Collapse
|
39
|
Welsh P, Grassia G, Botha S, Sattar N, Maffia P. Targeting inflammation to reduce cardiovascular disease risk: a realistic clinical prospect? Br J Pharmacol 2017; 174:3898-3913. [PMID: 28409825 PMCID: PMC5660005 DOI: 10.1111/bph.13818] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 03/28/2017] [Accepted: 03/30/2017] [Indexed: 12/16/2022] Open
Abstract
Data from basic science experiments is overwhelmingly supportive of the causal role of immune-inflammatory response(s) at the core of atherosclerosis, and therefore, the theoretical potential to manipulate the inflammatory response to prevent cardiovascular events. However, extrapolation to humans requires care and we still lack definitive evidence to show that interfering in immune-inflammatory processes may safely lessen clinical atherosclerosis. In this review, we discuss key therapeutic targets in the treatment of vascular inflammation, placing basic research in a wider clinical perspective, as well as identifying outstanding questions. LINKED ARTICLES This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.
Collapse
Affiliation(s)
- Paul Welsh
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Gianluca Grassia
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Shani Botha
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom campus, South Africa
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Pasquale Maffia
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.,Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.,Department of Pharmacy, University of Naples Federico II, Naples, Italy
| |
Collapse
|
40
|
Impact of 8-week linoleic acid intake in soy oil on Lp-PLA 2 activity in healthy adults. Nutr Metab (Lond) 2017; 14:32. [PMID: 28503188 PMCID: PMC5422895 DOI: 10.1186/s12986-017-0186-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 04/28/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND No intervention follow-up study has examined the association between plasma n-6 polyunsaturated fatty acids (PUFAs) and lipoprotein-associated phospholipase A2 (Lp-PLA2), which is a risk factor for cardiovascular disease (CVD). We aimed to determine whether the administration of linoleic acid (LA, 18:2n-6) in soy oil affected Lp-PLA2 activity in healthy adults. METHODS Self-reported healthy participants (n = 150) were randomly assigned to three groups: a low LA group, in which 10 mL soy oil was replaced with one apple; a medium LA group, in which the typical food intake was maintained; and a high LA group, in which 1/3 cup of cooked refined rice was replaced with 9.9 g of soy oil capsules daily. Plasma fatty acids and Lp-PLA2 activity were measured along with other CVD risk factors. RESULTS After 8 weeks of treatment, plasma LA levels decreased in the low LA group and increased in the high LA group. The high LA group showed greater increases in apolipoprotein B (apoB) and oxidized low-density lipoprotein (ox-LDL) than those in the low LA group. Plasma LA levels and Lp-PLA2 activities demonstrated greater increases in the high LA group than those in the medium and low LA groups. Changes in plasma LA positively and independently correlated with changes in Lp-PLA2 activity, which was negatively correlated with changes in collagen-epinephrine closure time (CEPI-CT). CONCLUSIONS An increase in plasma LA following intake of soy oil was independently associated with Lp-PLA2 activity, which was also related to apoB, ox-LDL and CEPI-CT. TRIAL REGISTRATION ClinicalTrail.gov Identifier: NCT02753907, registered 25 April 2016 (retrospectively registered).
Collapse
|
41
|
Gregson JM, Freitag DF, Surendran P, Stitziel NO, Chowdhury R, Burgess S, Kaptoge S, Gao P, Staley JR, Willeit P, Nielsen SF, Caslake M, Trompet S, Polfus LM, Kuulasmaa K, Kontto J, Perola M, Blankenberg S, Veronesi G, Gianfagna F, Männistö S, Kimura A, Lin H, Reilly DF, Gorski M, Mijatovic V, Munroe PB, Ehret GB, Thompson A, Uria-Nickelsen M, Malarstig A, Dehghan A, Vogt TF, Sasaoka T, Takeuchi F, Kato N, Yamada Y, Kee F, Müller-Nurasyid M, Ferrières J, Arveiler D, Amouyel P, Salomaa V, Boerwinkle E, Thompson SG, Ford I, Wouter Jukema J, Sattar N, Packard CJ, Shafi Majumder AA, Alam DS, Deloukas P, Schunkert H, Samani NJ, Kathiresan S, Nordestgaard BG, Saleheen D, Howson JMM, Di Angelantonio E, Butterworth AS, Danesh J. Genetic invalidation of Lp-PLA 2 as a therapeutic target: Large-scale study of five functional Lp-PLA 2-lowering alleles. Eur J Prev Cardiol 2017; 24:492-504. [PMID: 27940953 PMCID: PMC5460752 DOI: 10.1177/2047487316682186] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 10/24/2016] [Indexed: 01/12/2023]
Abstract
Aims Darapladib, a potent inhibitor of lipoprotein-associated phospholipase A2 (Lp-PLA2), has not reduced risk of cardiovascular disease outcomes in recent randomized trials. We aimed to test whether Lp-PLA2 enzyme activity is causally relevant to coronary heart disease. Methods In 72,657 patients with coronary heart disease and 110,218 controls in 23 epidemiological studies, we genotyped five functional variants: four rare loss-of-function mutations (c.109+2T > C (rs142974898), Arg82His (rs144983904), Val279Phe (rs76863441), Gln287Ter (rs140020965)) and one common modest-impact variant (Val379Ala (rs1051931)) in PLA2G7, the gene encoding Lp-PLA2. We supplemented de-novo genotyping with information on a further 45,823 coronary heart disease patients and 88,680 controls in publicly available databases and other previous studies. We conducted a systematic review of randomized trials to compare effects of darapladib treatment on soluble Lp-PLA2 activity, conventional cardiovascular risk factors, and coronary heart disease risk with corresponding effects of Lp-PLA2-lowering alleles. Results Lp-PLA2 activity was decreased by 64% ( p = 2.4 × 10-25) with carriage of any of the four loss-of-function variants, by 45% ( p < 10-300) for every allele inherited at Val279Phe, and by 2.7% ( p = 1.9 × 10-12) for every allele inherited at Val379Ala. Darapladib 160 mg once-daily reduced Lp-PLA2 activity by 65% ( p < 10-300). Causal risk ratios for coronary heart disease per 65% lower Lp-PLA2 activity were: 0.95 (0.88-1.03) with Val279Phe; 0.92 (0.74-1.16) with carriage of any loss-of-function variant; 1.01 (0.68-1.51) with Val379Ala; and 0.95 (0.89-1.02) with darapladib treatment. Conclusions In a large-scale human genetic study, none of a series of Lp-PLA2-lowering alleles was related to coronary heart disease risk, suggesting that Lp-PLA2 is unlikely to be a causal risk factor.
Collapse
Affiliation(s)
- John M Gregson
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Daniel F Freitag
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Praveen Surendran
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Nathan O Stitziel
- Departments of Medicine and Genetics, Washington University School of Medicine, St Louis, USA
| | - Rajiv Chowdhury
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Stephen Burgess
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Stephen Kaptoge
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Pei Gao
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - James R Staley
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Peter Willeit
- Department of Public Health and Primary Care, University of Cambridge, UK
- Department of Neurology, Innsbruck Medical University, Austria
| | - Sune F Nielsen
- Copenhagen University Hospital, University of Copenhagen, Denmark
| | | | | | | | - Kari Kuulasmaa
- THL-National Institute for Health and Welfare, Helsinki, Finland
| | - Jukka Kontto
- THL-National Institute for Health and Welfare, Helsinki, Finland
| | - Markus Perola
- Institute of Molecular Medicine FIMM, University of Helsinki, Finland
- Department of Health, National Institute for Health and Welfare, Helsinki, Finland
| | - Stefan Blankenberg
- Department of General and Interventional Cardiology, University Heart Centre Hamburg, Germany
- University Medical Centre Hamburg Eppendorf, Hamburg, Germany
| | - Giovanni Veronesi
- Research Centre, Department of Clinical and Experimental Medicine, University of Insubria, Varese, Italy
| | - Francesco Gianfagna
- Research Centre, Department of Clinical and Experimental Medicine, University of Insubria, Varese, Italy
- Department of Epidemiology and Prevention, IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy
| | - Satu Männistö
- THL-National Institute for Health and Welfare, Helsinki, Finland
| | - Akinori Kimura
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Japan
| | - Honghuang Lin
- Section of Computational Biomedicine, Department of Medicine, Boston University School of Medicine, USA
- The NHLBI’s Framingham Heart Study, Framingham, USA
| | - Dermot F Reilly
- Merck Research Laboratories, Genetics and Pharmacogenomics, Boston, USA
| | - Mathias Gorski
- Department of Genetic Epidemiology, University of Regensburg, Germany
- Department of Nephrology, University Hospital Regensburg, Germany
| | - Vladan Mijatovic
- Department of Life and Reproduction Sciences, University of Verona, Italy
| | | | - Patricia B Munroe
- Clinical Pharmacology and The Genome Centre, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
- NIHR Barts Cardiovascular Biomedical Research Unit, Queen Mary University of London, UK
| | - Georg B Ehret
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, USA
- Cardiology, Department of Medicine, Geneva University Hospital, Switzerland
- Institute of Social and Preventive Medicine (IUMSP), Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | - Abbas Dehghan
- Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | | | - Thomas F Vogt
- Merck Research Laboratories, Cardiometabolic Disease, Kenilworth, USA
- CHDI Management/CHDI Foundation, Princeton, USA
| | - Taishi Sasaoka
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Japan
| | - Fumihiko Takeuchi
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Centre for Global Health and Medicine, Tokyo, Japan
| | - Norihiro Kato
- Department of Gene Diagnostics and Therapeutics, Research Institute, National Centre for Global Health and Medicine, Tokyo, Japan
| | - Yoshiji Yamada
- Department of Human Functional Genomics, Life Science Research Centre, Mie University, Japan
| | - Frank Kee
- UKCRC Centre of Excellence for Public Health, Queens University, Belfast, Ireland
| | - Martina Müller-Nurasyid
- Institute of Genetic Epidemiology, Helmholtz Zentrum München - German Research Centre for Environmental Health, Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
- Department of Medicine I, Ludwig-Maximilians-University Munich, Germany
| | - Jean Ferrières
- Department of Epidemiology, UMR 1027-INSERM, Toulouse University-CHU Toulouse, France
| | - Dominique Arveiler
- Department of Epidemiology and Public Health, EA 3430, University of Strasbourg and Strasbourg University Hospital, France
| | - Philippe Amouyel
- Department of Epidemiology and Public Health, Institut Pasteur de Lille, France
| | - Veikko Salomaa
- THL-National Institute for Health and Welfare, Helsinki, Finland
| | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Science Center at Houston, USA
| | - Simon G Thompson
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | | | | | | | | | | | - Dewan S Alam
- Centre for Global Health Research, St Michael Hospital, Toronto, Canada
| | - Panos Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Heribert Schunkert
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
- Deutsches Herzzentrum München, Technische Universität München, Germany
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Cardiovascular Biomedical Research Unit, UK
| | - Sekar Kathiresan
- Broad Institute, Cambridge and Massachusetts General Hospital, Boston, USA
| | | | | | | | - Joanna MM Howson
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Emanuele Di Angelantonio
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - Adam S Butterworth
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
| | - John Danesh
- MRC/BHF Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, UK
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
- British Heart Foundation Cambridge Centre of Excellence, University of Cambridge, Cambridge, UK
- National Institute of Health Research Blood and Transplant Research Unit in Donor Health and Genomics, University of Cambridge, Cambridge, UK
| |
Collapse
|
42
|
Esenwa CC, Elkind MS. Inflammatory risk factors, biomarkers and associated therapy in ischaemic stroke. Nat Rev Neurol 2016; 12:594-604. [DOI: 10.1038/nrneurol.2016.125] [Citation(s) in RCA: 166] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
43
|
Okopień B, Bułdak Ł, Bołdys A. Current and future trends in the lipid lowering therapy. Pharmacol Rep 2016; 68:737-47. [DOI: 10.1016/j.pharep.2016.03.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 02/25/2016] [Accepted: 03/25/2016] [Indexed: 12/31/2022]
|
44
|
Wallentin L, Held C, Armstrong PW, Cannon CP, Davies RY, Granger CB, Hagström E, Harrington RA, Hochman JS, Koenig W, Krug-Gourley S, Mohler ER, Siegbahn A, Tarka E, Steg PG, Stewart RAH, Weiss R, Östlund O, White HD. Lipoprotein-Associated Phospholipase A2 Activity Is a Marker of Risk But Not a Useful Target for Treatment in Patients With Stable Coronary Heart Disease. J Am Heart Assoc 2016; 5:e003407. [PMID: 27329448 PMCID: PMC4937279 DOI: 10.1161/jaha.116.003407] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/19/2016] [Indexed: 01/20/2023]
Abstract
BACKGROUND We evaluated lipoprotein-associated phospholipase A2 (Lp-PLA2) activity in patients with stable coronary heart disease before and during treatment with darapladib, a selective Lp-PLA2 inhibitor, in relation to outcomes and the effects of darapladib in the STABILITY trial. METHODS AND RESULTS Plasma Lp-PLA2 activity was determined at baseline (n=14 500); at 1 month (n=13 709); serially (n=100) at 3, 6, and 18 months; and at the end of treatment. Adjusted Cox regression models evaluated associations between Lp-PLA2 activity levels and outcomes. At baseline, the median Lp-PLA2 level was 172.4 μmol/min per liter (interquartile range 143.1-204.2 μmol/min per liter). Comparing the highest and lowest Lp-PLA2 quartile groups, the hazard ratios were 1.50 (95% CI 1.23-1.82) for the primary composite end point (cardiovascular death, myocardial infarction, or stroke), 1.95 (95% CI 1.29-2.93) for hospitalization for heart failure, 1.42 (1.07-1.89) for cardiovascular death, and 1.37 (1.03-1.81) for myocardial infarction after adjustment for baseline characteristics, standard laboratory variables, and other prognostic biomarkers. Treatment with darapladib led to a ≈65% persistent reduction in median Lp-PLA2 activity. There were no associations between on-treatment Lp-PLA2 activity or changes of Lp-PLA2 activity and outcomes, and there were no significant interactions between baseline and on-treatment Lp-PLA2 activity or changes in Lp-PLA2 activity levels and the effects of darapladib on outcomes. CONCLUSIONS Although high Lp-PLA2 activity was associated with increased risk of cardiovascular events, pharmacological lowering of Lp-PLA2 activity by ≈65% did not significantly reduce cardiovascular events in patients with stable coronary heart disease, regardless of the baseline level or the magnitude of change of Lp-PLA2 activity. CLINICAL TRIAL REGISTRATION URL: https://www.clinicaltrials.gov. Unique identifier: NCT00799903.
Collapse
Affiliation(s)
- Lars Wallentin
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden Uppsala Clinical Research Center (UCR), Uppsala University, Uppsala, Sweden
| | - Claes Held
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden Uppsala Clinical Research Center (UCR), Uppsala University, Uppsala, Sweden
| | | | - Christopher P Cannon
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA Harvard Clinical Research Institute, Boston, MA
| | - Richard Y Davies
- Metabolic Pathways and Cardiovascular Therapeutic Area, GlaxoSmithKline, King of Prussia, PA
| | | | - Emil Hagström
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden Uppsala Clinical Research Center (UCR), Uppsala University, Uppsala, Sweden
| | | | - Judith S Hochman
- Department of Medicine, NYU Langone Medical Center, New York, NY
| | - Wolfgang Koenig
- Department of Internal Medicine II-Cardiology, University of Ulm Medical Center, Ulm, Germany Deutsches Herzzentrum München, Technische Universität München, Munich, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Sue Krug-Gourley
- Metabolic Pathways and Cardiovascular Therapeutic Area, GlaxoSmithKline, King of Prussia, PA
| | - Emile R Mohler
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Agneta Siegbahn
- Uppsala Clinical Research Center (UCR), Uppsala University, Uppsala, Sweden Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Elizabeth Tarka
- Former Employee of Metabolic Pathways and Cardiovascular Therapeutic Area, GlaxoSmithKline, King of Prussia, PA
| | - Philippe Gabriel Steg
- FACT (French Alliance for Cardiovascular Trials), Paris, France DHU FIRE, Université Paris-Diderot, Sorbonne Paris-Cité, Paris, France Hôpital Bichat, INSERUM U-1148, Paris, France NHLI, ICMS, Imperial College, Royal Brompton Hospital, London, UK
| | - Ralph A H Stewart
- Green Lane Cardiovascular Service, Auckland City Hospital, Auckland, New Zealand University of Auckland, New Zealand
| | | | - Ollie Östlund
- Uppsala Clinical Research Center (UCR), Uppsala University, Uppsala, Sweden
| | - Harvey D White
- Green Lane Cardiovascular Service, Auckland City Hospital, Auckland, New Zealand University of Auckland, New Zealand
| |
Collapse
|
45
|
Nus M, Mallat Z. Immune-mediated mechanisms of atherosclerosis and implications for the clinic. Expert Rev Clin Immunol 2016; 12:1217-1237. [PMID: 27253721 DOI: 10.1080/1744666x.2016.1195686] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION A large body of evidence supports the inflammatory hypothesis of atherosclerosis, and both innate and adaptive immune responses play important roles in all disease stages. Areas covered: Here, we review our understanding of the role of the immune response in atherosclerosis, focusing on the pathways currently amenable to therapeutic modulation. We also discuss the advantages or undesirable effects that may be foreseen from targeting the immune response in patients at high cardiovascular risk, suggesting new avenues for research. Expert commentary: There is an extraordinary opportunity to directly test the inflammatory hypothesis of atherosclerosis in the clinic using currently available therapeutics. However, a more balanced interpretation of the experimental and translational data is needed, which may help address and identify in more detail the appropriate settings where an immune pathway can be targeted with minimal risk.
Collapse
Affiliation(s)
- Meritxell Nus
- a Division of Cardiovascular Medicine, Department of Medicine , University of Cambridge , Cambridge , UK
| | - Ziad Mallat
- a Division of Cardiovascular Medicine, Department of Medicine , University of Cambridge , Cambridge , UK
| |
Collapse
|
46
|
Millwood IY, Bennett DA, Walters RG, Clarke R, Waterworth D, Johnson T, Chen Y, Yang L, Guo Y, Bian Z, Hacker A, Yeo A, Parish S, Hill MR, Chissoe S, Peto R, Cardon L, Collins R, Li L, Chen Z. A phenome-wide association study of a lipoprotein-associated phospholipase A2 loss-of-function variant in 90 000 Chinese adults. Int J Epidemiol 2016; 45:1588-1599. [PMID: 27301456 PMCID: PMC5100610 DOI: 10.1093/ije/dyw087] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2016] [Indexed: 12/20/2022] Open
Abstract
Background: Lipoprotein-associated phospholipase A2 (Lp-PLA2) has been implicated in development of atherosclerosis; however, recent randomized trials of Lp-PLA2 inhibition reported no beneficial effects on vascular diseases. In East Asians, a loss-of-function variant in the PLA2G7 gene can be used to assess the effects of genetically determined lower Lp-PLA2. Methods:PLA2G7 V279F (rs76863441) was genotyped in 91 428 individuals randomly selected from the China Kadoorie Biobank of 0.5 M participants recruited in 2004–08 from 10 regions of China, with 7 years’ follow-up. Linear regression was used to assess effects of V279F on baseline traits. Logistic regression was conducted for a range of vascular and non-vascular diseases, including 41 ICD-10 coded disease categories. Results:PLA2G7 V279F frequency was 5% overall (range 3–7% by region), and 9691 (11%) participants had at least one loss-of-function variant. V279F was not associated with baseline blood pressure, adiposity, blood glucose or lung function. V279F was not associated with major vascular events [7141 events; odds ratio (OR) = 0.98 per F variant, 95% confidence interval (CI) 0.90-1.06] or other vascular outcomes, including major coronary events (922 events; 0.96, 0.79-1.18) and stroke (5967 events; 1.00, 0.92-1.09). Individuals with V279F had lower risks of diabetes (7031 events; 0.91, 0.84-0.98) and asthma (182 events; 0.53, 0.28-0.98), but there was no association after adjustment for multiple testing. Conclusions: Lifelong lower Lp-PLA2 activity was not associated with major risks of vascular or non-vascular diseases in Chinese adults. Using functional genetic variants in large-scale prospective studies with linkage to a range of health outcomes is a valuable approach to inform drug development and repositioning.
Collapse
Affiliation(s)
- Iona Y Millwood
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Derrick A Bennett
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Robin G Walters
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Robert Clarke
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Dawn Waterworth
- GlaxoSmithKline (GSK) Medicines Research Centre, GSK, Stevenage, UK, Research Triangle Park, NC, USA and King of Prussia, PA, USA
| | - Toby Johnson
- GlaxoSmithKline (GSK) Medicines Research Centre, GSK, Stevenage, UK, Research Triangle Park, NC, USA and King of Prussia, PA, USA
| | - Yiping Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Ling Yang
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Yu Guo
- Chinese Academy of Medical Sciences, Dong Cheng District, Beijing, China and
| | - Zheng Bian
- Chinese Academy of Medical Sciences, Dong Cheng District, Beijing, China and
| | - Alex Hacker
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Astrid Yeo
- GlaxoSmithKline (GSK) Medicines Research Centre, GSK, Stevenage, UK, Research Triangle Park, NC, USA and King of Prussia, PA, USA
| | - Sarah Parish
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Michael R Hill
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Stephanie Chissoe
- GlaxoSmithKline (GSK) Medicines Research Centre, GSK, Stevenage, UK, Research Triangle Park, NC, USA and King of Prussia, PA, USA
| | - Richard Peto
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Lon Cardon
- GlaxoSmithKline (GSK) Medicines Research Centre, GSK, Stevenage, UK, Research Triangle Park, NC, USA and King of Prussia, PA, USA
| | - Rory Collins
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | - Liming Li
- Chinese Academy of Medical Sciences, Dong Cheng District, Beijing, China and.,Department of Epidemiology & Biostatistics, Peking University Health Science Centre, Beijing, China
| | - Zhengming Chen
- Clinical Trial Service Unit & Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, UK
| | | |
Collapse
|
47
|
Abstract
Cardiovascular disease (CVD) is the most common cause of death and disability worldwide. Therefore, great importance has been placed on the discovery of novel risk factors and metabolic pathways relevant in the prevention and management of CVD. Such research is ongoing and may continue to lead to better risk stratification of individuals and/or the development of new intervention targets and treatment options. This review highlights emerging biomarkers related to lipid metabolism, glycemia, inflammation, and cardiac damage, some of which show promising associations with CVD risk and provide further understanding of the underlying pathophysiology. However, their measurement methodology and assays will require validation and standardization, and it will take time to accumulate evidence of their role in CVD in various population settings in order to fully assess their clinical utility. Several of the novel biomarkers represent intriguing, potentially game-changing targets for therapy.
Collapse
Affiliation(s)
- Leah E Cahill
- Department of Medicine, Dalhousie University, 5790 University Ave, Halifax, NS, B3H 1V7, Canada.
- Department of Nutrition, Harvard T. H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA.
| | - Monica L Bertoia
- Department of Nutrition, Harvard T. H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA.
| | - Sarah A Aroner
- Department of Nutrition, Harvard T. H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA.
| | - Kenneth J Mukamal
- Beth Israel Deaconess Medical Center, 1309 Beacon Street, 2nd Floor, Brookline, Boston, MA, USA.
| | - Majken K Jensen
- Department of Nutrition, Harvard T. H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA.
| |
Collapse
|
48
|
Passacquale G, Di Giosia P, Ferro A. The role of inflammatory biomarkers in developing targeted cardiovascular therapies: lessons from the cardiovascular inflammation reduction trials. Cardiovasc Res 2015; 109:9-23. [PMID: 26410367 DOI: 10.1093/cvr/cvv227] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 08/06/2015] [Indexed: 01/04/2023] Open
Abstract
Anti-inflammatory add-on therapy to conventional cardiovascular prophylaxis has been proposed as a novel therapeutic approach to potentially reduce residual cardiovascular risk. This hypothesis has been challenged by a series of unsuccessful Phase III studies testing the impact on clinical outcomes of novel agents with immunomodulatory actions. Specifically, the apparent ability of phospholipase A2 (PLA2) inhibitors and of antioxidants to ameliorate inflammation and to reduce coronary disease in Phase II trials did not translate into improved secondary cardiovascular prevention in larger population-based studies. Other anti-inflammatory agents are still under scrutiny. However, studies to date have lacked information on the inflammatory profile of the participants, both at baseline and at follow-up, thereby limiting the possibility of identifying subgroups of patients in whom 'residual inflammation' can be detected despite optimal conventional therapy, and who could therefore benefit from a cardiovascular prevention strategy specifically targeting inflammation. This has also rendered it difficult to interpret the results as a conclusive demonstration of inefficacy of the tested anti-inflammatory strategies in the treatment of atherosclerosis. We here discuss the importance of better patient characterization to minimize heterogeneity of the study population, so that effectiveness of different anti-inflammatory strategies can be evaluated in targeted subgroups of patients. We also illustrate how specific inflammatory biomarkers could assist in this process, with particular emphasis on the roles of high-sensitivity C-reactive protein and circulating monocyte phenotype.
Collapse
Affiliation(s)
- Gabriella Passacquale
- Department of Clinical Pharmacology, BHF Centre of Research Excellence, Cardiovascular Division, King's College London, London, UK
| | - Paolo Di Giosia
- Department of Clinical Pharmacology, BHF Centre of Research Excellence, Cardiovascular Division, King's College London, London, UK
| | - Albert Ferro
- Department of Clinical Pharmacology, BHF Centre of Research Excellence, Cardiovascular Division, King's College London, London, UK
| |
Collapse
|
49
|
Campos CM, Suwannasom P, Koenig W, Serruys PW, Garcia-Garcia HM. Darapladib for the treatment of cardiovascular disease. Expert Rev Cardiovasc Ther 2015; 13:33-48. [PMID: 25521799 DOI: 10.1586/14779072.2015.986466] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Elevated levels of phospholipase A2 have been linked to atherosclerotic plaque progression, instability via promoting inflammation and subsequent acute coronary events. Epidemiological studies have demonstrated the correlation between elevated levels associated phospholipase A2 and cardiovascular events. Therefore, specific inhibition of lipoprotein-associated phospholipase A2 with darapladib has been tested as a therapeutic option for atherosclerosis. The aim of this profile is to review the physiologic aspects of lipoprotein-associated phospholipase A2 and to revisit the clinical evidence of darapladib as therapeutic option for atherosclerosis.
Collapse
Affiliation(s)
- Carlos M Campos
- Department of Interventional Cardiology, Erasmus University Medical Centre, Thoraxcenter, s-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands
| | | | | | | | | |
Collapse
|
50
|
Magee MH, Shaddinger B, Collins D, Siddiqi S, Soffer J. The pharmacokinetics and safety of darapladib in subjects with severe renal impairment. Br J Clin Pharmacol 2015; 80:654-61. [PMID: 25953363 DOI: 10.1111/bcp.12661] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 04/03/2015] [Accepted: 04/12/2015] [Indexed: 01/15/2023] Open
Abstract
AIM Darapladib is a potent and reversible orally active inhibitor of lipoprotein-associated phospholipase A2 (Lp-PLA2 ). The aim of the study was to assess the effects of severe renal impairment on the pharmacokinetics and safety/tolerability of darapladib compared with normal renal function. METHODS This was an open label, parallel group study of darapladib following 10 day once daily 160 mg oral dosing in subjects with normal (n = 8) and severe renal impairment (estimated glomerular filtration rate <30 ml min(-1) 1.73 m(-2) , n = 8). Plasma concentrations of total and unbound darapladib as well as total darapladib metabolites were determined in samples obtained over 24 h on day 10. RESULTS Plasma concentrations of total and unbound darapladib as well as all three metabolites were higher in subjects with severe renal impairment. Area under the plasma concentration vs. time curve between time zero and 24 h (AUC(0,24 h) and maximum plasma concentration (Cmax ) of total darapladib in severely renally impaired subjects were 52% and 59% higher than those in the matched healthy subjects, respectively. Similar results were found with the darapladib metabolites. Darapladib was highly plasma protein bound with 0.047% and 0.034% unbound circulating in plasma in severely renally impaired and healthy subjects, respectively. Unbound plasma darapladib exposures were more than two-fold higher in severely renally impaired subjects than in healthy controls. Adverse events (AE) were reported in 38% of healthy subjects and 75% of severely renally impaired subjects, most of which were mild or moderate in intensity. CONCLUSIONS The results of this study showed that darapladib exposure was increased in subjects with severe renal impairment compared with healthy controls. However, darapladib was generally well tolerated in both groups.
Collapse
Affiliation(s)
- Mindy He Magee
- Clinical Pharmacology Modeling and Simulation, GlaxoSmithKline, King of Prussia, PA
| | - Bonnie Shaddinger
- Metabolic Pathways and Cardiovascular Therapeutic Area, GlaxoSmithKline, King of Prussia, PA
| | - David Collins
- Clinical Statistics, GlaxoSmithKline, Research Triangle Park, NC
| | - Shabana Siddiqi
- Clincal Pharmacology Sciences and Study Operations, GlaxoSmithKline, King of Prussia, PA, USA
| | - Joseph Soffer
- Metabolic Pathways and Cardiovascular Therapeutic Area, GlaxoSmithKline, King of Prussia, PA
| |
Collapse
|